1
|
Elieh-Ali-Komi D, Shafaghat F, Alipoor SD, Kazemi T, Atiakshin D, Pyatilova P, Maurer M. Immunomodulatory Significance of Mast Cell Exosomes (MC-EXOs) in Immune Response Coordination. Clin Rev Allergy Immunol 2025; 68:20. [PMID: 39976807 PMCID: PMC11842441 DOI: 10.1007/s12016-025-09033-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 02/23/2025]
Abstract
Mast cells (MCs) communicate with other cells by direct cell-to-cell interaction, secreting mediators, and releasing exosomes (EXOs). MC-exosomes (MC-EXOs) contain proteins, lipids, mRNAs, and noncoding RNAs (ncRNAs), exhibit typical EXO markers such as heat shock proteins, tetraspanins, tumor susceptibility gene 101 protein (TSG101), and ALG-2-interacting protein X (ALIX), and are released constitutively or following MC degranulation. MC-EXOs also have signature MC markers like FcεRI and KIT (CD117), which allows for their identification and comparison with other EXO populations. Following their release, MC-EXOs may interact with the recipient cell(s) directly or be internalized and then release their protein and nucleic acid content. This may contribute to the regulation of immune responses and other biological processes and reprogramming of recipient cells. MC-EXO proteins may integrate and become a functional part of the recipient cell membrane. The mRNA transferred by MC-EXOs is functional and the transfer of exosomal RNA to other MCs results in the expression of donor MC proteins in the recipient MCs. Moreover, MCs may function as the recipients of EXOs that are released by other non-immune and immune cells, altering the secretome of MCs. In this review, we focus on how MC-EXOs modulate the biology of other cells and vice versa; and we highlight the role of MC-EXOs in the pathogenesis of allergic and non-allergic diseases.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany.
| | - Farzaneh Shafaghat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shamila D Alipoor
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dmitrii Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-Structural Analysis Innovative Technologies, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St, 117198, Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036, Voronezh, Russia
| | - Polina Pyatilova
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| |
Collapse
|
2
|
Pérez-Pons A, Henriques A, Contreras Sanfeliciano T, Jara-Acevedo M, Navarro-Navarro P, García-Montero AC, Álvarez-Twose I, Lecrevisse Q, Fluxa R, Sánchez-Muñoz L, Caldas C, Pozo J, González-López Ó, Pérez-Andrés M, Mayado A, Orfao A. Altered B-cell, plasma cell, and antibody immune profiles in blood of patients with systemic mastocytosis. J Allergy Clin Immunol 2025; 155:628-639. [PMID: 39423877 DOI: 10.1016/j.jaci.2024.10.005] [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: 04/30/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a heterogeneous disease characterized by an expansion of KIT-mutated constitutively activated mast cells (MCs) that release MC mediators, which might act on the tumor microenvironment including other immune cells. OBJECTIVE To investigate the blood distribution of B-cell, plasma cell (PC), and antibody isotype compartments in patients with SM. METHODS We used spectral flow cytometry and the EuroFlow Immunomonitoring panel and Lymphocyte Screening Tube to quantify B cells, PCs, and their subsets in blood of 108 patients with SM (35 bone marrow mastocytosis [BMM] cases, 64 indolent SM [ISM] cases, 9 aggressive SM [ASM] cases) versus 117 age-matched healthy donors and paired bone marrow samples of 31 patients with SM versus 17 controls, respectively. In parallel, IgM, IgD, IgG, IgA, and IgE plasma levels were measured. RESULTS Compared with healthy donors, patients with SM showed increased immature B-cell production in bone marrow (P = .003) associated with greater release of pre-germinal center immature (P < .001) and naive CD5+ B lymphocytes (P < .001) to blood, but a pronounced decrease in PC counts of all different IgH isotypes and subclasses (P ≤ .001) together with overall increased IgM (P = .001) and IgD (P < .001) plasma levels. Different immune profiles were found per diagnostic subtype of disease with progressively greater counts in blood of immature B lymphocytes together with decreased IgMD+, IgG2+, IgA1+, and IgA2+ memory B cells (P ≤ .032) and elevated IgM (P = .017) plasma levels in cases of ASM, increased IgM (P = .001) and IgD (P = .001) plasma levels in ISM cases, and exacerbated IgE (P < .001) with decreased IgG (P = .008) plasma levels in BMM cases. CONCLUSIONS Our results reveal a significant dysregulation of the B-cell and PC compartments in blood of patients with SM, consistent with distinctly altered antibody isotype profiles in plasma of patients with BMM versus ISM versus ASM.
Collapse
Affiliation(s)
- Alba Pérez-Pons
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Ana Henriques
- Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | | | - María Jara-Acevedo
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Sequencing Service, University of Salamanca, Salamanca, Spain
| | - Paula Navarro-Navarro
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Sequencing Service, University of Salamanca, Salamanca, Spain
| | - Andrés C García-Montero
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Iván Álvarez-Twose
- Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | - Quentin Lecrevisse
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain
| | | | - Laura Sánchez-Muñoz
- Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | - Carolina Caldas
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Julio Pozo
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain
| | - Óscar González-López
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Martín Pérez-Andrés
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain
| | - Andrea Mayado
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain.
| |
Collapse
|
3
|
Hao G, Han S, Xiao Z, Shen J, Zhao Y, Hao Q. Synovial mast cells and osteoarthritis: Current understandings and future perspectives. Heliyon 2024; 10:e41003. [PMID: 39720069 PMCID: PMC11665477 DOI: 10.1016/j.heliyon.2024.e41003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disease worldwide that significantly impacts the quality of life of individuals, particularly those in middle-aged and elderly populations. OA was initially considered as non-inflammatory arthritis, but recent studies have identified a substantial number of immune responses in OA, leading to the recognition of inflammation as a key factor in its pathogenesis. An increasing number of studies have found that mast cell (MC) and MC-secreted inflammatory mediators and cytokines are notably increased in the synovial fluid of OA patients, indicating a potential association between MCs and the onset and progression of synovial inflammation. The present review aims to summarize the significance and mechanism of MCs in the pathogenesis of OA. Meanwhile, we also discuss the clinical potential of using MCs as therapeutic target for OA therapy. Modulating the activities of MCs or the mediators of MCs in the synovial fluid inflammatory microenvironment will be promising new options for the treatment of OA.
Collapse
Affiliation(s)
- Guanghui Hao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shanqian Han
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Qi Hao
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Third People's Hospital of Longmatan District, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Song R, Zhang H, Liang Z. Research progress in OX40/OX40L in allergic diseases. Int Forum Allergy Rhinol 2024; 14:1921-1928. [PMID: 39404736 DOI: 10.1002/alr.23469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/24/2024] [Accepted: 10/01/2024] [Indexed: 12/13/2024]
Abstract
OX40/OX40L are costimulatory molecules in the tumor necrosis factor superfamily. Numerous studies have shown that OX40/OX40L are involved in immune regulation, especially in the proliferation and differentiation of T cells and the generation of memory T cells, which play important roles in allergic diseases. In recent years, the use of OX40/OX40L as therapeutic targets for treating T-cell-mediated diseases has attracted the interest of scholars. This paper reviews the role of OX40/OX40L in allergic diseases and the progress in clinical treatments targeting this signaling pathway.
