1
|
Ebo DG, Bahri R, Eggel A, Sabato V, Tontini C, Elst J. Flow cytometry-based basophil and mast cell activation tests in allergology: State of the art. J Allergy Clin Immunol 2024:S0091-6749(24)01242-9. [PMID: 39581294 DOI: 10.1016/j.jaci.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
The major challenge in allergy diagnosis is development of accessible and reliable diagnostics that can predict the clinical outcome following exposure to culprit allergen(s) or cross-reactive molecules and identification of safer alternatives than the current state-of-the-art methods. There is accumulating evidence that flow-based analyses for the quantification of activated basophils and mast cells subsequent to in vitro challenge (the basophil and mast cell activation test [BAT/MAT] or basophil activation test [BAT] and mast cell activation test [MAT]) could meet the diagnostic requirements for IgE-dependent allergies, drug hypersensitivities, and subsets of autoimmune urticaria. Furthermore, the BAT and MAT have found application in research and other nondiagnostic fields. However, appropriate use of the BAT and MAT requires understanding of the diversity of the source materials used and degranulation metrics to ensure correct test performance and interpretation of results. In this review, we provide the main applications and limitations of the BAT and MAT, as performed thus far.
Collapse
Affiliation(s)
- Didier G Ebo
- Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital, and Infla-Med Centre of Excellence Antwerp University, Antwerp, Belgium; Immunology-Allergology AZ Jan Palfijn Ghent, Ghent, Belgium.
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Core Technology Facility, University of Manchester, Manchester, United Kingdom
| | - Alexander Eggel
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland; Department for BioMedical Research, Lung Precision Medicine, University of Bern, Bern, Switzerland
| | - Vito Sabato
- Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital, and Infla-Med Centre of Excellence Antwerp University, Antwerp, Belgium
| | - Chiara Tontini
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Core Technology Facility, University of Manchester, Manchester, United Kingdom
| | - Jessy Elst
- Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital, and Infla-Med Centre of Excellence Antwerp University, Antwerp, Belgium
| |
Collapse
|
2
|
Li H, Yang L, Li J, Gao Q, Liu T, Zou Y, Chen X, Li H, Yu J. Allergenicity evaluation of an extensively hydrolyzed infant formula based on cow milk protein. Food Funct 2024; 15:11036-11046. [PMID: 39431858 DOI: 10.1039/d4fo03582h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Cow milk protein allergy (CMPA) is the most common food allergy in infants. Currently, hypoallergenic infant formulas on the market are mainly divided into extensively hydrolyzed whey protein formulas and extensively hydrolyzed casein formulas. There are few extensively hydrolyzed infant formulas (EHFs) with a similar protein composition to breast milk. Therefore, we developed a hypoallergenic infant formula based on extensively hydrolyzed cow milk protein (whey protein-to-casein ratio of 6 : 4) and evaluated its allergenicity in vitro and in vivo. The results showed that the antigenicity of EHF was significantly decreased. The levels of Treg and Th1 cells were increased, while the levels of Th2 cells, IgE and IgG1, plasma histamine and serum mast cell enzymes were significantly decreased. At the same time, the allergic symptoms of the jejunum and lungs of mice were relieved. This study provides a solution for the development of cow milk protein based hypoallergenic infant formulas.
Collapse
Affiliation(s)
- Hongbo Li
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Lin Yang
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Jiayi Li
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Quan Gao
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Tiantian Liu
- School of Food Science, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yang Zou
- Tianjin Haihe Dairy Co., Ltd, No. 158, Jingwu Road, Tianjin Airport Economic Zone, Tianjin Economic-Technological Development Area (TEDA), Tianjin, 300457, China
| | - Xiaohong Chen
- Tianjin Haihe Dairy Co., Ltd, No. 158, Jingwu Road, Tianjin Airport Economic Zone, Tianjin Economic-Technological Development Area (TEDA), Tianjin, 300457, China
| | - Hongjuan Li
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Jinghua Yu
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| |
Collapse
|
3
|
Lin Y, Lu H, Jia Q, Han S. Screening anti-anaphylactoid components in Polygonum cuspidatum via cell membrane chromatography with LC-MS targeting Mas-related G protein-coupled receptor X2. J Sep Sci 2024; 47:e2300924. [PMID: 38819784 DOI: 10.1002/jssc.202300924] [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: 12/15/2023] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
Mas-related G protein-coupled receptor X2 (MrgprX2) is acknowledged as a mast cell-specific receptor, playing a crucial role in orchestrating anaphylactoid responses through mast cell degranulation. It holds promise as a target for regulating allergic and inflammatory diseases mediated by mast cells. Polygonum cuspidatum (PC) has shown notable anti-anaphylactoid effects, while its pharmacologically active components remain unclear. In this study, we successfully utilized MrgprX2 high-expressing cell membrane chromatography (CMC), in conjunction with liquid chromatography-mass spectrometry (LC-MS), to identify active anti-anaphylactoid components in PC. Our study pinpointed polydatin, resveratrol, and emodin-8-O-β-d-glucoside as potential anti-anaphylactoid compounds in PC. Their anti-anaphylactoid activities were evaluated through β-aminohexosidase and histamine release assays, demonstrating a concentration-dependent inhibition for both β-aminohexosidase and histamine release. This approach, integrating MrgprX2 high-expression CMC with LC-MS, proves effective in screening potential anti-anaphylactoid ingredients in natural herbal medicines. The findings from this study illuminated the anti-anaphylactoid properties of specific components in PC and provided an efficient method for the drug development of natural products.
Collapse
Affiliation(s)
- Yuanyuan Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, P. R. China
| | - Huaqiu Lu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, P. R. China
- Linyi Traditional Chinese Medical Hospital, Linyi, P. R. China
| | - Qianqian Jia
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
- Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an, P. R. China
- Shaanxi Institute for Food and Drug Control, Xi'an, P. R. China
| | - Shengli Han
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
- Institute of Pharmaceutical Science and Technology, Western China Science &Technology Innovation Harbour, Xi'an, P. R. China
| |
Collapse
|
4
|
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
|
5
|
Xie CL, Xiao HX, Song PF, Liu QM, Wei H, Wu L, Zhu GH, Liu GM, Zhang Y, Wang P, Yang XW. Lead Optimization of Butyrolactone I as an Orally Bioavailable Antiallergic Agent Targeting FcγRIIB. J Med Chem 2024. [PMID: 38640354 DOI: 10.1021/acs.jmedchem.4c00354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Food allergy (FA) poses a growing global food safety concern, yet no effective cure exists in clinics. Previously, we discovered a potent antifood allergy compound, butyrolactone I (BTL-I, 1), from the deep sea. Unfortunately, it has a very low exposure and poor pharmacokinetic (PK) profile in rats. Therefore, a series of structural optimizations toward the metabolic pathways of BTL-I were conducted to provide 18 derives (2-19). Among them, BTL-MK (19) showed superior antiallergic activity and favorable pharmacokinetics compared to BTL-I, being twice as potent with a clearance (CL) rate of only 0.5% that of BTL-I. By oral administration, Cmax and area under the concentration-time curve (AUC0-∞) were 565 and 204 times higher than those of BTL-I, respectively. These findings suggest that butyrolactone methyl ketone (BTL-BK) could serve as a drug candidate for the treatment of FAs and offer valuable insights into optimizing the druggability of lead compounds.
Collapse
Affiliation(s)
- Chun-Lan Xie
- School of Pharmacy, Hainan Medical University, Hainan Academy of Medical Sciences, No. 3 Xueyuan Road, Haikou 571199, China
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
| | - Hong-Xiu Xiao
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory of Chemical Biology of Fujian Province, iCHEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Pei-Fang Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing-Mei Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Haoxiang Wei
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory of Chemical Biology of Fujian Province, iCHEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Liang Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guang-Hao Zhu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guang-Ming Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yandong Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory of Chemical Biology of Fujian Province, iCHEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xian-Wen Yang
- School of Pharmacy, Hainan Medical University, Hainan Academy of Medical Sciences, No. 3 Xueyuan Road, Haikou 571199, China
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian 361005, China
| |
Collapse
|
6
|
Wetten PA, Arismendi Sosa AC, Mariani ML, Vargas PM, Michaut MA, Penissi AB. Dehydroleucodine and xanthatin, two natural anti-inflammatory lactones, inhibit mast cell degranulation by affecting the actin cytoskeleton. Cytoskeleton (Hoboken) 2024; 81:215-226. [PMID: 37929805 DOI: 10.1002/cm.21805] [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: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
Actin remodeling is a critical regulator of mast cell secretion. In previous work, we have shown that dehydroleucodine and xanthatin, two natural α,β-unsaturated lactones, exhibit anti-inflammatory and mast cell stabilizing properties. Based on this background, this study aimed to determine whether the mast cell stabilizing action of these lactones is associated with changes in the actin cytoskeleton. Rat peritoneal mast cells were preincubated in the presence of dehydroleucodine or xanthatin before incubation with compound 48/80. Comparative studies with sodium cromoglycate and latrunculin B were also made. After treatments, different assays were performed on mast cell samples: β-hexosaminidase release, cell viability studies, quantification of mast cells and their state of degranulation by light microscopy, transmission electron microscopy, and actin staining for microscopy observation. Results showed that dehydroleucodine and xanthatin inhibited mast cell degranulation, evidenced by the inhibition of β-hexosaminidase release and decreased degranulated mast cell percentage. At the same time, both lactones altered the F-actin cytoskeleton in mast cells resulting, similarly to Latrunculin B, in a higher concentration of nuclear F-actin when activated by compound 48/80. For the first time, this study describes the biological properties of dehydroleucodine and xanthatin concerning to the rearrangement of actin filaments during stimulated exocytosis in mast cells. These data have important implications for developing new anti-inflammatory and mast cell stabilizing drugs and for designing new small molecules that may interact with the actin cytoskeleton.
Collapse
Affiliation(s)
- Paula A Wetten
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | | | - María Laura Mariani
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Patricia M Vargas
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marcela Alejandra Michaut
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Alicia Beatriz Penissi
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
7
|
Oka M, Akaki S, Ohno O, Terasaki M, Hamaoka-Tamura Y, Saito M, Kato S, Inoue A, Aoki J, Matsuno K, Furuta K, Tanaka S. Suppression of Mast Cell Activation by GPR35: GPR35 Is a Primary Target of Disodium Cromoglycate. J Pharmacol Exp Ther 2024; 389:76-86. [PMID: 38290974 DOI: 10.1124/jpet.123.002024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
Mast cell stabilizers, including disodium cromoglycate (DSCG), were found to have potential as the agonists of an orphan G protein-coupled receptor, GPR35, although it remains to be determined whether GPR35 is expressed in mast cells and involved in suppression of mast cell degranulation. Our purpose in this study is to verify the expression of GPR35 in mast cells and to clarify how GPR35 modulates the degranulation. We explored the roles of GPR35 using an expression system, a mast cell line constitutively expressing rat GPR35, peritoneal mast cells, and bone marrow-derived cultured mast cells. Immediate allergic responses were assessed using the IgE-mediated passive cutaneous anaphylaxis (PCA) model. Various known GPR35 agonists, including DSCG and newly designed compounds, suppressed IgE-mediated degranulation. GPR35 was expressed in mature mast cells but not in immature bone marrow-derived cultured mast cells and the rat mast cell line. Degranulation induced by antigens was significantly downmodulated in the mast cell line stably expressing GPR35. A GPR35 agonist, zaprinast, induced a transient activation of RhoA and a transient decrease in the amount of filamentous actin. GPR35 agonists suppressed the PCA responses in the wild-type mice but not in the GPR35-/- mice. These findings suggest that GPR35 should prevent mast cells from undergoing degranulation induced by IgE-mediated antigen stimulation and be the primary target of mast cell stabilizers. SIGNIFICANCE STATEMENT: The agonists of an orphan G protein-coupled receptor, GPR35, including disodium cromoglycate, were found to suppress degranulation of rat and mouse mature mast cells, and their antiallergic effects were abrogated in the GPR35-/- mice, indicating that the primary target of mast cell stabilizers should be GPR35.
