1
|
Kannen V, Grant DM, Matthews J. The mast cell-T lymphocyte axis impacts cancer: Friend or foe? Cancer Lett 2024; 588:216805. [PMID: 38462035 DOI: 10.1016/j.canlet.2024.216805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/01/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Crosstalk between mast cells (MCs) and T lymphocytes (TLs) releases specific signals that create an environment conducive to tumor development. Conversely, they can protect against cancer by targeting tumor cells for destruction. Although their role in immunity and cancer is complex, their potential in anticancer strategies is often underestimated. When peripheral MCs are activated, they can affect cancer development. Tumor-infiltrating TLs may malfunction and contribute to aggressive cancer and poor prognoses. One promising approach for cancer patients is TL-based immunotherapies. Recent reports suggest that MCs modulate TL activity in solid tumors and may be a potential therapeutic layer in multitargeting anticancer strategies. Pharmacologically modulating MC activity can enhance the anticancer cytotoxic TL response in tumors. By identifying tumor-specific targets, it has been possible to genetically alter patients' cells into fully humanized anticancer cellular therapies for autologous transplantation, including the engineering of TLs and MCs to target and kill cancer cells. Hence, recent scientific evidence provides a broader understanding of MC-TL activity in cancer.
Collapse
Affiliation(s)
- Vinicius Kannen
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| | - Denis M Grant
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jason Matthews
- Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Nutrition, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Ou Q, Power R, Griffin MD. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front Immunol 2023; 14:1287465. [PMID: 37928540 PMCID: PMC10623442 DOI: 10.3389/fimmu.2023.1287465] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Regulatory T cells (Treg) are known to be critical for the maintenance of immune homeostasis by suppressing the activation of auto- or allo-reactive effector T cells through a diverse repertoire of molecular mechanisms. Accordingly, therapeutic strategies aimed at enhancing Treg numbers or potency in the setting of autoimmunity and allogeneic transplants have been energetically pursued and are beginning to yield some encouraging outcomes in early phase clinical trials. Less well recognized from a translational perspective, however, has been the mounting body of evidence that Treg directly modulate most aspects of innate immune response under a range of different acute and chronic disease conditions. Recognizing this aspect of Treg immune modulatory function provides a bridge for the application of Treg-based therapies to common medical conditions in which organ and tissue damage is mediated primarily by inflammation involving myeloid cells (mononuclear phagocytes, granulocytes) and innate lymphocytes (NK cells, NKT cells, γδ T cells and ILCs). In this review, we comprehensively summarize pre-clinical and human research that has revealed diverse modulatory effects of Treg and specific Treg subpopulations on the range of innate immune cell types. In each case, we emphasize the key mechanistic insights and the evidence that Treg interactions with innate immune effectors can have significant impacts on disease severity or treatment. Finally, we discuss the opportunities and challenges that exist for the application of Treg-based therapeutic interventions to three globally impactful, inflammatory conditions: type 2 diabetes and its end-organ complications, ischemia reperfusion injury and atherosclerosis.
Collapse
Affiliation(s)
- Qifeng Ou
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Rachael Power
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| |
Collapse
|
3
|
Yang G, Li J, Liu Y, Wu G, Mo L, Xu Z, Liao Y, Huang Q, Yang P. Targeting the RhoA-GEF-H1 pathway of mast cells attenuates experimental airway allergy. Arch Biochem Biophys 2023; 741:109597. [PMID: 37054768 DOI: 10.1016/j.abb.2023.109597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
Mast cells are the major effector cells in allergic diseases. RhoA and its downstream pathway is associated with the pathogenesis of airway allergy. The objective of this study is to test a hypothesis that modulating the RhoA-GEF-H1 axis in mast cells can attenuate airway allergy. An airway allergic disorder (AAD) mouse model was employed. Mast cells were isolated from AAD mouse airway tissues to be analyzed by RNA sequencing. We observed that mast cells isolated from the respiratory tract of AAD mice were resistant to apoptosis. Mast cell mediator levels in nasal lavage fluid were correlated with apoptosis resistance in AAD mice. Activation of RhoA in AAD mast cells was related to resistance to apoptosis. Mast cells isolated from the airway tissues in AAD mouse exhibited strong RhoA-GEF-H1 expression. The RhoA-GEF-H1 axis was associated with the lower FasL expression in AAD mast cells. Activation of the RhoA-GEF-H1 axis promoted the production of mediators in mast cells. Inhibition of GEF-H1 facilitated the SIT-induced mast cell apoptosis and enhanced the therapeutic efficacy of AAD. In conclusion, RhoA-GEF-H1 activities are associated with resistance to apoptosis in mast cells isolated from sites of allergic lesions. The state of apoptosis resistance in mast cells is associated with the state of AAD disease. Inhibition of GEF-H1 restores the sensitivity of mast cells to apoptosis inducers, and alleviates experimental AAD in mice.
