1
|
Crozier RWE, Fajardo VA, MacNeil AJ. Targeting glycogen synthase kinase 3 with CHIR99021 negatively regulates allergen-induced mast cell activation. Eur J Immunol 2023; 53:e2250104. [PMID: 36349527 DOI: 10.1002/eji.202250104] [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: 07/28/2022] [Revised: 09/16/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Mast cells are granulated immune sentinels responsible for allergic inflammation. Allergen-induced FcεRI-signaling leads to rapid degranulation in the early-phase and sustained production and release of pro-inflammatory mediators in the late phase. Glycogen synthase kinase 3 (GSK3) is a constitutively active serine/threonine kinase and a central molecular convergence point for several pro-inflammatory pathways. GSK3 inhibition has been shown to reduce inflammation but has not yet been fully characterized in mast cell activation. Therefore, the objective of this study was to evaluate GSK3 as a putative therapeutic target in allergic inflammation using the GSK3 inhibitor, CHIR99021. Here, we found that GSK3 inhibition impaired ROS production and degranulation. Through modulation of MKK4-JNK, c-jun, and NF-κB signaling, GSK3 inhibition reduced the production/release of IL-6, IL-13, TNF, and CCL1, while only the release of CCL2 and CCL3 was impaired. Furthermore, CHIR99021-mediated GSK3 inhibition altered the pro-inflammatory phenotype of mast cells, reducing c-kit receptor levels. This implicated GSK3 in FcεRI signaling, reducing release of IL-6, TNF, and CCL1 when stimulated through FcεRI, while CCL2 and CCL3 remained unaffected, and were increased when stimulated with SCF only. These results identify GSK3 as a potential therapeutic target of utility warranting further consideration in contexts of pathological mast cell activation.
Collapse
Affiliation(s)
- Robert W E Crozier
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St Catharines, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
2
|
Yin Y, Bai Y, Olivera A, Metcalfe DD. Demonstration and implications of IL-3 upregulation of CD25 expression on human mast cells. J Allergy Clin Immunol 2021; 149:1392-1401.e6. [PMID: 34506850 DOI: 10.1016/j.jaci.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/11/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND CD25+ human mast cells (huMCs) have been reported in patients with monoclonal mast cell diseases and in rare association with inflammation. However, the regulation of CD25 expression on huMCs and the possible biologic consequences remain poorly understood. OBJECTIVE We sought to identify conditions that would upregulate CD25 expression on huMCs and to explore possible functional implications. METHODS huMCs were cultured from peripheral blood progenitor cells over 6 to 8 weeks. Expression of CD25 was determined by fluorescence-activated cell sorting and soluble CD25 by ELISA. Signal transducer and activator of transcription 5 (STAT5) phosphorylation induced by IL-2 in huMCs, regulatory T (Treg) cells, or in cocultured huMCs and Treg cells was examined by fluorescence-activated cell sorting. RESULTS Addition of IL-3 to CD34+ progenitors at the initiation of huMC cultures in the presence of stem cell factor and IL-6 upregulated the expression of CD25 in developing huMCs and resulted in shedding of soluble CD25 into the media. Removal of IL-3 after the first week of culture did not affect subsequent expression of CD25. Furthermore, addition of IL-3 14 days after the initiation of the culture did not induce significant CD25 expression. Treatment with anti-IL-3 antibody or the Janus kinase inhibitor tofacitinib blocked IL-3-induced CD25 upregulation. Binding of IL-2 to CD25+ huMCs did not induce STAT5 phosphorylation. However, coincubation of Treg cells with CD25+ huMCs pretreated with IL-2 was sufficient to result in STAT5 phosphorylation in Treg cells. CONCLUSIONS IL-3 promotes CD25 expression and shedding by huMCs. Although CD25+ huMCs do not respond to IL-2, they bind IL-2 and may act as a reservoir of IL-2 to then activate lymphocytes.
