1
|
Pajulas A, Fu Y, Cheung CCL, Chu M, Cannon A, Alakhras N, Zhang J, Ulrich BJ, Nelson AS, Zhou B, Kaplan MH. Interleukin-9 promotes mast cell progenitor proliferation and CCR2-dependent mast cell migration in allergic airway inflammation. Mucosal Immunol 2023; 16:432-445. [PMID: 37172907 PMCID: PMC10482122 DOI: 10.1016/j.mucimm.2023.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Allergic asthma is a chronic lung disease characterized by airway hyperresponsiveness and cellular infiltration that is exacerbated by immunoglobulin E-dependent mast cell (MC) activation. Interleukin-9 (IL-9) promotes MC expansion during allergic inflammation but precisely how IL-9 expands tissue MCs and promotes MC function is unclear. In this report, using multiple models of allergic airway inflammation, we show that both mature MCs (mMCs) and MC progenitors (MCp) express IL-9R and respond to IL-9 during allergic inflammation. IL-9 acts on MCp in the bone marrow and lungs to enhance proliferative capacity. Furthermore, IL-9 in the lung stimulates the mobilization of CCR2+ mMC from the bone marrow and recruitment to the allergic lung. Mixed bone marrow chimeras demonstrate that these are intrinsic effects in the MCp and mMC populations. IL-9-producing T cells are both necessary and sufficient to increase MC numbers in the lung in the context of allergic inflammation. Importantly, T cell IL-9-mediated MC expansion is required for the development of antigen-induced and MC-dependent airway hyperreactivity. Collectively, these data demonstrate that T cell IL-9 induces lung MC expansion and migration by direct effects on the proliferation of MCp and the migration of mMC to mediate airway hyperreactivity.
Collapse
Affiliation(s)
- Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Yongyao Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Cherry C L Cheung
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Michelle Chu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Anthony Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Nada Alakhras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Andrew S Nelson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Baohua Zhou
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
2
|
Hesse L, Oude Elberink J, van Oosterhout AJ, Nawijn MC. Allergen immunotherapy for allergic airway diseases: Use lessons from the past to design a brighter future. Pharmacol Ther 2022; 237:108115. [DOI: 10.1016/j.pharmthera.2022.108115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 10/19/2022]
|
3
|
Lam HY, Tergaonkar V, Kumar AP, Ahn KS. Mast cells: Therapeutic targets for COVID-19 and beyond. IUBMB Life 2021; 73:1278-1292. [PMID: 34467628 PMCID: PMC8652840 DOI: 10.1002/iub.2552] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 01/22/2023]
Abstract
Mast cells (MCs) are innate immune cells that widely distribute throughout all tissues and express a variety of cell surface receptors. Upon activation, MCs can rapidly release a diverse array of preformed mediators residing within their secretory granules and newly synthesize a broad spectrum of inflammatory and immunomodulatory mediators. These unique features of MCs enable them to act as sentinels in response to rapid changes within their microenvironment. There is increasing evidence now that MCs play prominent roles in other pathophysiological processes besides allergic inflammation. In this review, we highlight the recent findings on the emerging roles of MCs in the pathogenesis of coronavirus disease-2019 (COVID-19) and discuss the potential of MCs as novel therapeutic targets for COVID-19 and other non-allergic inflammatory diseases.
Collapse
Affiliation(s)
- Hiu Yan Lam
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
- Laboratory of NF‐κB SignalingInstitute of Molecular and Cell Biology (IMCB)SingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Vinay Tergaonkar
- Laboratory of NF‐κB SignalingInstitute of Molecular and Cell Biology (IMCB)SingaporeSingapore
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Pathology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingaporeSingapore
| | - Kwang Seok Ahn
- Department of Science in Korean MedicineKyung Hee UniversitySeoulRepublic of Korea
| |
Collapse
|
4
|
Kwiatkowska D, Reich A. Role of Mast Cells in the Pathogenesis of Pruritus in Mastocytosis. Acta Derm Venereol 2021; 101:adv00583. [PMID: 34642766 DOI: 10.2340/actadv.v101.350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pruritus can be defined as an unpleasant sensation that evokes a desire to scratch and significantly impairs patients' quality of life. Pruritus is widely observed in many dermatoses, including mastocytosis, a rare disease characterized by abnormal accumulation of mast cells, which can involve skin, bone marrow, and other organs. Increasing evidence highlights the role of mast cells in neurogenic inflammation and itching. Mast cells release various pruritogenic mediators, initiating subsequent mutual communication with specific nociceptors on sensory nerve fibres. Among important mediators released by mast cells that induce pruritus, one can distinguish histamine, serotonin, proteases, as well as various cytokines. During neuronal-induced inflammation, mast cells may respond to numerous mediators, including neuropeptides, such as substance P, neurokinin A, calcitonin gene-related peptide, endothelin 1, and nerve growth factor. Currently, treatment of pruritus in mastocytosis is focused on alleviating the effects of mediators secreted by mast cells. However, a deeper understanding of the intricacies of the neurobiology of this disease could help to provide better treatment options for patients.