Collapse
Affiliation(s)
- Rongrong Song
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Huanlei Zhang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Zhuoping Liang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
5
|
Zhang H, Li Q, Li Y, Guan J, Li K, Chen Y. Effects of Huang-Lian-Jie-Du decoction on improving skin barrier function and modulating T helper cell differentiation in 1-chloro-2,4-dinitrobenzene-induced atopic dermatitis mice. Front Pharmacol 2024; 15:1487402. [PMID: 39640480 PMCID: PMC11618541 DOI: 10.3389/fphar.2024.1487402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
Background: Atopic dermatitis (AD) is among the most frequently encountered skin diseases, bothering a considerable number of patients. Today, corticosteroids and antihistamines are among the numerous drugs applied for the therapy of AD. However, lengthy use of them contributes to side effects, such as physiological changes in skin. As an alternative and supplementary therapy, traditional Chinese medicine has become a trend for AD treatment. Huang-Lian-Jie-Du decoction (HLJDD), a renowned herbal formula has been employed to treat inflammatory diseases such as AD. However, its role in regulating immunity in AD remains unclear. The object of this study was to elucidate the efficacy of HLJDD and reveal the implicit mechanism from an immunological perspective in AD-like mice. Methods: In brief, 1-chloro-2,4-dinitrobenzene (DNCB) for the sensitization phase (1% DNCB) and stimulation phase (1.5% DNCB) were applied for BALB/c mice. HLJDD and dexamethasone (DXMS) were administered orally to the mice. Mice skin and spleens were collected to evaluate the efficacy of HLJDD. 16S rRNA sequencing was applied to evaluate the commensal microbiota changes in skin and fecal. In vitro, spleen CD4+ T cells and bone marrow-derived mast cells (BMMCs) were co-cultured to explore the modulation of HLJDD in T helper (Th) cells phenotyping. Results: HLJDD showcased a substantial amelioration in skin through the upregulation of FLG, LOR, AQP3, and reducing scratching behaviors in AD-like mice, Also, the quantity of infiltrated mast cells (MCs), pruritus-related mRNA were decreased. In addition, the expression of OX40/OX40L was decreased by HLJDD, which was critical in Th-cell phenotyping. With the treatment of HLJDD, Th1/Th2 and Th17/Treg ratios in AD-like mice became balanced. The structure of commensal microbiota in AD-like mice was affected by HLJDD. HLJDD could also improve the imbalance of Th17/Treg in vitro. Conclusion: HLJDD could improve the symptoms of AD-like mice by alleviating the scratching behaviors via decreased Th2 and pruritus-related mRNA expression. HLJDD also enhanced the relative diversity of skin microbiota and changed the structure of intestinal microbiota. An in-depth study found that HLJDD could balance the ratio of Th1/Th2, Th17/Treg in AD-like mice, and Th17/Treg in vitro by regulating the OX40/OX40L signaling pathway.
Collapse
Affiliation(s)
- Huiyuan Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Quanbin Li
- Hubei College of Chinese Medicine, Jing Zhou, Hubei Province, China
| | - Yaxing Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianhua Guan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kaidi Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunlong Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
6
|
Gupta A, Taneja V, Moreno JR, Abhimanyu, Ahmed M, Naqvi N, Chauhan KS, de León DTP, Ramírez-Martínez G, Jiménez-Alvarez L, Luna-Rivero C, Zuniga J, Kaushal D, Khader SA. Mast cells promote pathology and susceptibility in tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611333. [PMID: 39314389 PMCID: PMC11418949 DOI: 10.1101/2024.09.04.611333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis (Mtb), infects approximately one-fourth of the world's population. While most infected individuals are asymptomatic, latent TB infection (LTBI) can progress to cause pulmonary TB (PTB). We recently reported an increased accumulation of mast cells (MCs) in lungs of macaques with PTB, compared with LTBI in macaques. MCs respond in vitro to Mtb exposure via degranulation and by inducing proinflammatory cytokines. In the current study, we show the dominant production of chymase by MCs in granulomas of humans and macaques with PTB. Using scRNA seq analysis, we show that MCs found in LTBI and healthy lungs in macaques are enriched in genes involved in tumor necrosis factor alpha, cholesterol and transforming growth factor beta signaling. In contrast, MCs clusters found in PTB express transcriptional signatures associated with interferon gamma, oxidative phosphorylation, and MYC signaling. Additionally, MC deficiency in the mouse model showed improved control of Mtb infection that coincided with reduced accumulation of lung myeloid cells and diminished inflammation at chronic stages. Thus, these collective results provide novel evidence for the pathological contribution of MCs during Mtb infection and may represent a novel target for host directive therapy for TB.
Collapse
Affiliation(s)
- Ananya Gupta
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Vibha Taneja
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
| | - Javier Rangel Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Abhimanyu
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
| | - Mushtaq Ahmed
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
| | - Nilofer Naqvi
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
| | - Kuldeep S. Chauhan
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
| | - Daniela Trejo-Ponce de León
- Technologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Gustavo Ramírez-Martínez
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Luis Jiménez-Alvarez
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Cesar Luna-Rivero
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Joaquin Zuniga
- Technologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Deepak Kaushal
- Laboratory of Immunobiology and Genetics and Department of Pathology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico
| | - Shabaana A. Khader
- The University of Chicago, Department of Microbiology, 920 East 58 Street, CLSC 1117, Chicago, IL 60637
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Pérez-Pons A, Teodosio C, Jara-Acevedo M, Henriques A, Navarro-Navarro P, García-Montero AC, Álvarez-Twose I, Lecrevisse Q, Fluxa R, Sánchez-Muñoz L, Caldas C, Pozo J, Martín S, Sanfeliciano TC, Pedreira CE, Botafogo V, González-López O, Mayado A, Orfao A. T-cell immune profile in blood of systemic mastocytosis: Association with disease features. Allergy 2024; 79:1921-1937. [PMID: 38299742 DOI: 10.1111/all.16043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a heterogeneous disease characterized by an expansion of KIT-mutated mast cells (MC). KIT-mutated MC display activated features and release MC mediators that might act on the tumour microenvironment and other immune cells. Here, we investigated the distribution of lymphocyte subsets in blood of patients with distinct subtypes of SM and determined its association with other disease features. METHODS We studied the distribution of TCD4+ and TCD4- cytotoxic cells and their subsets, as well as total NK- and B cells, in blood of 115 SM patients-38 bone marrow mastocytosis (BMM), 67 indolent SM (ISM), 10 aggressive SM (ASM)- and 83 age-matched healthy donors (HD), using spectral flow cytometry and the EuroFlow Immunomonitoring panel, and correlated it with multilineage KITD816V, the alpha-tryptasemia genotype (HαT) and the clinical manifestations of the disease. RESULTS SM patients showed decreased counts (vs. HD) of TCD4- cytotoxic cells, NK cells and several functional subsets of TCD4+ cells (total Th1, Th2-effector memory, Th22-terminal effector and Th1-like Tregs), together with increased T-follicular-helper and Th1/Th17-like Treg counts, associated with different immune profiles per diagnostic subtype of SM, in multilineal versus MC-restricted KITD816V and in cases with a HαT+ versus HαT- genotype. Unique immune profiles were found among BMM and ISM patients with MC-restricted KITD816V who displayed HαT, anaphylaxis, hymenoptera venom allergy, bone disease, pruritus, flushing and GI symptoms. CONCLUSION Our results reveal altered T- and NK-cell immune profiles in blood of SM, which vary per disease subtype, the pattern of involvement of haematopoiesis by KITD816V, the HαT genotype and specific clinical manifestations of the disease.