Collapse
Affiliation(s)
- Yuzhi Yin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
3
|
Li Y, Zheng J, Gong C, Lan K, Shen Y, Ding X. Development of an immunogenomic landscape for the competing endogenous RNAs network of peri-implantitis. BMC MEDICAL GENETICS 2020; 21:208. [PMID: 33081707 PMCID: PMC7576812 DOI: 10.1186/s12881-020-01145-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Peri-implantitis is an inflammation that occurs around the implant, resulting in varying degrees of inflammatory damage to the soft and hard tissues. The characteristic criterion is the loss of the supporting bone in an inflammatory environment. However, the specific mechanisms and biomarkers involved in peri-implantitis remain to be further studied. Recently, competing endogenous RNAs (ceRNA) and immune microenvironment have been found to play a more important role in the inflammatory process. In our study, we analyzed the expression of immune related microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and message RNAs (mRNAs) in peri-implantitis by analyzing GSE33774 and GSE57631. METHODS In this study, we explored the expression profile data of immune-related lncRNAs, miRNAs and mRNAs, and constructed immune-related ceRNA network involved in the pathogenesis of peri-implantitis. In addition, the CIBERSORT was used to evaluate the content of immune cells in normal tissues and peri-implantitis to detect the immune microenvironment of peri-implantitis. RESULTS In the analysis, 14 DElncRNAs, 16 DEmiRNAs, and 18 DEmRNAs were used to establish an immune related ceRNA network and the immune infiltration patterns associated with peri-implantitis was discovered. Through the mutual verification of the two datasets, we found that GSK3B and miR-1297 may have important significance in the immune microenvironment and pathogenesis of peri-implantitis and GSK3B was closely related to four types of immune cells, especially with the highest correlation with resting mast cells (P = 0.0003). CONCLUSIONS Through immune-related ceRNA network, immune-related genes (IRGs) and immune cell infiltration can further comprehensively understand the pathogenesis of peri-implantitis, which built up an immunogenomic landscape with clinical significance for peri-implantitis.
Collapse
Affiliation(s)
- Yang Li
- Department of Stomatology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.,State key laboratory of molecular engineering of polymers, Fudan University, Shanghai, P.R. China
| | - Jina Zheng
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Chanjuan Gong
- Department of Stomatology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Kengfu Lan
- Department of Stomatology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Yuqing Shen
- Department of Stomatology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Xiaojun Ding
- Department of Stomatology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China. .,State key laboratory of molecular engineering of polymers, Fudan University, Shanghai, P.R. China.
| |
Collapse
|
4
|
Hazzan T, Eberle J, Worm M, Babina M. Thymic Stromal Lymphopoietin Interferes with the Apoptosis of Human Skin Mast Cells by a Dual Strategy Involving STAT5/Mcl-1 and JNK/Bcl-x L. Cells 2019; 8:E829. [PMID: 31387206 PMCID: PMC6721763 DOI: 10.3390/cells8080829] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
Mast cells (MCs) play critical roles in allergic and inflammatory reactions and contribute to multiple pathologies in the skin, in which they show increased numbers, which frequently correlates with severity. It remains ill-defined how MC accumulation is established by the cutaneous microenvironment, in part because research on human MCs rarely employs MCs matured in the tissue, and extrapolations from other MC subsets have limitations, considering the high level of MC heterogeneity. Thymic stromal lymphopoietin (TSLP)-released by epithelial cells, like keratinocytes, following disturbed homeostasis and inflammation-has attracted much attention, but its impact on skin MCs remains undefined, despite the vast expression of the TSLP receptor by these cells. Using several methods, each detecting a distinct component of the apoptotic process (membrane alterations, DNA degradation, and caspase-3 activity), our study pinpoints TSLP as a novel survival factor of dermal MCs. TSLP confers apoptosis resistance via concomitant activation of the TSLP/ signal transducer and activator of transcription (STAT)-5 / myeloid cell leukemia (Mcl)-1 route and a newly uncovered TSLP/ c-Jun-N-terminal kinase (JNK)/ B-cell lymphoma (Bcl)-xL axis, as evidenced by RNA interference and pharmacological inhibition. Our findings highlight the potential contribution of TSLP to the MC supportive niche of the skin and, vice versa, highlight MCs as crucial responders to TSLP in the context of TSLP-driven disorders.
Collapse
Affiliation(s)
- Tarek Hazzan
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Eberle
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Margitta Worm
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Magda Babina
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
5
|
Kim MM, Audet J. On-demand serum-free media formulations for human hematopoietic cell expansion using a high dimensional search algorithm. Commun Biol 2019; 2:48. [PMID: 30729186 PMCID: PMC6358607 DOI: 10.1038/s42003-019-0296-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Substitution of serum and other clinically incompatible reagents is requisite for controlling product quality in a therapeutic cell manufacturing process. However, substitution with chemically defined compounds creates a complex, large-scale optimization problem due to the large number of possible factors and dose levels, making conventional process optimization methods ineffective. We present a framework for high-dimensional optimization of serum-free formulations for the expansion of human hematopoietic cells. Our model-free approach utilizes evolutionary computing principles to drive an experiment-based feedback control platform. We validate this method by optimizing serum-free formulations for first, TF-1 cells and second, primary T-cells. For each cell type, we successfully identify a set of serum-free formulations that support cell expansions similar to the serum-containing conditions commonly used to culture these cells, by experimentally testing less than 1 × 10-5 % of the total search space. We also demonstrate how this iterative search process can provide insights into factor interactions that contribute to supporting cell expansion.