Collapse
Affiliation(s)
- Masumi Oka
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Sohta Akaki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Osamu Ohno
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Maho Terasaki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Yuho Hamaoka-Tamura
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Michiko Saito
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Shinichi Kato
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Asuka Inoue
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Junken Aoki
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Kenji Matsuno
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Kazuyuki Furuta
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| | - Satoshi Tanaka
- Laboratory of Pharmacology, Division of Pathological Sciences (M.O., M.T., Y.H.-T., S.T.), Bioscience Research Center (M.S.), and Laboratory of Pharmacology and Experimental Therapeutics, Division of Pathological Sciences (S.K.), Kyoto Pharmaceutical University, Kyoto, Japan; Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan (S.A., K.F.); Laboratory of Medicinal Chemistry, Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan (O.O.); Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (A.I.); Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (J.A.); and Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan (K.M.)
| |
Collapse
|
8
|
Lu W, Huang J, Flores J, Li P, Wang W, Liu S, Zhang JH, Tang J. GW0742 reduces mast cells degranulation and attenuates neurological impairments via PPAR β/δ/CD300a/SHP1 pathway after GMH in neonatal rats. Exp Neurol 2024; 372:114615. [PMID: 37995951 PMCID: PMC10842885 DOI: 10.1016/j.expneurol.2023.114615] [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: 08/01/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Activation of mast cells plays an important role in brain inflammation. CD300a, an inhibitory receptor located on mast cell surfaces, has been reported to reduce the production of pro-inflammatory cytokines and exert protective effects in inflammation-related diseases. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, activation upregulates the transcription of CD300a. In this study, we aim to investigate the role of PPARβ/δ in the attenuation of germinal matrix hemorrhage (GMH)-induced mast cell activation via CD300a/SHP1 pathway. METHODS GMH model was induced by intraparenchymal injection of bacterial collagenase into the right hemispheric ganglionic eminence in P7 Sprague Dawley rats. GW0742, a PPARβ/δ agonist, was administered intranasally at 1 h post-ictus. CD300a small interfering RNA (siRNA) and PPARβ/δ siRNA were injected intracerebroventricularly 5 days and 2 days before GMH induction. Behavioral tests, Western blot, immunofluorescence, Toluidine Blue staining, and Nissl staining were applied to assess post-GMH evaluation. RESULTS Results demonstrated that endogenous protein levels of PPARβ/δ and CD300a were decreased, whereas chymase, tryptase, IL-17A and transforming growth factor β1 (TGF-β1) were elevated after GMH. GMH induced significant short- and long-term neurobehavioral deficits in rat pups. GW0742 decreased mast cell degranulation, improved neurological outcomes, and attenuated ventriculomegaly after GMH. Additionally, GW0742 increased expression of PPARβ/δ, CD300a and phosphorylation of SHP1, decreased phosphorylation of Syk, chymase, tryptase, IL-17A and TGF-β1 levels. PPARβ/δ siRNA and CD300a siRNA abolished the beneficial effects of GW0742. CONCLUSIONS GW0742 inhibited mast cell-induced inflammation and improved neurobehavior after GMH, which is mediated by PPARβ/δ/CD300a/SHP1 pathway. GW0742 may serve as a potential treatment to reduce brain injury for GMH patients.
Collapse
Affiliation(s)
- Weitian Lu
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing 400016, China; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Juan Huang
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing 400016, China; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jerry Flores
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Peng Li
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Wenna Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
9
|
Tiligada E, Gafarov D, Zaimi M, Vitte J, Levi-Schaffer F. Novel Immunopharmacological Drugs for the Treatment of Allergic Diseases. Annu Rev Pharmacol Toxicol 2024; 64:481-506. [PMID: 37722722 DOI: 10.1146/annurev-pharmtox-051623-091038] [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: 09/20/2023]
Abstract
The exponential rise in the prevalence of allergic diseases since the mid-twentieth century has led to a genuine public health emergency and has also fostered major progress in research on the underlying mechanisms and potential treatments. The management of allergic diseases benefits from the biological revolution, with an array of novel immunomodulatory therapeutic and investigational tools targeting players of allergic inflammation at distinct pathophysiological steps. Prominent examples include therapeutic monoclonal antibodies against cytokines, alarmins, and their receptors, as well as small-molecule modifiers of signal transduction mainly mediated by Janus kinases and Bruton's tyrosine kinases. However, the first-line therapeutic options have yet to switch from symptomatic to disease-modifying interventions. Here we present an overview of available drugs in the context of our current understanding of allergy pathophysiology, identify potential therapeutic targets, and conclude by providing a selection of candidate immunopharmacological molecules under investigation for potential future use in allergic diseases.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Daria Gafarov
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Maria Zaimi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Joana Vitte
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
- Desbrest Institute of Epidemiology and Public Health, University of Montpellier, INSERM
- Montpellier, France
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| |
Collapse
|
10
|
Storni F, Vogel M, Bachmann MF, Engeroff P. IgG in the control of FcεRI activation: a battle on multiple fronts. Front Immunol 2024; 14:1339171. [PMID: 38274816 PMCID: PMC10808611 DOI: 10.3389/fimmu.2023.1339171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
The rising global incidence of IgE-mediated allergic reactions poses a significant challenge to the quality of life of affected individuals and to healthcare systems, with current treatments being limited in effectiveness, safety, and disease-modifying capabilities. IgE acts by sensitizing the high-affinity IgE receptor FcεRI expressed by mast cells and basophils, tuning these cells for inflammatory degranulation in response to future allergen encounters. In recent years, IgG has emerged as an essential negative regulator of IgE-dependent allergic inflammation. Mechanistically, studies have proposed different pathways by which IgG can interfere with the activation of IgE-mediated inflammation. Here, we briefly summarize the major proposed mechanisms of action by which IgG controls the IgE-FcεRI inflammatory axis and how those mechanisms are currently applied as therapeutic interventions for IgE-mediated inflammation.
Collapse
Affiliation(s)
- Federico Storni
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Monique Vogel
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| | - Paul Engeroff
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
11
|
Lei Y, Guo X, Luo Y, Niu X, Xi Y, Xiao L, He D, Bian Y, Zhang Y, Wang L, Peng X, Wang Z, Chen G. Synovial microenvironment-influenced mast cells promote the progression of rheumatoid arthritis. Nat Commun 2024; 15:113. [PMID: 38168103 PMCID: PMC10761862 DOI: 10.1038/s41467-023-44304-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Mast cells are phenotypically and functionally heterogeneous, and their state is possibly controlled by local microenvironment. Therefore, specific analyses are needed to understand whether mast cells function as powerful participants or dispensable bystanders in specific diseases. Here, we show that degranulation of mast cells in inflammatory synovial tissues of patients with rheumatoid arthritis (RA) is induced via MAS-related G protein-coupled receptor X2 (MRGPRX2), and the expression of MHC class II and costimulatory molecules on mast cells are upregulated. Collagen-induced arthritis mice treated with a combination of anti-IL-17A and cromolyn sodium, a mast cell membrane stabilizer, show significantly reduced clinical severity and decreased bone erosion. The findings of the present study suggest that synovial microenvironment-influenced mast cells contribute to disease progression and may provide a further mast cell-targeting therapy for RA.
Collapse
Affiliation(s)
- Yunxuan Lei
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xin Guo
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Yanping Luo
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xiaoyin Niu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Yebin Xi
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Lianbo Xiao
- Department of Joint Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqin Bian
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yong Zhang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Li Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xiaochun Peng
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhaojun Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China.
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China.
| |
Collapse
|
12
|
Zhu L, Jian X, Zhou B, Liu R, Muñoz M, Sun W, Xie L, Chen X, Peng C, Maurer M, Li J. Gut microbiota facilitate chronic spontaneous urticaria. Nat Commun 2024; 15:112. [PMID: 38168034 PMCID: PMC10762022 DOI: 10.1038/s41467-023-44373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Chronic spontaneous urticaria (CSU) comes with gut dysbiosis, but its relevance remains elusive. Here we use metagenomics sequencing and short-chain fatty acids metabolomics and assess the effects of human CSU fecal microbial transplantation, Klebsiella pneumoniae, Roseburia hominis, and metabolites in vivo. CSU gut microbiota displays low diversity and short-chain fatty acids production, but high gut Klebsiella pneumoniae levels, negatively correlates with blood short-chain fatty acids levels and links to high disease activity. Blood lipopolysaccharide levels are elevated, link to rapid disease relapse, and high gut levels of conditional pathogenic bacteria. CSU microbiome transfer and Klebsiella pneumoniae transplantation facilitate IgE-mediated mast cell(MC)-driven skin inflammatory responses and increase intestinal permeability and blood lipopolysaccharide accumulation in recipient mice. Transplantation of Roseburia hominis and caproate administration protect recipient mice from MC-driven skin inflammation. Here, we show gut microbiome alterations, in CSU, may reduce short-chain fatty acids and increase lipopolysaccharide levels, respectively, and facilitate MC-driven skin inflammation.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Furong Labratory, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingxing Jian
- Bioinformatics Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bingjing Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Furong Labratory, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Runqiu Liu
- Department of Dermatology, the First people's Hospital of Yancheng, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Melba Muñoz
- Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Wan Sun
- BGI, Complex building, Beishan Industrial Zone, Yantian District, Shenzhen, China
| | - Lu Xie
- Bioinformatics Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Furong Labratory, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Furong Labratory, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Marcus Maurer
- Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Furong Labratory, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
13
|
Martínez M, Mariani ML, García C, Ceñal JP, Penissi AB. A one-pot and eco-friendly synthesis of novel β-substituted-α-halomethyl acrylates and the bioactivity of these compounds in an in vitro model of mast cell degranulation induced by pro-inflammatory stimuli. Biomed Pharmacother 2024; 170:116009. [PMID: 38134632 DOI: 10.1016/j.biopha.2023.116009] [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: 08/28/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
The goal of the present work was to develop novel β-substituted-α-halomethyl acrylates from a methodology in an aqueous phase and to evaluate their bioactivity as potential inhibitors of mast cell activation. Eleven β-substituted-α-halomethyl acrylates were synthesized through a modified Horner-Wadsworth-Emmons reaction. Compound 48/80 and the calcium ionophore A23187 stimulated the release of β-hexosaminidase from mast cells. The effect induced by compound 48/80 was inhibited by compound 5 (320 µM) and compound 9 (160 and 320 µM) without causing cytotoxic effects. The effect induced by A23187 was inhibited by compound 5 (40, 80, 160, and 320 µM) without affecting cell viability. The inhibitory effects exhibited by compounds 5 and 9 were more potent than those of the reference compound sodium cromoglycate at the same concentrations. The biochemical results were consistent with the morphological findings obtained by light and transmission electron microscopy. This study reports, for the first time, that the new synthetic compounds methyl (Z)- 2-bromo-3-(furan-3-yl)acrylate (compound 5) and methyl (E)- 2-bromo-3-(3-bromophenyl)acrylate (compound 9) strongly inhibit mast cell degranulation, without affecting cell viability. The implications of these results are relevant as a basis for developing new anti-inflammatory and mast cell stabilizing drugs.