Collapse
Affiliation(s)
- Alba Pérez-Pons
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
| | - Cristina Teodosio
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - María Jara-Acevedo
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
- Sequencing Service (NUCLEUS), Universidad de Salamanca, Salamanca, Spain
| | - Ana Henriques
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Virgen del Valle Hospital, CIBERONC, Toledo, Madrid, Spain
- Cytognos SL, Salamanca, Spain
| | - Paula Navarro-Navarro
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
- Sequencing Service (NUCLEUS), Universidad de Salamanca, Salamanca, Spain
| | - Andrés C García-Montero
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
| | - Iván Álvarez-Twose
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Virgen del Valle Hospital, CIBERONC, Toledo, Madrid, Spain
| | - Quentin Lecrevisse
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | | | - Laura Sánchez-Muñoz
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Virgen del Valle Hospital, CIBERONC, Toledo, Madrid, Spain
| | - Carolina Caldas
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
| | - Julio Pozo
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Silvia Martín
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | | | - Carlos E Pedreira
- Systems and Computing Department (PESC), COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Vitor Botafogo
- Department of Hematology and Hemotherapy, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Oscar González-López
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
| | - Andrea Mayado
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service (NUCLEUS), Cancer Research Center (IBMCC, USAL-CSIC), Universidad de Salamanca, Salamanca, Spain
- Biomedical Research Networking Center Consortium (CIBERONC; CB16/12/00400), Madrid, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Salamanca, Spain
| |
Collapse
|
8
|
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
|
9
|
Elieh-Ali-Komi D, Bot I, Rodríguez-González M, Maurer M. Cellular and Molecular Mechanisms of Mast Cells in Atherosclerotic Plaque Progression and Destabilization. Clin Rev Allergy Immunol 2024; 66:30-49. [PMID: 38289515 DOI: 10.1007/s12016-024-08981-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Mast cells (MCs) are commonly recognized for their crucial involvement in the pathogenesis of allergic diseases, but over time, it has come to light that they also play a role in the pathophysiology of non-allergic disorders including atherosclerosis. The involvement of MCs in the pathology of atherosclerosis is supported by their accumulation in atherosclerotic plaques upon their progression and the association of intraplaque MC numbers with acute cardiovascular events. MCs that accumulate within the atherosclerotic plaque release a cocktail of mediators through which they contribute to neovascularization, plaque progression, instability, erosion, rupture, and thrombosis. At a molecular level, MC-released proteases, especially cathepsin G, degrade low-density lipoproteins (LDL) and mediate LDL fusion and binding of LDL to proteoglycans (PGs). Through a complicated network of chemokines including CXCL1, MCs promote the recruitment of among others CXCR2+ neutrophils, therefore, aggravating the inflammation of the plaque environment. Additionally, MCs produce extracellular traps which worsen inflammation and contribute to atherothrombosis. Altogether, evidence suggests that MCs actively, via several underlying mechanisms, contribute to atherosclerotic plaque destabilization and acute cardiovascular syndromes, thus, making the study of interventions to modulate MC activation an interesting target for cardiovascular medicine.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Marcus Maurer
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany.
| |
Collapse
|
10
|
Lei Y, Guo X, Luo Y, Niu X, Xi Y, Xiao L, He D, Bian Y, Zhang Y, Wang L, Peng X, Wang Z, Chen G. Synovial microenvironment-influenced mast cells promote the progression of rheumatoid arthritis. Nat Commun 2024; 15:113. [PMID: 38168103 PMCID: PMC10761862 DOI: 10.1038/s41467-023-44304-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Mast cells are phenotypically and functionally heterogeneous, and their state is possibly controlled by local microenvironment. Therefore, specific analyses are needed to understand whether mast cells function as powerful participants or dispensable bystanders in specific diseases. Here, we show that degranulation of mast cells in inflammatory synovial tissues of patients with rheumatoid arthritis (RA) is induced via MAS-related G protein-coupled receptor X2 (MRGPRX2), and the expression of MHC class II and costimulatory molecules on mast cells are upregulated. Collagen-induced arthritis mice treated with a combination of anti-IL-17A and cromolyn sodium, a mast cell membrane stabilizer, show significantly reduced clinical severity and decreased bone erosion. The findings of the present study suggest that synovial microenvironment-influenced mast cells contribute to disease progression and may provide a further mast cell-targeting therapy for RA.
Collapse
Affiliation(s)
- Yunxuan Lei
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xin Guo
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Yanping Luo
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xiaoyin Niu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Yebin Xi
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Lianbo Xiao
- Department of Joint Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqin Bian
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yong Zhang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Li Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China
| | - Xiaochun Peng
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhaojun Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China.
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Shanghai, China.
| |
Collapse
|
11
|
Lan Y, Zou S, Wang W, Chen Q, Zhu Y. Progress in cancer neuroscience. MedComm (Beijing) 2023; 4:e431. [PMID: 38020711 PMCID: PMC10665600 DOI: 10.1002/mco2.431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer of the central nervous system (CNS) can crosstalk systemically and locally in the tumor microenvironment and has become a topic of attention for tumor initiation and advancement. Recently studied neuronal and cancer interaction fundamentally altered the knowledge about glioma and metastases, indicating how cancers invade complex neuronal networks. This review systematically discussed the interactions between neurons and cancers and elucidates new therapeutic avenues. We have overviewed the current understanding of direct or indirect communications of neuronal cells with cancer and the mechanisms associated with cancer invasion. Besides, tumor-associated neuronal dysfunction and the influence of cancer therapies on the CNS are highlighted. Furthermore, interactions between peripheral nervous system and various cancers have also been discussed separately. Intriguingly and importantly, it cannot be ignored that exosomes could mediate the "wireless communications" between nervous system and cancer. Finally, promising future strategies targeting neuronal-brain tumor interactions were reviewed. A great deal of work remains to be done to elucidate the neuroscience of cancer, and future more research should be directed toward clarifying the precise mechanisms of cancer neuroscience, which hold enormous promise to improve outcomes for a wide range of malignancies.
Collapse
Affiliation(s)
- Yu‐Long Lan
- Department of NeurosurgerySecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Shuang Zou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
| | - Wen Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
| | - Yongjian Zhu
- Department of NeurosurgerySecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
12
|
Li L, Wang C, Li Q, Guan Y, Zhang X, Kong F, Feng Z, Lu Y, Wang D, Wang N. Exosomes as a modulator of immune resistance in human cancers. Cytokine Growth Factor Rev 2023; 73:135-149. [PMID: 37543438 DOI: 10.1016/j.cytogfr.2023.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
In the tumor microenvironment (TME), exosomes secreted by cells form interactive networks between the tumor cells and immune cells, thereby regulating immune signaling cascades in the TME. As key messengers of cell-to-cell communication in the TME, exosomes not only take charge of tumor cell antigen presentation to the immune cells, but also regulate the activities of immune cells, inhibit immune function, and, especially, promote immune resistance, all of which affects the therapeutic outcomes of tumors. Exosomes, which are small-sized vesicles, possess some remarkable advantages, including strong biological activity, a lack of immunogenicity and toxicity, and a strong targeting ability. Based on these characteristics, research on exosomes as biomarkers or carriers of tumor therapeutic drugs has become a research hotspot in related fields. This review describes the role of exosomes in cell communications in the TME, summarizes the effectiveness of exosome-based immunotherapy in overcoming immune resistance in cancer treatment, and systematically summarizes and discusses the characteristics of exosomes from different cell sources. Furthermore, the prospects and challenges of exosome-related therapies are discussed.
