1
|
Stevenson M, Hebron ML, Liu X, Balaraman K, Wolf C, Moussa C. c-KIT inhibitors reduce pathology and improve behavior in the Tg(SwDI) model of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402625. [PMID: 39009412 PMCID: PMC11249953 DOI: 10.26508/lsa.202402625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Treatments for Alzheimer's disease have primarily focused on removing brain amyloid plaques to improve cognitive outcomes in patients. We developed small compounds, known as BK40143 and BK40197, and we hypothesize that these drugs alleviate microglial-mediated neuroinflammation and induce autophagic clearance of neurotoxic proteins to improve behavior in models of neurodegeneration. Specificity binding assays of BK40143 and BK40197 showed primary binding to c-KIT/Platelet Derived Growth Factor Receptors (PDGFR)α/β, whereas BK40197 also differentially binds to FYVE finger-containing phosphoinositide kinase (PIKFYVE). Both compounds penetrate the CNS, and treatment with these drugs inhibited the maturation of peripheral mast cells in transgenic mice, correlating with cognitive improvements on measures of memory and anxiety. In the brain, microglial activation was profoundly attenuated and amyloid-beta and tau were reduced via autophagy. Multi-kinase inhibition, including c-KIT, exerts multifunctional effects to reduce neurodegenerative pathology via autophagy and microglial activity and may represent a potential therapeutic option for neurodegeneration.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Kaluvu Balaraman
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Christian Wolf
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
2
|
Yin J, Wang Y, Han W, Ge W, Yu Q, Jing Y, Yan W, Liu Q, Gong L, Yan S, Wang S, Li X, Li Y, Hu H. Oxytocin Attenuates Sympathetic Innervation with Inhibition of Cardiac Mast Cell Degranulation in Rats after Myocardial Infarction. J Pharmacol Exp Ther 2024; 390:240-249. [PMID: 38902033 DOI: 10.1124/jpet.124.002064] [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: 01/02/2024] [Revised: 05/05/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Sympathetic hyperinnervation is the leading cause of fatal ventricular arrhythmia (VA) after myocardial infarction (MI). Cardiac mast cells cause arrhythmias directly through degranulation. However, the role and mechanism of mast cell degranulation in sympathetic remodeling remain unknown. We investigated the role of oxytocin (OT) in stabilizing cardiac mast cells and improving sympathetic innervation in rats. MI was induced by coronary artery ligation. Western blotting, immunofluorescence, and toluidine staining of mast cells were performed to determine the expression and location of target protein. Mast cells accumulated significantly in peri-infarcted tissues and were present in a degranulated state. They expressed OT receptor (OTR), and OT infusion reduced the number of degranulated cardiac mast cells post-MI. Sympathetic hyperinnervation was attenuated as assessed by immunofluorescence for tyrosine hydroxylase (TH). Seven days post-MI, the arrhythmia score of programmed electrical stimulation was higher in vehicle-treated rats with MI than in rats treated with OT. An in vitro study showed that OT stabilized mast cells via the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Further in vivo studies on OTR-deficient mice showed worsening mast cell degranulation and worsening sympathetic innervation. OT pretreatment inhibited cardiac mast cell degranulation post-MI and prevented sympathetic hyperinnervation, along with mast cell stabilization via the PI3K/Akt pathway. SIGNIFICANCE STATEMENT: This is the first study to elucidate the role and mechanism of oxytocin (OT) in inflammatory-sympathetic communication mediated sympathetic hyperinnervation after myocardial infarction (MI), providing new approaches to prevent fatal arrhythmias.
Collapse
Affiliation(s)
- Jie Yin
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Ye Wang
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Weizhong Han
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Weili Ge
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Qingxia Yu
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Yanyan Jing
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Wenju Yan
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Qian Liu
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Liping Gong
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Suhua Yan
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Shuanglian Wang
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Xiaolu Li
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Yan Li
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| | - Hesheng Hu
- Department of Cardiology (J.Y., Y.W., S.Y., H.H.), Department of Emergency Medicine (X.L.), and Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine (Y.L.), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China; Department of Cardiology, Shandong Provincial Hospital affiliated with Shandong First Medical University, Jinan, China (J.Y., W.H.); Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated with Wenzhou Medical University, Zhejiang, China (W.G.); School of Medicine, Shandong University, Jinan, China (Q.Y., Y.J., W.Y., Q.L.); Department of Infectious Disease and Hepatology, the Second Hospital of Shandong University, Shandong University, Jinan, China (L.G.); and Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (S.W.)
| |
Collapse
|
3
|
Villanueva CR, Barksdale K, Owolabi T, Bridges D, Chichester K, Saini S, Oliver ET. Functional human skin explants as tools for assessing mast cell activation and inhibition. FRONTIERS IN ALLERGY 2024; 5:1373511. [PMID: 38601026 PMCID: PMC11004268 DOI: 10.3389/falgy.2024.1373511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/04/2024] [Indexed: 04/12/2024] Open
Abstract
Mast cells are activated through a variety of different receptors to release preformed granules and mediators synthesized de novo. However, the physiology and function of mast cells are not fully understood. Traditional studies of mast cell activation in humans have utilized cultures of tissue-derived mast cells including CD34+ progenitor cells or well-characterized commercially available cell lines. One limitation of these methods is that mast cells are no longer in a natural state. Therefore, their applicability to human skin disorders may be limited. Human skin explant models have been utilized to investigate the short-term effects of cell mediators, drugs, and irritants on skin while avoiding the ethical concerns surrounding in vivo stimulation studies with non-approved agents. Nonetheless, few studies have utilized intact human tissue to study mast cell degranulation. This "Methods" paper describes the development and application of an intact skin explant model to study human mast cell activation. In this manuscript, we share our protocol for setting up ex vivo human skin explants and describe the results of stimulation experiments and techniques to minimize trauma-induced histamine release. Skin explants were generated using de-identified, full-thickness, non-diseased skin specimens from plastic and reconstructive surgeries. Results were reproducible and demonstrated FcɛRI- and MRGPRX2-induced mediator release which was inhibited with the use of a BTK inhibitor and QWF, respectively. Thus, this explant model provides a quick and accessible method of assessing human skin mast cell activation and inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric T. Oliver
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
P2X4 receptor stimulation enhances MrgprB2-mediated mast cell activation and pseudoallergic reactions in mice. Sci Rep 2022; 12:18613. [PMID: 36329102 PMCID: PMC9633816 DOI: 10.1038/s41598-022-21667-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Pseudoallergies caused by drugs make disease treatment difficult. Mas-relate G protein-coupled receptor X2 (MRGPRX2), which is specifically expressed in mast cells (MCs), has been implicated in pseudoallergies. High concentrations of therapeutic agents are typically required to stimulate MRGPRX2. Although regulatory mechanisms may enhance this response, the factors involved in this regulation are not well-understood. In this study, the effects of extracellular ATP on MC activation induced by MrgprB2, the mouse ortholog of human MRGPRX2, were examined in mouse peritoneal MCs (PMCs). ATP alone induced minimal PMC degranulation but markedly enhanced degranulation induced by the MrgprB2 agonist compound 48/80 (CP48/80), substance P, PAMP-12, and vancomycin. ATP promoted CP48/80-induced increase in intracellular Ca2+ in PMCs. This enhancement effect of ATP was absent in PMCs prepared from P2X4 receptor (P2X4R)-deficient mice and inhibited by the PI3K inhibitor wortmannin. In addition, P2X4R deficiency reduced the skin-specific and systemic anaphylactic responses to CP48/80 in vivo. In MC-deficient KitW-sh/W-sh mice, reconstitution with MCs obtained from wild-type mice led to a more severe anaphylactic response to CP48/80 compared to that from P2X4R-deficient mice. P2X4R-mediated effect may be involved in MrgprB2-mediated MC activation in vivo and is a potential target for alleviating pseudoallergic reactions.
Collapse
|
5
|
Matsuoka I, Yoshida K, Ito MA. Purinergic regulation of mast cell function: P2X4 receptor-mediated enhancement of allergic responses. J Pharmacol Sci 2022; 150:94-99. [PMID: 36055757 DOI: 10.1016/j.jphs.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 10/15/2022] Open
Abstract
Adenosine triphosphate (ATP) initially attracted attention as a neurotransmitter, with much research conducted on the regulation of neurotransmission in the autonomic and central nervous systems. ATP is also abundant as an energy currency in all living cells and is released into extracellular spaces by various regulated mechanisms. The role of ATP and related purine and pyrimidine nucleotides as extracellular signaling molecules in the regulation of immune cell functions has been reported as evidence for purinergic signaling and has become the focus of attention as therapeutic targets for various diseases. Mast cells (MCs) are distributed in tissues in contact with the outside environment and are the first immune cells to respond to non-microbial environmental antigens. Although extracellular ATP is known as an activator of MCs, the details remain to be investigated. Based on our series of studies, this review describes the unique features of ionotropic P2X4 receptor signals in MC functions. The role of purinergic signaling may exist in combination with various physiological, chemical and physical stimuli. The characteristics of P2X4 receptor-mediated action in MCs described in this article may provide clues to reveal the previously unknown effects induced by purinergic signaling.
Collapse
Affiliation(s)
- Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Kazuki Yoshida
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Masa-Aki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| |
Collapse
|
6
|
Wang X, Yi W, He L, Luo S, Wang J, Jiang L, Long H, Zhao M, Lu Q. Abnormalities in Gut Microbiota and Metabolism in Patients With Chronic Spontaneous Urticaria. Front Immunol 2021; 12:691304. [PMID: 34721374 PMCID: PMC8554312 DOI: 10.3389/fimmu.2021.691304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/29/2021] [Indexed: 01/09/2023] Open
Abstract
Background Increasing evidence suggests that the gut microbiome plays a role in the pathogenesis of allergy and autoimmunity. The association between abnormalities in the gut microbiota and chronic spontaneous urticaria (CSU) remains largely undefined. Methods Fecal samples were obtained from 39 patients with CSU and 40 healthy controls (HCs). 16S ribosomal RNA (rRNA) gene sequencing (39 patients with CSU and 40 HCs) and untargeted metabolomics (12 patients with CSU and 12 HCs) were performed to analyze the compositional and metabolic alterations of the gut microbiome in CSU patients and HCs. Results The 16S rRNA gene sequencing results showed a significant difference in the β-diversity of the gut microbiota, presented as the Jaccard distance, between CSU patients and HCs. No significant differences were found in the α-diversity of the gut microbiota between patients and HCs. At the phylum level, the major bacteria in the gut microbiome of patients with CSU were Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. At the genus level, Lactobacillus, Turicibacter, and Lachnobacterium were significantly increased and Phascolarctobacterium was decreased in patients with CSU. PICRUSt and correlation analysis indicated that Lactobacillus, Turicibacter, and Phascolarctobacterium were positively related to G protein-coupled receptors. Metabolomic analysis showed that α-mangostin and glycyrrhizic acid were upregulated and that 3-indolepropionic acid, xanthine, and isobutyric acid were downregulated in patients with CSU. Correlation analysis between the intestinal microbiota and metabolites suggested that there was a positive correlation between Lachnobacterium and α-mangostin. Conclusions This study suggests that disturbances in the gut microbiome composition and metabolites and their crosstalk or interaction may participate in the pathogenesis of CSU.