Collapse
Affiliation(s)
- Gui Yang
- Department of Otolaryngology and Allergy, Longgang Central Hospital, Shenzhen, China
| | - Jianxiang Li
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yu Liu
- Department of Otolaryngology, Jinjiang Municipal Hospital, Jinjiang, China
| | - Gaohui Wu
- Department of Otolaryngology, Jinjiang Municipal Hospital, Jinjiang, China
| | - Lihua Mo
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Ziyi Xu
- Vanke Meisha Academy, Shenzhen, China
| | - Yun Liao
- Department of Otolaryngology and Allergy, Longgang Central Hospital, Shenzhen, China
| | - Qinmiao Huang
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Pingchang Yang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China.
| |
Collapse
|
4
|
Krajewska NM, Fiancette R, Oo YH. Interplay between Mast Cells and Regulatory T Cells in Immune-Mediated Cholangiopathies. Int J Mol Sci 2022; 23:5872. [PMID: 35682552 PMCID: PMC9180565 DOI: 10.3390/ijms23115872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 01/10/2023] Open
Abstract
Immune-mediated cholangiopathies are characterised by the destruction of small and large bile ducts causing bile acid stasis, which leads to subsequent inflammation, fibrosis, and eventual cirrhosis of the liver tissue. A breakdown of peripheral hepatic immune tolerance is a key feature of these diseases. Regulatory T cells (Tregs) are a major anti-inflammatory immune cell subset, and their quantities and functional capacity are impaired in autoimmune liver diseases. Tregs can undergo phenotypic reprogramming towards pro-inflammatory Th1 and Th17 profiles. The inflamed hepatic microenvironment influences and can impede normal Treg suppressive functions. Mast cell (MC) infiltration increases during liver inflammation, and active MCs have been shown to be an important source of pro-inflammatory mediators, thus driving pathogenesis. By influencing the microenvironment, MCs can indirectly manipulate Treg functions and inhibit their suppressive and proliferative activity. In addition, direct cell-to-cell interactions have been identified between MCs and Tregs. It is critical to consider the effects of MCs on the inflammatory milieu of the liver and their influence on Treg functions. This review will focus on the roles and crosstalk of Tregs and MCs during autoimmune cholangiopathy pathogenesis progression.
Collapse
Affiliation(s)
- Natalia M. Krajewska
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Rémi Fiancette
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Ye H. Oo
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
- Advanced Cellular Therapy Facility, University of Birmingham, Birmingham B15 2TT, UK
- Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| |
Collapse
|
5
|
Khan N, Hu Y, Lowell CA, Rothstein TL. Signal Integration by Translocation and Phosphorylation of PKCδ in the B Cell Alternate Pathway. THE JOURNAL OF IMMUNOLOGY 2021; 207:2288-2296. [PMID: 34588218 DOI: 10.4049/jimmunol.2100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022]
Abstract
B cell signaling for activation via the BCR occurs as an isolated event only in vitro; in real life, BCR signaling takes place within a complex milieu that involves interactions with agents that trigger additional receptors. Chief among these is IL-4. We have shown that BCR signaling is reprogrammed by IL-4 receptor engagement and that this reprogramming involves creation of a new, signalosome-independent, Lyn-dependent alternate signaling pathway in B cells isolated from BALB/cByJ mice. A unique aspect of the alternate pathway is protein kinase Cδ (PKCδ) phosphorylation. In dissecting this pathway, we unexpectedly found that Lyn is associated with IL-4Rα, that IL-4 induces Lyn activation, and that Lyn immunoprecipitated from IL-4-treated B cells capably phosphorylates PKCδ in a cell-free system. However, PKCδ phosphorylation does not occur in the absence of BCR triggering in vivo. This raised the question of why IL-4 alone failed to produce PKCδ phosphorylation. We considered the possibility that Lyn and PKCδ may be spatially separated. As expected, before any treatment, Lyn is located primarily in the membrane fraction, whereas PKCδ is located mainly in the cytosol fraction. However, when anti-Ig follows IL-4 treatment, PKCδ is found in the membrane fraction and phosphorylated. This translocation of PKCδ to the membrane fraction is not affected by loss of Lyn, although PKCδ phosphorylation requires Lyn. Thus, PKCδ phosphorylation through the alternate pathway represents the result of signal integration, whereby neither IL-4 nor anti-Ig working alone produces this outcome, but together they achieve this result by Lyn activation (IL-4) and PKCδ translocation (IL-4 followed by anti-Ig).