Collapse
Affiliation(s)
- Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China
| | - Chunyue Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China
| | - Qiucheng Li
- School of Chinese Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Yue Guan
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China
| | - Xin Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China
| | - Zixin Feng
- School of Chinese Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Yuanjun Lu
- School of Chinese Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, PR China.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam 999077, Hong Kong Special Administrative Region of China.
| |
Collapse
|
13
|
Atiakshin D, Kostin A, Shishkina V, Burtseva A, Buravleva A, Volodkin A, Elieh-Ali-Komi D, Buchwalow I, Tiemann M. Space-Flight- and Microgravity-Dependent Alteration of Mast Cell Population and Protease Expression in Digestive Organs of Mongolian Gerbils. Int J Mol Sci 2023; 24:13604. [PMID: 37686410 PMCID: PMC10488096 DOI: 10.3390/ijms241713604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
Mast cell (MC)-specific proteases are of particular interest for space biology and medicine due to their biological activity in regulating targets of a specific tissue microenvironment. MC tryptase and chymase obtain the ability to remodel connective tissue through direct and indirect mechanisms. Yet, MC-specific protease expression under space flight conditions has not been adequately investigated. Using immunohistochemical stainings, we analyzed in this study the protease profile of the jejunal, gastric, and hepatic MC populations in three groups of Mongolian gerbils-vivarium control, synchronous experiment, and 12-day orbital flight on the Foton-M3 spacecraft-and in two groups-vivarium control and anti-orthostatic suspension-included in the experiment simulating effects of weightlessness in the ground-based conditions. After a space flight, there was a decreased number of MCs in the studied organs combined with an increased proportion of chymase-positive MCs and MCs with a simultaneous content of tryptase and chymase; the secretion of specific proteases into the extracellular matrix increased. These changes in the expression of proteases were observed both in the mucosal and connective tissue MC subpopulations of the stomach and jejunum. Notably, the relative content of tryptase-positive MCs in the studied organs of the digestive system decreased. Space flight conditions simulated in the synchronous experiment caused no similar significant changes in the protease profile of MC populations. The space flight conditions resulted in an increased chymase expression combined with a decreased total number of protease-positive MCs, apparently due to participating in the processes of extracellular matrix remodeling and regulating the state of the cardiovascular system.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-structural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (D.A.); (A.K.); (A.V.)
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia; (V.S.); (A.B.); (A.B.)
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-structural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (D.A.); (A.K.); (A.V.)
| | - Viktoriya Shishkina
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia; (V.S.); (A.B.); (A.B.)
| | - Alexandra Burtseva
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia; (V.S.); (A.B.); (A.B.)
| | - Anastasia Buravleva
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia; (V.S.); (A.B.); (A.B.)
| | - Artem Volodkin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-structural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (D.A.); (A.K.); (A.V.)
| | - Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, 12203 Berlin, Germany
| | - Igor Buchwalow
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultra-structural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (D.A.); (A.K.); (A.V.)
- Institute for Hematopathology, 22547 Hamburg, Germany;
| | | |
Collapse
|
14
|
Faiz A, Pavlidis S, Kuo CH, Rowe A, Hiemstra PS, Timens W, Berg M, Wisman M, Guo YK, Djukanović R, Sterk P, Meyer KB, Nawijn MC, Adcock I, Chung KF, van den Berge M. Th2 high and mast cell gene signatures are associated with corticosteroid sensitivity in COPD. Thorax 2023; 78:335-343. [PMID: 36598042 PMCID: PMC10086461 DOI: 10.1136/thorax-2021-217736] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/27/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE Severe asthma and chronic obstructive pulmonary disease (COPD) share common pathophysiological traits such as relative corticosteroid insensitivity. We recently published three transcriptome-associated clusters (TACs) using hierarchical analysis of the sputum transcriptome in asthmatics from the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort comprising one Th2-high inflammatory signature (TAC1) and two Th2-low signatures (TAC2 and TAC3). OBJECTIVE We examined whether gene expression signatures obtained in asthma can be used to identify the subgroup of patients with COPD with steroid sensitivity. METHODS Using gene set variation analysis, we examined the distribution and enrichment scores (ES) of the 3 TACs in the transcriptome of bronchial biopsies from 46 patients who participated in the Groningen Leiden Universities Corticosteroids in Obstructive Lung Disease COPD study that received 30 months of treatment with inhaled corticosteroids (ICS) with and without an added long-acting β-agonist (LABA). The identified signatures were then associated with longitudinal clinical variables after treatment. Differential gene expression and cellular convolution were used to define key regulated genes and cell types. MEASUREMENTS AND MAIN RESULTS Bronchial biopsies in patients with COPD at baseline showed a wide range of expression of the 3 TAC signatures. After ICS±LABA treatment, the ES of TAC1 was significantly reduced at 30 months, but those of TAC2 and TAC3 were unaffected. A corticosteroid-sensitive TAC1 signature was developed from the TAC1 ICS-responsive genes. This signature consisted of mast cell-specific genes identified by single-cell RNA-sequencing and positively correlated with bronchial biopsy mast cell numbers following ICS±LABA. Baseline levels of gene transcription correlated with the change in RV/TLC %predicted following 30-month ICS±LABA. CONCLUSION Sputum-derived transcriptomic signatures from an asthma cohort can be recapitulated in bronchial biopsies of patients with COPD and identified a signature of airway mast cells as a predictor of corticosteroid responsiveness.