Collapse
Affiliation(s)
- Xin Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wanyu Yi
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liting He
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuaihantian Luo
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaqi Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Jiang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
7
|
Klewer T, Bakic L, Müller-Reichert T, Kiewisz R, Jessberger G, Kiessling N, Roers A, Jessberger R. E-Cadherin restricts mast cell degranulation in mice. Eur J Immunol 2021; 52:44-53. [PMID: 34606636 DOI: 10.1002/eji.202049087] [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: 12/02/2020] [Revised: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 11/11/2022]
Abstract
Crosslinking of FcεRI-bound IgE triggers the release of a large number of biologically active, potentially anaphylactic compounds by mast cells. FcεRI activation ought to be well-controlled to restrict adverse activation. As mast cells are embedded in tissues, adhesion molecules may contribute to limiting premature activation. Here, we report that E-Cadherin serves that purpose. Having confirmed that cultured mast cells express E-Cadherin, a mast-cell-specific E-Cadherin deficiency, Mcpt5-Cre E-Cdhfl/fl mice, was used to analyze mast cell degranulation in vitro and in vivo. Cultured peritoneal mast cells from Mcpt5-Cre E-Cdhfl/fl mice were normal with respect to many parameters but showed much-enhanced degranulation in three independent assays. Soluble E-Cadherin reduced the degranulation of control cells. The release of some newly synthesized inflammatory cytokines was decreased by E-Cadherin deficiency. Compared to controls, Mcpt5-Cre E-Cdhfl/fl mice reacted much stronger to IgE-dependent stimuli, developing anaphylactic shock. We suggest E-Cadherin-mediated tissue interactions restrict mast cell degranulation to prevent their precocious activation.
Collapse
Affiliation(s)
- Theres Klewer
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ljubica Bakic
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thomas Müller-Reichert
- Core Facility Cellular Imaging, Experimental Center, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Robert Kiewisz
- Core Facility Cellular Imaging, Experimental Center, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gregor Jessberger
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Research Institute of Molecular Pathology, Vienna, Austria
| | - Nadine Kiessling
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Axel Roers
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Rolf Jessberger
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
8
|
Liang HY, Chen Y, Wei X, Ma GG, Ding J, Lu C, Zhou RP, Hu W. Immunomodulatory functions of TRPM7 and its implications in autoimmune diseases. Immunology 2021; 165:3-21. [PMID: 34558663 DOI: 10.1111/imm.13420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022] Open
Abstract
An autoimmune disease is an inappropriate response to one's tissues due to a break in immune tolerance and exposure to self-antigens. It often leads to structural and functional damage to organs and systemic disorders. To date, there are no effective interventions to prevent the progression of autoimmune diseases. Hence, there is an urgent need for new treatment targets. TRPM7 is an enzyme-coupled, transient receptor ion channel of the subfamily M that plays a vital role in pathologic and physiologic conditions. While TRPM7 is constitutively activated under certain conditions, it can regulate cell migration, polarization, proliferation and cytokine secretion. However, a growing body of evidence highlights the critical role of TRPM7 in autoimmune diseases, including rheumatoid arthritis, multiple sclerosis and diabetes. Herein, we present (a) a review of the channel kinase properties of TRPM7 and its pharmacological properties, (b) discuss the role of TRPM7 in immune cells (neutrophils, macrophages, lymphocytes and mast cells) and its upstream immunoreactive substances, and (c) highlight TRPM7 as a potential therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Hong-Yu Liang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xin Wei
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Gang-Gang Ma
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chao Lu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Unlocking the Non-IgE-Mediated Pseudo-Allergic Reaction Puzzle with Mas-Related G-Protein Coupled Receptor Member X2 (MRGPRX2). Cells 2021; 10:cells10051033. [PMID: 33925682 PMCID: PMC8146469 DOI: 10.3390/cells10051033] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/17/2022] Open
Abstract
Mas-related G-protein coupled receptor member X2 (MRGPRX2) is a class A GPCR expressed on mast cells. Mast cells are granulated tissue-resident cells known for host cell response, allergic response, and vascular homeostasis. Immunoglobulin E receptor (FcεRI)-mediated mast cell activation is a well-studied and recognized mechanism of allergy and hypersensitivity reactions. However, non-IgE-mediated mast cell activation is less explored and is not well recognized. After decades of uncertainty, MRGPRX2 was discovered as the receptor responsible for non-IgE-mediated mast cells activation. The puzzle of non-IgE-mediated pseudo-allergic reaction is unlocked by MRGPRX2, evidenced by a plethora of reported endogenous and exogenous MRGPRX2 agonists. MRGPRX2 is exclusively expressed on mast cells and exhibits varying affinity for many molecules such as antimicrobial host defense peptides, neuropeptides, and even US Food and Drug Administration-approved drugs. The discovery of MRGPRX2 has changed our understanding of mast cell biology and filled the missing link of the underlying mechanism of drug-induced MC degranulation and pseudo-allergic reactions. These non-canonical characteristics render MRGPRX2 an intriguing player in allergic diseases. In the present article, we reviewed the emerging role of MRGPRX2 as a non-IgE-mediated mechanism of mast cell activation in pseudo-allergic reactions. We have presented an overview of mast cells, their receptors, structural insight into MRGPRX2, MRGPRX2 agonists and antagonists, the crucial role of MRGPRX2 in pseudo-allergic reactions, current challenges, and the future research direction.
Collapse
|
10
|
Yashima M, Sato Y, Kazama I. Catechin synergistically potentiates mast cell-stabilizing property of caffeine. Allergy Asthma Clin Immunol 2021; 17:1. [PMID: 33407842 PMCID: PMC7789391 DOI: 10.1186/s13223-020-00502-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/08/2020] [Indexed: 08/30/2023] Open
Abstract
Caffeine and catechin, contained in coffee and tea, are commonly consumed substances worldwide. Studies revealed their health promoting functions, such as anti-oxidant, anti-cancer and anti-bacterial properties. Additionally, studies also revealed their roles in ameliorating the symptoms of allergic disorders, indicating their anti-allergic properties. In the present study, using the differential-interference contrast (DIC) microscopy, we examined the effects of caffeine and catechin on the degranulation from rat peritoneal mast cells. Both caffeine and catechin dose-dependently decreased the numbers of degranulating mast cells. At concentrations equal to or higher than 25 mM, caffeine and catechin markedly suppressed the numbers of degranulating mast cells. In contrast, at relatively lower concentrations, both substances did not significantly affect the numbers of degranulating mast cells. However, surprisingly enough, low concentrations of catechin (1, 2.5 mM) synergistically enhanced the suppressive effect of 10 mM caffeine on mast cell degranulation. These results provided direct evidence for the first time that caffeine and catechin dose-dependently inhibited the process of exocytosis. At relatively lower concentrations, caffeine or catechin alone did not stabilize mast cells. However, low concentrations of catechin synergistically potentiated the mast cell-stabilizing property of caffeine.
Collapse
Affiliation(s)
- Misaki Yashima
- Miyagi University, School of Nursing, 1-1 Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, 981-3298, Japan
| | - Yukine Sato
- Miyagi University, School of Nursing, 1-1 Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, 981-3298, Japan
| | - Itsuro Kazama
- Miyagi University, School of Nursing, 1-1 Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, 981-3298, Japan.
| |
Collapse
|
11
|
Grattan CE, Mahler V. Immediate Contact Reactions: Pathomechanisms and Clinical Presentation. Contact Dermatitis 2021. [DOI: 10.1007/978-3-030-36335-2_60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
12
|
Wang X, Ilarraza R, Tancowny BP, Alam SB, Kulka M. Disrupted Lipid Raft Shuttling of FcεRI by n-3 Polyunsaturated Fatty Acid Is Associated With Ligation of G Protein-Coupled Receptor 120 (GPR120) in Human Mast Cell Line LAD2. Front Nutr 2020; 7:597809. [PMID: 33330598 PMCID: PMC7732685 DOI: 10.3389/fnut.2020.597809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022] Open
Abstract
n-3 polyunsaturated fatty acids (PUFA) influences a variety of disease conditions, such as hypertension, heart disease, diabetes, cancer and allergic diseases, by modulating membrane constitution, inhibiting production of proinflammatory eicosanoids and cytokines, and binding to cell surface and nuclear receptors. We have previously shown that n-3 PUFA inhibit mast cell functions by disrupting high affinity IgE receptor (FcεRI) lipid raft partitioning and subsequent suppression of FcεRI signaling in mouse bone marrow-derived mast cells. However, it is still largely unknown how n-3 PUFA modulate human mast cell function, which could be attributed to multiple mechanisms. Using a human mast cell line (LAD2), we have shown similar modulating effects of n-3 PUFA on FcεRI lipid raft shuttling, FcεRI signaling, and mediator release after cell activation through FcεRI. We have further shown that these effects are at least partially associated with ligation of G protein-coupled receptor 120 expressed on LAD2 cells. This observation has advanced our mechanistic knowledge of n-3 PUFA's effect on mast cells and demonstrated the interplay between n-3 PUFA, lipid rafts, FcεRI, and G protein-coupled receptor 120. Future research in this direction may present new targets for nutritional intervention and therapeutic agents.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Ramses Ilarraza
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Brian P Tancowny
- Department of Biochemistry, Prion Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Syed Benazir Alam
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,National Research Council Canada, Nanotechnology Research Centre, Edmonton, AB, Canada
| | - Marianna Kulka
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,National Research Council Canada, Nanotechnology Research Centre, Edmonton, AB, Canada
| |
Collapse
|
13
|
Naranjo AN, Bandara G, Bai Y, Smelkinson MG, Tobío A, Komarow HD, Boyden SE, Kastner DL, Metcalfe DD, Olivera A. Critical Signaling Events in the Mechanoactivation of Human Mast Cells through p.C492Y-ADGRE2. J Invest Dermatol 2020; 140:2210-2220.e5. [PMID: 32222457 PMCID: PMC7529699 DOI: 10.1016/j.jid.2020.03.936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022]
Abstract
A role for the adhesion G-protein coupled receptor ADGRE2 or EMR2 in mechanosensing was revealed by the finding of a missense substitution (p.C492Y) associated with familial vibratory urticaria. In these patients, friction of the skin induces mast cell hyper-degranulation through p.C492Y-ADGRE2, causing localized hives, flushing, and hypotension. We have now characterized the responses and intracellular signals elicited by mechanical activation in human mast cells expressing p.C492Y-ADGRE2 and attached to dermatan sulfate, a ligand for ADGRE2. The presence of p.C492Y-ADGRE2 reduced the threshold to activation and increased the extent of degranulation along with the percentage of mast cells responding. Vibration caused phospholipase C activation, transient increases in cytosolic calcium, and downstream activation of phosphoinositide 3-kinase and extracellular signal-regulated kinases 1 and 2 by Gβγ, Gαq/11, and Gαi/o-independent mechanisms. Degranulation induced by vibration was dependent on phospholipase C pathways, including calcium, protein kinase C, and phosphoinositide 3-kinase but not extracellular signal-regulated kinases 1/2 pathways, along with pertussis toxin-sensitive signals. In addition, mechanoactivation of mast cells stimulated the synthesis and release of prostaglandin D2, to our knowledge a previously unreported mediator in vibratory urticaria, and extracellular signal-regulated kinases 1/2 activation was required for this response together with calcium, protein kinase C, and to some extent, phosphoinositide 3-kinase. Our studies thus identified critical molecular events initiated by mechanical forces and potential therapeutic targets for patients with vibratory urticaria.