Collapse
Affiliation(s)
| | - Adam Reich
- Department of Dermatology, University of Rzeszow, Ul. Szopena 2, PL-35-055 Rzeszów, Poland.
| |
Collapse
|
5
|
Luo Y, Fernandez Vallone V, He J, Frischbutter S, Kolkhir P, Moñino-Romero S, Stachelscheid H, Streu-Haddad V, Maurer M, Siebenhaar F, Scheffel J. A novel approach for studying mast cell-driven disorders: Mast cells derived from induced pluripotent stem cells. J Allergy Clin Immunol 2021; 149:1060-1068.e4. [PMID: 34371081 DOI: 10.1016/j.jaci.2021.07.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/08/2021] [Accepted: 07/21/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mast cells (MCs) are considered the main effectors in allergic reactions and well known for their contribution to the pathogenesis of various inflammatory diseases, urticaria, and mastocytosis. To study their functions in vitro, human primary MCs are isolated directly from several tissues or differentiated from hematopoietic progenitors. However, these techniques bear several disadvantages and challenges including low proliferation capacity, donor-dependent heterogeneity, and the lack of a continuous cell source. OBJECTIVE To address this, we developed a novel strategy for the rapid and efficient differentiation of MCs from human-induced pluripotent stem cells (hiPSCs). METHODS A 4-step protocol for the generation of hiPSC-derived MCs, based on the use of 3 hiPSC lines, was established and validated by comparison with human skin MCs and peripheral hematopoietic stem cell-derived MCs. RESULTS hiPSC-MCs share phenotypic and functional characteristics of human skin MCs and peripheral hematopoietic stem cell-derived MCs. They display stable expression of the MC-associated receptors CD117, FcεRIα, and Mas-related G protein-coupled receptor X2 and degranulate in response to IgE/anti-IgE and substance P. CONCLUSIONS This novel hiPSC-based approach provides a sustainable and homogeneous source for a rapid and highly productive generation of phenotypically mature, functional MCs, and its principle allows for the investigation of disease- and patient-specific MC populations.
Collapse
Affiliation(s)
- Yanyan Luo
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Valeria Fernandez Vallone
- Charité-BIH Centrum Therapy and Research, BIH Stem Cell Core Facility, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jiajun He
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, China; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Stefan Frischbutter
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Pavel Kolkhir
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; I.M. Sechenov First Moscow State Medical University (Sechenov University), Division of Immune-mediated Skin Diseases, Moscow, Russia; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Sherezade Moñino-Romero
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Harald Stachelscheid
- Charité-BIH Centrum Therapy and Research, BIH Stem Cell Core Facility, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Viktoria Streu-Haddad
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Marcus Maurer
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| | - Frank Siebenhaar
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Jörg Scheffel
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| |
Collapse
|
6
|
Panda D, Chatterjee G, Khanka T, Ghogale S, Badrinath Y, Deshpande N, Sardana R, Chaturvedi A, Rajpal S, Shetty D, Patkar NV, Gujral S, Subramanian PG, Tembhare PR. Mast cell differentiation of leukemic blasts in diverse myeloid neoplasms: A potential pre-myelomastocytic leukemia condition. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2021; 100:331-344. [PMID: 32738100 DOI: 10.1002/cyto.b.21938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/20/2020] [Accepted: 06/30/2020] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Myeloid neoplasm with blasts showing mast cell (MC)-differentiation and MC-component less than 10% of all nucleated cells but not fulfilling the criteria for systemic mastocytosis with associated hematological neoplasm (SM-AHN) or myelomastocytic leukemia (MML) has not been described in the literature. Herein, we report a study of diverse myeloid malignancies with blasts showing MC-differentiation but not meeting the criteria for SM-AHN or MML. We also evaluated the utility of flow-cytometric immunophenotyping (FCI) in the characterization of immature-MCs (iMCs). METHODS We identified nine patients of