Collapse
Affiliation(s)
- Michelle M. Kim
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5S 3G9 Canada
| | - Julie Audet
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, ON M5S 3E5 Canada
| |
Collapse
|
6
|
Chen LC, Chen MH, Su TP, Tsai SJ, Bai YM, Li CT, Yang AC, Chang WH, Chen TJ. Atopic diseases/diathesis and subsequent ischemic stroke among patients with bipolar disorder: A nationwide longitudinal study. PLoS One 2018; 13:e0200682. [PMID: 30114239 PMCID: PMC6095510 DOI: 10.1371/journal.pone.0200682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/07/2018] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Previous studies have suggested that both bipolar disorder and atopy are associated with an increased risk of stroke. However, the role of atopic diseases/diathesis in the risk of stroke among patients with bipolar disorder remains unclear. METHODS Using Taiwan's National Health Insurance Research Database, we selected 55,593 patients with bipolar disorder between 2002 and 2008, divided them into patients with atopic diseases/diathesis (n = 21,050) and patients without atopic diseases/diathesis (n = 34,543), and observed them until the end of 2011. Patients who experienced a stroke during the follow-up period were identified. RESULTS Patients with bipolar disorder and atopic diseases/diathesis had an elevated risk of ischemic stroke (hazard ratio [HR]: 1.44, 95% confidence interval [CI]: 1.25-1.59) compared with patients with only bipolar disorder; a dose-dependent relationship was observed between the number of allergic comorbidities and the risk of ischemic stroke (1 atopic disease, HR: 1.30, 95% CI: 1.13-1.49; 2 atopic diseases, HR: 1.59, 95% CI: 1.33-1.91; ≥ 3 atopic diseases, HR: 2.09, 95% CI: 1.50-2.91).The role of atopic diseases in the risk of hemorrhagic stroke among patients with bipolar disorder was nonsignificant (HR: 0.84, 95% CI: 0.64-1.09). CONCLUSIONS Patients with bipolar disorder and atopic diseases/diathesis are more prone to ischemic stroke later in life than are those without atopic diseases/diathesis. Further study is required to investigate the underlying mechanism linking atopy, bipolar disorder, and stroke.
Collapse
Affiliation(s)
- Li-Chi Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (TPS); (SJT)
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (TPS); (SJT)
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Albert C. Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Wen-Han Chang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Hospital and Health Care Administration, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
7
|
Apoptotic resistance of human skin mast cells is mediated by Mcl-1. Cell Death Discov 2017; 3:17048. [PMID: 28845295 PMCID: PMC5563844 DOI: 10.1038/cddiscovery.2017.48] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/03/2017] [Indexed: 12/16/2022] Open
Abstract
Mast cells (MCs) are major effector cells of allergic reactions and contribute to multiple other pathophysiological processes. MCs are long-lived in the tissue microenvironment, in which they matured, but it remains ill-defined how longevity is established by the natural habitat, as research on human MCs chiefly employs cells generated and expanded in culture. In this study, we report that naturally differentiated skin MCs exhibit substantial resilience to cell death with considerable portions surviving up to 3 days in the complete absence of growth factors (GF). This was evidenced by kinetic resolution of membrane alterations (Annexin-V, YoPro), DNA degradation (propidium iodide), mitochondrial membrane disruption (Depsipher), and Caspase-3 activity. Because of the high basal survival, further protection by SCF was modest. Conversely, survival was severely compromised by staurosporine, implying functional caspase machinery. Contrary to the resistance of freshly purified MCs, their culture-expanded counterpart readily underwent cell death upon GF deprivation. Searching for the molecular underpinnings explaining the difference, we identified Mcl-1 as a critical protector. In fact, silencing Mcl-1 by RNAi led to impaired survival in skin MCs ex vivo, but not their cultured equivalent. Therefore, MCs matured in the skin have not only higher expression of Mcl-1 than proliferating MCs, but also greater reliance on Mcl-1 for their survival. Collectively, we report that human skin MCs display low susceptibility to cell death through vast expression of Mcl-1, which protects from mortality and may contribute to MC longevity in the tissue.