Collapse
Affiliation(s)
- Andrea N Naranjo
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Geethani Bandara
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Margery G Smelkinson
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Araceli Tobío
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hirsh D Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven E Boyden
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
14
|
Jin JR, Gogvadze E, Xavier AR, Bohnacker T, Voelzmann J, Wymann MP. PI3K γ Regulatory Protein p84 Determines Mast Cell Sensitivity to Ras Inhibition-Moving Towards Cell Specific PI3K Targeting? Front Immunol 2020; 11:585070. [PMID: 33193405 PMCID: PMC7655736 DOI: 10.3389/fimmu.2020.585070] [Citation(s) in RCA: 4] [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: 08/10/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022] Open
Abstract
Mast cells are the major effector cells in immunoglobulin E (IgE)-mediated allergy. The high affinity IgE receptor FcεRI, as well as G protein-coupled receptors (GPCRs) on the mast cell surface signals to phosphoinositide 3-kinase γ (PI3Kγ) to initiate degranulation, cytokine release, and chemotaxis. PI3Kγ is therefore considered as a target for treatment of allergic disorders. However, leukocyte PI3Kγ is key to many functions in innate and adaptive immunity, and attenuation of host defense mechanisms is an expected adverse effect that complicates treatment of chronic illnesses. PI3Kγ operates as a p110γ/p84 or p110γ/p101 complex, where p110γ/p84 requires Ras activation. Here we investigated if modulation of Ras-isoprenylation could target PI3Kγ activity to attenuate PI3Kγ-dependent mast cell responses without impairment of macrophage functions. In murine bone marrow-derived mast cells, GPCR stimulation triggers activation of N-Ras and H-Ras isoforms, which is followed by the phosphorylation of protein kinase B (PKB/Akt) relayed through PI3Kγ. Although K-Ras is normally not activated in Ras wild-type cells, it is able to compensate for genetically deleted N- and H-Ras isoforms. Inhibition of Ras isoprenylation with farnesyltransferase inhibitor FTI-277 leads to a significant reduction of mast cell degranulation, cytokine production, and migration. Complementation experiments expressing PI3Kγ adaptor proteins p84 or p101 demonstrated a differential sensitivity towards Ras-inhibition depending on PI3Kγ complex composition. Mast cell responses are exclusively p84-dependent and were effectively controlled by FTI-277. Similar results were obtained when GTP-Ras was inactivated by overexpression of the GAP-domain of Neurofibromin-1 (NF-1). Unlike mast cells, macrophages express p84 and p101 but are p101-dominated and thus remain functional under treatment with FTI-277. Our work demonstrates that p101 and p84 have distinct physiological roles, and that Ras dependence of PI3Kγ signaling differs between cell types. FTI-277 reduces GPCR-activated PI3Kγ responses in p84-expressing but not p101-containing bone marrow derived cells. However, prenylation inhibitors have pleiotropic effects beyond Ras and non-tolerable side-effects that disfavor further clinical validation. Statins are, however, clinically well-established drugs that have previously been proposed to block mast cell degranulation by interference with protein prenylation. We show here that Simvastatin inhibits mast cell degranulation, but that this does not occur via Ras-PI3Kγ pathway alterations.
Collapse
|
15
|
Molinari G, Molinari L, Nervo E. Environmental and Endogenous Acids Can Trigger Allergic-Type Airway Reactions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4688. [PMID: 32610702 PMCID: PMC7370125 DOI: 10.3390/ijerph17134688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Inflammatory allergic and nonallergic respiratory disorders are spreading worldwide and often coexist. The root cause is not clear. This review demonstrates that, from a biochemical point of view, it is ascribable to protons (H+) released into cells by exogenous and endogenous acids. The hypothesis of acids as the common cause stems from two considerations: (a) it has long been known that exogenous acids present in air pollutants can induce the irritation of epithelial surfaces, particularly the airways, inflammation, and bronchospasm; (b) according to recent articles, endogenous acids, generated in cells by phospholipases, play a key role in the biochemical mechanisms of initiation and progression of allergic-type reactions. Therefore, the intracellular acidification and consequent Ca2+ increase, induced by protons generated by either acid pollutants or endogenous phospholipases, may constitute the basic mechanism of the multimorbidity of these disorders, and environmental acidity may contribute to their spread.
Collapse
Affiliation(s)
- Giuliano Molinari
- Studio Tecnico Ing. Laura Molinari, Environmental Health and Safety Via Quarto Ponte 17, 37138 Verona, Italy;
| | - Laura Molinari
- Studio Tecnico Ing. Laura Molinari, Environmental Health and Safety Via Quarto Ponte 17, 37138 Verona, Italy;
| | - Elsa Nervo
- Elsa Nervo, Società Chimica Italiana, 00198 Rome, Italy;
| |
Collapse
|
16
|
Abe N, Toyama H, Ejima Y, Saito K, Tamada T, Yamauchi M, Kazama I. α 1-Adrenergic Receptor Blockade by Prazosin Synergistically Stabilizes Rat Peritoneal Mast Cells. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3214186. [PMID: 32461978 PMCID: PMC7243011 DOI: 10.1155/2020/3214186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/03/2020] [Accepted: 04/17/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Adrenaline quickly inhibits the release of histamine from mast cells. Besides β 2-adrenergic receptors, several in vitro studies also indicate the involvement of α-adrenergic receptors in the process of exocytosis. Since exocytosis in mast cells can be detected electrophysiologically by the changes in the membrane capacitance (Cm), its continuous monitoring in the presence of drugs would determine their mast cell-stabilizing properties. METHODS Employing the whole-cell patch-clamp technique in rat peritoneal mast cells, we examined the effects of adrenaline on the degranulation of mast cells and the increase in the Cm during exocytosis. We also examined the degranulation of mast cells in the presence or absence of α-adrenergic receptor agonists or antagonists. RESULTS Adrenaline dose-dependently suppressed the GTP-γ-S-induced increase in the Cm and inhibited the degranulation from mast cells, which was almost completely erased in the presence of butoxamine, a β 2-adrenergic receptor antagonist. Among α-adrenergic receptor agonists or antagonists, high-dose prazosin, a selective α 1-adrenergic receptor antagonist, significantly reduced the ratio of degranulating mast cells and suppressed the increase in the Cm. Additionally, prazosin augmented the inhibitory effects of adrenaline on the degranulation of mast cells. CONCLUSIONS This study provided electrophysiological evidence for the first time that adrenaline dose-dependently inhibited the process of exocytosis, confirming its usefulness as a potent mast cell stabilizer. The pharmacological blockade of α 1-adrenergic receptor by prazosin synergistically potentiated such mast cell-stabilizing property of adrenaline, which is primarily mediated by β 2-adrenergic receptors.
Collapse
Affiliation(s)
- Nozomu Abe
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Hiroaki Toyama
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Yutaka Ejima
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Kazutomo Saito
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Masanori Yamauchi
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Itsuro Kazama
- Miyagi University, School of Nursing, Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, Japan
| |
Collapse
|
17
|
Yoshida K, Ito MA, Sato N, Obayashi K, Yamamoto K, Koizumi S, Tanaka S, Furuta K, Matsuoka I. Extracellular ATP Augments Antigen-Induced Murine Mast Cell Degranulation and Allergic Responses via P2X4 Receptor Activation. THE JOURNAL OF IMMUNOLOGY 2020; 204:3077-3085. [PMID: 32358018 DOI: 10.4049/jimmunol.1900954] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/06/2020] [Indexed: 01/09/2023]
Abstract
Extracellular ATP released from stimulated and/or damaged cells modulates physiological responses via stimulation of various purinoceptors. We previously showed that ATP potentiated the Ag-induced mast cell (MC) degranulation via purinoceptors pharmacologically similar to the ionotropic P2X4 receptor. In this study, we investigated the role of P2X4 receptor in MC degranulation induced by stimulation of IgE-FcεRI complex with Ag, using bone marrow-derived MCs (BMMCs) prepared from wild type and P2X4 receptor-deficient (P2rx4-/- ) mice. ATP significantly increased Ag-induced degranulation in BMMCs prepared from wild type mice. This effect of ATP was reduced in BMMCs prepared from P2rx4-/- mice. The potentiating effect of ATP was restored by expressing P2X4 receptor in P2rx4-/- BMMCs. The P2X4 receptor-mediated effects were maintained even after differentiating into the connective tissue-type MCs. P2X4 receptor stimulation did not affect the Ag-induced Ca2+ response but enhanced Ag-induced early signals, such as tyrosine phosphorylation of Syk and phospholipase C-γ. Interestingly, these effects of ATP on Syk phosphorylation were not impaired by pretreatment with Cu2+, an inhibitor of the P2X4 receptor channel, or removal of external Ca2+, suggesting that a mechanisms other than Ca2+ influx through ion channel activity may be involved. In vivo experiments revealed that systemic and intradermal passive anaphylaxis responses were significantly alleviated in P2rx4-/- mice. Taken together, the present data suggest that the P2X4 receptor plays an essential role in ATP-induced upregulation of MC degranulation in response to Ag, and also contributes to the Ag-induced allergic response in vivo.
Collapse
Affiliation(s)
- Kazuki Yoshida
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Masa-Aki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Naoko Sato
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Kosuke Obayashi
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Kimiko Yamamoto
- Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Satoshi Tanaka
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; and
| | - Kazuyuki Furuta
- Department of Immunobiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan;
| |
Collapse
|
18
|
Grattan CE, Mahler V. Immediate Contact Reactions: Pathomechanisms and Clinical Presentation. Contact Dermatitis 2020. [DOI: 10.1007/978-3-319-72451-5_60-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
19
|
Yoshida K, Tajima M, Nagano T, Obayashi K, Ito M, Yamamoto K, Matsuoka I. Co-Stimulation of Purinergic P2X4 and Prostanoid EP3 Receptors Triggers Synergistic Degranulation in Murine Mast Cells. Int J Mol Sci 2019; 20:E5157. [PMID: 31627451 PMCID: PMC6829402 DOI: 10.3390/ijms20205157] [Citation(s) in RCA: 14] [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: 10/04/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 01/21/2023] Open
Abstract
Mast cells (MCs) recognize antigens (Ag) via IgE-bound high affinity IgE receptors (FcεRI) and trigger type I allergic reactions. FcεRI-mediated MC activation is regulated by various G protein-coupled receptor (GPCR) agonists. We recently reported that ionotropic P2X4 receptor (P2X4R) stimulation enhanced FcεRI-mediated degranulation. Since MCs are involved in Ag-independent hypersensitivity, we investigated whether co-stimulation with ATP and GPCR agonists in the absence of Ag affects MC degranulation. Prostaglandin E2 (PGE2) induced synergistic degranulation when bone marrow-derived MCs (BMMCs) were co-stimulated with ATP, while pharmacological analyses revealed that the effects of PGE2 and ATP were mediated by EP3 and P2X4R, respectively. Consistently, this response was absent in BMMCs prepared from P2X4R-deficient mice. The effects of ATP and PGE2 were reduced by PI3 kinase inhibitors but were insensitive to tyrosine kinase inhibitors which suppressed the enhanced degranulation induced by Ag and ATP. MC-dependent PGE2-triggered vascular hyperpermeability was abrogated in a P2X4R-deficient mouse ear edema model. Collectively, our results suggest that P2X4R signaling enhances EP3R-mediated MC activation via a different mechanism to that involved in enhancing Ag-induced responses. Moreover, the cooperative effects of the common inflammatory mediators ATP and PGE2 on MCs may be involved in Ag-independent hypersensitivity in vivo.