Collapse
Affiliation(s)
- Maricel Martínez
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Investigaciones en Tecnología Química (INTEQUI-CONICET), Universidad Nacional de San Luis, San Luis, Argentina
| | - María Laura Mariani
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Celina García
- Instituto Universitario de Bio-Organica "Antonio Gonzalez", Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Juan Pedro Ceñal
- Instituto de Investigaciones en Tecnología Química (INTEQUI-CONICET), Universidad Nacional de San Luis, San Luis, Argentina; Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Alicia Beatriz Penissi
- Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM, UNCUYO-CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
14
|
Xu X, Delves PJ, Huang J, Shao W, Qiu X. Comparison of Non B-Ig and B-Ig. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1445:73-88. [PMID: 38967751 DOI: 10.1007/978-981-97-0511-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Immunoglobulin (Ig) has been widely acknowledged to be produced solely by B-lineage cells. However, growing evidence has demonstrated the expression of Ig in an array of cancer cells, as well as normal cells including epithelial cells, epidermal cells, mesangial cells, monocytes, and neutrophils. Ig has even been found to be expressed in non-B cells at immune-privileged sites such as neurons and spermatogenic cells. Despite these non-B cell-derived Igs (non-B-Igs) sharing the same symmetric structures with conventional Igs (B-Igs), further studies have revealed unique characteristics of non-B-Ig, such as restricted variable region and aberrant glycosylation. Moreover, non-B-Ig exhibits properties of promoting malignant behaviours of cancer cells, therefore it could be utilised in the clinic as a potential therapeutic biomarker or target. The elucidation of the generation and regulation of non-B-Ig will certainly broaden our understanding of immunology.
Collapse
Affiliation(s)
- Xiaojun Xu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Peter J Delves
- Division of Infection and Immunity, Department of Immunology, UCL (University College London), London, UK
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
15
|
Guo X, Sun M, Yang P, Meng X, Liu R. Role of mast cells activation in the tumor immune microenvironment and immunotherapy of cancers. Eur J Pharmacol 2023; 960:176103. [PMID: 37852570 DOI: 10.1016/j.ejphar.2023.176103] [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: 06/08/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023]
Abstract
The mast cell is an important cellular component that plays a crucial role in the crosstalk between innate and adaptive immune responses within the tumor microenvironment (TME). Recently, numerous studies have indicated that mast cells related to tumors play a dual role in regulating cancers, with conflicting results seemingly determined by the degranulation medium. As such, mast cells are an ignored but very promising potential target for cancer immunotherapy based on their immunomodulatory function. In this review, we present a comprehensive overview of the roles and mechanisms of mast cells in diverse cancer types. Firstly, we evaluated the infiltration density and location of mast cells on tumor progression. Secondly, mast cells are activated by the TME and subsequently release a range of inflammatory mediators, cytokines, chemokines, and lipid products that modulate their pro-or anti-tumor functions. Thirdly, activated mast cells engage in intercellular communication with other immune or stromal cells to modulate the immune status or promote tumor development. Finally, we deliberated on the clinical significance of targeting mast cells as a therapeutic approach to restrict tumor initiation and progression. Overall, our review aims to provide insights for future research on the role of mast cells in tumors and their potential as therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China; Xiangnan University, Chenzhou, China
| | - Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Peiyan Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xingchen Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.
| |
Collapse
|
16
|
Conesa MPB, Blixt FW, Peesh P, Khan R, Korf J, Lee J, Jagadeesan G, Andersohn A, Das TK, Tan C, Di Gesu C, Colpo GD, Moruno-Manchón JF, McCullough LD, Bryan R, Ganesh BP. Stabilizing histamine release in gut mast cells mitigates peripheral and central inflammation after stroke. J Neuroinflammation 2023; 20:230. [PMID: 37805585 PMCID: PMC10560441 DOI: 10.1186/s12974-023-02887-7] [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: 04/12/2023] [Accepted: 08/31/2023] [Indexed: 10/09/2023] Open
Abstract
Stroke is the most common cause of long-term disability and places a high economic burden on the global healthcare system. Functional outcomes from stroke are largely determined by the extent of ischemic injury, however, there is growing recognition that systemic inflammatory responses also contribute to outcomes. Mast cells (MCs) rapidly respond to injury and release histamine (HA), a pro-inflammatory neurotransmitter that enhances inflammation. The gut serves as a major reservoir of HA. We hypothesized that cromolyn, a mast cell stabilizer that prevents the release of inflammatory mediators, would decrease peripheral and central inflammation, reduce MC trafficking to the brain, and improve stroke outcomes. We used the transient middle cerebral artery occlusion (MCAO) model of ischemic stroke in aged (18 mo) male mice to investigate the role of MC in neuroinflammation post-stroke. After MCAO we treated mice with 25 mg/kg body weight of cromolyn (MC stabilizer) by oral gavage. Cromolyn was administered at 3 h, 10 h, 24 h and every 24 h for 3 days post-stroke. Three control groups were used. One group underwent a sham surgery and was treated with cromolyn, one received sham surgery with PBS vehicle and the third underwent MCAO with PBS vehicle. Mice were euthanized at 24 h and 3 days post-stroke. Cromolyn administration significantly reduced MC numbers in the brain at both 24 h and 3 days post-stroke. Infarct volume was not significantly different between groups, however improved functional outcomes were seen at 3 days post-stroke in mice that received cromolyn. Treatment with cromolyn reduced plasma histamine and IL-6 levels in both the 24-h and 3-day cohorts. Gut MCs numbers were significantly reduced after cromolyn treatment at 24 h and 3 days after stroke. To determine if MC trafficking from the gut to the brain occurred after injury, GFP+MCs were adoptively transferred to c-kit-/- MC knock-out animals prior to MCAO. 24 h after stroke, elevated MC recruitment was seen in the ischemic brain. Preventing MC histamine release by cromolyn improved gut barrier integrity and an improvement in stroke-induced dysbiosis was seen with treatment. Our results show that preventing MC histamine release possesses prevents post-stroke neuroinflammation and improves neurological and functional outcomes.
Collapse
Affiliation(s)
- Maria P Blasco Conesa
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Frank W Blixt
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Pedram Peesh
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Romeesa Khan
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Janelle Korf
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Juneyoung Lee
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Gayathri Jagadeesan
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Alexander Andersohn
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Tushar K Das
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Chunfeng Tan
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Claudia Di Gesu
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Gabriela Delevati Colpo
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | | | - Louise D McCullough
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Robert Bryan
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Theoharides TC, Kempuraj D. Potential Role of Moesin in Regulating Mast Cell Secretion. Int J Mol Sci 2023; 24:12081. [PMID: 37569454 PMCID: PMC10418457 DOI: 10.3390/ijms241512081] [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: 06/27/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Mast cells have existed for millions of years in species that never suffer from allergic reactions. Hence, in addition to allergies, mast cells can play a critical role in homeostasis and inflammation via secretion of numerous vasoactive, pro-inflammatory and neuro-sensitizing mediators. Secretion may utilize different modes that involve the cytoskeleton, but our understanding of the molecular mechanisms regulating secretion is still not well understood. The Ezrin/Radixin/Moesin (ERM) family of proteins is involved in linking cell surface-initiated signaling to the actin cytoskeleton. However, how ERMs may regulate secretion from mast cells is still poorly understood. ERMs contain two functional domains connected through a long α-helix region, the N-terminal FERM (band 4.1 protein-ERM) domain and the C-terminal ERM association domain (C-ERMAD). The FERM domain and the C-ERMAD can bind to each other in a head-to-tail manner, leading to a closed/inactive conformation. Typically, phosphorylation on the C-terminus Thr has been associated with the activation of ERMs, including secretion from macrophages and platelets. It has previously been shown that the ability of the so-called mast cell "stabilizer" disodium cromoglycate (cromolyn) to inhibit secretion from rat mast cells closely paralleled the phosphorylation of a 78 kDa protein, which was subsequently shown to be moesin, a member of ERMs. Interestingly, the phosphorylation of moesin during the inhibition of mast cell secretion was on the N-terminal Ser56/74 and Thr66 residues. This phosphorylation pattern could lock moesin in its inactive state and render it inaccessible to binding to the Soluble NSF attachment protein receptors (SNAREs) and synaptosomal-associated proteins (SNAPs) critical for exocytosis. Using confocal microscopic imaging, we showed moesin was found to colocalize with actin and cluster around secretory granules during inhibition of secretion. In conclusion, the phosphorylation pattern and localization of moesin may be important in the regulation of mast cell secretion and could be targeted for the development of effective inhibitors of secretion of allergic and inflammatory mediators from mast cells.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| |
Collapse
|
18
|
Sulsenti R, Jachetti E. Frenemies in the Microenvironment: Harnessing Mast Cells for Cancer Immunotherapy. Pharmaceutics 2023; 15:1692. [PMID: 37376140 DOI: 10.3390/pharmaceutics15061692] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor development, progression, and resistance to therapies are influenced by the interactions between tumor cells and the surrounding microenvironment, comprising fibroblasts, immune cells, and extracellular matrix proteins. In this context, mast cells (MCs) have recently emerged as important players. Yet, their role is still controversial, as MCs can exert pro- or anti-tumor functions in different tumor types depending on their location within or around the tumor mass and their interaction with other components of the tumor microenvironment. In this review, we describe the main aspects of MC biology and the different contribution of MCs in promoting or inhibiting cancer growth. We then discuss possible therapeutic strategies aimed at targeting MCs for cancer immunotherapy, which include: (1) targeting c-Kit signaling; (2) stabilizing MC degranulation; (3) triggering activating/inhibiting receptors; (4) modulating MC recruitment; (5) harnessing MC mediators; (6) adoptive transferring of MCs. Such strategies should aim to either restrain or sustain MC activity according to specific contexts. Further investigation would allow us to better dissect the multifaceted roles of MCs in cancer and tailor novel approaches for an "MC-guided" personalized medicine to be used in combination with conventional anti-cancer therapies.