Collapse
|
8
|
Wyse Jackson AC, Cotter TG. The synthetic progesterone Norgestrel is neuroprotective in stressed photoreceptor-like cells and retinal explants, mediating its effects via basic fibroblast growth factor, protein kinase A and glycogen synthase kinase 3β signalling. Eur J Neurosci 2016; 43:899-911. [PMID: 26750157 DOI: 10.1111/ejn.13166] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/17/2015] [Accepted: 12/29/2015] [Indexed: 01/19/2023]
Abstract
The synthetic progesterone Norgestrel has been shown to have proven neuroprotective efficacy in two distinct models of retinitis pigmentosa: the rd10/rd10 (B6.CXBI-Pde6b(rd10)/J) mouse model and the Balb/c light-damage model. However, the cellular mechanism underlying this neuroprotection is still largely unknown. Therefore, this study aimed to examine the downstream signalling pathways associated with Norgestrel both in vitro and ex vivo. In this work, we identify the potential of Norgestrel to rescue stressed 661W photoreceptor-like cells and ex vivo retinal explants from cell death over 24 h. Norgestel is thought to work through an upregulation of neuroprotective basic fibroblast growth factor (bFGF). Analysis of 661W cells in vitro by real-time polymerase chain reaction (rt-PCR), enzyme-linked immunosorbent assay (ELISA) and Western blotting revealed an upregulation of bFGF in response to Norgestrel over 6 h. Specific siRNA knockdown of bFGF abrogated the protective properties of Norgestrel on damaged photoreceptors, thus highlighting the crucial importance of bFGF in Norgestrel-mediated protection. Furthermore, Norgestrel initiated a bFGF-dependent inactivation of glycogen synthase kinase 3β (GSK3β) through phosphorylation at serine 9. The effects of Norgestrel on GSK3β were dependent on protein kinase A (PKA) pathway activation. Specific inhibition of both the PKA and GSK3β pathways prevented Norgestrel-mediated neuroprotection of stressed photoreceptor cells in vitro. Involvement of the PKA pathway following Norgestrel treatment was also confirmed ex vivo. Therefore, these results indicate that the protective efficacy of Norgestrel is, at least in part, due to the bFGF-mediated activation of the PKA pathway, with subsequent inactivation of GSK3β.
Collapse
Affiliation(s)
- Alice C Wyse Jackson
- Biochemistry Department, Cell Development and Disease Laboratory, Bioscience Research Institute, University College Cork, College Road, Cork City Centre, Cork, Ireland
| | - Thomas G Cotter
- Biochemistry Department, Cell Development and Disease Laboratory, Bioscience Research Institute, University College Cork, College Road, Cork City Centre, Cork, Ireland
| |
Collapse
|
9
|
Zhang Y, Wahl LM. Cytokine-induced monocyte MMP-1 is negatively regulated by GSK-3 through a p38 MAPK-mediated decrease in ERK1/2 MAPK activation. J Leukoc Biol 2015; 97:921-927. [PMID: 25714802 DOI: 10.1189/jlb.3a0413-235r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 01/02/2015] [Accepted: 01/30/2015] [Indexed: 12/28/2022] Open
Abstract
Elucidation of the signal transduction events leading to the production of MMPs by monocytes/macrophages may provide insights into the mechanisms involved in the destruction of connective tissue associated with chronic inflammatory lesions. Here, we show that GSK-3 is a negative regulator of cytokine-induced MMP-1 production by monocytes. Inhibition of monocyte GSK-3 pharmacologically with SB216763 or GSK-3β siRNA caused a significant enhancement of MMP-1 by TNF-α- and GM-CSF-activated monocytes, indicating that induction of MMP-1 by TNF-α and GM-CSF involved phosphorylation/inactivation of GSK-3. TNF-α- and GM-CSF-induced phosphorylation of GSK-3 and subsequent MMP-1 production was blocked with the PKC inhibitor Gö6976 but not by the AKT1/2 inhibitor AKT VIII, showing that cytokine phosphorylation of GSK-3 occurs primarily through a PKC pathway. Inhibition of GSK-3 resulted in decreased phosphorylation of p38 MAPK with a corresponding increase in phosphorylation of ERK1/2 MAPK. Enhanced MMP-1 production by treatment with SB216763 was a result of increased ERK1/2 activation, as demonstrated by inhibition of MMP-1 by PD98059, a specific ERK1/2 inhibitor. Conversely, the p38 MAPK inhibitor SB203580 enhanced cytokine activation of ERK1/2 and the production of MMP-1 similar to that of SB216763. These findings demonstrate that the degree of cytokine-mediated phosphorylation/inhibition of GSK-3 determines the level of MMP-1 production through a mechanism involving decreased activation of p38 MAPK, a negative regulator of ERK1/2 required for cytokine-induced production of MMP-1 by monocytes.