Collapse
Affiliation(s)
- Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Ultimo, New South Wales, Australia
- Pulmonary Diseases, UMCG, Groningen, The Netherlands
- GRAIC, University of Groningen, Groningen, The Netherlands
| | - Stelios Pavlidis
- Department of Computing and Data Science Institute, Imperial College London, London, UK
| | - Chih-Hsi Kuo
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Anthony Rowe
- Discovery IT, Janssen Research and Development LLC, High Wycombe, UK
| | - Pieter S Hiemstra
- Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim Timens
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marijn Berg
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marissa Wisman
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Yi-Ke Guo
- Department of Computing and Data Science Institute, Imperial College London, London, UK
| | - Ratko Djukanović
- Academic Unit of Clinical and Experimental Sciences, Southampton University Faculty of Medicine, Southampton, UK
| | - Peter Sterk
- Respiratory Medicine, Amsterdam UMC-Locatie AMC, Amsterdam, The Netherlands
| | - Kerstin B Meyer
- Gene expression genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Martijn C Nawijn
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ian Adcock
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Kian Fan Chung
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Maarten van den Berge
- Pulmonary Diseases, UMCG, Groningen, The Netherlands
- GRAIC, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Zheng X, Zhu J, Zheng C, Tan Z, Ji Z, Tao J, Zhao Y, Ji Z, Hu Y. Dissolving Microneedle Arrays as a Hepatitis B Vaccine Delivery System Adjuvanted by APC-Targeted Poly (Lactic-co-Glycolic Acid) (PLGA) Nanoparticles. AAPS PharmSciTech 2023; 24:42. [PMID: 36697935 DOI: 10.1208/s12249-022-02473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/17/2022] [Indexed: 01/26/2023] Open
Abstract
The objective of this study is to develop a new hepatitis B surface antigen (HBsAg) delivery system by coating soluble microneedle arrays with mannose-modified PLGA nanoparticles (MNPs). MNPs of different sizes were synthesized. The effects these nanoparticles on the maturation of dendritic cells were studied by flow cytometry. HBsAg-containing MNPs (HBsAg/MNPs) of the appropriate sizes were coated into water-soluble microneedle arrays. The in vitro characteristics of microneedles arrays and the immune responses after subcutaneous administration in mice were studied. The results showed that PLGA nanoparticles with an average size of about 800 nm showed the most significant effects in stimulating the maturation of dendritic cells. In the water-soluble microneedle array, the targeted PLGA nanoparticles containing HBsAg were distributed discretely with a maximum distribution height of about 280 μm with a drug load of 0.98 ± 0.05 μg/mg. The drug-containing microneedle arrays exhibited excellent mechanical properties and improved biosafety. The results of immune responses in vivo showed that the subcutaneous administration of the microneedle arrays induced the proliferation of splenocyte, secreted specific IL-12 and IFN-γ, and promote the production of IgG in mice. This study verifies the feasibility of soluble composited microneedles administration in hepatitis B immunization, and provides new ideas for the development and application of non-injectable vaccine delivery systems.
Collapse
Affiliation(s)
- Xiaoling Zheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiahuan Zhu
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China.,School of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Caihong Zheng
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zeng Tan
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China
| | - Zhonghua Ji
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China
| | - Jin Tao
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China
| | - Yunchun Zhao
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhuwa Ji
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China
| | - Ying Hu
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, 315000, China.
| |
Collapse
|
16
|
Chen Y, Griffiths CEM, Bulfone-Paus S. Exploring Mast Cell-CD8 T Cell Interactions in Inflammatory Skin Diseases. Int J Mol Sci 2023; 24:1564. [PMID: 36675078 PMCID: PMC9861959 DOI: 10.3390/ijms24021564] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
The skin is exposed to environmental challenges and contains skin-resident immune cells, including mast cells (MCs) and CD8 T cells that act as sentinels for pathogens and environmental antigens. Human skin MCs and their mediators participate in the maintenance of tissue homeostasis and regulate the recruitment and activity of immune cells involved in the pathogenesis of skin diseases. The cutaneous CD8 T cell compartment is comprised of long-persisting resident memory T cells (TRM) and migratory or recirculating cells; both populations provide durable site immune surveillance. Several lines of evidence indicate that MC-derived products, such as CCL5 and TNF-α, modulate the migration and function of CD8 T cells. Conversely, activated CD8 T cells induce the upregulation of MC costimulatory molecules. Moreover, the close apposition of MCs and CD8 T cells has been recently identified in the skin of several dermatoses, such as alopecia areata. This review outlines the current knowledge about bidirectional interactions between human MCs and CD8 T cells, analyses the alteration of their communication in the context of three common skin disorders in which these cells have been found altered in number or function-psoriasis, atopic dermatitis, and vitiligo-and discusses the current unanswered questions.
Collapse
Affiliation(s)
| | | | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Dermatology Research Centre, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
17
|
van der Elst G, Varol H, Hermans M, Baan CC, Duong-van Huyen JP, Hesselink DA, Kramann R, Rabant M, Reinders MEJ, von der Thüsen JH, van den Bosch TPP, Clahsen-van Groningen MC. The mast cell: A Janus in kidney transplants. Front Immunol 2023; 14:1122409. [PMID: 36891297 PMCID: PMC9986315 DOI: 10.3389/fimmu.2023.1122409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Mast cells (MCs) are innate immune cells with a versatile set of functionalities, enabling them to orchestrate immune responses in various ways. Aside from their known role in allergy, they also partake in both allograft tolerance and rejection through interaction with regulatory T cells, effector T cells, B cells and degranulation of cytokines and other mediators. MC mediators have both pro- and anti-inflammatory actions, but overall lean towards pro-fibrotic pathways. Paradoxically, they are also seen as having potential protective effects in tissue remodeling post-injury. This manuscript elaborates on current knowledge of the functional diversity of mast cells in kidney transplants, combining theory and practice into a MC model stipulating both protective and harmful capabilities in the kidney transplant setting.
Collapse
Affiliation(s)
- G van der Elst
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - H Varol
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M Hermans
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - C C Baan
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - D A Hesselink
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - R Kramann
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.,Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - M Rabant
- Department of Pathology, Necker Hospital, APHP, Paris, France
| | - M E J Reinders
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands
| | - M C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
18
|
Sarandy MM, Pelinsari SM, de Souza LM, Novaes RD, Zanuncio VV, Gonçalves RV. l-arginine and l-citrulline supplementation accelerates second intention wound healing in iNOS knockout mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
19
|
Sheng W, Liu A, Peng H, Wang J, Guan L. A time-series analysis on generalized additive model for atmospheric pollen concentration and the number of visits of allergic conjunctivitis, Beijing, China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:61522-61533. [PMID: 35445302 DOI: 10.1007/s11356-022-19897-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
The objective of this study is to investigate the correlation between atmospheric pollen concentration and daily visits for allergic conjunctivitis. Daily counts of outpatient visits for allergic conjunctivitis, atmospheric pollen concentration, and meteorological data during pollen season of 2018 and 2019 were collected from Beijing Shijitan Hospital, China. A time-series analysis on generalized additive model with Poisson distribution was used to estimate the relationship between pollen concentration and visits for allergic conjunctivitis, after controlling for the time trend, weather variables, day of the week, and holiday effect. The RStudio was used to generate Spearman correlation coefficients and then to plot the lag-response curves for specific and incremental cumulative effects of relative risk (RR). There was a moderate positive correlation between pollen concentration and visits for allergic conjunctivitis, and Spearman's correlation coefficient was 0.521 in 2018 and 0.515 in 2019 (P<0.01). The specific cumulative effect peak associated with per 10 grains/kmm2 increases of atmospheric pollen concentration was within 0 day, and the lag time was 8 days(2018, 2019). The incremental cumulative effect peak associated with per 10 grains/kmm2 increases of atmospheric pollen concentration occurred on lag day 10 (2018) and lag day 8 (2019), and the lag time was 14 days (2018) and 20 days (2019). Elevated concentrations of pollen increase the risk of allergic conjunctivitis with a time lag effect.