Collapse
Affiliation(s)
- Roberta Sulsenti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| |
Collapse
|
19
|
Aziz SN, Badawy AA, Nessem DI, Abd El Malak NS, Naguib MJ. Chitosan-coated alginate (CCA) nanoparticles for augmentation of topical antihistaminic activity of diphenhydramine: in-vitro optimization, skin histopathology and pharmacodynamic studies with in vitro/in vivo correlation. Drug Dev Ind Pharm 2023:1-12. [PMID: 37158038 DOI: 10.1080/03639045.2023.2211672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
OBJECTIVE The aim of the present study was to formulate chitosan-coated alginate nanoparticles containing the drug diphenhydramine hydrochloride (DHH). SIGNIFICANCE Diphenhydramine hydrochloride (DHH) is the prototype of H1-antihistaminic drugs. It is a lipophilic drug, that easily crosses the blood-brain barrier when taken orally causing decrements in alertness and performance. Multiple applications of topical drug product are required. Thus, drug incorporation in nanocarriers would increase the skin penetration powers increasing the drug efficacy. METHODS Chitosan coated alginate (CCA) nanoparticles were prepared via polyelectrolyte complex technique adopting 23 full factorial design. Three factors, namely, alginate concentration, drug to alginate ratio and CaCl2 volume, each in two levels were studied. The prepared formulae were evaluated utilizing entrapment efficiency (EE), particle size (PS), polydispersity index (PDI), zeta potential (ZP) and in vitro release. Characterization process was then followed by optimization. RESULTS At alginate conc. of 1%, drug to alginate ratio of 2:1 and CaCl2 volume of 4 mL, NP8 was chosen as a candidate formula. Histopathological examination on shaved rat dorsal skin disclosed the safety of NP8 with no signs of necrosis or even inflammation. The enhanced topical delivery of diphenhydramine hydrochloride enclosed in the developed nanoparticles was further proved by induction of allergic reaction using intradermal histamine injection. The results revealed the superior ability of NP8 to decrease the diameter of the formed wheal in comparison to marketed DHH product. CONCLUSION Thus, CCA nanoparticles are considered as a candidate nanocarriers for fortifying the topical antihistaminic activity of DHH.
Collapse
Affiliation(s)
- Sandy N Aziz
- Physicochemical lab, Central Administration of Drug Control, Egyptian Drug Authority, Egypt
| | - Alia A Badawy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | - Nevine S Abd El Malak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- School of pharmacy, New Giza University NGU, New Giza, Egypt
| | - Marianne J Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Franke K, Bal G, Li Z, Zuberbier T, Babina M. Clorfl86/RHEX Is a Negative Regulator of SCF/KIT Signaling in Human Skin Mast Cells. Cells 2023; 12:cells12091306. [PMID: 37174705 PMCID: PMC10177086 DOI: 10.3390/cells12091306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Mast cells (MCs) are key effector cells in allergic and inflammatory diseases, and the SCF/KIT axis regulates most aspects of the cells' biology. Using terminally differentiated skin MCs, we recently reported on proteome-wide phosphorylation changes initiated by KIT dimerization. C1orf186/RHEX was revealed as one of the proteins to become heavily phosphorylated. Its function in MCs is undefined and only some information is available for erythroblasts. Using public databases and our own data, we now report that RHEX exhibits highly restricted expression with a clear dominance in MCs. While expression is most pronounced in mature MCs, RHEX is also abundant in immature/transformed MC cell lines (HMC-1, LAD2), suggesting early expression with further increase during differentiation. Using RHEX-selective RNA interference, we reveal that RHEX unexpectedly acts as a negative regulator of SCF-supported skin MC survival. This finding is substantiated by RHEX's interference with KIT signal transduction, whereby ERK1/2 and p38 both were more strongly activated when RHEX was attenuated. Comparing RHEX and capicua (a recently identified repressor) revealed that each protein preferentially suppresses other signaling modules elicited by KIT. Induction of immediate-early genes strictly requires ERK1/2 in SCF-triggered MCs; we now demonstrate that RHEX diminution translates to this downstream event, and thereby enhances NR4A2, JUNB, and EGR1 induction. Collectively, our study reveals RHEX as a repressor of KIT signaling and function in MCs. As an abundant and selective lineage marker, RHEX may have various roles in the lineage, and the provided framework will enable future work on its involvement in other crucial processes.
Collapse
Affiliation(s)
- Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
21
|
Chen B, Song Y, Yang X, Yang J, Hao F. Bacterial DNA promoting inflammation via the Sgk1/Nedd4L/Syk pathway in mast cells contributes to antihistamine-nonresponsive CSU. J Leukoc Biol 2023; 113:461-470. [PMID: 36857592 DOI: 10.1093/jleuko/qiad025] [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: 08/09/2022] [Revised: 12/31/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
Inflammation centered on non-IgE-mediated mast cell activation characterizes chronic spontaneous urticaria resistant to nonsedating H1-antihistamines. We recently uncovered a strong positive association between inflammation and the fecal Escherichia. To further explore the actions of bacterial DNA derived from Escherichia on mast cells, intestinal permeability of patients with chronic spontaneous urticaria with or without nonsedating H1-antihistamine resistance and healthy controls were determined, and LAD2 cells with knockdown of Syk, Nedd4L, or Sgk1 or with incubation of inhibitors GS9973, GSK650394, and MG132 were posttreated with btDNA. We found that (i) serum intestinal permeability indices and bacterial DNA markedly increased in patients with chronic spontaneous urticaria with nonsedating H1-antihistamine resistance compared with those without (all P < 0.001), and bacterial DNA positively correlated with the degree of inflammation; (ii) IL-6 and TNF-α levels were time- and dose-dependently upregulated in bacterial DNA-stimulated LAD2 cells, which relied on unmethylated CpG in bacterial DNA and Toll-like receptor 9 protein in cells; (iii) Syk knockdown or inhibition of Syk Tyr525/526 phosphorylation blocked bacterial DNA-initiated cytokine production; (iv) Nedd4L interacted with Tyr525/526-phosphorylated Syk, and inhibition of Nedd4L Ser448 phosphorylation induced by bacterial DNA-activated Sgk1 was mandatory for bacterial DNA's proinflammatory property; and (v) Sgk1 suppression showed an inhibitory effect on bacterial DNA-induced inflammation by ensuring Nedd4L-mediated ubiquitination of Tyr525/526-phosphorylated Syk. Collectively, we identified previously unknown contributory roles of bacterial translocation and serum bacterial DNA on the inflammation phenotype in patients with chronic spontaneous urticaria with nonsedating H1-antihistamine resistance and further uncovered a vital negative regulatory role for the Sgk1/Nedd4L/Syk pathway in bacterial DNA-induced inflammation in LAD2 cells.
Collapse
Affiliation(s)
- Bangtao Chen
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, No.165, Xincheng Road, Wanzhou District, Chongqing 400030, China
| | - Yao Song
- Department of Pediatrics, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Road, Yubei District, Chongqing 401120, China
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Road, Yubei District, Chongqing 401120, China
| | - Xiongbo Yang
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, No.165, Xincheng Road, Wanzhou District, Chongqing 400030, China
| | - Jing Yang
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, No.165, Xincheng Road, Wanzhou District, Chongqing 400030, China
| | - Fei Hao
- Department of Pediatrics, The Third Affiliated Hospital of Chongqing Medical University, No.1, Shuanghu Road, Yubei District, Chongqing 401120, China
| |
Collapse
|
22
|
Parente R, Giudice V, Cardamone C, Serio B, Selleri C, Triggiani M. Secretory and Membrane-Associated Biomarkers of Mast Cell Activation and Proliferation. Int J Mol Sci 2023; 24:ijms24087071. [PMID: 37108232 PMCID: PMC10139107 DOI: 10.3390/ijms24087071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Mast cells (MCs) are immune cells distributed in many organs and tissues and involved in the pathogenesis of allergic and inflammatory diseases as a major source of pro-inflammatory and vasoactive mediators. MC-related disorders are heterogeneous conditions characterized by the proliferation of MC within tissues and/or MC hyper-reactivity that leads to the uncontrolled release of mediators. MC disorders include mastocytosis, a clonal disease characterized by tissue MC proliferation, and MC activation syndromes that can be primary (clonal), secondary (related to allergic disorders), or idiopathic. Diagnosis of MC disorders is difficult because symptoms are transient, unpredictable, and unspecific, and because these conditions mimic many other diseases. Validation of markers of MC activation in vivo will be useful to allow faster diagnosis and better management of MC disorders. Tryptase, being the most specific MC product, is a widely used biomarker of proliferation and activation. Other mediators, such as histamine, cysteinyl leukotrienes, and prostaglandin D2, are unstable molecules and have limitations in their assays. Surface MC markers, detected by flow cytometry, are useful for the identification of neoplastic MC in mastocytosis but, so far, none of them has been validated as a biomarker of MC activation. Further studies are needed to identify useful biomarkers of MC activation in vivo.
Collapse
Affiliation(s)
- Roberta Parente
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| | - Valentina Giudice
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Chiara Cardamone
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| | - Bianca Serio
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Carmine Selleri
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
23
|
Gimeno R, Ribas‐Llauradó C, Pesque D, Andrades E, Cenni B, Ambros B, Pujol R, Giménez‐Arnau AM. Remibrutinib inhibits hives effector cells stimulated by serum from chronic urticaria patients independently of FcεR1 expression level and omalizumab clinical response. Clin Transl Allergy 2023; 13:e12227. [PMID: 36973953 PMCID: PMC9985467 DOI: 10.1002/clt2.12227] [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: 11/01/2022] [Revised: 01/22/2023] [Accepted: 02/07/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Despite advances in the treatment of chronic urticaria, in a significant percentage of the patients symptoms are not fully controlled with conventional approaches. New strategies under development include blocking intracellular mediators of mast cell and basophil activation. OBJECTIVE We aim to investigate the effects of the Bruton's tyrosine kinase (BTK) inhibitor remibrutinib on human blood basophils and CD34+ -derived mast cells activation induced by serum obtained from chronic urticaria patients. METHODS Twenty-two patients with chronic spontaneous urticaria (mean age 52 years, 27% women) and 22 patients with chronic inducible urticaria (46 years, 27% women) were included in the study together with a sex-matched control group. Patients were classified as responders or non-responders to anti-IgE therapy on the basis of their clinical data, FcεR1a expression on blood basophils and total IgE levels. Changes on CD63 expression-as an activation marker-, were used to evaluate in vitro the response of basophils and mast cells to serum exposure and the inhibitory effects of remibrutinib. RESULTS Remibrutinib inhibits degranulation induced by IgE cross-linking in mast cells and basophils and also the activation triggered by factors present in the sera of spontaneous and inducible chronic urticaria patients. Patient's serum induces a greater degranulation of effector cells than controls. Activation of mast cells and basophils by patient sera and remibrutinib effects were not related to omalizumab responsiveness. CONCLUSION Remibrutinib inhibits activation of human basophils and mast cells induced in vitro by exposure to the serum of chronic urticaria patients independently of their response to omalizumab.