Collapse
Affiliation(s)
- Yahong Zhang
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Larry M Wahl
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Ogawa K, Tanaka Y, Uruno T, Duan X, Harada Y, Sanematsu F, Yamamura K, Terasawa M, Nishikimi A, Côté JF, Fukui Y. DOCK5 functions as a key signaling adaptor that links FcεRI signals to microtubule dynamics during mast cell degranulation. ACTA ACUST UNITED AC 2014; 211:1407-19. [PMID: 24913231 PMCID: PMC4076576 DOI: 10.1084/jem.20131926] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Mast cells play a key role in the induction of anaphylaxis, a life-threatening IgE-dependent allergic reaction, by secreting chemical mediators that are stored in secretory granules. Degranulation of mast cells is triggered by aggregation of the high-affinity IgE receptor, FcεRI, and involves dynamic rearrangement of microtubules. Although much is known about proximal signals downstream of FcεRI, the distal signaling events controlling microtubule dynamics remain elusive. Here we report that DOCK5, an atypical guanine nucleotide exchange factor (GEF) for Rac, is essential for mast cell degranulation. As such, we found that DOCK5-deficient mice exhibit resistance to systemic and cutaneous anaphylaxis. The Rac GEF activity of DOCK5 is surprisingly not required for mast cell degranulation. Instead, DOCK5 associated with Nck2 and Akt to regulate microtubule dynamics through phosphorylation and inactivation of GSK3β. When DOCK5-Nck2-Akt interactions were disrupted, microtubule formation and degranulation response were severely impaired. Our results thus identify DOCK5 as a key signaling adaptor that orchestrates remodeling of the microtubule network essential for mast cell degranulation.
Collapse
Affiliation(s)
- Kana Ogawa
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Yoshihiko Tanaka
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Xuefeng Duan
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Yosuke Harada
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Fumiyuki Sanematsu
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Kazuhiko Yamamura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Masao Terasawa
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Akihiko Nishikimi
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Quebec H2W 1R7, Canada
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, JapanDivision of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102-0076, Japan
| |
Collapse
|
11
|
Fukuishi N, Igawa Y, Kunimi T, Hamano H, Toyota M, Takahashi H, Kenmoku H, Yagi Y, Matsui N, Akagi M. Generation of mast cells from mouse fetus: analysis of differentiation and functionality, and transcriptome profiling using next generation sequencer. PLoS One 2013; 8:e60837. [PMID: 23573287 PMCID: PMC3616098 DOI: 10.1371/journal.pone.0060837] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/03/2013] [Indexed: 12/31/2022] Open
Abstract
While gene knockout technology can reveal the roles of proteins in cellular functions, including in mast cells, fetal death due to gene manipulation frequently interrupts experimental analysis. We generated mast cells from mouse fetal liver (FLMC), and compared the fundamental functions of FLMC with those of bone marrow-derived mouse mast cells (BMMC). Under electron microscopy, numerous small and electron-dense granules were observed in FLMC. In FLMC, the expression levels of a subunit of the FcεRI receptor and degranulation by IgE cross-linking were comparable with BMMC. By flow cytometry we observed surface expression of c-Kit prior to that of FcεRI on FLMC, although on BMMC the expression of c-Kit came after FcεRI. The surface expression levels of Sca-1 and c-Kit, a marker of putative mast cell precursors, were slightly different between bone marrow cells and fetal liver cells, suggesting that differentiation stage or cell type are not necessarily equivalent between both lineages. Moreover, this indicates that phenotypically similar mast cells may not have undergone an identical process of differentiation. By comprehensive analysis using the next generation sequencer, the same frequency of gene expression was observed for 98.6% of all transcripts in both cell types. These results indicate that FLMC could represent a new and useful tool for exploring mast cell differentiation, and may help to elucidate the roles of individual proteins in the function of mast cells where gene manipulation can induce embryonic lethality in the mid to late stages of pregnancy.
Collapse
Affiliation(s)
- Nobuyuki Fukuishi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Current world literature. Curr Opin Nephrol Hypertens 2012; 21:557-66. [PMID: 22874470 DOI: 10.1097/mnh.0b013e3283574c3b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|