Collapse
Affiliation(s)
- Kazuki Yoshida
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Makoto Tajima
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Tomoki Nagano
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Kosuke Obayashi
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Masaaki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| | - Kimiko Yamamoto
- Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| |
Collapse
|
20
|
Activation mechanisms and multifaceted effects of mast cells in ischemia reperfusion injury. Exp Cell Res 2019; 376:227-235. [DOI: 10.1016/j.yexcr.2019.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/20/2019] [Accepted: 01/31/2019] [Indexed: 12/16/2022]
|
21
|
Halova I, Rönnberg E, Draberova L, Vliagoftis H, Nilsson GP, Draber P. Changing the threshold-Signals and mechanisms of mast cell priming. Immunol Rev 2019; 282:73-86. [PMID: 29431203 DOI: 10.1111/imr.12625] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mast cells play a key role in allergy and other inflammatory diseases involving engagement of multivalent antigen with IgE bound to high-affinity IgE receptors (FcεRIs). Aggregation of FcεRIs on mast cells initiates a cascade of signaling events that eventually lead to degranulation, secretion of leukotrienes and prostaglandins, and cytokine and chemokine production contributing to the inflammatory response. Exposure to pro-inflammatory cytokines, chemokines, bacterial and viral products, as well as some other biological products and drugs, induces mast cell transition from the basal state into a primed one, which leads to enhanced response to IgE-antigen complexes. Mast cell priming changes the threshold for antigen-mediated activation by various mechanisms, depending on the priming agent used, which alone usually do not induce mast cell degranulation. In this review, we describe the priming processes induced in mast cells by various cytokines (stem cell factor, interleukins-4, -6 and -33), chemokines, other agents acting through G protein-coupled receptors (adenosine, prostaglandin E2 , sphingosine-1-phosphate, and β-2-adrenergic receptor agonists), toll-like receptors, and various drugs affecting the cytoskeleton. We will review the current knowledge about the molecular mechanisms behind priming of mast cells leading to degranulation and cytokine production and discuss the biological effects of mast cell priming induced by several cytokines.
Collapse
Affiliation(s)
- Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Elin Rönnberg
- Immunology and Allergy Unit, Department of Medicine, Karolinska Institutet and Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Harissios Vliagoftis
- Immunology and Allergy Unit, Department of Medicine, Karolinska Institutet and Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.,Alberta Respiratory Center and Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gunnar P Nilsson
- Immunology and Allergy Unit, Department of Medicine, Karolinska Institutet and Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
22
|
Halova I, Bambouskova M, Draberova L, Bugajev V, Draber P. The transmembrane adaptor protein NTAL limits mast cell chemotaxis toward prostaglandin E2. Sci Signal 2018; 11:11/556/eaao4354. [DOI: 10.1126/scisignal.aao4354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotaxis of mast cells is one of the crucial steps in their development and function. Non–T cell activation linker (NTAL) is a transmembrane adaptor protein that inhibits the activation of mast cells and B cells in a phosphorylation-dependent manner. Here, we studied the role of NTAL in the migration of mouse mast cells stimulated by prostaglandin E2 (PGE2). Although PGE2 does not induce the tyrosine phosphorylation of NTAL, unlike IgE immune complex antigens, we found that loss of NTAL increased the chemotaxis of mast cells toward PGE2. Stimulation of mast cells that lacked NTAL with PGE2 enhanced the phosphorylation of AKT and the production of phosphatidylinositol 3,4,5-trisphosphate. In resting NTAL-deficient mast cells, phosphorylation of an inhibitory threonine in ERM family proteins accompanied increased activation of β1-containing integrins, which are features often associated with increased invasiveness in tumors. Rescue experiments indicated that only full-length, wild-type NTAL restored the chemotaxis of NTAL-deficient cells toward PGE2. Together, these data suggest that NTAL is a key inhibitor of mast cell chemotaxis toward PGE2, which may act through the RHOA/ERM/β1-integrin and PI3K/AKT axes.
Collapse
|
23
|
Arizmendi N, Kulka M. Adenosine activates Gα s proteins and inhibits C3a-induced activation of human mast cells. Biochem Pharmacol 2018; 156:157-167. [PMID: 30099007 DOI: 10.1016/j.bcp.2018.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/08/2018] [Indexed: 01/14/2023]
Abstract
Anaphylatoxin C3a and adenosine receptors (AR) are implicated in the inflammatory process associated with allergic rhinitis and asthma by modifying mast cell (MC) responses. Possible interactions between these G-protein coupled receptor (GPCR) pathways in MCs have not yet been demonstrated. LAD2 human MC were stimulated with C3a in the presence or absence of AR agonists and antagonists and their adhesion, chemotaxis and mediator release were measured. The pan-specific AR agonist, 5'-N-Ethylcarboxamidoadenosine (NECA) inhibited C3a-induced LAD2 cell migration, adhesion, degranulation, production of CCL2, and ERK1/2 phosphorylation. The selective A2A receptor agonist CGS 21680 inhibited C3a-mediated degranulation, while the A2B and A3 receptor agonists BAY 60-6583 and IB-MECA, respectively, had no effect. Moreover, an A2A receptor antagonist SCH 58261 blocked the inhibitory effect of NECA on C3a-induced degranulation, suggesting that inhibition of degranulation was mediated through the A2A receptor. NECA increased intracellular cAMP in C3a-activated mast cells, suggesting that Gαs protein signals are required for adenosine-induced inhibition of C3a-mediated human mast cell activation. The adenylyl cyclase inhibitor SQ 22536 attenuated the inhibitory effect of NECA on C3a-activated degranulation, and the A2A agonist CSG 21680 potentiated the inhibition of mast cell activation mediated by the A2A receptor. Our results suggest that adenosine inhibits C3a-mediated activation of human mast cells, possibly through a Gαs protein-dependent pathway.
Collapse
Affiliation(s)
- Narcy Arizmendi
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB, Canada
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB, Canada; Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
24
|
Hermans MAW, Schrijver B, van Holten-Neelen CCPA, Gerth van Wijk R, van Hagen PM, van Daele PLA, Dik WA. The JAK1/JAK2- inhibitor ruxolitinib inhibits mast cell degranulation and cytokine release. Clin Exp Allergy 2018; 48:1412-1420. [PMID: 29939445 DOI: 10.1111/cea.13217] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mastocytosis is characterized by the accumulation of aberrant mast cells (MC). Patients suffering from mastocytosis suffer from a wide range of symptoms due to increased levels of MC mediators. It would therefore be of great benefit to inhibit MC mediator release. However, to date there are few drugs available that are known to effectively lower MC mediator levels. The evidence for the involvement of the janus kinase 2 (JAK2)-signal transducer and activation of transcription 5 (STAT5) signalling pathway in MC activation is slowly accumulating. Interference with the JAK2-STAT5 pathway might inhibit MC mediator release. Ruxolitinib, a JAK1/JAK2 inhibitor, indeed decreases symptoms like pruritus and fatigue in patients with myeloproliferative neoplasms. Yet, detailed studies on how ruxolitinib affects human mast cell activity are lacking. OBJECTIVE To investigate the effect of JAK1/2-inhibition with ruxolitinib in the human mast cell lines LAD2 and HMC1. METHODS LAD2 and HMC1 were stimulated with substance P, codeine or the calcium ionophore A23817. The effect of ruxolitinib on mast cell degranulation (via measurement of β-hexosaminidase, histamine release and CD63 membrane expression) and IL-6, IL-13, MCP-1 and TNF-α production was investigated. The involvement of STAT5 activation was explored using the selective STAT5 inhibitor pimozide. RESULTS Ruxolitinib effectively inhibited codeine- and substance P-induced degranulation in a concentration-dependent manner. Ruxolitinib also significantly inhibited the production of IL-6, TNF-α and MCP-1 as induced by A23817 and substance P. Selective STAT5 inhibition with pimozide resulted in diminished degranulation and inhibition of cytokine production as induced by A23817 and substance P. CONCLUSIONS & CLINICAL RELEVANCE This study demonstrates that the JAK1/JAK2 inhibitor ruxolitinib can inhibit MCactivity, possibly through prevention of STAT5 activation. This renders the JAK-STAT pathway as an interesting target for therapy to release symptom burden in mastocytosis and many other MC mediator-related diseases.
Collapse
Affiliation(s)
- Maud A W Hermans
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Benjamin Schrijver
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Conny C P A van Holten-Neelen
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Roy Gerth van Wijk
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands.,Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Paul L A van Daele
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands.,Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Willem A Dik
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Abstract
Food allergies manifest in a variety of clinical conditions within the gastrointestinal tract, skin and lungs, with the most dramatic and sometimes fatal manifestation being anaphylactic shock. Major progress has been made in basic, translational and clinical research, leading to a better understanding of the underlying immunological mechanisms that lead to the breakdown of clinical and immunological tolerance against food antigens, which can result in either immunoglobulin E (IgE)-mediated reactions or non-IgE-mediated reactions. Lifestyle factors, dietary habits and maternal-neonatal interactions play a pivotal part in triggering the onset of food allergies, including qualitative and quantitative composition of the microbiota. These factors seem to have the greatest influence early in life, an observation that has led to the generation of hypotheses to explain the food allergy epidemic, including the dual-allergen exposure hypothesis. These hypotheses have fuelled research in preventive strategies that seek to establish desensitization to allergens and/or tolerance to allergens in affected individuals. Allergen-nonspecific therapeutic strategies have also been investigated in a number of clinical trials, which will eventually improve the treatment options for patients with food allergy.
Collapse
|
26
|
Wajdner HE, Farrington J, Barnard C, Peachell PT, Schnackenberg CG, Marino JP, Xu X, Affleck K, Begg M, Seward EP. Orai and TRPC channel characterization in Fc εRI-mediated calcium signaling and mediator secretion in human mast cells. Physiol Rep 2017; 5:5/5/e13166. [PMID: 28292887 PMCID: PMC5350174 DOI: 10.14814/phy2.13166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/23/2016] [Accepted: 12/28/2016] [Indexed: 12/23/2022] Open
Abstract
Inappropriate activation of mast cells via the FcεRI receptor leads to the release of inflammatory mediators and symptoms of allergic disease. Calcium influx is a critical regulator of mast cell signaling and is required for exocytosis of preformed mediators and for synthesis of eicosanoids, cytokines and chemokines. Studies in rodent and human mast cells have identified Orai calcium channels as key contributors to FcεRI-initiated mediator release. However, until now the role of TRPC calcium channels in FcεRI-mediated human mast cell signaling has not been published. Here, we show evidence for the expression of Orai 1,2, and 3 and TRPC1 and 6 in primary human lung mast cells and the LAD2 human mast cell line but, we only find evidence of functional contribution of Orai and not TRPC channels to FcεRI-mediated calcium entry. Calcium imaging experiments, utilizing an Orai selective antagonist (Synta66) showed the contribution of Orai to FcεRI-mediated signaling in human mast cells. Although, the use of a TRPC3/6 selective antagonist and agonist (GSK-3503A and GSK-2934A, respectively) did not reveal evidence for TRPC6 contribution to FcεRI-mediated calcium signaling in human mast cells. Similarly, inactivation of STIM1-regulated TRPC1 in human mast cells (as tested by transfecting cells with STIM1-KK684-685EE - TRPC1 gating mutant) failed to alter FcεRI-mediated calcium signaling in LAD2 human mast cells. Mediator release assays confirm that FcεRI-mediated calcium influx through Orai is necessary for histamine and TNFα release but is differentially involved in the generation of cytokines and eicosanoids.