Collapse
Affiliation(s)
- Ramón Gimeno
- Laboratory of ImmunologyDepartment of PathologyHospital del MarBarcelonaSpain
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Clara Ribas‐Llauradó
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
| | - David Pesque
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Evelyn Andrades
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Bruno Cenni
- Department of ResearchBC Novartis Institutes for BioMedical Research, NovartisBaselSwitzerland
| | - Barbara Ambros
- Department of Clinical DevelopmentBA Global Drug Development, NovartisBaselSwitzerland
| | - Ramon Pujol
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| | - Ana M. Giménez‐Arnau
- Department ImmnologyHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
- Department of DermatologyHospital del MarIMIMUniversitat Pompeu FabraBarcelonaSpain
| |
Collapse
|
24
|
Steiner P, Arlt E, Boekhoff I, Gudermann T, Zierler S. TPC Functions in the Immune System. Handb Exp Pharmacol 2023; 278:71-92. [PMID: 36639434 DOI: 10.1007/164_2022_634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels (TPCs) are novel intracellular cation channels, which play a key role in numerous (patho-)physiological and immunological processes. In this chapter, we focus on their function in immune cells and immune reactions. Therefore, we first give an overview of the cellular immune response and the partaking immune cells. Second, we concentrate on ion channels which in the past have been shown to play an important role in the regulation of immune cells. The main focus is then directed to TPCs, which are primarily located in the membranes of acidic organelles, such as lysosomes or endolysosomes but also certain other vesicles. They regulate Ca2+ homeostasis and thus Ca2+ signaling in immune cells. Due to this important functional role, TPCs are enjoying increasing attention within the field of immunology in the last few decades but are also becoming more pertinent as pharmacological targets for the treatment of pro-inflammatory diseases such as allergic hypersensitivity. However, to uncover the precise molecular mechanism of TPCs in immune cell responses, further molecular, genetic, and ultrastructural investigations on TPCs are necessary, which then may pave the way to develop novel therapeutic strategies to treat diseases such as anaphylaxis more specifically.
Collapse
Affiliation(s)
- Philip Steiner
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Susanna Zierler
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria.
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
25
|
Shao M, Liu J, Luo H. Colitis aggravated by Mrgprb2 knockout is associated with altered immune response, intestinal barrier function and gut microbiota. Exp Physiol 2023; 108:63-75. [PMID: 36440681 PMCID: PMC10103767 DOI: 10.1113/ep090635] [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: 06/20/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the role of mas-related G protein-coupled receptor X2 (MRGPRX2/Mrgprb2) in ulcerative colitis in relation to the intestinal flora, intestinal barrier and immune response? What is the main finding and its importance? Knockout of mouse Mrgprb2 aggravates dextran sulfate sodium (DSS)-induced colitis, which is associated with altered gut microbiota and immune response and disruption of the intestinal barrier. MRGPRB2 may have a protective effect on DSS-induced colitis. ABSTRACT Ulcerative colitis (UC) is a chronic immune-related disease, and changes in the intestinal microbiota and damage to the intestinal barrier contribute to its pathogenesis. Mast cells (MCs) are widely distributed in the gastrointestinal tract and are thought to be related to the pathogenesis of UC. Human mas-related G protein-coupled receptor X2 (MRGPRX2) and its mouse homologue, Mrgprb2, are selectively expressed on MCs to recruit immune cells and modulate host defence against microbial infection. To investigate the role of Mrgprb2 in UC in mice, we compared the differences between Mrgprb2 knockout (b2KO) male mice and wild-type (WT) male mice with dextran sulfate sodium (DSS)-induced colitis in the severity of clinical symptoms, inflammatory cell infiltration, degree of intestinal barrier damage and composition of the intestinal flora. The results showed that weight loss, disease activity index score, colon shortening and colonic pathological damage were significantly increased in b2KO mice while MC activation, cytokine and chemokine secretion, and inflammatory cell infiltration were decreased. In addition, the abundance and diversity of the intestinal microbiota were reduced in b2KO mice. B2KO mice also exhibited a reduction of probiotics such as norank_f_Muribaculaceae and Lactobacillus and increase of harmful bacteria like Escherichia-Shigella. Intestinal mucosal barrier damage of b2KO mice was more severe than that of WT mice due to the attenuated expression of mucin-2 and occludin. These results demonstrated that MRGPRB2 may have a protective effect on DSS-induced colitis by altering the intestinal flora, participating in barrier repair and recruiting inflammatory cells to eliminate pathogens.
Collapse
Affiliation(s)
- Ming Shao
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of GastroenterologyHubei Key Laboratory of Digestive DiseasesWuhanChina
| | - Jingwen Liu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of GastroenterologyHubei Key Laboratory of Digestive DiseasesWuhanChina
| | - Hesheng Luo
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
26
|
van der Elst G, Varol H, Hermans M, Baan CC, Duong-van Huyen JP, Hesselink DA, Kramann R, Rabant M, Reinders MEJ, von der Thüsen JH, van den Bosch TPP, Clahsen-van Groningen MC. The mast cell: A Janus in kidney transplants. Front Immunol 2023; 14:1122409. [PMID: 36891297 PMCID: PMC9986315 DOI: 10.3389/fimmu.2023.1122409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Mast cells (MCs) are innate immune cells with a versatile set of functionalities, enabling them to orchestrate immune responses in various ways. Aside from their known role in allergy, they also partake in both allograft tolerance and rejection through interaction with regulatory T cells, effector T cells, B cells and degranulation of cytokines and other mediators. MC mediators have both pro- and anti-inflammatory actions, but overall lean towards pro-fibrotic pathways. Paradoxically, they are also seen as having potential protective effects in tissue remodeling post-injury. This manuscript elaborates on current knowledge of the functional diversity of mast cells in kidney transplants, combining theory and practice into a MC model stipulating both protective and harmful capabilities in the kidney transplant setting.
Collapse
Affiliation(s)
- G van der Elst
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - H Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M Hermans
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - C C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - D A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - R Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - M Rabant
- Department of Pathology, Necker Hospital, APHP, Paris, France
| | - M E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
27
|
Wang Y, Yu M, Matsushita K, Liu C, Ishihara N, Nomura M, Tsai M, Galli SJ. Dynamin-related protein 1 differentially regulates FcεRI- and substance P-induced mast cell activation. J Allergy Clin Immunol 2022; 150:1228-1231.e5. [PMID: 35561839 PMCID: PMC9643595 DOI: 10.1016/j.jaci.2022.04.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The mitochondrial fission protein dynamin-related protein 1 (Drp1) has been suggested to regulate mast cell (MC) activation by certain stimuli in vitro, but its functions in MCs activated by various stimuli in vivo have not yet been examined. OBJECTIVE We sought to analyze Drp1 function in both mouse and human MCs. METHODS We used human peripheral blood-derived cultured MCs and 2 genetic mouse models in which MCs were depleted of Drp1: Drp1fl/flMcpt5cre+/- mice and Drp1fl/flCpa3cre+/- mice. RESULTS In mice, Drp1 depletion enhanced FcεRI-induced MC activation while suppressing substance P-stimulated MC activation in vitro and in vivo. This was also true in human peripheral blood-derived cultured MCs in vitro after pharmacologic inhibition of Drp1. CONCLUSION Drp1 differentially regulates MC activation by various stimuli. Promoting Drp1 activation might therefore represent a novel therapy for suppressing IgE-dependent MC activation. Further, inhibiting Drp1 activation might mitigate other MC-dependent responses, such as those induced by substance P.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif
| | - Mang Yu
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif
| | - Kazufumi Matsushita
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Chen Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Naotada Ishihara
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
28
|
Babina M, Franke K, Bal G. How "Neuronal" Are Human Skin Mast Cells? Int J Mol Sci 2022; 23:ijms231810871. [PMID: 36142795 PMCID: PMC9505265 DOI: 10.3390/ijms231810871] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Mast cells are evolutionarily old cells and the principal effectors in allergic responses and inflammation. They are seeded from the yolk sac during embryogenesis or are derived from hematopoietic progenitors and are therefore related to other leukocyte subsets, even though they form a separate clade in the hematopoietic system. Herein, we systematically bundle information from several recent high-throughput endeavors, especially those comparing MCs with other cell types, and combine such information with knowledge on the genes’ functions to reveal groups of neuronal markers specifically expressed by MCs. We focus on recent advances made regarding human tissue MCs, but also refer to studies in mice. In broad terms, genes hyper-expressed in MCs, but largely inactive in other myelocytes, can be classified into subcategories such as traffic/lysosomes (MLPH and RAB27B), the dopamine system (MAOB, DRD2, SLC6A3, and SLC18A2), Ca2+-related entities (CALB2), adhesion molecules (L1CAM and NTM) and, as an overall principle, the transcription factors and modulators of transcriptional activity (LMO4, PBX1, MEIS2, and EHMT2). Their function in MCs is generally unknown but may tentatively be deduced by comparison with other systems. MCs share functions with the nervous system, as they express typical neurotransmitters (histamine and serotonin) and a degranulation machinery that shares features with the neuronal apparatus at the synapse. Therefore, selective overlaps are plausible, and they further highlight the uniqueness of MCs within the myeloid system, as well as when compared with basophils. Apart from investigating their functional implications in MCs, a key question is whether their expression in the lineage is due to the specific reactivation of genes normally silenced in leukocytes or whether the genes are not switched off during mastocytic development from early progenitors.
Collapse
Affiliation(s)
- Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
- Correspondence:
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Allergology, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
29
|
Tang X, Hou Y, Schwartz TW, Haeggström JZ. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids. Biochem Pharmacol 2022; 204:115208. [PMID: 35963340 DOI: 10.1016/j.bcp.2022.115208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Eicosanoids are a family of bioactive compounds derived from arachidonic acid (AA) that play pivotal roles in physiology and disease, including inflammatory conditions of multiple organ systems. The biosynthesis of eicosanoids requires a series of catalytic steps that are controlled by designated enzymes, which can be regulated by inflammatory and stress signals via transcriptional and translational mechanisms. In the past decades, evidence have emerged indicating that G-protein coupled receptors (GPCRs) can sense extracellular metabolites, and regulate inflammatory responses including eicosanoid production. This review focuses on the recent advances of metabolite GPCRs research, their role in regulation of eicosanoid biosynthesis, and the link to pathophysiological conditions.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| | - Yaolin Hou
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Thue W Schwartz
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department for Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| |
Collapse
|
30
|
El Ansari YS, Kanagaratham C, Burton OT, Santos JV, Hollister BMA, Lewis OL, Renz H, Oettgen HC. Allergen-Specific IgA Antibodies Block IgE-Mediated Activation of Mast Cells and Basophils. Front Immunol 2022; 13:881655. [PMID: 35865546 PMCID: PMC9294179 DOI: 10.3389/fimmu.2022.881655] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mast cells and basophils have long been implicated in the pathogenesis of IgE-mediated hypersensitivity reactions. They express the high-affinity IgE receptor, FcϵRI, on their surface. Antigen-induced crosslinking of IgE antibodies bound to that receptor triggers a signaling cascade that results in activation, leading to the release of an array of preformed vasoactive mediators and rapidly synthesized lipids, as well as the de novo production of inflammatory cytokines. In addition to bearing activating receptors like FcεRI, these effector cells of allergy express inhibitory ones including FcγR2b, an IgG Fc receptor with a cytosolic inhibitory motif that activates protein tyrosine phosphatases that suppress IgE-mediated activation. We and others have shown that food allergen-specific IgG antibodies strongly induced during the course of oral immunotherapy (OIT), signal via FcγR2b to suppress IgE-mediated mast cell and basophil activation triggered by food allergen challenge. However, the potential inhibitory effects of IgA antibodies, which are also produced in response to OIT and are present at high levels at mucosal sites, including the intestine where food allergens are encountered, have not been well studied. Here we uncover an inhibitory function for IgA. We observe that IgA binds mouse bone marrow-derived mast cells (BMMCs) and peritoneal mast cells. Binding to BMMCs is dependent on calcium and sialic acid. We also found that IgA antibodies inhibit IgE-mediated mast cell degranulation in an allergen-specific fashion. Antigen-specific IgA inhibits IgE-mediated mast cell activation early in the signaling cascade, suppressing the phosphorylation of Syk, the proximal protein kinase mediating FcεRI signaling, and suppresses mast cell production of cytokines. Furthermore, using basophils from a peanut allergic donor we found that IgA binds to basophils and that activation by exposure to peanuts is effectively suppressed by IgA. We conclude that IgA serves as a regulator of mast cell and basophil degranulation, suggesting a physiologic role for IgA in the maintenance of immune homeostasis at mucosal sites.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Oliver T. Burton
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Jenna V. Santos
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | | | - Owen L. Lewis
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Harald Renz
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, German Center for Lung Research (DZL), 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
- *Correspondence: Hans C. Oettgen,
| |
Collapse
|
31
|
Afify SM, Hassan G, Seno A, Seno M. Cancer-inducing niche: the force of chronic inflammation. Br J Cancer 2022; 127:193-201. [PMID: 35292758 PMCID: PMC9296522 DOI: 10.1038/s41416-022-01775-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
The growth of cancer tissue is thought to be considered driven by a small subpopulation of cells, so-called cancer stem cells (CSCs). CSCs are located at the apex of a hierarchy in a cancer tissue with self-renewal, differentiation and tumorigenic potential that produce the progeny in the tissue. Although CSCs are generally believed to play a critical role in the growth, metastasis, and recurrence of cancers, the origin of CSCs remains to be reconsidered. We hypothesise that, chronic diseases, including obesity and diabetes, establish the cancer-inducing niche (CIN) that drives the undifferentiated/progenitor cells into CSCs, which then develop malignant tumours in vivo. In this context, a CIN could be traced to chronic inflammation that involves long-lasting tissue damage and repair after being exposed to factors such as cytokines and growth factors. This must be distinguished from the cancer microenvironment, which is responsible for cancer maintenance. The concept of a CIN is most important for cancer prevention as well as cancer therapy.