Collapse
Affiliation(s)
- Naeem Khan
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Yongmei Hu
- Department of Laboratory Medicine, University of California at San Francisco School of Medicine, San Francisco, CA; and
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California at San Francisco School of Medicine, San Francisco, CA; and
| | - Thomas L Rothstein
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI; .,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| |
Collapse
|
6
|
Tontini C, Bulfone-Paus S. Novel Approaches in the Inhibition of IgE-Induced Mast Cell Reactivity in Food Allergy. Front Immunol 2021; 12:613461. [PMID: 34456900 PMCID: PMC8387944 DOI: 10.3389/fimmu.2021.613461] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 06/23/2021] [Indexed: 01/21/2023] Open
Abstract
Allergy is an IgE-dependent type-I hypersensitivity reaction that can lead to life-threatening systemic symptoms such as anaphylaxis. In the pathogenesis of the allergic response, the common upstream event is the binding of allergens to specific IgE, inducing cross-linking of the high-affinity FcεRI on mast cells, triggering cellular degranulation and the release of histamine, proteases, lipids mediators, cytokines and chemokines with inflammatory activity. A number of novel therapeutic options to curb mast cell activation are in the pipeline for the treatment of severe allergies. In addition to anti-IgE therapy and allergen-specific immunotherapy, monoclonal antibodies targeted against several key Th2/alarmin cytokines (i.e. IL-4Rα, IL-33, TSLP), active modification of allergen-specific IgE (i.e. inhibitory compounds, monoclonal antibodies, de-sialylation), engagement of inhibitory receptors on mast cells and allergen-specific adjuvant vaccines, are new promising options to inhibit the uncontrolled release of mast cell mediators upon allergen exposure. In this review, we critically discuss the novel approaches targeting mast cells limiting allergic responses and the immunological mechanisms involved, with special interest on food allergy treatment.
Collapse
Affiliation(s)
- Chiara Tontini
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
7
|
Bhuiyan P, Chen Y, Karim M, Dong H, Qian Y. Bidirectional communication between mast cells and the gut-brain axis in neurodegenerative diseases: Avenues for therapeutic intervention. Brain Res Bull 2021; 172:61-78. [PMID: 33892083 DOI: 10.1016/j.brainresbull.2021.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/02/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022]
Abstract
Although the global incidence of neurodegenerative diseases has been steadily increasing, especially in adults, there are no effective therapeutic interventions. Neurodegeneration is a heterogeneous group of disorders that is characterized by the activation of immune cells in the central nervous system (CNS) (e.g., mast cells and microglia) and subsequent neuroinflammation. Mast cells are found in the brain and the gastrointestinal tract and play a role in "tuning" neuroimmune responses. The complex bidirectional communication between mast cells and gut microbiota coordinates various dynamic neuro-cellular responses, which propagates neuronal impulses from the gastrointestinal tract into the CNS. Numerous inflammatory mediators from degranulated mast cells alter intestinal gut permeability and disrupt blood-brain barrier, which results in the promotion of neuroinflammatory processes leading to neurological disorders, thereby offsetting the balance in immune-surveillance. Emerging evidence supports the hypothesis that gut-microbiota exert a pivotal role in inflammatory signaling through the activation of immune and inflammatory cells. Communication between inflammatory cytokines and neurocircuits via the gut-brain axis (GBA) affects behavioral responses, activates mast cells and microglia that causes neuroinflammation, which is associated with neurological diseases. In this comprehensive review, we focus on what is currently known about mast cells and the gut-brain axis relationship, and how this relationship is connected to neurodegenerative diseases. We hope that further elucidating the bidirectional communication between mast cells and the GBA will not only stimulate future research on neurodegenerative diseases but will also identify new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Yinan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Mazharul Karim
- College of Pharmacy, Western University of Health Science, 309 East 2nd Street, Pomona, CA, 91766, USA
| | - Hongquan Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China.
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China.
| |
Collapse
|
8
|
El Ansari YS, Kanagaratham C, Lewis OL, Oettgen HC. IgE and mast cells: The endogenous adjuvant. Adv Immunol 2020; 148:93-153. [PMID: 33190734 DOI: 10.1016/bs.ai.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are most familiar as the effectors of type I hypersensitivity reactions including anaphylaxis. It is becoming clear however that this pair has important immunomodulatory effects on innate and adaptive cells of the immune system. In this purview, they act as endogenous adjuvants to ignite evolving immune responses, promote the transition of allergic disease into chronic illness and disrupt the development of active mechanisms of tolerance to ingested foods. Suppression of IgE-mediated mast cell activation can be exerted by molecules targeting IgE, FcɛRI or signaling kinases including Syk, or by IgG antibodies acting via inhibitory Fcγ receptors. In 2015 we reviewed the evidence for the adjuvant functions of mast cells. This update includes the original text, incorporates some important developments in the field over the past five years and discusses how interventions targeting these pathways might have promise in the development of strategies to treat allergic disease.