Collapse
Affiliation(s)
- Hannah E Wajdner
- Department of Biomedical Science, University of Sheffield Western Bank, Sheffield, UK
| | - Jasmine Farrington
- Department of Biomedical Science, University of Sheffield Western Bank, Sheffield, UK
| | - Claire Barnard
- Department of Biomedical Science, University of Sheffield Western Bank, Sheffield, UK
| | - Peter T Peachell
- Academic Unit of Respiratory Medicine, University of Sheffield The Royal Hallamshire Hospital, Sheffield, UK
| | | | - Joseph P Marino
- Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline, King of Prussia, Pennsylvania
| | - Xiaoping Xu
- Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline, King of Prussia, Pennsylvania
| | - Karen Affleck
- Respiratory Therapy Area Unit, GlaxoSmithKline, Stevenage, UK
| | - Malcolm Begg
- Respiratory Therapy Area Unit, GlaxoSmithKline, Stevenage, UK
| | - Elizabeth P Seward
- Department of Biomedical Science, University of Sheffield Western Bank, Sheffield, UK
| |
Collapse
|
27
|
Shah SA, Page CP, Pitchford SC. Platelet-Eosinophil Interactions As a Potential Therapeutic Target in Allergic Inflammation and Asthma. Front Med (Lausanne) 2017; 4:129. [PMID: 28848732 PMCID: PMC5550710 DOI: 10.3389/fmed.2017.00129] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/24/2017] [Indexed: 01/24/2023] Open
Abstract
The importance of platelet activation during hemostasis is well understood. An understanding of these mechanisms has led to the use of several classes of anti-platelet drugs to inhibit aggregation for the prevention of thrombi during cardiovascular disease. It is now also recognized that platelets can function very differently during inflammation, as part of their role in the innate immune response against pathogens. This dichotomy in platelet function occurs through distinct physiological processes and alternative signaling pathways compared to that of hemostasis (leading to platelet aggregation) and is manifested as increased rheological interactions with leukocytes, the ability to undergo chemotaxis, communication with antigen-presenting cells, and direct anti-pathogen responses. Mounting evidence suggests platelets are also critical in the pathogenesis of allergic diseases such as asthma, where they have been associated with antigen presentation, bronchoconstriction, bronchial hyperresponsiveness, airway inflammation, and airway remodeling in both clinical and experimental studies. In particular, platelets have been reported bound to eosinophils in the blood of patients with asthma and the incidence of these events increases after both spontaneous asthma attacks in a biphasic manner, or after allergen challenge in the clinic. Platelet depletion in animal models of allergic airway inflammation causes a profound reduction in eosinophil recruitment to the lung, suggesting that the association of platelets with eosinophils is indeed an important event during eosinophil activation. Furthermore, in cases of severe asthma, and in animal models of allergic airways inflammation, platelet–eosinophil complexes move into the lung through a platelet P-selectin-mediated, eosinophil β1-integrin activation-dependent process, while platelets increase adherence of eosinophils to the vascular endothelium in vitro, demonstrating a clear interaction between these cell types in allergic inflammatory diseases. This review will explore non-thrombotic platelet activation in the context of allergy and the association of platelets with eosinophils, to reveal how these phenomena may lead to the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Sajeel A Shah
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Simon C Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Ayyadurai S, Gibson AJ, D'Costa S, Overman EL, Sommerville LJ, Poopal AC, Mackey E, Li Y, Moeser AJ. Frontline Science: Corticotropin-releasing factor receptor subtype 1 is a critical modulator of mast cell degranulation and stress-induced pathophysiology. J Leukoc Biol 2017; 102:1299-1312. [PMID: 28684600 DOI: 10.1189/jlb.2hi0317-088rr] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/28/2017] [Accepted: 06/05/2017] [Indexed: 12/23/2022] Open
Abstract
Life stress is a major risk factor in the onset and exacerbation of mast cell-associated diseases, including allergy/anaphylaxis, asthma, and irritable bowel syndrome. Although it is known that mast cells are highly activated upon stressful events, the mechanisms by which stress modulates mast cell function and disease pathophysiology remains poorly understood. Here, we investigated the role of corticotropin-releasing factor receptor subtype 1 (CRF1) in mast cell degranulation and associated disease pathophysiology. In a mast cell-dependent model of IgE-mediated passive systemic anaphylaxis (PSA), prophylactic administration of the CRF1-antagonist antalarmin attenuated mast cell degranulation and hypothermia. Mast cell-deficient KitW-sh/W-sh mice engrafted with CRF1-/- bone marrow-derived mast cells (BMMCs) exhibited attenuated PSA-induced serum histamine, hypothermia, and clinical scores compared with wild-type BMMC-engrafted KitW-sh/W-sh mice. KitW-sh/W-sh mice engrafted with CRF1-/- BMMCs also exhibited suppressed in vivo mast cell degranulation and intestinal permeability in response to acute restraint stress. Genetic and pharmacologic experiments with murine BMMCs, rat RBL-2H3, and human LAD2 mast cells demonstrated that although CRF1 activation did not directly induce MC degranulation, CRF1 signaling potentiated the degranulation responses triggered by diverse mast cell stimuli and was associated with enhanced release of Ca2+ from intracellular stores. Taken together, our results revealed a prominent role for CRF1 signaling in mast cells as a positive modulator of stimuli-induced degranulation and in vivo pathophysiologic responses to immunologic and psychologic stress.
Collapse
Affiliation(s)
- Saravanan Ayyadurai
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | | | - Susan D'Costa
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Elizabeth L Overman
- Department of Biology, Methodist University, Fayetteville, North Carolina, USA
| | - Laura J Sommerville
- Department of Pathology, Duke University, School of Medicine, Durham, North Carolina, USA
| | - Ashwini C Poopal
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Emily Mackey
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA.,Comparative Biomedical Sciences Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Yihang Li
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Adam J Moeser
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA; .,Neuroscience Program, Michigan State University, East Lansing, Michigan, USA; and.,Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
29
|
SNAP23-Dependent Surface Translocation of Leukotriene B4 (LTB4) Receptor 1 Is Essential for NOX2-Mediated Exocytotic Degranulation in Human Mast Cells Induced by Trichomonas vaginalis-Secreted LTB4. Infect Immun 2016; 85:IAI.00526-16. [PMID: 27795355 DOI: 10.1128/iai.00526-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/16/2016] [Indexed: 11/20/2022] Open
Abstract
Trichomonas vaginalis is a sexually transmitted parasite that causes vaginitis in women and itself secretes lipid mediator leukotriene B4 (LTB4). Mast cells are important effector cells of tissue inflammation during infection with parasites. Membrane-bridging SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes are critical for fusion during exocytosis. Although T. vaginalis-derived secretory products (TvSP) have been shown to induce exocytosis in mast cells, information regarding the signaling mechanisms between mast cell activation and TvSP is limited. In this study, we found that SNAP23-dependent surface trafficking of LTB4 receptor 1 (BLT1) is required for nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2)-mediated exocytotic degranulation of mast cells induced by TvSP. First, stimulation with TvSP induced exocytotic degranulation and reactive oxygen species (ROS) generation in HMC-1 cells. Next, TvSP-induced ROS generation and exocytosis were strongly inhibited by transfection of BLT1 small interfering RNA (siRNA). TvSP induced trafficking of BLT1 from the cytosol to the plasma membrane. We also found that knockdown of SNAP23 abrogated TvSP-induced ROS generation, exocytosis, and surface trafficking of BLT1 in HMC-1 cells. By coimmunoprecipitation, there was a physical interaction between BLT1 and SNAP23 in TvSP-stimulated HMC-1 cells. Taken together, our results suggest that SNAP23-dependent surface trafficking of BLT1 is essential for exocytosis in human mast cells induced by T. vaginalis-secreted LTB4 Our data collectively demonstrate a novel regulatory mechanism for SNAP23-dependent mast cell activation of T. vaginalis-secreted LTB4 involving surface trafficking of BLT1. These results can help to explain how the cross talk mechanism between parasite and host can govern deliberately tissue inflammatory responses.
Collapse
|
30
|
Singh J, Shah R, Singh D. Targeting mast cells: Uncovering prolific therapeutic role in myriad diseases. Int Immunopharmacol 2016; 40:362-384. [PMID: 27694038 DOI: 10.1016/j.intimp.2016.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023]
Abstract
The mast cells are integral part of immune system and they have pleiotropic physiological functions in our body. Any type of abnormal stimuli causes the mast cells receptors to spur the otherwise innocuous mast cells to degranulate and release inflammatory mediators like histamine, cytokines, chemokines and prostaglandins. These mediators are involved in various diseases like allergy, asthma, mastocytosis, cardiovascular disorders, etc. Herein, we describe the receptors involved in degranulation of mast cells and are broadly divided into four categories: G-protein coupled receptors, ligand gated ion channels, immunoreceptors and pattern recognition receptors. Although, activation of pattern recognition receptors do not cause mast cell degranulation, but result in cytokines production. Degranulation itself is a complex process involving cascade of events like membrane fusion events and various proteins like VAMP, Syntaxins, DOCK5, SNAP-23, MARCKS. Furthermore, we described these mast cell receptors antagonists or agonists useful in treatment of myriad diseases. Like, omalizumab anti-IgE antibody is highly effective in asthma, allergic disorders treatment and recently mechanistic insight of IgE uncovered; matrix mettaloprotease inhibitor marimistat is under phase III trial for inflammation, muscular dystrophy diseases; ZPL-389 (H4 receptor antagonist) is in Phase 2a Clinical Trial for atopic dermatitis and psoriasis; JNJ3851868 an oral H4 receptor antagonist is in phase II clinical development for asthma, rheumatoid arthritis. Therefore, research is still in inchoate stage to uncover mast cell biology, mast cell receptors, their therapeutic role in myriad diseases.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Ramanpreet Shah
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India.
| |
Collapse
|
31
|
Subramanian H, Gupta K, Ali H. Roles of Mas-related G protein-coupled receptor X2 on mast cell-mediated host defense, pseudoallergic drug reactions, and chronic inflammatory diseases. J Allergy Clin Immunol 2016; 138:700-710. [PMID: 27448446 DOI: 10.1016/j.jaci.2016.04.051] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/21/2016] [Accepted: 04/06/2016] [Indexed: 12/11/2022]
Abstract
Mast cells (MCs), which are granulated tissue-resident cells of hematopoietic lineage, contribute to vascular homeostasis, innate/adaptive immunity, and wound healing. However, MCs are best known for their roles in allergic and inflammatory diseases, such as anaphylaxis, food allergy, rhinitis, itch, urticaria, atopic dermatitis, and asthma. In addition to the high-affinity IgE receptor (FcεRI), MCs express numerous G protein-coupled receptors (GPCRs), which are the largest group of membrane receptor proteins and the most common targets of drug therapy. Antimicrobial host defense peptides, neuropeptides, major basic protein, eosinophil peroxidase, and many US Food and Drug Administration-approved peptidergic drugs activate human MCs through a novel GPCR known as Mas-related G protein-coupled receptor X2 (MRGPRX2; formerly known as MrgX2). Unique features of MRGPRX2 that distinguish it from other GPCRs include their presence both on the plasma membrane and intracellular sites and their selective expression in MCs. In this article we review the possible roles of MRGPRX2 on host defense, drug-induced anaphylactoid reactions, neurogenic inflammation, pain, itch, and chronic inflammatory diseases, such as urticaria and asthma. We propose that host defense peptides that kill microbes directly and activate MCs through MRGPRX2 could serve as novel GPCR targets to modulate host defense against microbial infection. Furthermore, mAbs or small-molecule inhibitors of MRGPRX2 could be developed for the treatment of MC-dependent allergic and inflammatory disorders.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa
| | - Kshitij Gupta
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa
| | - Hydar Ali
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa.