Collapse
Affiliation(s)
- Said M Afify
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.
- Division of Biochemistry, Chemistry Department, Faculty of Science, Menoufia University, Shebin El Koum-Menoufia, 32511, Egypt.
| | - Ghmkin Hassan
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Akimasa Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
- Okayama University Research Laboratory of Stem Cell Engineering in Detroit, IBio, Wayne State University, Detroit, MI, USA
| | - Masaharu Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.
- Okayama University Research Laboratory of Stem Cell Engineering in Detroit, IBio, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
32
|
Laggner M, Acosta GS, Kitzmüller C, Copic D, Gruber F, Altenburger LM, Vorstandlechner V, Gugerell A, Direder M, Klas K, Bormann D, Peterbauer A, Shibuya A, Bohle B, Ankersmit HJ, Mildner M. The secretome of irradiated peripheral blood mononuclear cells attenuates activation of mast cells and basophils. EBioMedicine 2022; 81:104093. [PMID: 35671621 PMCID: PMC9168057 DOI: 10.1016/j.ebiom.2022.104093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND IgE-mediated hypersensitivity is becoming increasingly prevalent and activation of mast cells and basophils represent key events in the pathophysiology of allergy. We have previously reported that the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCsec) exerts beneficial anti-inflammatory effects. Yet, its ability to alleviate allergic symptoms has not been investigated so far. METHODS Several experimental in vitro and in vivo models have been used in this basic research study. A murine ear swelling model was used to study the effects of PBMCsec on 48/80-induced mast cell degranulation in vivo. The transcriptional profile of murine mast cells was analysed by single cell RNA sequencing (scRNAseq). Mast cell activation was studied in vitro using primary skin mast cells. Basophils from individuals allergic to birch pollens were used to investigate basophile activation by allergens. Transcriptomic and lipidomic analyses were used to identify mRNA expression and lipid species present in PBMCsec, respectively. FINDINGS Topical application of PBMCsec on mouse ears (C57BL/6) significantly reduced tissue swelling following intradermal injection of compound 48/80, an inducer of mast cell degranulation. Single cell RNA sequencing of PBMCsec-treated murine dermal mast cells (Balb/c) revealed a downregulation of genes involved in immune cell degranulation and Fc-receptor signalling. In addition, treatment of primary human dermal mast cells with PBMCsec strongly inhibited compound 48/80- and α-IgE-induced mediator release in vitro. Furthermore, PBMCsec remarkably attenuated allergen driven activation of basophils from allergic individuals. Transcriptomic analysis of these basophils showed that PBMCsec downregulated a distinct gene battery involved in immune cell degranulation and Fc-receptor signalling, corroborating results obtained from dermal mast cells. Finally, we identified the lipid fraction of PBMCsec as the major active ingredient involved in effector cell inhibition. INTERPRETATION Collectively, our data demonstrate that PBMCsec is able to reduce activation of mast cells and basophils, encouraging further studies on the potential use of PBMCsec for treating allergy. FUNDING Austrian Research Promotion Agency (852748 and 862068, 2015-2019), Vienna Business Agency (2343727, 2018-2020), Aposcience AG, Austrian Federal Ministry of Education, Science and Research (SPA06/055), Danube Allergy Research Cluster, Austrian Science Fund (I4437 and P32953).
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria
| | - Gabriela Sánchez Acosta
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Claudia Kitzmüller
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | | | - Vera Vorstandlechner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria; Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Martin Direder
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Katharina Klas
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria
| | - Anja Peterbauer
- Aposcience AG, Vienna, Austria; Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, Austria; Aposcience AG, Vienna, Austria.
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria.
| |
Collapse
|
33
|
Allantoin Inhibits Compound 48/80-Induced Pseudoallergic Reactions In Vitro and In Vivo. Molecules 2022; 27:molecules27113473. [PMID: 35684410 PMCID: PMC9182162 DOI: 10.3390/molecules27113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
Pseudoallergic reactions are hypersensitivity reactions mediated by an IgE-independent mechanism. Since allantoin (AT)-mediated pseudoallergy has not been studied, in this study, our objective is to investigate the anti-pseudoallergy effect of AT and its underlying mechanism. In vitro, β-hexosaminidase (β-Hex) and histamine (HIS) release assays, inflammatory cytokine assays, toluidine blue staining, and F-actin microfilament staining were used to evaluate the inhibitory effect of AT in RBL-2H3 cells stimulated with Compound 48/80 (C48/80). Western blot analysis is further performed to investigate intracellular calcium fluctuation-related signaling pathways. In vivo, Evans Blue extraction, paw swelling, and the diameter of Evans Blue extravasation were evaluated, and skin tissues are examined for histopathological examination in mice with passive cutaneous anaphylaxis (PCA) induced by C48/80. Body temperature is measured, and the levels of cytokines are further determined by ELISA kits in mice with active systemic anaphylaxis (ASA) induced by C48/80. The results show that AT dose-dependently inhibited degranulation in C48/80-stimulated RBL-2H3 cells by inhibiting β-Hex and HIS release, reducing the levels of TNF-α, IL-8, and MCP-1, inhibiting shape changes due to degranulation and disassembling the F-actin cytoskeleton. Furthermore, AT dose-dependently inhibits the phosphorylation of PLCγ and IP3R. In vivo, AT decreased Evans Blue extravasation, paw swelling, and the diameter of Evans Blue extravasation and significantly ameliorate pathological changes and mast cell degranulation in C48/80-induced PCA. Furthermore, AT help the mice recover from the C48/80-induced decrease in body temperature and decreased the levels of cytokines in C48/80-treated ASA mice. Our results indicate that allantoin inhibits compound 48/80-induced pseudoallergic reactions. AT has the potential to be used in IgE-independent anti-allergic and anti-inflammatory therapies.
Collapse
|
34
|
Steiner P, Arlt E, Boekhoff I, Gudermann T, Zierler S. Two-Pore Channels Regulate Inter-Organellar Ca 2+ Homeostasis in Immune Cells. Cells 2022; 11:1465. [PMID: 35563771 PMCID: PMC9103377 DOI: 10.3390/cells11091465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 02/01/2023] Open
Abstract
Two-pore channels (TPCs) are ligand-gated cation-selective ion channels that are preserved in plant and animal cells. In the latter, TPCs are located in membranes of acidic organelles, such as endosomes, lysosomes, and endolysosomes. Here, we focus on the function of these unique ion channels in mast cells, which are leukocytes that mature from myeloid hematopoietic stem cells. The cytoplasm of these innate immune cells contains a large number of granules that comprise messenger substances, such as histamine and heparin. Mast cells, along with basophil granulocytes, play an essential role in anaphylaxis and allergic reactions by releasing inflammatory mediators. Signaling in mast cells is mainly regulated via the release of Ca2+ from the endoplasmic reticulum as well as from acidic compartments, such as endolysosomes. For the crosstalk of these organelles TPCs seem essential. Allergic reactions and anaphylaxis were previously shown to be associated with the endolysosomal two-pore channel TPC1. The release of histamine, controlled by intracellular Ca2+ signals, was increased upon genetic or pharmacologic TPC1 inhibition. Conversely, stimulation of TPC channel activity by one of its endogenous ligands, namely nicotinic adenine dinucleotide phosphate (NAADP) or phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), were found to trigger the release of Ca2+ from the endolysosomes; thereby improving the effect of TPC1 on regulated mast cell degranulation. In this review we discuss the importance of TPC1 for regulating Ca2+ homeostasis in mast cells and the overall potential of TPC1 as a pharmacological target in anti-inflammatory therapy.
Collapse
Affiliation(s)
- Philip Steiner
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria;
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Susanna Zierler
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria;
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| |
Collapse
|
35
|
Wang Z, Franke K, Bal G, Li Z, Zuberbier T, Babina M. MRGPRX2-Mediated Degranulation of Human Skin Mast Cells Requires the Operation of Gαi, Gαq, Ca++ Channels, ERK1/2 and PI3K—Interconnection between Early and Late Signaling. Cells 2022; 11:cells11060953. [PMID: 35326404 PMCID: PMC8946553 DOI: 10.3390/cells11060953] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
The recent discovery of MRGPRX2 explains mast cell (MC)-dependent symptoms independently of FcεRI-activation. Because of its novelty, signaling cascades triggered by MRGPRX2 are rudimentarily understood, especially in cutaneous MCs, by which MRGPRX2 is chiefly expressed. Here, MCs purified from human skin were used following preculture or ex vivo and stimulated by FcεRI-aggregation or MRGPRX2 agonists (compound 48/80, Substance P) in the presence/absence of inhibitors. Degranulation was assessed by β-hexosaminidase or histamine release. Phosphorylation events were studied by immunoblotting. As a G protein-coupled receptor, MRGPRX2 signals by activating G proteins; however, their nature has remained controversial. In skin MCs, Gαi and Gαq were required for degranulation, but Gαi was clearly more relevant. Ca++ channels were likewise crucial. Downstream, PI3K was essential for granule discharge initiated by MRGPRX2 or FcεRI. ERK1/2 and JNK were additional participants, especially in the allergic route. Addressing possible points of intersection between early and later events, pERK1/2 and pAKT were found to depend on Gαi, further highlighting its significance. Gαq and Ca++ channels made some contributions to the phosphorylation of ERK. Ca++ differentially affected PI3K activation in FcεRI- vis-à-vis MRGPRX2-signaling, as channel inhibition increased pAKT only when triggered via FcεRI. Collectively, our study significantly extends our understanding of the molecular framework behind granule secretion from skin MCs.