myeloid neoplasms and studied their morphological, FCI, immunohistochemistry, cytogenetic and molecular characteristics. We also compared the immunophenotypic features of MCs from patient samples with control samples. RESULTS The study included patients with newly-diagnosed acute myeloid leukemia (n = 4), chronic myelomonocytic leukemia (n = 1), and chronic myeloid leukemia on follow-up (n = 4) showing MC differentiation in leukemic-blasts. These patients had mildly increased MCs (range, 0.5%-3%) in bone-marrow morphology, including immature-forms and did not meet the criteria for either SM-AHN or MML. On FCI, iMCs were positive for bright-CD117, heterogeneous-CD34, dim-to-negative-HLADR, and moderate-CD203c expression. Expression-levels of CD123 and CD38 were higher (p < 0.001) but CD33 and CD45 were lower in iMCs compared to mature-MC from control samples (p = 0.019 and p = 0.0037). CONCLUSION We reported a rare finding of MC differentiation of leukemic blasts in diverse myeloid neoplasms and proposed it as a potential pre-myelomastocytic leukemia condition. We described the distinct immunophenotypic signature of immature-MCs using commonly used markers and highlighted the utility of FCI for the diagnosis of this entity.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Antigens, CD/metabolism
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Cell Differentiation/physiology
- Child
- Female
- Hematologic Neoplasms/metabolism
- Hematologic Neoplasms/pathology
- Humans
- Immunophenotyping/methods
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelomonocytic, Chronic/metabolism
- Leukemia, Myelomonocytic, Chronic/pathology
- Male
- Mast Cells/metabolism
- Mast Cells/pathology
- Mastocytosis, Systemic/metabolism
- Mastocytosis, Systemic/pathology
- Middle Aged
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Primary Myelofibrosis/metabolism
- Primary Myelofibrosis/pathology
Collapse
Affiliation(s)
- Devasis Panda
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Twinkle Khanka
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Yajamanam Badrinath
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Rohan Sardana
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Anumeha Chaturvedi
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Sweta Rajpal
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Sumeet Gujral
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
- Department of Pathology, Tata Memorial Center, Mumbai, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| | - Prashant R Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial Center, HBNI University, Navi Mumbai, India
| |
Collapse
|
7
|
Domagala M, Laplagne C, Leveque E, Laurent C, Fournié JJ, Espinosa E, Poupot M. Cancer Cells Resistance Shaping by Tumor Infiltrating Myeloid Cells. Cancers (Basel) 2021; 13:E165. [PMID: 33418996 PMCID: PMC7825276 DOI: 10.3390/cancers13020165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Interactions between malignant cells and neighboring stromal and immune cells profoundly shape cancer progression. New forms of therapies targeting these cells have revolutionized the treatment of cancer. However, in order to specifically address each population, it was essential to identify and understand their individual roles in interaction between malignant cells, and the formation of the tumor microenvironment (TME). In this review, we focus on the myeloid cell compartment, a prominent, and heterogeneous group populating TME, which can initially exert an anti-tumoral effect, but with time actively participate in disease progression. Macrophages, dendritic cells, neutrophils, myeloid-derived suppressor cells, mast cells, eosinophils, and basophils act alone or in concert to shape tumor cells resistance through cellular interaction and/or release of soluble factors favoring survival, proliferation, and migration of tumor cells, but also immune-escape and therapy resistance.
Collapse
Affiliation(s)
- Marcin Domagala
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Chloé Laplagne
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Edouard Leveque
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Camille Laurent
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
- IUCT-O, 31000 Toulouse, France
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Eric Espinosa
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse, Inserm UMR1037, 31037 Toulouse, France; (M.D.); (C.L.); (E.L.); (C.L.); (J.-J.F.); (E.E.)