| |
Collapse
|
32
|
Yu Y, Blokhuis BR, Garssen J, Redegeld FA. Non-IgE mediated mast cell activation. Eur J Pharmacol 2016; 778:33-43. [DOI: 10.1016/j.ejphar.2015.07.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/15/2015] [Accepted: 07/07/2015] [Indexed: 12/28/2022]
|
33
|
Yu Y, Huang Z, Mao Z, Zhang Y, Jin M, Chen W, Zhang W, Yu B, Zhang W, Alaster Lau HY. Go is required for the release of IL-8 and TNF-α, but not degranulation in human mast cells. Eur J Pharmacol 2016; 780:115-21. [PMID: 27025291 DOI: 10.1016/j.ejphar.2016.03.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/13/2016] [Accepted: 03/23/2016] [Indexed: 01/16/2023]
Abstract
Mast cells activated by IgE-dependent and -independent mechanisms play important roles in innate and acquired immune responses. Activation of pertussis toxin (PTX)-sensitive Gi/o proteins is the key step in mast cell degranulation and release of de novo synthesized inflammatory mediators through IgE-independent mechanism. However, the roles of Gi and Go proteins in mast cells activation have not yet been differentiated. In the current study, the functional roles of Go proteins in the activities of LAD2 cells, a human mast cell line, are identified. Knockdown of Gαo expression significantly inhibited the synthesis of IL-8 and TNF-α from substance P activated LAD2 cells but demonstrated no effect on degranulation. This effect was associated with the activation of Erk and JNK/MAPKs signaling, whereas PI3K-Akt, calcium mobilization and NFAT translocation remained unchanged. These results suggest that Gi and Go proteins differentially regulate human mast cells activities through activating distinct signaling cascades.
Collapse
Affiliation(s)
- Yangyang Yu
- School of medicine, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Zhenhe Huang
- Department of Geriatrics, The Sixth People's Hospital of Shenzhen, Guangdong Province, China
| | - Zhuo Mao
- School of medicine, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Yarui Zhang
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| | - Meiling Jin
- School of medicine, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Wenwen Chen
- School of medicine, Shenzhen University, Shenzhen, Guangdong Province 518060, China
| | - Wei Zhang
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| | - Bo Yu
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Weizhen Zhang
- School of medicine, Shenzhen University, Shenzhen, Guangdong Province 518060, China.
| | - Hang Yung Alaster Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
34
|
Zierler S, Sumoza-Toledo A, Suzuki S, Dúill FÓ, Ryazanova LV, Penner R, Ryazanov AG, Fleig A. TRPM7 kinase activity regulates murine mast cell degranulation. J Physiol 2016; 594:2957-70. [PMID: 26660477 DOI: 10.1113/jp271564] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/10/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The Mg(2+) and Ca(2+) conducting transient receptor potential melastatin 7 (TRPM7) channel-enzyme (chanzyme) has been implicated in immune cell function. Mice heterozygous for a TRPM7 kinase deletion are hyperallergic, while mice with a single point mutation at amino acid 1648, silencing kinase activity, are not. As mast cell mediators trigger allergic reactions, we here determine the function of TRPM7 in mast cell degranulation and histamine release. Our data establish that TRPM7 kinase activity regulates mast cell degranulation and release of histamine independently of TRPM7 channel function. Our findings suggest a regulatory role of TRPM7 kinase activity on intracellular Ca(2+) and extracellular Mg(2+) sensitivity of mast cell degranulation. ABSTRACT Transient receptor potential melastatin 7 (TRPM7) is a divalent ion channel with a C-terminally located α-kinase. Mice heterozygous for a TRPM7 kinase deletion (TRPM7(+/∆K) ) are hypomagnesaemic and hyperallergic. In contrast, mice carrying a single point mutation at amino acid 1648, which silences TRPM7 kinase activity (TRPM7(KR) ), are not hyperallergic and are resistant to systemic magnesium (Mg(2+) ) deprivation. Since allergic reactions are triggered by mast cell-mediated histamine release, we investigated the function of TRPM7 on mast cell degranulation and histamine release using wild-type (TRPM7(+/+) ), TRPM7(+/∆K) and TRPM7(KR) mice. We found that degranulation and histamine release proceeded independently of TRPM7 channel function. Furthermore, extracellular Mg(2+) assured unperturbed IgE-DNP-dependent exocytosis, independently of TRPM7. However, impairment of TRPM7 kinase function suppressed IgE-DNP-dependent exocytosis, slowed the cellular degranulation rate, and diminished the sensitivity to intracellular calcium (Ca(2+) ) in G protein-induced exocytosis. In addition, G protein-coupled receptor (GPCR) stimulation revealed strong suppression of histamine release, whereas removal of extracellular Mg(2+) caused the phenotype to revert. We conclude that the TRPM7 kinase activity regulates murine mast cell degranulation by changing its sensitivity to intracellular Ca(2+) and affecting granular mobility and/or histamine contents.
Collapse
Affiliation(s)
- Susanna Zierler
- Center for Biomedical Research, The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, HI, 96813, USA.,Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, 80336, Munich, Germany
| | - Adriana Sumoza-Toledo
- Center for Biomedical Research, The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, HI, 96813, USA.,Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Colonia Centro CP 91700, Veracruz, Mexico
| | - Sayuri Suzuki
- Center for Biomedical Research, The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, HI, 96813, USA
| | - Fionán Ó Dúill
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, 80336, Munich, Germany
| | - Lillia V Ryazanova
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Reinhold Penner
- Center for Biomedical Research, The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, HI, 96813, USA
| | - Alexey G Ryazanov
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Andrea Fleig
- Center for Biomedical Research, The Queen's Medical Center and University of Hawaii John A. Burns School of Medicine and Cancer Center, Honolulu, HI, 96813, USA
| |
Collapse
|
35
|
Krystel-Whittemore M, Dileepan KN, Wood JG. Mast Cell: A Multi-Functional Master Cell. Front Immunol 2016; 6:620. [PMID: 26779180 PMCID: PMC4701915 DOI: 10.3389/fimmu.2015.00620] [Citation(s) in RCA: 445] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/27/2015] [Indexed: 12/24/2022] Open
Abstract
Mast cells are immune cells of the myeloid lineage and are present in connective tissues throughout the body. The activation and degranulation of mast cells significantly modulates many aspects of physiological and pathological conditions in various settings. With respect to normal physiological functions, mast cells are known to regulate vasodilation, vascular homeostasis, innate and adaptive immune responses, angiogenesis, and venom detoxification. On the other hand, mast cells have also been implicated in the pathophysiology of many diseases, including allergy, asthma, anaphylaxis, gastrointestinal disorders, many types of malignancies, and cardiovascular diseases. This review summarizes the current understanding of the role of mast cells in many pathophysiological conditions.
Collapse
Affiliation(s)
- Melissa Krystel-Whittemore
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center , Kansas City, KS , USA
| | - Kottarappat N Dileepan
- Department of Medicine, Division of Allergy, Clinical Immunology and Rheumatology, University of Kansas Medical Center , Kansas City, KS , USA
| | - John G Wood
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
36
|
Efergan A, Azouz NP, Klein O, Noguchi K, Rothenberg ME, Fukuda M, Sagi-Eisenberg R. Rab12 Regulates Retrograde Transport of Mast Cell Secretory Granules by Interacting with the RILP-Dynein Complex. THE JOURNAL OF IMMUNOLOGY 2016; 196:1091-101. [PMID: 26740112 DOI: 10.4049/jimmunol.1500731] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 11/20/2015] [Indexed: 12/19/2022]
Abstract
Secretory granule (SG) transport is a critical step in regulated exocytosis including degranulation of activated mast cells. The latter process results in the release of multiple inflammatory mediators that play key roles in innate immunity, as well as in allergic responses. In this study, we identified the small GTPase Rab12 as a novel regulator of mast cell SG transport, and we provide mechanistic insights into its mode of action. We show that Rab12 is activated in a stimulus-dependent fashion and promotes microtubule-dependent retrograde transport of the SGs in the activated cells. We also show that this minus end transport of the SGs is mediated by the RILP-dynein complex and identify RILP as a novel effector of Rab12. Finally, we show that Rab12 negatively regulates mast cell degranulation. Taken together, our results identify Rab12 as a novel regulator of mast cell responses and disclose for the first time, to our knowledge, the mechanism of retrograde transport of the mast cell SGs.
Collapse
Affiliation(s)
- Adi Efergan
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nurit P Azouz
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ofir Klein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Kenta Noguchi
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan; and
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan; and
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel;
| |
Collapse
|
37
|
Pundir P, MacDonald CA, Kulka M. The Novel Receptor C5aR2 Is Required for C5a-Mediated Human Mast Cell Adhesion, Migration, and Proinflammatory Mediator Production. THE JOURNAL OF IMMUNOLOGY 2015; 195:2774-87. [PMID: 26283482 DOI: 10.4049/jimmunol.1401348] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/08/2015] [Indexed: 12/31/2022]
Abstract
C5a generated during complement activation possesses proinflammatory and immunoregulatory properties critical for the development and modulation of allergic immune responses. In immune cells, C5a mediates its effects through binding to two G protein-coupled receptors, C5aR1 and C5aR2. Mast cells are key effectors in allergic reactions, and decades of research have suggested that the majority of C5a effects on mast cells are mediated through C5aR1, whereas the expression and function of C5aR2 have not been explored. We demonstrated that the human mast cell line Laboratory of Allergic Diseases 2 (LAD2) expresses surface C5aR2 but not C5aR1, whereas CD34(+) cell-derived primary mast cells do not express surface C5aR1 or C5aR2. Stem cell factor and IL-4 upregulated C5aR2 expression on LAD2 cells. Furthermore, C5a caused internalization of LAD2 cell-surface C5aR2. We therefore used LAD2 cells as a model to study C5a/C5aR2-induced biological responses and signaling in human mast cells. We found that whereas C5a was unable to induce degranulation, it stimulated GM-CSF, TNF, CXCL10, and CCL2 production. C5a caused ERK phosphorylation, a signaling molecule important in cytokine and chemokine generation. In addition, C5a stimulated adhesion and chemotaxis of mast cells. Wortmannin, an inhibitor of PI3K, and small interfering RNA against β-arrestin-2 blocked C5a-induced adhesion. Silencing of C5aR2 using lentiviral short hairpin RNA rendered the cells unresponsive to C5a-induced adhesion, chemotaxis, and mediator release, as well as ERK phosphorylation. Overall, this study reveals a novel role for C5aR2 in C5a-mediated activation of mast cells and demonstrates that C5aR2 ligation initiates a β-arrestin-2-, PI3K-, and ERK-dependent signaling pathway in these cells.