Collapse
Affiliation(s)
- Zhao Wang
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Department of Dermatology, The Second Affiliated Hospital, Northwest Hospital, Xi’an Jiaotong University, Xi’an 710004, China
| | - Kristin Franke
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Gürkan Bal
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Zhuoran Li
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Magda Babina
- Institute for Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (Z.W.); (K.F.); (G.B.); (Z.L.); (T.Z.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
- Correspondence:
| |
Collapse
|
36
|
Mast Cells and Acupuncture Analgesia. Cells 2022; 11:cells11050860. [PMID: 35269483 PMCID: PMC8909752 DOI: 10.3390/cells11050860] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
Mast cells are widely distributed in various parts of the human body and play a vital role in the progression of many diseases. Recently, the close relationship between mast cells and acupoints was elucidated, and the role of mast cells in acupuncture analgesia has attracted the attention of researchers worldwide. Using mast cells, acupuncture analgesia and acupoint as key words to search CNKI, PubMed, Web of Science and other databases, combining the representative articles in these databases with the published research papers of our group, we summarized: The enrichment of mast cells and the dense arrangement of collagen fibers, microvessels, and nerves form the basis for acupoints as the reaction sites of acupuncture; acupuncture can cause the deformation of collagen fibers and activate TRPV channels on mast cells membrane, so as to stimulate mast cells to release bioactive substances and activate nerve receptors to generate analgesic effect; system biology models are set up to explain the quantitative process of information initiation and transmission at acupuncture points, and indicate that the acupuncture effect depends on the local mast cells density. In a conclusion, this review will give a scientific explanation of acupuncture analgesia from the material basis of acupoints, the local initiation, and afferent biological mechanism.
Collapse
|
37
|
Yang BC, Castells MC. The Who, What, Where, When, Why, and How of Drug Desensitization. Immunol Allergy Clin North Am 2022; 42:403-420. [DOI: 10.1016/j.iac.2021.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
38
|
Wan X, Zhang Y, Tang H, Li M, Jiang T, He J, Bao C, Wang J, Song Y, Xiao P, Liu Y, Lai L, Wang Q. IL‐27 signaling negatively regulates FcɛRI‐mediated mast cell activation and allergic response. J Leukoc Biol 2022; 112:411-424. [PMID: 35075687 DOI: 10.1002/jlb.2ma1221-637r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 11/29/2021] [Accepted: 12/28/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Xiaopeng Wan
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
- State Key Laboratory of Veterinary Biotechnology, Harbin Veternary Research Institute Chinese Academy of Agricultural Sciences Harbin China
| | - Yuanyuan Zhang
- Department of Pulmonology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health Hangzhou China
| | - Huanna Tang
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Mengyao Li
- Department of Pulmonology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health Hangzhou China
| | - Tianqi Jiang
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Jia He
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Chunjing Bao
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Junkai Wang
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Yinjing Song
- Department of Dermatology and Venereology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Peng Xiao
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Yang Liu
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| | - Lihua Lai
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
- Department of Pharmacology Zhejiang University School of Medicine Hangzhou China
| | - Qingqing Wang
- Institute of Immunology Zhejiang University School of Medicine Hangzhou China
| |
Collapse
|
39
|
Jang HY, Ha DH, Rah SY, Lee DH, Lee SM, Park BH. Sirtuin 6 is a negative regulator of FcεRI signaling and anaphylactic responses. J Allergy Clin Immunol 2022; 149:156-167.e7. [PMID: 34051221 DOI: 10.1016/j.jaci.2021.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Binding IgE to a cognate allergen causes aggregation of Fcε receptor I (FcεRI) in mast cells, resulting in activation of receptor-associated Src family tyrosine kinases, including Lyn and Syk. Protein tyrosine phosphatase, receptor type C (PTPRC), also known as CD45, has emerged as a positive regulator of FcεRI signaling by dephosphorylation of the inhibitory tyrosine of Lyn. OBJECTIVE Sirtuin 6 (Sirt6), a NAD+-dependent deacetylase, exhibits an anti-inflammatory property. It remains to be determined, however, whether Sirt6 attenuates mast cell-associated diseases, including anaphylaxis. METHODS FcεRI signaling and mast cell degranulation were measured after IgE cross-linking in murine bone marrow-derived mast cells (BMMCs) and human cord blood-derived mast cells. To investigate the function of Sirt6 in mast cell activation in vivo, we used mast cell-dependent animal models of passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA). RESULTS Sirt6-deficient BMMCs augmented IgE-FcεRI-mediated signaling and degranulation compared to wild-type BMMCs. Reconstitution of mast cell-deficient KitW-sh/W-sh mice with BMMCs received from Sirt6 knockout mice developed more severe PSA and PCA compared to mice engrafted with wild-type BMMCs. Similarly, genetic overexpression or pharmacologic activation of Sirt6 suppressed mast cell degranulation and blunted responses to PCA. Mechanistically, Sirt6 deficiency increased PTPRC transcription via acetylating histone H3, leading to enhanced aggregation of FcεRI in BMMCs. Finally, we recapitulated the Sirt6 regulation of PTPRC and FcεRI signaling in human mast cells. CONCLUSIONS Sirt6 acts as a negative regulator of FcεRI signaling cascade in mast cells by suppressing PTPRC transcription. Activation of Sirt6 may therefore represent a promising and novel therapeutic strategy for anaphylaxis.
Collapse
Affiliation(s)
- Hyun-Young Jang
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea
| | - Do Hyun Ha
- Division of Biotechnology, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan, Korea
| | - So-Young Rah
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea
| | - Dong-Hyun Lee
- Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju, Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
40
|
Lopez-Perez D, Redruello-Romero A, Garcia-Rubio J, Arana C, Garcia-Escudero LA, Tamayo F, Salmeron J, Galvez J, Leon J, Carazo Á. In Obese Patients With Type 2 Diabetes, Mast Cells in Omental Adipose Tissue Decrease the Surface Expression of CD45, CD117, CD203c, and FcϵRI. Front Endocrinol (Lausanne) 2022; 13:818388. [PMID: 35370964 PMCID: PMC8965342 DOI: 10.3389/fendo.2022.818388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
The paradigm of mast cells in type 2 diabetes is changing. Although they were first considered deleterious inflammatory cells, now they seem to be important players driving adipose tissue homeostasis. Here we have employed a flow cytometry-based approach for measuring the surface expression of 4 proteins (CD45, CD117, CD203c, and FcϵRI) on mast cells of omental (o-WAT) and subcutaneous white adipose tissue (s-WAT) in a cohort of 96 patients with morbid obesity. The cohort was split into three groups: non-T2D, pre-T2D, and T2D. Noteworthy, patients with T2D have a mild condition (HbA1c <7%). In o-WAT, mast cells of patients with T2D have a decrease in the surface expression of CD45 (p=0.0013), CD117 (p=0.0066), CD203c (p=0.0025), and FcϵRI (p=0.043). Besides, in s-WAT, the decrease was seen only in CD117 (p=0.046). These results indicate that T2D affects more to mast cells in o-WAT than in s-WAT. The decrease in these four proteins has serious effects on mast cell function. CD117 is critical for mast cell survival, while CD45 and FcϵRI are important for mast cell activation. Additionally, CD203c is only present on the cell surface after granule release. Taking together these observations, we suggest that mast cells in o-WAT of patients with T2D have a decreased survival, activation capacity, and secretory function.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Anaïs Redruello-Romero
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | | - Carlos Arana
- Endocrinology and nutrition department, Virgen de la Luz University Hospital, Cuenca, Spain
| | - Luis A. Garcia-Escudero
- Department of Statistics and Operative Research, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | | | - Javier Salmeron
- Gastroenterology Unit, San Cecilio University Hospital, Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, Granada, Spain
- *Correspondence: Julio Galvez, ; Ángel Carazo,
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
| | - Ángel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
- *Correspondence: Julio Galvez, ; Ángel Carazo,
| |
Collapse
|
41
|
Dahlin JS, Maurer M, Metcalfe DD, Pejler G, Sagi‐Eisenberg R, Nilsson G. The ingenious mast cell: Contemporary insights into mast cell behavior and function. Allergy 2022; 77:83-99. [PMID: 33955017 DOI: 10.1111/all.14881] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
Mast cells are (in)famous for their role in allergic diseases, but the physiological and pathophysiological roles of this ingenious cell are still not fully understood. Mast cells are important for homeostasis and surveillance of the human system, recognizing both endogenous and exogenous agents, which induce release of a variety of mediators acting on both immune and non-immune cells, including nerve cells, fibroblasts, endothelial cells, smooth muscle cells, and epithelial cells. During recent years, clinical and experimental studies on human mast cells, as well as experiments using animal models, have resulted in many discoveries that help decipher the function of mast cells in health and disease. In this review, we focus particularly on new insights into mast cell biology, with a focus on mast cell development, recruitment, heterogeneity, and reactivity. We also highlight the development in our understanding of mast cell-driven diseases and discuss the development of novel strategies to treat such conditions.
Collapse
Affiliation(s)
- Joakim S. Dahlin
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Marcus Maurer
- Department of Dermatology and Allergy Dermatological Allergology Allergie‐Centrum‐Charité Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, Berlin Institute of Health Berlin Germany
| | - Dean D. Metcalfe
- Mast Cell Biology Section Laboratory of Allergic Diseases NIAID, NIH Bethesda MD USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
- Department of Anatomy, Physiology and Biochemistry Swedish University of Agricultural Sciences Uppsala Sweden
| | - Ronit Sagi‐Eisenberg
- Department of Cell and Developmental Biology Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Gunnar Nilsson
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
- Department of Medical Sciences Uppsala University Uppsala Sweden
| |
Collapse
|
42
|
Buelow LM, Hoji A, Tat K, Schroeder-Carter LM, Carroll DJ, Cook-Mills JM. Mechanisms for Alternaria alternata Function in the Skin During Induction of Peanut Allergy in Neonatal Mice With Skin Barrier Mutations. FRONTIERS IN ALLERGY 2021; 2:677019. [PMID: 35387035 PMCID: PMC8974772 DOI: 10.3389/falgy.2021.677019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/16/2021] [Indexed: 01/07/2023] Open
Abstract
Neonatal mice with heterozygous mutations in genes encoding the skin barrier proteins filaggrin and mattrin (flaky tail mice [FT+/-]) exhibit oral peanut-induced anaphylaxis after skin sensitization. As we have previously reported, sensitization in this model is achieved via skin co- exposure to the environmental allergen Alternaria alternata (Alt), peanut extract (PNE), and detergent. However, the function of Alt in initiation of peanut allergy in this model is little understood. The purpose of this study was to investigate candidate cytokines induced by Alt in the skin and determine the role of these cytokines in the development of food allergy, namely oncostatin M (Osm), amphiregulin (Areg), and IL-33. RT-qPCR analyses demonstrated that skin of FT+/- neonates expressed Il33 and Osm following Alt or Alt/PNE but not PNE exposure. By contrast, expression of Areg was induced by either Alt, PNE, or Alt/PNE sensitization in FT+/- neonates. In scRNAseq analyses, Osm, Areg, and Il33 were expressed by several cell types, including a keratinocyte cluster that was expanded in the skin of Alt/PNE-exposed FT+/- pups as compared to Alt/PNE-exposed WT pups. Areg and OSM were required for oral PNE-induced anaphylaxis since anaphylaxis was inhibited by administration of neutralizing anti-Areg or anti-OSM antibodies prior to each skin sensitization with Alt/PNE. It was then determined if intradermal injection of recombinant IL33 (rIL33), rAreg, or rOSM in the skin could substitute for Alt during skin sensitization to PNE. PNE skin sensitization with intradermal rIL33 was sufficient for oral PNE-induced anaphylaxis, whereas skin sensitization with intradermal rAreg or rOSM during skin exposure to PNE was not sufficient for anaphylaxis to oral PNE challenge. Based on these studies a pathway for IL33, Areg and OSM in Alt/PNE sensitized FT+/- skin was defined for IgE induction and anaphylaxis. Alt stimulated two pathways, an IL33 pathway and a pathway involving OSM and Areg. These two pathways acted in concert with PNE to induce food allergy in pups with skin barrier mutations.