- Université Toulouse III Paul-Sabatier, 31400 Toulouse, France
- ERL 5294 CNRS, 31037 Toulouse, France
| |
Collapse
|
8
|
Peng H, Liao B, Zhu X, Liu Y, Jiang Y, Wu S. CCR3-shRNA promotes apoptosis and inhibits chemotaxis and degranulation of mouse mast cells. Exp Ther Med 2020; 20:1030-1038. [PMID: 32742345 PMCID: PMC7388334 DOI: 10.3892/etm.2020.8737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 03/17/2020] [Indexed: 11/09/2022] Open
Abstract
Mast cells (MCs) are the major effector cells of allergic rhinitis (AR). The present study aimed to investigate the effects of C-C chemokine receptor type 3 (CCR3) on the proliferation, apoptosis, chemotaxis and activated degranulation of mouse MCs. Mouse bone marrow-derived MCs were cultured in vitro, purified and identified using toluidine blue staining and flow cytometry. Three different CCR3-short hairpin (shRNA) lentiviral vectors were constructed and transfected into MCs, and the mRNA and protein expression levels of CCR3 were assessed by reverse transcription-quantitative PCR and western blotting. Proliferation and apoptosis of the MCs were measured using Cell Counting kit-8 (CCK-8) assays and flow cytometry, respectively. MC chemotaxis was assessed by Transwell assay and quantified using flow cytometry. The activation of MC degranulation was examined using ELISAs. The results demonstrated that MCs were appropriately isolated, and identified that CCR3-shRNA2 presented the higher knockdown effect among the three shRNAs tested. Following 96 h of transfection, the results of CCK-8 and flow cytometry assays demonstrated that CCR3-shRNA2 inhibited MC proliferation and promoted MC apoptosis. The results from the Transwell assay indicated that CCR3-shRNA2 restrained MC chemotaxis, whereas ELISA results demonstrated that CCR3-shRNA2 suppressed MC degranulation. In conclusion, CCR3-shRNA2 effectively downregulated CCR3 mRNA and protein expression levels in mouse MCs. In addition, CCR3-shRNA2 promoted MC apoptosis and suppressed the proliferation, chemotaxis and degranulation of mouse MCs, suggesting that CCR3-shRNA2 may serve as a therapeutic tool for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Haisen Peng
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bing Liao
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xinhua Zhu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuehui Liu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yinli Jiang
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuhong Wu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
9
|
Drissen R, Thongjuea S, Theilgaard-Mönch K, Nerlov C. Identification of two distinct pathways of human myelopoiesis. Sci Immunol 2020; 4:4/35/eaau7148. [PMID: 31126997 DOI: 10.1126/sciimmunol.aau7148] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/27/2019] [Indexed: 12/12/2022]
Abstract
Human myelopoiesis has been proposed to occur through oligopotent common myeloid progenitor (CMP) and lymphoid-primed multipotent progenitor (LMPP) populations. However, other studies have proposed direct commitment of multipotent cells to unilineage fates, without specific intermediary lineage cosegregation patterns. We here show that distinct human myeloid progenitor populations generate the neutrophil/monocyte and mast cell/basophil/eosinophil lineages as previously shown in mouse. Moreover, we find that neutrophil/monocyte potential selectively cosegregates with lymphoid lineage and mast cell/basophil/eosinophil potentials with megakaryocyte/erythroid potential early during lineage commitment. Furthermore, after this initial commitment step, mast cell/basophil/eosinophil and megakaryocyte/erythroid potentials colocalize at the single-cell level in restricted oligopotent progenitors. These results show that human myeloid lineages are generated through two distinct cellular pathways defined by complementary oligopotent cell populations.
Collapse
Affiliation(s)
- Roy Drissen
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| | - Supat Thongjuea
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Kim Theilgaard-Mönch
- Department of Hematology and Finsen Laboratory, National University Hospital, Denmark.,Biotech Research and Innovation Centre and Program for Translational Hematology - The Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Nerlov
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
10
|
Wagner N, Staubach P. Mastocytosis - pathogenesis, clinical manifestation and treatment. J Dtsch Dermatol Ges 2019; 16:42-57. [PMID: 29314691 DOI: 10.1111/ddg.13418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/14/2017] [Indexed: 01/08/2023]
Abstract
The term mastocytosis designates a group of rare disorders characterized by typical skin lesions, frequently associated episodes of anaphylaxis, and clinical symptoms related to the release of various mediators. Dermatologists/allergists are frequently the first to establish the diagnosis. The condition is based on clonal mast cell proliferation, usually in the skin or bone marrow and only rarely in the gastrointestinal tract or other tissues. In general, mastocytosis has a good prognosis in terms of life expectancy. Rare variants - including mast cell leukemia, aggressive mastocytosis, and the exceedingly rare mast cell sarcoma - require cytoreductive therapy. In cases associated with hematological neoplasms, the prognosis depends on the underlying hematologic disorder.