Collapse
Affiliation(s)
- Yasmeen S El Ansari
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Owen L Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
9
|
The Alternate Pathway for BCR Signaling Induced by IL-4 Requires Lyn Tyrosine Kinase. J Mol Biol 2020; 433:166667. [PMID: 33058880 DOI: 10.1016/j.jmb.2020.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
BCR signaling triggers a cascade of intracellular mediators that eventuates in transcription factor activation. Signaling is proximally mediated by Src family tyrosine kinases, the most abundant being Lyn. Key mediators are grouped together as the signalosome, and failure of any single member of this group leads to failure of signaling via this classical pathway. Recent work has revealed an alternate pathway for BCR signaling, in which signalosome elements are bypassed for downstream events such as ERK and PKCδ phosphorylation. This pathway is created by B cell treatment with IL-4 prior to BCR triggering. After IL-4 treatment, the alternate pathway for pERK operates in parallel with the classical pathway for pERK, whereas PKCδ phosphorylation is specific to the alternate pathway. Remarkably, Lyn is not required for B cell activation via the classical pathway; however, Lyn is indispensable and irreplaceable for B cell activation via the alternate pathway. Thus, Lyn operates at a branch point that determines the nature of the B cell response to BCR activation. The mechanism underlying the absolute dependence of alternate pathway signaling on Lyn is unknown. Here, our current understanding of receptor crosstalk between IL-4R and BCR is summarized along with several possible mechanisms for the role of Lyn in alternate pathway signaling. Further dissection of alternate pathway signaling and the role of Lyn is likely to provide important information relating to normal B cell responses, malignant B cell expansion, and generic principles relating to receptor interactions and crosstalk.
Collapse
|
10
|
Zhang Y, Saradna A, Ratan R, Ke X, Tu W, Do DC, Hu C, Gao P. RhoA/Rho-kinases in asthma: from pathogenesis to therapeutic targets. Clin Transl Immunology 2020; 9:e01134. [PMID: 32355562 PMCID: PMC7190398 DOI: 10.1002/cti2.1134] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Asthma is a chronic and heterogeneous disease characterised by airway inflammation and intermittent airway narrowing. The key obstacle in the prevention and treatment of asthma has been our incomplete understanding of its aetiology and biological mechanisms. The ras homolog family member A (RhoA) of the Rho family GTPases has been considered to be one of the most promising and novel therapeutic targets for asthma. It is well known that RhoA/Rho-kinases play an important role in the pathophysiology of asthma, including airway smooth muscle contraction, airway hyper-responsiveness, β-adrenergic desensitisation and airway remodelling. However, recent advances have suggested novel roles for RhoA in regulating allergic airway inflammation. Specifically, RhoA has been shown to regulate allergic airway inflammation through controlling Th2 or Th17 cell differentiation and to regulate airway remodelling through regulating mesenchymal stem cell (MSC) differentiation. In this review, we evaluate the literature regarding the recent advances in the activation of RhoA/Rho-kinase, cytokine and epigenetic regulation of RhoA/Rho-kinase, and the role of RhoA/Rho-kinase in regulating major features of asthma, such as airway hyper-responsiveness, remodelling and inflammation. We also discuss the importance of the newly identified role of RhoA/Rho-kinase signalling in MSC differentiation and bronchial epithelial barrier dysfunction. These findings indicate the functional significance of the RhoA/Rho-kinase pathway in the pathophysiology of asthma and suggest that RhoA/Rho-kinase signalling may be a promising therapeutic target for the treatment of asthma.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Arjun Saradna
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Division of PulmonaryCritical Care and Sleep MedicineState University of New York at BuffaloBuffaloNYUSA
| | - Rhea Ratan
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Xia Ke
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of OtorhinolaryngologyFirst Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wei Tu
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Respirology and AllergyThird Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Danh C Do
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Chengping Hu
- Department of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Peisong Gao
- Division of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
11
|