Collapse
Affiliation(s)
- Weixuan Sheng
- Department of Anesthesiology, Capital Medical University Affiliated Beijing Shijitan Hospital, No. 10 Yangfangdian Railway Hospital Road, Haidian District, Beijing, 100038, China
| | - Aizhu Liu
- Department of Otolaryngology Head and Neck Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Hong Peng
- Department of Otolaryngology Head and Neck Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Jia Wang
- Department of Otolaryngology Head and Neck Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Lei Guan
- Department of Anesthesiology, Capital Medical University Affiliated Beijing Shijitan Hospital, No. 10 Yangfangdian Railway Hospital Road, Haidian District, Beijing, 100038, China.
| |
Collapse
|
20
|
Wang Y, Chen A. Mast cell-derived exosomal miR-181a-5p modulated trophoblast cell viability, migration, and invasion via YY1/MMP-9 axis. J Clin Lab Anal 2022; 36:e24549. [PMID: 35698293 PMCID: PMC9280008 DOI: 10.1002/jcla.24549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background Mast cells regulate the process of preeclampsia (PE). Since we previously identified mast cells specifically expressing miR‐181a‐5p in the placenta of PE patients, it is plausible to examine the effect and mechanism of mast cell‐derived exosomal miR‐181a‐5p on trophoblast cells. Methods The miR‐181a‐5p and YY1 levels were determined by quantitative real‐time reverse transcription‐polymerase chain reaction. Exosomes were identified by transmission electron microscopy, Western blot, and PKH‐26 labeling. Mast cells or trophoblast cell malignant phenotype were detected using 3‐(4,5‐dimethyl‐2‐thiazolyl)‐2,5‐diphenyl‐2‐H‐tetrazolium bromide, wound healing, and Transwell assays. Quantification of YY1 and metastasis‐related proteins was performed using Western blot. TargetScan, JASPAR, dual‐luciferase reporter genes, and chromatin immunoprecipitation were exploited to verify the relationship between miR‐181a‐5p, YY1, and MMP‐9. Results MiR‐181a‐5p was overexpressed in mast cells of PE patients. Overexpressed miR‐181a‐5p restrained mast cell viability. Mast cell exosomes were successfully isolated, containing high expressions of CD63 and HSP70 and low expression of Calnexin and could be transported to the cytoplasm of trophoblast cells. Mast cell exosomes attenuated the viability, migration, and invasion of HTR‐8/SVneo cells, inhibited YY1, N‐cadherin, Vimentin, and MMP‐9 protein expressions, and promoted E‐cadherin protein expression. The effect of exosomes was enhanced by miR‐181a‐5p mimic but was reversed by miR‐181a‐5p inhibitor. MiR‐181a‐5p targeted YY1 which bound to the MMP‐9 promoter. Overexpressed YY1 in HTR‐8/SVneo cells accelerated the malignant phenotype of the cells and reversed the regulatory effects of exosomal miR‐181a‐5p. Conclusion Mast cell‐derived exosomal miR‐181a‐5p modulates HTR‐8/SVneo cell viability, migration, and invasion via YY1/MMP‐9.
Collapse
Affiliation(s)
- Yinfen Wang
- Maternity Department, Ningbo Women & Children's Hospital, Ningbo City, Zhejiang Province, China
| | - Aner Chen
- Maternity Department, Ningbo Women & Children's Hospital, Ningbo City, Zhejiang Province, China
| |
Collapse
|
21
|
Zhou B, Li J, Liu R, Zhu L, Peng C. The Role of Crosstalk of Immune Cells in Pathogenesis of Chronic Spontaneous Urticaria. Front Immunol 2022; 13:879754. [PMID: 35711438 PMCID: PMC9193815 DOI: 10.3389/fimmu.2022.879754] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic spontaneous urticaria (CSU) is defined as recurrent episodes of spontaneous wheal development and/or angioedema for more than six weeks and at least twice a week. The core link in the pathogenesis of CSU is the activation of mast cells, T cells, eosinophils, and other immune cells infiltrating around the small venules of the lesion. Increased vascular permeability, vasodilatation, and recruitment of inflammatory cells directly depend on mast cell mediators’ release. Complex regulatory systems tightly influence the critical roles of mast cells in the local microenvironment. The bias toward Th2 inflammation and autoantibodies derived from B cells, histamine expressed by basophils, and initiation of the extrinsic coagulation pathway by eosinophils or monocytes exerts powerful modulatory influences on mast cells. Cell-to-cell interactions between mast cells and eosinophils/T cells also are regulators of their function and may involve CSU’s pathomechanism. This review summarizes up-to-date knowledge regarding the crosstalk between mast cells and other immune cells, providing the impetus to develop new research concepts and treatment strategies for CSU.
Collapse
Affiliation(s)
- Bingjing Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Runqiu Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Cong Peng,
| |
Collapse
|
22
|
Gan X, Gu J, Ju Z, Lu L. Diverse Roles of Immune Cells in Transplant Rejection and Immune Tolerance. ENGINEERING 2022; 10:44-56. [DOI: 10.1016/j.eng.2021.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
23
|
Chuchkova NN, Smetanina MV, Shklyaev AE, Pazinenko KA, Kormilina NV, Kanunnikova OM. Morphofunctional characterization of rat thymus mast cells after administration of magnesium orotate mechanically activated forms. RUDN JOURNAL OF MEDICINE 2021. [DOI: 10.22363/2313-0245-2021-25-3-248-255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Relevance. The topicality of the work is determined by the wide spread of hypomagnesemia among the people, which makes it necessary to correct it. The aim of the work is to elucidate the cell-mediated response of the thymus mastocytic link to magnesium deficiency and its correction by the mechanoactivated form of magnesium orotate. Materials and Methods . Animals with drug-induced magnesium deficiency (administration of furosemide 30 mg/kg for 14 days) were administered either the initial preparation Magnerot (Magnerot, Vervag Pharma, Germany), or its mechanoactivated form. The level of magnesium in the blood was determined by test systems ARKREY (Japan). The concentration of magnesium in the thymus tissue was determined by the method of emission spectroscopy with inductively coupled (argon) plasma on an atomic emission spectrometer. Density of mastocytes and the indices of degranulation and granulolosis were calculated on paraffin sections of the thymus after coloration with toluidine blue. Results and Discussion . It was shown that furosemide administration the amount of magnesium decreased in the blood (from 1,750,08 to 0,9020,18 mmol/l, p0,05), but increased in the thymus (from 1,60,6 in the control to 3,71,2 mg/l); in the gland tissue, the number of mastocytes of morphotype A decreased and the number of mastocytes of morphotype D, after active degranulation, increased (by 7,1 times, p0,05). The type of mastocyte secretion in hypomagnesemia is represented by the merocrine variant. The administration of the initial magnesium orotate led to an increase in the concentration of magnesium in the blood to 1,150,25 mmol/l, which is 65,7% of the initial level, the amount of magnesium in the thymus remained elevated (3,41,1 mg/l), the number of actively degranulating cells (morphotype D) was increased. Mechanoactivated magnesium orotate restored the concentration of Mg2+ in the blood to 89,1% (1,560,18 mmol/l, p0,05) and decreased in the thymus (to 2,30,7 mg/l), restored the subpopulation of mastocytes saturated with heparin (type A), reduced the number of mastocytes of morphotype D. Conclusion . The mechanoactivated form of magnesium orotate has a normalizing effect on the population of thymic mastocytes, shows pronounced immunomodulatory activity, which allows us to consider it as a potential therapeutic agent for clinical testing in the complex therapy of hypomagnesemia and associated immunodeficiency.