Collapse
Affiliation(s)
- Nicola Wagner
- Department of Dermatology, University Medical Center, Erlangen, Germany
| | - Petra Staubach
- Department of Dermatology, University Medical Center, Mainz, Germany
| |
Collapse
|
11
|
Robida PA, Puzzovio PG, Pahima H, Levi-Schaffer F, Bochner BS. Human eosinophils and mast cells: Birds of a feather flock together. Immunol Rev 2019; 282:151-167. [PMID: 29431215 DOI: 10.1111/imr.12638] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
While the origin of the phrase "birds of a feather flock together" is unclear, it has been in use for centuries and is typically employed to describe the phenomenon that people with similar tastes or interests tend to seek each other out and congregate together. In this review, we have co-opted this phrase to compare innate immune cells of related origin, the eosinophil and mast cell, because they very often accumulate together in tissue sites under both homeostatic and inflammatory conditions. To highlight overlapping yet distinct features, their hematopoietic development, cell surface phenotype, mediator release profiles and roles in diseases have been compared and contrasted. What emerges is a sense that these two cell types often interact with each other and their tissue environment to provide synergistic contributions to a variety of normal and pathologic immune responses.
Collapse
Affiliation(s)
- Piper A Robida
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Pahima
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Gross AR, Theoharides TC. Chondroitin sulfate inhibits secretion of TNF and CXCL8 from human mast cells stimulated by IL-33. Biofactors 2019; 45:49-61. [PMID: 30521103 DOI: 10.1002/biof.1464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/14/2018] [Accepted: 08/21/2018] [Indexed: 01/19/2023]
Abstract
Glycosaminoglycans (GAGs) are linear, highly negatively charged carbohydrate chains present in connective tissues. Chondroitin sulfate (CS) and heparin (Hep) are also found in the numerous secretory granules of mast cells (MC), tissue immune cells involved in allergic and inflammatory reactions. CS and Hep may inhibit secretion of histamine from rat connective tissue MC, but their effect on human MC remains unknown. Human LAD2 MC were pre-incubated with CS, Hep, or dermatan sulfate (DS) before being stimulated by either the peptide substance P (SP, 2 μM) or the cytokine IL-33 (10 ng/mL). Preincubation with CS had no effect on MC degranulation stimulated by SP, but inhibited TNF (60%) and CXCL8 (45%) secretion from LAD2 cells stimulated by IL-33. Fluorescein-conjugated CS (CS-F) was internalized by LAD2 cells only at 37 °C, but not 4 °C, indicating it occurred by endocytosis. DS and Hep inhibited IL-33-stimulated secretion of TNF and CXCL8 to a similar extent as CS. None of the GAGs tested inhibited IL-33-stimulated gene expression of either TNF or CXCL8. There was no effect of CS on ionomycin-stimulated calcium influx. There was also no effect of CS on surface expression of the IL-33 receptor, ST2. Neutralization of the hyaluronan receptor CD44 did not affect the internalization of CS-F. The findings in this article show that CS inhibits secretion of TNF and CXCL8 from human cultured MC stimulated by IL-33. CS could be formulated for systemic or topical treatment of allergic or inflammatory diseases, such as atopic dermatitis, cutaneous mastocytosis, and psoriasis. © 2018 BioFactors, 45(1):49-61, 2019.
Collapse
Affiliation(s)
- Amanda R Gross
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
13
|
Vorup-Jensen T, Jensen RK. Structural Immunology of Complement Receptors 3 and 4. Front Immunol 2018; 9:2716. [PMID: 30534123 PMCID: PMC6275225 DOI: 10.3389/fimmu.2018.02716] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/05/2018] [Indexed: 01/10/2023] Open
Abstract
Complement receptors (CR) 3 and 4 belong to the family of beta-2 (CD18) integrins. CR3 and CR4 are often co-expressed in the myeloid subsets of leukocytes, but they are also found in NK cells and activated T and B lymphocytes. The heterodimeric ectodomain undergoes considerable conformational change in order to switch the receptor from a structurally bent, ligand-binding in-active state into an extended, ligand-binding active state. CR3 binds the C3d fragment of C3 in a way permitting CR2 also to bind concomitantly. This enables a hand-over of complement-opsonized antigens from the cell surface of CR3-expressing macrophages to the CR2-expressing B lymphocytes, in consequence acting as an antigen presentation mechanism. As a more enigmatic part of their functions, both CR3 and CR4 bind several structurally unrelated proteins, engineered peptides, and glycosaminoglycans. No consensus motif in the proteinaceous ligands has been established. Yet, the experimental evidence clearly suggest that the ligands are primarily, if not entirely, recognized by a single site within the receptors, namely the metal-ion dependent adhesion site (MIDAS). Comparison of some recent identified ligands points to CR3 as inclined to bind positively charged species, while CR4, by contrast, binds strongly negative-charged species, in both cases with the critical involvement of deprotonated, acidic groups as ligands for the Mg2+ ion in the MIDAS. These properties place CR3 and CR4 firmly within the realm of modern molecular medicine in several ways. The expression of CR3 and CR4 in NK cells was recently demonstrated to enable complement-dependent cell cytotoxicity toward antibody-coated cancer cells as part of biological therapy, constituting a significant part of the efficacy of such treatment. With the flexible principles of ligand recognition, it is also possible to propose a response of CR3 and CR4 to existing medicines thereby opening a possibility of drug repurposing to influence the function of these receptors. Here, from advances in the structural and cellular immunology of CR3 and CR4, we review insights on their biochemistry and functions in the immune system.