Qian F, Zhang L, Lu S, Mao G, Guo F, Liu P, Xu J, Li Y. Scrodentoid A Inhibits Mast Cell-Mediated Allergic Response by Blocking the Lyn-FcεRIβ Interaction. Front Immunol 2019; 10:1103. [PMID: 31156646 PMCID: PMC6532554 DOI: 10.3389/fimmu.2019.01103] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Mast cells are considered an attractive therapeutic target for treating allergic diseases, and the Lyn–FcεRIβ interaction is essential for mast cell activation. This study investigated the antiallergic effect of scrodentoid A (SA) on mast cells and mast cell–mediated anaphylaxis. Methods: For in vitro experiments, mast cells were treated with SA. Cell proliferation was tested using the XTT assay. The mRNA expression of various cytokines and chemokines was measured using qPCR. The levels of histamine, eicosanoids (PGD2, LTC4), and cytokines were measured using enzyme immunoassay kits. Signaling was investigated using Western blotting and immunoprecipitation. For in vivo experiments, the antiallergic activity of SA was evaluated using two mouse models of passive anaphylaxis as passive cutaneous and systemic anaphylaxis. The mechanism was investigated through immunohistochemistry and immunofluorescence. Results: SA considerably inhibited immunoglobulin (Ig) E-mediated mast cell activation, including β-hexosaminidase release, mRNA and protein expression of various cytokines, and PGD2 and LTC4 release. Oral administration of SA effectively and dose-dependently suppressed mast cell–mediated passive cutaneous and systemic anaphylaxis. SA significantly attenuated the activation of Lyn, Syk, LAT, PLCγ, JNK, Erk1/2, and Ca2+ mobilization without Fyn, Akt, and P38 activation by blocking the Lyn–FcεRIβ interaction. Conclusions: SA suppresses mast cell–mediated allergic response by blocking the Lyn–FcεRIβ interaction in vitro and in vivo. SA may be a promising therapeutic agent for allergic and other mast cell–related diseases.
Collapse
Affiliation(s)
- Fei Qian
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liuqiang Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shaodong Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gaohui Mao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinwen Xu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Jiang S, Da Y, Han S, He Y, Che H. Notch ligand Delta-like1 enhances degranulation and cytokine production through a novel Notch/Dok-1/MAPKs pathway in vitro. Immunol Res 2019; 66:87-96. [PMID: 29181775 DOI: 10.1007/s12026-017-8977-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Food allergy includes sensitization phase and effect phase, and effect cells degranulate and secrete cytokines in the effect phase, causing allergic clinical symptoms. We have demonstrated that Notch signaling plays an important role in the sensitization phase, but its role in effect phases still remains unclear. In this study, we investigated the role of Notch signaling in degranulation and cytokine production of the effect phase response. A RBL-2H3 cell model was used and Notch signaling was induced by priming with Notch ligands. Our results showed after priming with Notch ligand, Delta-like1(Dll1)-Fc, β-hexosaminidase release, and cytokines production, including TGF-β, IL-1β, IL-4, IL-6, and IL-13, were increased significantly, and the enhancement was abolished after DAPT treatment, a γ-secretase inhibitor, indicating that Dll1 Notch signaling enhanced RBL-2H3 cell degranulation and cytokine production. Western blot analysis showed that Dll1 Notch signaling augmented high-affinity IgE receptors-mediated phosphorylation of MAPKs through suppressing the expression of downstream tyrosine kinases 1 (Dok-1). Besides, a passive systemic anaphylaxis mouse model was used to confirm the role of Notch signaling. And our data showed that allergic clinical features of mice were alleviated, and the level of degranulation was decreased significantly after inhibiting Notch signaling in vivo. Therefore, we demonstrated Notch ligand Dll1 enhanced RBL-2H3 cell degranulation and cytokine production through a novel Notch/Dok-1/MAPKs pathway, suggesting Notch signaling played a key role in the effect phase of food allergy.
Collapse
Affiliation(s)
- Songsong Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Yifan Da
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Shiwen Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Yahong He
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China
| | - Huilian Che
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua Donglu, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
13
|
Sibilano R, Gaudenzio N, DeGorter MK, Reber LL, Hernandez JD, Starkl PM, Zurek OW, Tsai M, Zahner S, Montgomery SB, Roers A, Kronenberg M, Yu M, Galli SJ. A TNFRSF14-FcɛRI-mast cell pathway contributes to development of multiple features of asthma pathology in mice. Nat Commun 2016; 7:13696. [PMID: 27982078 PMCID: PMC5171877 DOI: 10.1038/ncomms13696] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/26/2016] [Indexed: 01/07/2023] Open
Abstract
Asthma has multiple features, including airway hyperreactivity, inflammation and remodelling. The TNF superfamily member TNFSF14 (LIGHT), via interactions with the receptor TNFRSF14 (HVEM), can support TH2 cell generation and longevity and promote airway remodelling in mouse models of asthma, but the mechanisms by which TNFSF14 functions in this setting are incompletely understood. Here we find that mouse and human mast cells (MCs) express TNFRSF14 and that TNFSF14:TNFRSF14 interactions can enhance IgE-mediated MC signalling and mediator production. In mouse models of asthma, TNFRSF14 blockade with a neutralizing antibody administered after antigen sensitization, or genetic deletion of Tnfrsf14, diminishes plasma levels of antigen-specific IgG1 and IgE antibodies, airway hyperreactivity, airway inflammation and airway remodelling. Finally, by analysing two types of genetically MC-deficient mice after engrafting MCs that either do or do not express TNFRSF14, we show that TNFRSF14 expression on MCs significantly contributes to the development of multiple features of asthma pathology.