Collapse
|
24
|
Association of Matrix Metalloproteinase-2 (MMP-2) and MMP-9 Promoter Polymorphisms, Their Serum Levels, and Activities with Coronary Artery Calcification (CAC) in an Iranian Population. Cardiovasc Toxicol 2021; 22:118-129. [PMID: 34731407 DOI: 10.1007/s12012-021-09707-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 10/19/2022]
Abstract
The serum levels and activity of matrix metalloproteinases (MMPs) are associated with the risk of coronary artery calcification (CAC). We sought to investigate the association between MMP-2 -1575G>A (rs243866) and MMP-9 -1562 C>T (rs3918242) SNPs with MMP-2 and MMP-9 serum levels and activity in individuals with CAC. One hundred and fifty-five cases with CAC and 155 healthy individuals as control group from West of Iran were included and frequency of genotypes and alleles of rs243866 and rs3918242 in MMP-2 and MMP-9 genes were determined using PCR-RFLP. We also investigated the serum levels of MMP-2 and MMP-9 and their activity using ELISA and gelatin zymography, respectively. Additionally, serum biochemical parameters including FBS (fasting blood sugar), urea, creatinine, cholesterol, triglyceride, HDL (high-density lipoprotein), LDL (low-density lipoprotein), calcium, and phosphorus as well as blood pressure (systolic blood pressure (SBP) and diastolic blood pressure (DBP)) were measured. Our results showed that both serum levels of MMP-2 and MMP-9 (P < 0.001) and their activity (P < 0.001) were higher in individuals with CAC when compared to the control group. Carrying A and T alleles in MMP-2 -1575G>A (rs243866) and MMP-9 -1562 C>T (rs3918242) SNPs, respectively, may predispose the individuals to CAC by acting as the risk factors. Serum levels and activity of MMP-2 and MMP-9 were found to be higher in CAC cases when compared to the healthy controls. Carriers of A allele in rs243866 SNP and T allele in rs3918242 SNP were shown to have higher MMP-2 and MMP-9 serum levels and activity that may result in increased ECM degradation and support the initiation and development of calcification.
Collapse
|
25
|
Wang X, Lv Z, Han B, Li S, Yang Q, Wu P, Li J, Han B, Deng N, Zhang Z. The aggravation of allergic airway inflammation with dibutyl phthalate involved in Nrf2-mediated activation of the mast cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 789:148029. [PMID: 34082215 DOI: 10.1016/j.scitotenv.2021.148029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/06/2021] [Accepted: 05/22/2021] [Indexed: 06/12/2023]
Abstract
Dibutyl phthalate (DBP)-an organic pollutant-is ubiquitous in the environment. DBP as an immune adjuvant is related to the development of multiple allergic diseases. However, the current research involving DBP-induced pulmonary toxicity remains poorly understood. Therefore, this research aimed to explore the adverse effect and potential mechanism of DBP exposure on the lungs in rats. In our study, ovalbumin was used to build a rat model of allergic airway inflammation to study any harmful effect of DBP exposure on lung tissues. Rats were treated by intragastric administration of DBP (500 mg kg-1 or 750 mg kg-1) and/or subcutaneous injection of SFN (4 mg kg-1). The results of histopathological analysis, cell count, and myeloperoxidase showed that DBP promoted the inflammatory damage of lungs. In the lung tissues, the detection of terminal deoxynucleotidyl transferase dUNT nick end labeling and oxidative stress indices showed that DBP significantly increased the level of apoptosis and oxidative stress. Western blot analysis indicated that DBP raised the expression level of thymic stromal lymphopoietin and reduced the nuclear expression level of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was further verified by quantitative real-time PCR. Meanwhile, DBP treatment markedly up-regulated the inflammatory cytokines such as IL-4 and IL-13, and rat mast cell protease-2, a marker secreted by mast cells (MCs). Conversely, sulforaphane, a Nrf2 inducer, ameliorated the pulmonary damage induced by DBP in the above. Altogether, our data provides a new insight into the impacts of the activation of MCs on the DBP-induced pulmonary toxicity as well as the safety evaluation of DBP.
Collapse
Affiliation(s)
- Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ning Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China.
| |
Collapse
|
26
|
Palma AM, Hanes MR, Marshall JS. Mast Cell Modulation of B Cell Responses: An Under-Appreciated Partnership in Host Defence. Front Immunol 2021; 12:718499. [PMID: 34566974 PMCID: PMC8460918 DOI: 10.3389/fimmu.2021.718499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Mast cells are well known to be activated via cross-linking of immunoglobulins bound to surface receptors. They are also recognized as key initiators and regulators of both innate and adaptive immune responses against pathogens, especially in the skin and mucosal surfaces. Substantial attention has been given to the role of mast cells in regulating T cell function either directly or indirectly through actions on dendritic cells. In contrast, the ability of mast cells to modify B cell responses has been less explored. Several lines of evidence suggest that mast cells can greatly modify B cell generation and activities. Mast cells co-localise with B cells in many tissue settings and produce substantial amounts of cytokines, such as IL-6, with profound impacts on B cell development, class-switch recombination events, and subsequent antibody production. Mast cells have also been suggested to modulate the development and functions of regulatory B cells. In this review, we discuss the critical impacts of mast cells on B cells using information from both clinical and laboratory studies and consider the implications of these findings on the host response to infections.
Collapse
Affiliation(s)
- Alejandro M Palma
- IWK Health Centre and Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Mark R Hanes
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
27
|
Elieh Ali Komi D, Jalili A. The emerging role of mast cells in skin cancers: involved cellular and molecular mechanisms. Int J Dermatol 2021; 61:792-803. [PMID: 34570900 DOI: 10.1111/ijd.15895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 02/04/2023]
Abstract
Skin cancers are the most common cancers worldwide. They can be divided into nonmelanoma skin cancers (NMSC) including basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and less common lymphomas and merkel cell carcinoma, and melanomas. Melanomas comprise less than 5% of skin cancer rate but are responsible for more than 90% of skin cancer death. Mast cells (MCs) are multifunctional cells that play an important role in inflammatory and allergic reactions. They attract other key players of the immune system by releasing cytokines. Healthy human skin comprises MCs under physiological status, and the number can increase under certain conditions including skin malignancies postulating their possible role in pathogenesis of and immunity against skin cancers. MCs respond to cytokines released by tumor stromal cells, release mediators (including histamine and tryptase), and induce the neovascularization, degradation of extracellular matrix (ECM), and induce mitogenesis. However, MCs may use molecular mechanisms to exert immunosuppressive activity including releasing complement C3, lower expression of CD40L, and overexpression of enzymes with vitamin D3 metabolizing activity including CYP27A1 and CYP27B1. This review summarizes the current knowledge on the role of MCs in pathogenesis and immunity against skin cancers.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Jalili
- Department of Dermatology, Bürgenstock Medical Center, Obbürgen, Switzerland
| |
Collapse
|
28
|
Yin Y, Bai Y, Olivera A, Metcalfe DD. Demonstration and implications of IL-3 upregulation of CD25 expression on human mast cells. J Allergy Clin Immunol 2021; 149:1392-1401.e6. [PMID: 34506850 DOI: 10.1016/j.jaci.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/11/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND CD25+ human mast cells (huMCs) have been reported in patients with monoclonal mast cell diseases and in rare association with inflammation. However, the regulation of CD25 expression on huMCs and the possible biologic consequences remain poorly understood. OBJECTIVE We sought to identify conditions that would upregulate CD25 expression on huMCs and to explore possible functional implications. METHODS huMCs were cultured from peripheral blood progenitor cells over 6 to 8 weeks. Expression of CD25 was determined by fluorescence-activated cell sorting and soluble CD25 by ELISA. Signal transducer and activator of transcription 5 (STAT5) phosphorylation induced by IL-2 in huMCs, regulatory T (Treg) cells, or in cocultured huMCs and Treg cells was examined by fluorescence-activated cell sorting. RESULTS Addition of IL-3 to CD34+ progenitors at the initiation of huMC cultures in the presence of stem cell factor and IL-6 upregulated the expression of CD25 in developing huMCs and resulted in shedding of soluble CD25 into the media. Removal of IL-3 after the first week of culture did not affect subsequent expression of CD25. Furthermore, addition of IL-3 14 days after the initiation of the culture did not induce significant CD25 expression. Treatment with anti-IL-3 antibody or the Janus kinase inhibitor tofacitinib blocked IL-3-induced CD25 upregulation. Binding of IL-2 to CD25+ huMCs did not induce STAT5 phosphorylation. However, coincubation of Treg cells with CD25+ huMCs pretreated with IL-2 was sufficient to result in STAT5 phosphorylation in Treg cells. CONCLUSIONS IL-3 promotes CD25 expression and shedding by huMCs. Although CD25+ huMCs do not respond to IL-2, they bind IL-2 and may act as a reservoir of IL-2 to then activate lymphocytes.