Collapse
|
43
|
Tontini C, Bulfone-Paus S. Novel Approaches in the Inhibition of IgE-Induced Mast Cell Reactivity in Food Allergy. Front Immunol 2021; 12:613461. [PMID: 34456900 PMCID: PMC8387944 DOI: 10.3389/fimmu.2021.613461] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 06/23/2021] [Indexed: 01/21/2023] Open
Abstract
Allergy is an IgE-dependent type-I hypersensitivity reaction that can lead to life-threatening systemic symptoms such as anaphylaxis. In the pathogenesis of the allergic response, the common upstream event is the binding of allergens to specific IgE, inducing cross-linking of the high-affinity FcεRI on mast cells, triggering cellular degranulation and the release of histamine, proteases, lipids mediators, cytokines and chemokines with inflammatory activity. A number of novel therapeutic options to curb mast cell activation are in the pipeline for the treatment of severe allergies. In addition to anti-IgE therapy and allergen-specific immunotherapy, monoclonal antibodies targeted against several key Th2/alarmin cytokines (i.e. IL-4Rα, IL-33, TSLP), active modification of allergen-specific IgE (i.e. inhibitory compounds, monoclonal antibodies, de-sialylation), engagement of inhibitory receptors on mast cells and allergen-specific adjuvant vaccines, are new promising options to inhibit the uncontrolled release of mast cell mediators upon allergen exposure. In this review, we critically discuss the novel approaches targeting mast cells limiting allergic responses and the immunological mechanisms involved, with special interest on food allergy treatment.
Collapse
Affiliation(s)
- Chiara Tontini
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
44
|
Galli SJ, Gaudenzio N, Tsai M. Mast Cells in Inflammation and Disease: Recent Progress and Ongoing Concerns. Annu Rev Immunol 2021; 38:49-77. [PMID: 32340580 DOI: 10.1146/annurev-immunol-071719-094903] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mast cells have existed long before the development of adaptive immunity, although they have been given different names. Thus, in the marine urochordate Styela plicata, they have been designated as test cells. However, based on their morphological characteristics (including prominent cytoplasmic granules) and mediator content (including heparin, histamine, and neutral proteases), test cells are thought to represent members of the lineage known in vertebrates as mast cells. So this lineage presumably had important functions that preceded the development of antibodies, including IgE. Yet mast cells are best known, in humans, as key sources of mediators responsible for acute allergic reactions, notably including anaphylaxis, a severe and potentially fatal IgE-dependent immediate hypersensitivity reaction to apparently harmless antigens, including many found in foods and medicines. In this review, we briefly describe the origins of tissue mast cells and outline evidence that these cells can have beneficial as well as detrimental functions, both innately and as participants in adaptive immune responses. We also discuss aspects of mast cell heterogeneity and comment on how the plasticity of this lineage may provide insight into its roles in health and disease. Finally, we consider some currently open questions that are yet unresolved.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; , .,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, California 94305, USA
| | - Nicolas Gaudenzio
- Unité de Différenciation Epithéliale et Autoimmunité Rhumatoïde (UDEAR), INSERM UMR 1056, Université de Toulouse, 31 059 Toulouse CEDEX 9, France;
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA; , .,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, California 94305, USA
| |
Collapse
|
45
|
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
|
46
|
Masini M, Suleiman M, Novelli M, Marselli L, Marchetti P, De Tata V. Mast Cells and the Pancreas in Human Type 1 and Type 2 Diabetes. Cells 2021; 10:cells10081875. [PMID: 34440644 PMCID: PMC8391487 DOI: 10.3390/cells10081875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
Mast cells are highly differentiated, widely distributed cells of the innate immune system, that are currently considered as key regulators of both innate and adaptive immunity. Mast cells play a key role in health and survival mechanisms, especially as sentinel cells that can stimulate protective immune responses. On the other hand, it has been shown that mast cells are involved in the pathogenesis of several diseases, and recently a possible pathogenetic role of mast cells in diabetes has been proposed. In this review we summarize the evidence on the increased presence of mast cells in the pancreas of subjects with type 1 diabetes, which is due to the autoimmune destruction of insulin secreting beta cells, and discuss the differences with type 2 diabetes, the other major form of diabetes. In addition, we describe some of the pathophysiological mechanisms through which mast cells might exert their actions, which could be targeted to potentially protect the beta cells in autoimmune diabetes.
Collapse
Affiliation(s)
- Matilde Masini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55-Scuola Medica, 56126 Pisa, Italy; (M.M.); (M.N.)
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, Pancreatic Islet Laboratory, University of Pisa, 56124 Pisa, Italy; (M.S.); (L.M.); (P.M.)
| | - Michela Novelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55-Scuola Medica, 56126 Pisa, Italy; (M.M.); (M.N.)
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Pancreatic Islet Laboratory, University of Pisa, 56124 Pisa, Italy; (M.S.); (L.M.); (P.M.)
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Pancreatic Islet Laboratory, University of Pisa, 56124 Pisa, Italy; (M.S.); (L.M.); (P.M.)
| | - Vincenzo De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55-Scuola Medica, 56126 Pisa, Italy; (M.M.); (M.N.)
- Centro Interdipartimentale di Microscopia Elettronica (C.I.M.E.), University of Pisa, 56126 Pisa, Italy
- Correspondence:
| |
Collapse
|
47
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 980] [Impact Index Per Article: 245.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
48
|
Zhang Z, Kurashima Y. Two Sides of the Coin: Mast Cells as a Key Regulator of Allergy and Acute/Chronic Inflammation. Cells 2021; 10:cells10071615. [PMID: 34203383 PMCID: PMC8308013 DOI: 10.3390/cells10071615] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/17/2022] Open
Abstract
It is well known that mast cells (MCs) initiate type I allergic reactions and inflammation in a quick response to the various stimulants, including—but not limited to—allergens, pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs). MCs highly express receptors of these ligands and proteases (e.g., tryptase, chymase) and cytokines (TNF), and other granular components (e.g., histamine and serotonin) and aggravate the allergic reaction and inflammation. On the other hand, accumulated evidence has revealed that MCs also possess immune-regulatory functions, suppressing chronic inflammation and allergic reactions on some occasions. IL-2 and IL-10 released from MCs inhibit excessive immune responses. Recently, it has been revealed that allergen immunotherapy modulates the function of MCs from their allergic function to their regulatory function to suppress allergic reactions. This evidence suggests the possibility that manipulation of MCs functions will result in a novel approach to the treatment of various MCs-mediated diseases.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- CU-UCSD Center for Mucosal Immunology, Department of Pathology/Medicine, Allergy and Vaccines, University of California, San Diego, CA 92093-0063, USA
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Correspondence: ; Tel.: +81-43-226-2848; Fax: +81-43-226-2183
| |
Collapse
|
49
|
Draberova L, Tumova M, Draber P. Molecular Mechanisms of Mast Cell Activation by Cholesterol-Dependent Cytolysins. Front Immunol 2021; 12:670205. [PMID: 34248949 PMCID: PMC8260682 DOI: 10.3389/fimmu.2021.670205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
Mast cells are potent immune sensors of the tissue microenvironment. Within seconds of activation, they release various preformed biologically active products and initiate the process of de novo synthesis of cytokines, chemokines, and other inflammatory mediators. This process is regulated at multiple levels. Besides the extensively studied IgE and IgG receptors, toll-like receptors, MRGPR, and other protein receptor signaling pathways, there is a critical activation pathway based on cholesterol-dependent, pore-forming cytolytic exotoxins produced by Gram-positive bacterial pathogens. This pathway is initiated by binding the exotoxins to the cholesterol-rich membrane, followed by their dimerization, multimerization, pre-pore formation, and pore formation. At low sublytic concentrations, the exotoxins induce mast cell activation, including degranulation, intracellular calcium concentration changes, and transcriptional activation, resulting in production of cytokines and other inflammatory mediators. Higher toxin concentrations lead to cell death. Similar activation events are observed when mast cells are exposed to sublytic concentrations of saponins or some other compounds interfering with the membrane integrity. We review the molecular mechanisms of mast cell activation by pore-forming bacterial exotoxins, and other compounds inducing cholesterol-dependent plasma membrane perturbations. We discuss the importance of these signaling pathways in innate and acquired immunity.
Collapse
Affiliation(s)
- Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magda Tumova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
50
|
Construction and validation of a scoring system to predict resistance to chemotherapeutic drugs using gene expression profiles in canine lymphoma. Res Vet Sci 2021; 137:208-216. [PMID: 34020336 DOI: 10.1016/j.rvsc.2021.05.007] [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: 12/20/2020] [Revised: 05/03/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022]
Abstract
The present study aimed to verify the changes in the expression levels of 13 candidate genes associated with chemotherapy resistance and to construct a scoring system to predict resistance to these drugs. The expression levels of the 13 candidate genes were compared between 20 dogs with lymphoma that were sensitive to drugs used in CHOP-based protocol and 16 dogs with lymphoma that were resistant to these drugs. The expression levels of six genes; ASNS, CCR3, CALCA, FCER1A, LOC448801, and EDNRB were significantly different between the two groups. A scoring system to predict resistance to cyclophosphamide, doxorubicin and vincristine, which are used in CHOP-based protocol, was constructed based on expression levels of the six genes in these 36 dogs using logistic regression models. After internal validation, sensitivity and specificity of the scoring system were 0.759 and 0.853, respectively. External validation was conducted in another cohort of 33 dogs with lymphoma, and sensitivity and specificity of the scoring system were 0.800 and 0.696, respectively. In conclusion, this study identified six genes associated with resistance to drugs used in CHOP-based protocol in canine lymphoma and proposed a novel scoring system to predict resistance to these drugs. This system might be beneficial in selecting the most appropriate chemotherapy protocol for individual dogs with lymphoma.
Collapse
|