Collapse
Affiliation(s)
- Riccardo Sibilano
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Nicolas Gaudenzio
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Marianne K. DeGorter
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Laurent L. Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, INSERM U1222, Institut Pasteur, Paris 75015, France
| | - Joseph D. Hernandez
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Philipp M. Starkl
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences and Department of Medicine I, Research Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Oliwia W. Zurek
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Sonja Zahner
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Stephen B. Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Axel Roers
- Institute for Immunology, Technische Universität Dresden, Dresden 01307, Germany
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Mang Yu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Stephen J. Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Microbiology and Immunology and Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
14
|
Reber LL, Sibilano R, Mukai K, Galli SJ. Potential effector and immunoregulatory functions of mast cells in mucosal immunity. Mucosal Immunol 2015; 8:444-63. [PMID: 25669149 PMCID: PMC4739802 DOI: 10.1038/mi.2014.131] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/27/2014] [Indexed: 02/04/2023]
Abstract
Mast cells (MCs) are cells of hematopoietic origin that normally reside in mucosal tissues, often near epithelial cells, glands, smooth muscle cells, and nerves. Best known for their contributions to pathology during IgE-associated disorders such as food allergy, asthma, and anaphylaxis, MCs are also thought to mediate IgE-associated effector functions during certain parasite infections. However, various MC populations also can be activated to express functional programs--such as secreting preformed and/or newly synthesized biologically active products--in response to encounters with products derived from diverse pathogens, other host cells (including leukocytes and structural cells), damaged tissue, or the activation of the complement or coagulation systems, as well as by signals derived from the external environment (including animal toxins, plant products, and physical agents). In this review, we will discuss evidence suggesting that MCs can perform diverse effector and immunoregulatory roles that contribute to homeostasis or pathology in mucosal tissues.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Riccardo Sibilano
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Kaori Mukai
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Stephen J Galli
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA,Department of Microbiology & Immunology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| |
Collapse
|
15
|
Sulimenko V, Hájková Z, Černohorská M, Sulimenko T, Sládková V, Dráberová L, Vinopal S, Dráberová E, Dráber P. Microtubule Nucleation in Mouse Bone Marrow–Derived Mast Cells Is Regulated by the Concerted Action of GIT1/βPIX Proteins and Calcium. THE JOURNAL OF IMMUNOLOGY 2015; 194:4099-111. [DOI: 10.4049/jimmunol.1402459] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/27/2015] [Indexed: 11/19/2022]
|
16
|
Nakano N, Nishiyama C, Yagita H, Hara M, Motomura Y, Kubo M, Okumura K, Ogawa H. Notch signaling enhances FcεRI-mediated cytokine production by mast cells through direct and indirect mechanisms. THE JOURNAL OF IMMUNOLOGY 2015; 194:4535-44. [PMID: 25821223 DOI: 10.4049/jimmunol.1301850] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 02/23/2015] [Indexed: 01/12/2023]
Abstract
Th2-type cytokines and TNF-α secreted by activated mast cells upon cross-linking of FcεRI contribute to the development and maintenance of Th2 immunity to parasites and allergens. We have previously shown that cytokine secretion by mouse mast cells is enhanced by signaling through Notch receptors. In this study, we investigated the molecular mechanisms by which Notch signaling enhances mast cell cytokine production induced by FcεRI cross-linking. FcεRI-mediated production of cytokines, particularly IL-4, was significantly enhanced in mouse bone marrow-derived mast cells by priming with Notch ligands. Western blot analysis showed that Notch signaling augmented and prolonged FcεRI-mediated phosphorylation of MAPKs, mainly JNK and p38 MAPK, through suppression of the expression of SHIP-1, a master negative regulator of FcεRI signaling, resulting in the enhanced production of multiple cytokines. The enhancing effect of Notch ligand priming on multiple cytokine production was abolished by knockdown of Notch2, but not Notch1, and FcεRI-mediated production of multiple cytokines was enhanced by retroviral transduction with the intracellular domain of Notch2. However, only IL-4 production was enhanced by both Notch1 and Notch2. The enhancing effect of Notch signaling on IL-4 production was lost in bone marrow-derived mast cells from mice lacking conserved noncoding sequence 2, which is located at the distal 3' element of the Il4 gene locus and contains Notch effector RBP-J binding sites. These results indicate that Notch2 signaling indirectly enhances the FcεRI-mediated production of multiple cytokines, and both Notch1 and Notch2 signaling directly enhances IL-4 production through the noncoding sequence 2 enhancer of the Il4 gene.