Collapse
Affiliation(s)
- Thomas Vorup-Jensen
- Biophysical Immunology Laboratory, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Rasmus Kjeldsen Jensen
- Department of Molecular Biology and Genetics-Structural Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Substance P and IL-33 administered together stimulate a marked secretion of IL-1β from human mast cells, inhibited by methoxyluteolin. Proc Natl Acad Sci U S A 2018; 115:E9381-E9390. [PMID: 30232261 DOI: 10.1073/pnas.1810133115] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mast cells are critical for allergic and inflammatory responses in which the peptide substance P (SP) and the cytokine IL-33 are involved. SP (0.01-1 μM) administered together with IL-33 (30 ng/mL) to human cultured LAD2 mast cells stimulates a marked increase (P < 0.0001) in secretion of the proinflammatory cytokine IL-1β. Preincubation of LAD2 (30 min) with the SP receptor (NK-1) antagonists L-733,060 (10 μM) or CP-96345 (10 µM) inhibits (P < 0.001) secretion of IL-1β stimulated by either SP (1 μM) or SP together with IL-33 (30 ng/mL). Surprisingly, secretion of IL-1β stimulated by IL-33 is inhibited (P < 0.001) by each NK-1 antagonist. Preincubation with an antibody against the IL-33 receptor ST2 inhibits (P < 0.0001) secretion of IL-1β stimulated either by IL-33 or together with SP. The combination of SP (1 μM) with IL-33 (30 ng/mL) increases IL-1β gene expression by 90-fold in LAD2 cells and by 200-fold in primary cultured mast cells from human umbilical cord blood. The combination of SP and IL-33 increases intracellular levels of IL-1β in LAD2 by 100-fold and gene expression of IL-1β and procaspase-1 by fivefold and pro-IL-1β by twofold. Active caspase-1 is present even in unstimulated cells and is detected extracellularly. Preincubation of LAD2 cells with the natural flavonoid methoxyluteolin (1-100 mM) inhibits (P < 0.0001) secretion and gene expression of IL-1β, procaspase-1, and pro-IL-1β. Mast cell secretion of IL-1β in response to SP and IL-33 reveals targets for the development of antiinflammatory therapies.
Collapse
|
15
|
Yu T, He Z, Yang M, Song J, Ma C, Ma S, Feng J, Liu B, Wang X, Wei Z, Li J. The development of methods for primary mast cells in vitro and ex vivo: An historical review. Exp Cell Res 2018; 369:179-186. [PMID: 29842878 DOI: 10.1016/j.yexcr.2018.05.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
Abstract
Mast cells (MCs) are tissue-based stationary effector cells that form the immune system's first-line defense against various challenges. They are developed from the bone marrow-derived progenitors to complete their differentiation and maturation in the tissues where they eventually establish residence. MCs have been implicated in many diseases, such as allergy, parasitic infection, and neoplastic disorders. Immortalized MC lines, such as RBL-2H3, HMC-1, and LAD-2, are useful for investigating the biological functions of MC only to some extents due to the restriction of degranulation evaluation, in vivo injection and other factors. Over the past few decades, technologies for acquiring primarily MCs have been continually optimized, and novel protocols have been proposed. However, no relevant publications have analyzed and summarized these techniques. In this review, the classical approaches for extracting MCs are generalized, and new methods with potential values are introduced. We also evaluate the advantages and applicability of diverse MC models. Since MCs exhibit substantial plasticity and functional diversity due to different origins, it is both necessary and urgent to select a reliable and suitable source of MCs for a particular study.