Collapse
Affiliation(s)
- Priyanka Pundir
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island C1A 4P3, Canada
| | - Clayton A MacDonald
- National Institute for Nanotechnology, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada; and
| | - Marianna Kulka
- National Institute for Nanotechnology, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada; and Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
38
|
Draber P, Halova I, Polakovicova I, Kawakami T. Signal transduction and chemotaxis in mast cells. Eur J Pharmacol 2015; 778:11-23. [PMID: 25941081 DOI: 10.1016/j.ejphar.2015.02.057] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 01/08/2023]
Abstract
Mast cells play crucial roles in both innate and adaptive arms of the immune system. Along with basophils, mast cells are essential effector cells for allergic inflammation that causes asthma, allergic rhinitis, food allergy and atopic dermatitis. Mast cells are usually increased in inflammatory sites of allergy and, upon activation, release various chemical, lipid, peptide and protein mediators of allergic reactions. Since antigen/immunoglobulin E (IgE)-mediated activation of these cells is a central event to trigger allergic reactions, innumerable studies have been conducted on how these cells are activated through cross-linking of the high-affinity IgE receptor (FcεRI). Development of mature mast cells from their progenitor cells is under the influence of several growth factors, of which the stem cell factor (SCF) seems to be the most important. Therefore, how SCF induces mast cell development and activation via its receptor, KIT, has been studied extensively, including a cross-talk between KIT and FcεRI signaling pathways. Although our understanding of the signaling mechanisms of the FcεRI and KIT pathways is far from complete, pharmaceutical applications of the knowledge about these pathways are underway. This review will focus on recent progresses in FcεRI and KIT signaling and chemotaxis.
Collapse
Affiliation(s)
- Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic.
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic
| | - Iva Polakovicova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, CZ 14220 Prague, Czech Republic
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle La Jolla, CA 92037, USA; Laboratory for Allergic Disease, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan
| |
Collapse
|
39
|
Wang X, Kulka M. n-3 Polyunsaturated fatty acids and mast cell activation. J Leukoc Biol 2015; 97:859-871. [DOI: 10.1189/jlb.2ru0814-388r] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 02/04/2015] [Accepted: 02/19/2015] [Indexed: 01/18/2023] Open
|
40
|
Smrz D, Cruse G, Beaven MA, Kirshenbaum A, Metcalfe DD, Gilfillan AM. Rictor negatively regulates high-affinity receptors for IgE-induced mast cell degranulation. THE JOURNAL OF IMMUNOLOGY 2014; 193:5924-32. [PMID: 25378594 DOI: 10.4049/jimmunol.1303495] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rictor is a regulatory component of the mammalian target of rapamycin (mTOR) complex 2 (mTORC2). We have previously demonstrated that rictor expression is substantially downregulated in terminally differentiated mast cells as compared with their immature or transformed counterparts. However, it is not known whether rictor and mTORC2 regulate mast cell activation. In this article, we show that mast cell degranulation induced by aggregation of high-affinity receptors for IgE (FcεRI) is negatively regulated by rictor independently of mTOR. We found that inhibition of mTORC2 by the dual mTORC1/mTORC2 inhibitor Torin1 or by downregulation of mTOR by short hairpin RNA had no impact on FcεRI-induced degranulation, whereas downregulation of rictor itself resulted in an increased sensitivity (∼50-fold) of cells to FcεRI aggregation with enhancement of degranulation. This was linked to a similar enhancement in calcium mobilization and cytoskeletal rearrangement attributable to increased phosphorylation of LAT and PLCγ1. In contrast, degranulation and calcium responses elicited by the G protein-coupled receptor ligand, C3a, or by thapsigargin, which induces a receptor-independent calcium signal, was unaffected by rictor knockdown. Overexpression of rictor, in contrast with knockdown, suppressed FcεRI-mediated degranulation. Taken together, these data provide evidence that rictor is a multifunctional signaling regulator that can regulate FcεRI-mediated degranulation independently of mTORC2.
Collapse
Affiliation(s)
- Daniel Smrz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Glenn Cruse
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Michael A Beaven
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Arnold Kirshenbaum
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Dean D Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Alasdair M Gilfillan
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
41
|
Amison RT, Momi S, Morris A, Manni G, Keir S, Gresele P, Page CP, Pitchford SC. RhoA signaling through platelet P2Y₁ receptor controls leukocyte recruitment in allergic mice. J Allergy Clin Immunol 2014; 135:528-38. [PMID: 25445826 DOI: 10.1016/j.jaci.2014.09.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 07/21/2014] [Accepted: 09/19/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Clinical studies reveal platelet activation in patients with asthma, allergic rhinitis, and eczema. This is distinct from platelet aggregation, which is critical for the maintenance of hemostasis and in which a role for platelet purinergic receptors is well documented. However, purines are also essential for inflammatory cell trafficking in animal models of allergic lung inflammation, which are known to be platelet dependent, yet the role of purines in the platelet activation accompanying inflammation is unknown. OBJECTIVES We investigated whether the involvement of purine activation of platelets during allergic inflammation is distinct from purine involvement in platelet aggregation. METHODS BALB/c mice were sensitized to ovalbumin and subsequent airway ovalbumin challenge. Bronchoalveolar lavage fluid was analyzed for inflammatory cells, and blood samples were assessed for platelet activation. The role of platelet purinergic receptors and associated signaling mechanisms (RhoA) were assessed. RESULTS P2Y₁, but not P2Y₁₂ or P2X₁, antagonism inhibited pulmonary leukocyte recruitment. The formation of platelet-leukocyte complexes in vivo and platelet/P-selectin-dependent polymorphonuclear cell migration in vitro were exclusively platelet P2Y₁ receptor dependent. Furthermore, platelet P2Y₁ activation resulted in RhoA activity in vivo after allergen challenge, and RhoA signaling in platelets through P2Y₁ stimulation was required for platelet-dependent leukocyte chemotaxis in vitro. Leukocyte recruitment in thrombocytopenic mice remained suppressed after reinfusion of platelets pretreated with a P2Y₁ antagonist or a Rho-associated kinase 1 inhibitor, confirming the crucial role of platelet P2Y₁ receptor and subsequent activation of RhoA. CONCLUSION RhoA signaling downstream of platelet P2Y₁, but not P2Y₁₂, represents a clear dichotomy in platelet activation during allergic inflammation versus hemostasis.
Collapse
Affiliation(s)
- Richard T Amison
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Stefania Momi
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Abigail Morris
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Giorgia Manni
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Sandra Keir
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Simon C Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom.
| |
Collapse
|
42
|
Torres-Atencio I, Ainsua-Enrich E, de Mora F, Picado C, Martín M. Prostaglandin E2 prevents hyperosmolar-induced human mast cell activation through prostanoid receptors EP2 and EP4. PLoS One 2014; 9:e110870. [PMID: 25329458 PMCID: PMC4203853 DOI: 10.1371/journal.pone.0110870] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/24/2014] [Indexed: 11/26/2022] Open
Abstract
Background Mast cells play a critical role in allergic and inflammatory diseases, including exercise-induced bronchoconstriction (EIB) in asthma. The mechanism underlying EIB is probably related to increased airway fluid osmolarity that activates mast cells to the release inflammatory mediators. These mediators then act on bronchial smooth muscle to cause bronchoconstriction. In parallel, protective substances such as prostaglandin E2 (PGE2) are probably also released and could explain the refractory period observed in patients with EIB. Objective This study aimed to evaluate the protective effect of PGE2 on osmotically activated mast cells, as a model of exercise-induced bronchoconstriction. Methods We used LAD2, HMC-1, CD34-positive, and human lung mast cell lines. Cells underwent a mannitol challenge, and the effects of PGE2 and prostanoid receptor (EP) antagonists for EP1–4 were assayed on the activated mast cells. Beta-hexosaminidase release, protein phosphorylation, and calcium mobilization were assessed. Results Mannitol both induced mast cell degranulation and activated phosphatidyl inositide 3-kinase and mitogen-activated protein kinase (MAPK) pathways, thereby causing de novo eicosanoid and cytokine synthesis. The addition of PGE2 significantly reduced mannitol-induced degranulation through EP2 and EP4 receptors, as measured by beta-hexosaminidase release, and consequently calcium influx. Extracellular-signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 phosphorylation were diminished when compared with mannitol activation alone. Conclusions Our data show a protective role for the PGE2 receptors EP2 and EP4 following osmotic changes, through the reduction of human mast cell activity caused by calcium influx impairment and MAP kinase inhibition.
Collapse
MESH Headings
- Asthma, Exercise-Induced/genetics
- Asthma, Exercise-Induced/metabolism
- Asthma, Exercise-Induced/pathology
- Cell Degranulation
- Cell Line
- Dinoprostone/genetics
- Dinoprostone/metabolism
- Diuretics, Osmotic/pharmacology
- Extracellular Signal-Regulated MAP Kinases/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Humans
- Lung/metabolism
- Lung/pathology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- Mannitol/pharmacology
- Mast Cells/metabolism
- Mast Cells/pathology
- Osmotic Pressure
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
Collapse
Affiliation(s)
- Ivonne Torres-Atencio
- Unidad de Farmacología, Facultad de Medicina, Universidad de Panamá, Panama, Panama Republic
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erola Ainsua-Enrich
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Unitat de Bioquímica i Biologia Molecular, Department de Ciències Fisològiques I, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Fernando de Mora
- Department de Farmacologia, Terapéutica i Toxicologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - César Picado
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Margarita Martín
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Unitat de Bioquímica i Biologia Molecular, Department de Ciències Fisològiques I, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
43
|
da Silva EZM, Jamur MC, Oliver C. Mast cell function: a new vision of an old cell. J Histochem Cytochem 2014; 62:698-738. [PMID: 25062998 PMCID: PMC4230976 DOI: 10.1369/0022155414545334] [Citation(s) in RCA: 405] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
Since first described by Paul Ehrlich in 1878, mast cells have been mostly viewed as effectors of allergy. It has been only in the past two decades that mast cells have gained recognition for their involvement in other physiological and pathological processes. Mast cells have a widespread distribution and are found predominantly at the interface between the host and the external environment. Mast cell maturation, phenotype and function are a direct consequence of the local microenvironment and have a marked influence on their ability to specifically recognize and respond to various stimuli through the release of an array of biologically active mediators. These features enable mast cells to act as both first responders in harmful situations as well as to respond to changes in their environment by communicating with a variety of other cells implicated in physiological and immunological responses. Therefore, the critical role of mast cells in both innate and adaptive immunity, including immune tolerance, has gained increased prominence. Conversely, mast cell dysfunction has pointed to these cells as the main offenders in several chronic allergic/inflammatory disorders, cancer and autoimmune diseases. This review summarizes the current knowledge of mast cell function in both normal and pathological conditions with regards to their regulation, phenotype and role.