Collapse
Affiliation(s)
- Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan;
| | - Chiharu Nishiyama
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan; Department of Biological Science and Technology, Tokyo University of Science, Tokyo 125-8585, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Mutsuko Hara
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Yasutaka Motomura
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; and Laboratory for Cytokine Regulation, Research Center for Integrative Medical Science, RIKEN Research Center for Allergy and Immunology, RIKEN Yokohama Institute, Yokohama 230-0045, Japan
| | - Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; and Laboratory for Cytokine Regulation, Research Center for Integrative Medical Science, RIKEN Research Center for Allergy and Immunology, RIKEN Yokohama Institute, Yokohama 230-0045, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan; Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
17
|
Suzuki R, Scheffel J, Rivera J. New insights on the signaling and function of the high-affinity receptor for IgE. Curr Top Microbiol Immunol 2015; 388:63-90. [PMID: 25553795 DOI: 10.1007/978-3-319-13725-4_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Clustering of the high-affinity receptor for immunoglobulin E (FcεRI) through the interaction of receptor-bound immunoglobulin E (IgE) antibodies with their cognate antigen is required to couple IgE antibody production to cellular responses and physiological consequences. IgE-induced responses through FcεRI are well known to defend the host against certain infectious agents and to lead to unwanted allergic responses to normally innocuous substances. However, the cellular and/or physiological response of individuals that produce IgE antibodies may be markedly different and such antibodies (even to the same antigenic epitope) can differ in their antigen-binding affinity. How affinity variation in the interaction of FcεRI-bound IgE antibodies with antigen is interpreted into cellular responses and how the local environment may influence these responses is of interest. In this chapter, we focus on recent advances that begin to unravel how FcεRI distinguishes differences in the affinity of IgE-antigen interactions and how such discrimination along with surrounding environmental stimuli can shape the (patho) physiological response.
Collapse
Affiliation(s)
- Ryo Suzuki
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | |
Collapse
|
18
|
|
19
|
Sibilano R, Pucillo CE, Gri G. Allergic responses and aryl hydrocarbon receptor novel pathway of mast cell activation. Mol Immunol 2014; 63:69-73. [PMID: 24656327 DOI: 10.1016/j.molimm.2014.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/20/2014] [Accepted: 02/25/2014] [Indexed: 12/21/2022]
Abstract
The activation of the transcription factor aryl hydrocarbon receptor (AhR) is modulated by a wide variety of xenobiotics and ligands deriving from products of metabolism. The study of the contribution of AhR to allergic diseases has gained much interest in recent years. Here we discuss the role that environmental factors and metabolic products, particularly acting on AhR-expressing mast cells (MCs), could have in the development of local allergic/atopic response. Thus, this review will cover: a brief overview of the AhR mechanism of action in the immune system; a description of different AhR ligands and their effects to IgE-mediated MC activation in the allergic response, with particular attention to the role of IL-17; a discussion about the potential involvement of AhR in immune tolerance; and a conclusion on human diseases in which direct AhR activation of MC might have a major impact.
Collapse
Affiliation(s)
- Riccardo Sibilano
- Department of Pathology, CCSR 3255, 269 Campus Drive, Stanford, CA 94305, USA
| | - Carlo E Pucillo
- Department of Medical and Biological Sciences, University of Udine, P.le M. Kolbe 4, 33100 Udine, Italy
| | - Giorgia Gri
- Department of Medical and Biological Sciences, Section of Surgical Pathology, University of Udine, P.le S. Maria della Misericordia 15, 33100 Udine, Italy.