Collapse
Affiliation(s)
- Tianyu Yu
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China; Department of General Surgery, Shanghai Clinical Medical College, Anhui Medical University, China
| | - Zhigang He
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China
| | - Muqing Yang
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China
| | - Jian Song
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China
| | - Cheng Ma
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China; Department of General Surgery, Shanghai Clinical Medical College, Anhui Medical University, China
| | - Sunqiang Ma
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China
| | - Junlan Feng
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China
| | - Bin Liu
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China; Department of General Surgery, Shanghai Clinical Medical College, Anhui Medical University, China
| | - Xiaodong Wang
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China; Department of General Surgery, Shanghai Clinical Medical College, Anhui Medical University, China
| | - Zhubo Wei
- Cancer Center, Houston Methodist Research Institute, United States.
| | - Jiyu Li
- Department of General Surgery, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, China; Department of General Surgery, Shanghai Clinical Medical College, Anhui Medical University, China.
| |
Collapse
|
16
|
Mast Cell, the Neglected Member of the Tumor Microenvironment: Role in Breast Cancer. J Immunol Res 2018; 2018:2584243. [PMID: 29651440 PMCID: PMC5832101 DOI: 10.1155/2018/2584243] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/08/2017] [Accepted: 11/26/2017] [Indexed: 02/06/2023] Open
Abstract
Mast cells are unique tissue-resident immune cells that secrete a diverse array of biologically active compounds that can stimulate, modulate, or suppress the immune response. Although mounting evidence supports that mast cells are consistently infiltrating tumors, their role as either a driving or an opposite force for cancer progression is still controversial. Particularly, in breast cancer, their function is still under discussion. While some studies have shown a protective role, recent evidence indicates that mast cells enhance blood and lymphatic vessel formation. Interestingly, one of the most important components of the mast cell cargo, the serine protease tryptase, is a potent angiogenic factor, and elevated serum tryptase levels correlate with bad prognosis in breast cancer patients. Likewise, histamine is known to induce tumor cell proliferation and tumor growth. In agreement, mast cell depletion reduces the size of mammary tumors and metastasis in murine models that spontaneously develop breast cancer. In this review, we will discuss the evidence supporting protumoral and antitumoral roles of mast cells, emphasizing recent findings placing mast cells as important drivers of tumor progression, as well as the potential use of these cells or their mediators as therapeutic targets.
Collapse
|
17
|
Wagner N, Staubach P. Mastozytose - Pathogenese, Klinik und Therapie. J Dtsch Dermatol Ges 2018; 16:42-59. [PMID: 29314684 DOI: 10.1111/ddg.13418_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/14/2017] [Indexed: 01/08/2023]
|
18
|
Kovanen PT, Bot I. Mast cells in atherosclerotic cardiovascular disease – Activators and actions. Eur J Pharmacol 2017; 816:37-46. [DOI: 10.1016/j.ejphar.2017.10.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
19
|
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, Marck P, Branchereau M, Le Gall S, Renault N, Guerin C, Ranek MJ, Kervadec A, Danelli L, Gautier G, Blank U, Launay P, Camerer E, Bruneval P, Menasche P, Heymes C, Luche E, Casteilla L, Cousin B, Rodewald HR, Kass DA, Silvestre JS. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J Exp Med 2017; 213:1353-74. [PMID: 27353089 PMCID: PMC4925026 DOI: 10.1084/jem.20160081] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/12/2016] [Indexed: 11/24/2022] Open
Abstract
Ngkelo et al. use a mast cell–deficient mouse model to reveal a protective role of mast cells in myocardial infarction, through regulation of the cardiac contractile machinery. Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit–independent MC-deficient (Cpa3Cre/+) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca2+ desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force–Ca2+ interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.