Collapse
Affiliation(s)
- Elaine Zayas Marcelino da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| |
Collapse
|
44
|
Autocrine hemokinin-1 functions as an endogenous adjuvant for IgE-mediated mast cell inflammatory responses. J Allergy Clin Immunol 2014; 135:1019-1030.e8. [PMID: 25201259 DOI: 10.1016/j.jaci.2014.07.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 01/24/2023]
Abstract
BACKGROUND Efficient development of atopic diseases requires interactions between allergen and adjuvant to initiate and amplify the underlying inflammatory responses. Substance P (SP) and hemokinin-1 (HK-1) are neuropeptides that signal through the neurokinin-1 receptor (NK1R) to promote inflammation. Mast cells initiate the symptoms and tissue effects of atopic disorders, secreting TNF and IL-6 after FcεRI cross-linking by antigen-IgE complexes (FcεRI-activated mast cells [FcεRI-MCs]). Additionally, MCs express the NK1R, suggesting an adjuvant role for NK1R agonists in FcεRI-MC-mediated pathologies; however, in-depth research addressing this relevant aspect of MC biology is lacking. OBJECTIVE We sought to investigate the effect of NK1R signaling and the individual roles of SP and HK-1 as potential adjuvants for FcεRI-MC-mediated allergic disorders. METHODS Bone marrow-derived mast cells (BMMCs) from C57BL/6 wild-type (WT) or NK1R(-/-) mice were used to investigate the effects of NK1R signaling on FcεRI-MCs. BMMCs generated from Tac1(-/-) mice or after culture with Tac4 small interfering RNA were used to address the adjuvancy of SP and HK-1. WT, NK1R(-/-), and c-Kit(W-sh/W-sh) mice reconstituted with WT or NK1R(-/-) BMMCs were used to evaluate NK1R signaling on FcεRI-MC-mediated passive local and systemic anaphylaxis and on airway inflammation. RESULTS FcεRI-activated MCs upregulated NK1R and HK-1 transcripts and protein synthesis, without modifying SP expression. In a positive signaling loop HK-1 promoted TNF and IL-6 secretion by MC degranulation and protein synthesis, the latter through the phosphoinositide 3-kinase/Akt/nuclear factor κB pathways. In vivo NK1R signaling was necessary for the development of passive local and systemic anaphylaxis and airway inflammation. CONCLUSIONS FcεRI stimulation of MCs promotes autocrine secretion of HK-1, which signals through NK1R to provide adjuvancy for efficient development of FcεRI-MC-mediated disorders.
Collapse
|
45
|
The role of mast cells in ischemia and reperfusion injury. Inflamm Res 2014; 63:899-905. [PMID: 25108401 DOI: 10.1007/s00011-014-0763-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/04/2014] [Accepted: 07/24/2014] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Ischemia and reperfusion (IR) injury is a challenging clinical problem that is triggered by ischemia in an organ followed by subsequent restoration of the blood supply. The effects of mast cell (MC) in IR injury are not totally clear. MATERIALS AND METHODS We review the body of literature on the role of MCs in IR injury based on an unrestricted Pubmed search for the descriptors "mast cell", "ischemia" and "reperfusion injury", as well as discuss implications for treatment and future directions. RESULTS Shortly after IR, chemicals released by MC can trigger vasoactive substance formation, tissue leakage, upregulation of adhesive molecules followed by leukocyte recruitment and infiltration, and pronecrotic pathway activation, among other physiologic changes. In the long term, MCs may influence tissue remodeling and repair as well as blood restoration after IR. Consistent with these findings, methods and drugs that target MCs have been shown to attenuate IR injury. CONCLUSION It has been demonstrated that MCs play a role in IR injury, but the mechanisms are complex and need to be further studied.
Collapse
|
46
|
Acuña-Castroviejo D, Escames G, Venegas C, Díaz-Casado ME, Lima-Cabello E, López LC, Rosales-Corral S, Tan DX, Reiter RJ. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci 2014; 71:2997-3025. [PMID: 24554058 PMCID: PMC11113552 DOI: 10.1007/s00018-014-1579-2] [Citation(s) in RCA: 728] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/26/2014] [Accepted: 01/27/2014] [Indexed: 12/15/2022]
Abstract
Endogenous melatonin is synthesized from tryptophan via 5-hydroxytryptamine. It is considered an indoleamine from a biochemical point of view because the melatonin molecule contains a substituted indolic ring with an amino group. The circadian production of melatonin by the pineal gland explains its chronobiotic influence on organismal activity, including the endocrine and non-endocrine rhythms. Other functions of melatonin, including its antioxidant and anti-inflammatory properties, its genomic effects, and its capacity to modulate mitochondrial homeostasis, are linked to the redox status of cells and tissues. With the aid of specific melatonin antibodies, the presence of melatonin has been detected in multiple extrapineal tissues including the brain, retina, lens, cochlea, Harderian gland, airway epithelium, skin, gastrointestinal tract, liver, kidney, thyroid, pancreas, thymus, spleen, immune system cells, carotid body, reproductive tract, and endothelial cells. In most of these tissues, the melatonin-synthesizing enzymes have been identified. Melatonin is present in essentially all biological fluids including cerebrospinal fluid, saliva, bile, synovial fluid, amniotic fluid, and breast milk. In several of these fluids, melatonin concentrations exceed those in the blood. The importance of the continual availability of melatonin at the cellular level is important for its physiological regulation of cell homeostasis, and may be relevant to its therapeutic applications. Because of this, it is essential to compile information related to its peripheral production and regulation of this ubiquitously acting indoleamine. Thus, this review emphasizes the presence of melatonin in extrapineal organs, tissues, and fluids of mammals including humans.
Collapse
Affiliation(s)
- Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avda. del Conocimiento s/n, Armilla, 18100, Granada, Spain,
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kuhny M, Hochdörfer T, Ayata CK, Idzko M, Huber M. CD39 is a negative regulator of P2X7-mediated inflammatory cell death in mast cells. Cell Commun Signal 2014; 12:40. [PMID: 25184735 PMCID: PMC4110707 DOI: 10.1186/s12964-014-0040-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/13/2014] [Indexed: 02/08/2023] Open
Abstract
Background Mast cells (MCs) are major contributors to an inflammatory milieu. One of the most potent drivers of inflammation is the cytokine IL-1β, which is produced in the cytoplasm in response to danger signals like LPS. Several controlling mechanisms have been reported which limit the release of IL-1β. Central to this regulation is the NLRP3 inflammasome, activation of which requires a second danger signal with the capacity to subvert the homeostasis of lysosomes and mitochondria. High concentrations of extracellular ATP have the capability to perturb the plasma membrane by activation of P2X7 channels and serve as such a danger signal. In this study we investigate the role of P2X7 channels and the ecto-5´-nucleotidase CD39 in ATP-triggered release of IL-1β from LPS-treated mast cells. Results We report that in MCs CD39 sets an activation threshold for the P2X7-dependent inflammatory cell death and concomitant IL-1β release. Knock-out of CD39 or stimulation with non-hydrolysable ATP led to a lower activation threshold for P2X7-dependent responses. We found that stimulation of LPS-primed MCs with high doses of ATP readily induced inflammatory cell death. Yet, cell death-dependent release of IL-1β yielded only minute amounts of IL-1β. Intriguingly, stimulation with low ATP concentrations augmented the production of IL-1β in LPS-primed MCs in a P2X7-independent but caspase-1-dependent manner. Conclusion Our study demonstrates that the fine-tuned interplay between ATP and different surface molecules recognizing or modifying ATP can control inflammatory and cell death decisions.
Collapse
|
48
|
Haddock BJ, Zhu Y, Doyle SP, Abdullah LH, Davis CW. Role of MARCKS in regulated secretion from mast cells and airway goblet cells. Am J Physiol Lung Cell Mol Physiol 2014; 306:L925-36. [PMID: 24705720 DOI: 10.1152/ajplung.00213.2013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
MARCKS (myristoylated alanine-rich C kinase substrate) is postulated to regulate the passage of secretory granules through cortical actin in the early phase of exocytosis. There are, however, three proposed mechanisms of action, all of which were derived from studies using synthetic peptides representing either the central phosphorylation site domain or the upstream, NH2-terminal domain: it tethers actin to the plasma membrane and/or to secretory granules, and/or it sequesters PIP2. Using MARCKS-null mice, we probed for a loss of function secretory phenotype in mast cells harvested from embryonic livers and maturated in vivo [embryonic hepatic-derived mast cells (eHMCs)]. Both wild-type (WT) and MARCKS-null eHMCs exhibited full exocytic responses upon FcϵRI receptor activation with DNP-BSA (2,4-dinitrophenyl-BSA), whether they were in suspension or adherent. The secretory responses of MARCKS-null eHMCs were consistently higher than those of WT cells, but the differences had sporadic statistical significance. The MARCKS-null cells exhibited faster secretory kinetics, however, achieving the plateau phase of the response with a t½ ∼2.5-fold faster. Hence, MARCKS appears to be a nonessential regulatory protein in mast cell exocytosis but exerts a negative modulation. Surprisingly, the MARCKS NH2-terminal peptide, MANS, which has been reported to inhibit mucin secretion from airway goblet cells (Li Y, Martin LD, Spizz G, Adler KB. J Biol Chem 276: 40982-40990, 2001), inhibited hexosaminidase secretion from WT and MARCKS-null eHMCs, leading us to reexamine its effects on mucin secretion. Results from studies using peptide inhibitors with human bronchial epithelial cells and with binding assays using purified mucins suggested that MANS inhibited the mucin binding assay, rather than the secretory response.
Collapse
Affiliation(s)
- Brookelyn J Haddock
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Yunxiang Zhu
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina and
| | - Sean P Doyle
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina and
| | - Lubna H Abdullah
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina and
| | - C William Davis
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina and Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
49
|
Kan L, Mutso AA, McGuire TL, Apkarian AV, Kessler JA. Opioid signaling in mast cells regulates injury responses associated with heterotopic ossification. Inflamm Res 2013; 63:207-15. [PMID: 24327087 DOI: 10.1007/s00011-013-0690-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/18/2013] [Accepted: 11/20/2013] [Indexed: 10/25/2022] Open
Abstract
INTRODUCTION Previous studies found that neuron specific enolase promoter (Nse-BMP4) transgenic mice have increased expression of the nociceptive mediator, substance P and exaggerated local injury responses associated with heterotopic ossification (HO). It is of interest great to know the pain responses in these mice and how the opioid signaling is involved in the downstream events such as mast cell (MC) activation. MATERIALS AND METHODS This study utilized a transgenic mouse model of HO in which BMP4 is expressed under the control of the Nse-BMP4. The tactile sensitivity and the cold sensitivity of the mice were measured in a classic inflammatory pain model (carrageenan solution injected into the plantar surface of the left hind paw). The MC activation and the expression profiles of different components in the opioid signaling were demonstrated through routine histology and immunohistochemistry and Western blotting, in the superficial and deep muscle injury models. RESULTS We found that the pain responses in these mice were paradoxically attenuated or unchanged, and we also found increased expression of both Methionine Enkephalin (Met-Enk), and the μ-opioid receptor (MOR). Met-Enk and MOR both co-localized within activated MCs in limb tissues. Further, Nse-BMP4;MOR(-/-) double mutant mice showed attenuated MC activation and had a significant reduction in HO formation in response to injuries. CONCLUSIONS These observations suggest that opioid signaling may play a key role in MC activation and the downstream inflammatory responses associated with HO. In addition to providing insight into the role of MC activation and associated injury responses in HO, these findings suggest opioid signaling as a potential therapeutic target in HO and possibly others disorders involving MC activation.
Collapse
Affiliation(s)
- Lixin Kan
- Department of Neurology, Northwestern University, Ward Building 10-233, 303 East Chicago Avenue, Chicago, IL, 60611-3008, USA,
| | | | | | | | | |
Collapse
|
50
|
Abstract
Rapid progress has recently been made regarding how phospholipase C (PLC)-β functions downstream of G protein-coupled receptors and how PLC-β functions in the nucleus. PLC-β has also been shown to interplay with tyrosine kinase-based signaling pathways, specifically to inhibit Stat5 activation by recruiting the protein-tyrosine phosphatase SHP-1. In this regard, a new multimolecular signaling platform, named SPS complex, has been identified. The SPS complex has important regulatory roles in tumorigenesis and immune cell activation. Furthermore, a growing body of work suggests that PLC-β also participates in the differentiation and activation of immune cells that control both the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Wenbin Xiao
- Department of Pathology, University Hospital Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|