| |
Collapse
|
20
|
Abstract
OX40L is expressed by many cell types, including antigen presenting cells (APCs), T cells, vascular endothelial cells, mast cells (MCs), and natural killer cells. The importance of OX40L:OX40 interactions and the OX40L signaling is crucial for the homeostasis and for the modulation of the effector functions of the immune system. However, the lack of non-murine/non-IgG commercially available OX40L-triggering antibodies and the potential signal cross-contamination caused by the binding to the FcγRs co-expressed by several immune cells have limited the study of the OX40L-signaling cascade. We recently characterized the functions and described the molecular events, which follow the engagement of OX40L in MCs, by the use of the soluble OX40 molecule, able to mimic the regulatory T cell-driven engagement of MC-OX40L. This molecule enables signaling studies in MCs with any requirement for OX40-expressing cells. Using this unique reagent, we determined the modality and the extent by which the engagement of OX40L in MCs influences the IgE-dependent MC degranulation. This tool may find a potential application for signaling studies of other OX40L-expressing populations other than MCs, mainly APCs, with similar approaches we reported for the study of OX40L cascade.
Collapse
Affiliation(s)
- Riccardo Sibilano
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | |
Collapse
|
21
|
Bochner BS, Rothenberg ME, Boyce JA, Finkelman F. Advances in mechanisms of allergy and clinical immunology in 2012. J Allergy Clin Immunol 2013; 131:661-7. [PMID: 23352632 DOI: 10.1016/j.jaci.2012.12.676] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 12/11/2012] [Accepted: 12/12/2012] [Indexed: 10/27/2022]
Abstract
Manuscripts published in the "Mechanisms of allergy and clinical immunology" section of the Journal of Allergy and Clinical Immunology during 2012 enhanced our knowledge of the involvement of cytokines and other mediators in allergic disorders and described novel approaches for understanding mechanisms of allergic and immunologic diseases. Also published were articles focused on mechanisms of allergen-specific immunotherapy and the development of novel antiallergic treatments, as well as strategies to achieve tolerance to allergens. The highlights of these studies and their potential clinical implications are summarized in this review.
Collapse
Affiliation(s)
- Bruce S Bochner
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
22
|
Serra-Pages M, Olivera A, Torres R, Picado C, de Mora F, Rivera J. E-prostanoid 2 receptors dampen mast cell degranulation via cAMP/PKA-mediated suppression of IgE-dependent signaling. J Leukoc Biol 2012; 92:1155-65. [PMID: 22859831 PMCID: PMC3501892 DOI: 10.1189/jlb.0212109] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 01/11/2023] Open
Abstract
The experimental administration of PGE(2) for the treatment of asthma dampens clinical symptoms, and similar efficacy has been found in dust mite-induced hypersensitivity reactions in animal models. Here, we investigate the mechanism by which PGE(2) mediates suppression of MC degranulation. We find that the effect of PGE(2) on FcεRI-dependent MC degranulation varies from activating to suppressing, depending on the relative ratio of EP(2) to EP(3) expression on these cells with suppression evident only in cells having increased EP(2) to EP(3) expression. Consistent with a role for EP(2) in suppressing MC responses in vitro, we found that a selective EP(2) agonist, Butaprost, inhibited MC-mediated FcεRI-induced immediate hypersensitivity in a model of PCA. EP(2) engagement on MCs increased cAMP production and inhibited FcεRI-mediated calcium influx. In addition, it also decreased the extent of FcεRI-induced Fyn kinase activity, leading to decreased phosphorylation of key signaling molecules such as Gab2 and Akt. Treatment with an antagonist of cAMP or shRNA down-regulation of PKA (the principal intracellular target of cAMP) reversed the EP(2)-mediated inhibitory effect on MC degranulation and restored calcium influx and phosphorylation of Akt. Collectively, the findings demonstrate that EP(2) suppresses the Fyn-mediated signals that are central to FcεRI-dependent MC degranulation, suggesting that engagement of the EP(2) on MCs may be beneficial in dampening allergic responses.
Collapse
MESH Headings
- Alprostadil/analogs & derivatives
- Alprostadil/pharmacology
- Animals
- Calcium/metabolism
- Cell Degranulation/drug effects
- Cell Degranulation/genetics
- Cell Degranulation/immunology
- Cell Line
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dinoprostone/pharmacology
- Female
- Gene Silencing
- Humans
- Immunoglobulin E/immunology
- Immunoglobulin E/metabolism
- Mast Cells/drug effects
- Mast Cells/immunology
- Mast Cells/metabolism
- Mice
- Proto-Oncogene Proteins c-fyn/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/agonists
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/agonists
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Mariona Serra-Pages
- Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain; and
| | - Ana Olivera
- Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rosa Torres
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain; and
- Department of Pneumology and Respiratory Allergy, Hospital Clínic i Universitari de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - César Picado
- Department of Pneumology and Respiratory Allergy, Hospital Clínic i Universitari de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Fernando de Mora
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain; and
| | - Juan Rivera
- Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|