Collapse
Affiliation(s)
- Anta Ngkelo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Adèle Richart
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Jonathan A Kirk
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Philippe Bonnin
- INSERM, U965, Hôpital Lariboisière-Fernand-Widal, Assistance Publique Hôpitaux de Paris, F-75010 Paris, France
| | - Jose Vilar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Mathilde Lemitre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Pauline Marck
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Maxime Branchereau
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Sylvain Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Nisa Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Coralie Guerin
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Mark J Ranek
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Anaïs Kervadec
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Luca Danelli
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Gregory Gautier
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Ulrich Blank
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Pierre Launay
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Patrick Bruneval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Philippe Menasche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Christophe Heymes
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Elodie Luche
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Béatrice Cousin
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| |
Collapse
|
20
|
Patel AB, Theoharides TC. Methoxyluteolin Inhibits Neuropeptide-stimulated Proinflammatory Mediator Release via mTOR Activation from Human Mast Cells. J Pharmacol Exp Ther 2017; 361:462-471. [PMID: 28404689 DOI: 10.1124/jpet.117.240564] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/27/2017] [Indexed: 03/08/2025] Open
Abstract
Mast cells (MCs) are critical for allergic reactions but are also important in inflammatory processes. Stimulation by neuropeptides, such as substance P (SP) and neurotensin (NT), leads to release of preformed molecules stored in numerous MC secretory granules and newly synthesized proinflammatory mediators, including tumor necrosis factor, C-X-C motif chemokine ligand 8, and vascular endothelial growth factor. Here, we investigate the role of mammalian target of rapamycin (mTOR) signaling in the stimulation of cultured human LAD2 MCs by NT or SP, as well as the inhibitory effect of the natural flavonoids 3',4',5,7-tetrahydroxyflavone (luteolin) and its novel structural analog 3',4',5,7-tetramethoxyflavone (methoxyluteolin). Stimulation by NT (10 μM) or SP (1 μM) increases (P < 0.0001) gene expression (after 6 hours) and release (after 24 hours) of tumor necrosis factor, C-X-C motif chemokine ligand 8, and vascular endothelial growth factor. This occurs via activation of both mTOR complexes, as denoted by the increased phosphorylated (p) protein levels (P < 0.0001) of the downstream mTORC1 substrate pp70S6KThr389 and mTORC2 component pmTORSer2448. Pretreatment of human MCs using the mTORC1 inhibitor rapamycin, the mTORC1/mTORC2 inhibitor Torin1, or the two flavonoids decreases both gene expression and release (P < 0.0001) of all three mediators. Methoxyluteolin is a more potent human MC inhibitor than luteolin or Torin1, implicating other MC protein targets in addition to the mTOR complex. These findings indicate that mTOR is partially involved in the neuropeptide stimulation of MCs, but the novel flavonoid methoxyluteolin inhibits the response entirely, suggesting that it may be developed for treatment of allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Arti B Patel
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts (A.B.P. and T.C.T.); Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts (A.B.P.); Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| | - Theoharis C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts (A.B.P. and T.C.T.); Graduate Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts (A.B.P.); Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts (T.C.T.)
| |
Collapse
|
21
|
Naccache A, Louiset E, Duparc C, Laquerrière A, Patrier S, Renouf S, Gomez-Sanchez CE, Mukai K, Lefebvre H, Castanet M. Temporal and spatial distribution of mast cells and steroidogenic enzymes in the human fetal adrenal. Mol Cell Endocrinol 2016; 434:69-80. [PMID: 27302892 DOI: 10.1016/j.mce.2016.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/03/2016] [Accepted: 06/10/2016] [Indexed: 11/16/2022]
Abstract
Mast cells are present in the human adult adrenal with a potential role in the regulation of aldosterone secretion in both normal cortex and adrenocortical adenomas. We have investigated the human developing adrenal gland for the presence of mast cells in parallel with steroidogenic enzymes profile and serotonin signaling pathway. RT-QPCR and immunohistochemical studies were performed on adrenals at 16-41 weeks of gestation (WG). Tryptase-immunopositive mast cells were found from 18 WG in the adrenal subcapsular layer, close to 3βHSD- and CYP11B2-immunoreactive cells, firstly detected at 18 and 24 WG, respectively. Tryptophan hydroxylase and serotonin receptor type 4 expression increased at 30 WG before the CYP11B2 expression surge. In addition, HDL and LDL cholesterol receptors were expressed in the subcapsular zone from 24 WG. Altogether, our findings suggest the implication of mast cells and serotonin in the establishment of the mineralocorticoid synthesizing pathway during fetal adrenal development.
Collapse
Affiliation(s)
- Alexandre Naccache
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| | - Estelle Louiset
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| | - Céline Duparc
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| | | | - Sophie Patrier
- Pathology Laboratory, University Hospital of Rouen, Rouen, France.
| | - Sylvie Renouf
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| | - Celso E Gomez-Sanchez
- Endocrine Section, Department of Medicine, G.V. (Sonny) Montgomery VA Medical Center and University of Mississippi Medical Center, Jackson, MS, USA.
| | - Kuniaki Mukai
- Department of Biochemistry, School of Medicine, Keio University, Tokyo, 160-8582, Japan.
| | - Hervé Lefebvre
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| | - Mireille Castanet
- INSERM U982, Laboratory of Differentiation & Neuronal and Neuroendocrine Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France.
| |
Collapse
|
22
|
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
|