Collapse
Affiliation(s)
- Yuzhi Yin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
29
|
Zhang W, Zhang Y, Chen S, Zhang H, Yuan M, Xiao L, Lu Y, Xu H. Trigonelline, An Alkaloid From Leonurus japonicus Houtt., Suppresses Mast Cell Activation and OVA-Induced Allergic Asthma. Front Pharmacol 2021; 12:687970. [PMID: 34421593 PMCID: PMC8371462 DOI: 10.3389/fphar.2021.687970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/27/2021] [Indexed: 12/02/2022] Open
Abstract
Trigonelline, one of the active compounds from Leonurus japonicus Houtt., has been proven to have pharmacological value in diabetes, the central nervous system and cardiovascular diseases. Recent studies have shown that it may also be beneficial in controlling inflammation. However, the mechanism of the antiallergic effects of trigonelline has not been well studied. As the key effector cells participating in the development of allergies, mast cells have been linked to the pathogenesis of asthma for ages. In this study, we demonstrated the inhibitory effect of trigonelline on activated bone marrow-derived mast cells (BMMCs) and verified its anti-inflammatory properties using an ovalbumin (OVA)-induced asthma model. Trigonelline suppressed BMMC degranulation and decreased the production of the cytokines, prostaglandin D2 (PGD2) and leukotriene C4 (LTC4) in a dose-dependent manner. The potent mechanism is mainly through the suppression of the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. Trigonelline can alleviate pathological damage in lung tissue and reduce the levels of serum immunoglobulin E (IgE) and T helper 2 (Th2) cytokines. RNA-seq results revealed the HIF-1α to be a potential target for the allergic reaction. Taken together, our study demonstrated that trigonelline can inhibit allergic inflammation in vitro and in vivo, which may provide a basis for novel anti-inflammatory drug development.
Collapse
Affiliation(s)
- Wenhui Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingling Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Simin Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Yuan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lianbo Xiao
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Yue Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
30
|
Nishi H, Niyonsaba F, Pelleg A, Schulman ES. Enhancement of Mast Cell Degranulation Mediated by Purinergic Receptors' Activation and PI3K Type δ. THE JOURNAL OF IMMUNOLOGY 2021; 207:1001-1008. [PMID: 34330752 DOI: 10.4049/jimmunol.2001002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 05/28/2021] [Indexed: 11/19/2022]
Abstract
Mast cells express multiple metabotropic purinergic P2Y receptor (P2YR) subtypes. Few studies have evaluated their role in human mast cell (HMC) allergic response as quantified by degranulation induced by cross-linking the high-affinity IgE receptor (FcεRI). We have previously shown that extracellular nucleotides modify the FcεRI activation-dependent degranulation in HMCs derived from human lungs, but the mechanism of this action has not been fully delineated. This study was undertaken to determine the mechanism of activation of P2YRs on the degranulation of HMCs and elucidate the specific postreceptor pathways involved. Sensitized LAD2 cells, a human-derived mast cell line, were subjected to a weak allergic stimulation (WAS) using a low concentration of Ag in the absence and presence of P2YR agonists. Only the metabotropic purinergic P2Y11 receptor (P2Y11R) agonist, adenosine 5'-(3-thio)triphosphate (ATPγS), enhanced WAS-induced degranulation resulting in a net 7-fold increase in release (n = 4; p < 0.01). None of the P2YR agonists tested, including high concentrations of ATPγS (1000 μM), enhanced WAS-induced intracellular Ca2+ mobilization, an essential component of activated FcεRI-induced degranulation. Both a PI3K inhibitor and the relevant gene knockout decreased the ATPγS-induced enhancement. The effect of ATPγS was associated with enhanced phosphorylation of PI3K type δ and protein kinase B, but not the phosphoinositide-dependent kinase-1. The effects of ATPγS were dose dependently inhibited by NF157, a P2Y11R antagonist. To our knowledge, these data indicate for the first time that P2YR is linked to enhancement of allergic degranulation in HMC via the PI3K/protein kinase B pathway.
Collapse
Affiliation(s)
- Haruhisa Nishi
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan;
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
| | - Amir Pelleg
- Danmir Therapeutics, LLC, Haverford, PA; and
| | | |
Collapse
|
31
|
Abo Al Hayja M, Wahlström J, Kullberg S, Darlington P, Eklund A, Grunewald J. Bronchoalveolar lavage fluid cell subsets associate with the disease course in Löfgren's and non-Löfgren's sarcoidosis patients. Respir Med 2021; 186:106521. [PMID: 34198166 DOI: 10.1016/j.rmed.2021.106521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sarcoidosis is a multisystem granulomatous inflammatory disorder, that predominantly involves the lungs. Patients with Löfgren's syndrome (LS) are characterized by acute onset and usually have the HLA-DRB1*03 (DR3positive) allele and a good prognosis. Non-LS patients are usually DR3negative and are more likely to develop chronic disease. The study aimed to identify bronchoalveolar lavage fluid (BALF) cells that could associate with disease severity (reduced pulmonary function tests (PFTs), advanced chest radiographs, need for treatment) and/or chronicity (duration >2 years) in newly diagnosed LS and non-LS patients, respectively. METHODS We retrospectively included data from 955 non-LS patients, 477 LS patients, and 295 healthy controls (HC) in this study. Intra-group comparison of patients with resolving versus chronic disease was performed in LS and non-LS, respectively. Non-LS patients were divided into two subgroups according to the binary BALF cell concentrations for intra-group comparison (i.e. higher or lower than the 95th percentile of the BALF cells references in healthy individuals). RESULTS LS patients with a non-resolving disease course had higher BALF lymphocytes, neutrophils, and eosinophils than LS with a favourable outcome. In non-LS subjects increased BALF of the same cells and in addition also of basophils and mast cells were more likely associated with more severe disease course. CONCLUSION Increased BALF cells display prognostic significance in sarcoidosis. Certain BALF profiles should promote the clinician to monitor these patients more closely as they may associate non-resolving disease, in turn, resulting in future irreversible functional impairment.
Collapse
Affiliation(s)
- Muntasir Abo Al Hayja
- Respiratory Medicine Division, Department of Medicine, Solna, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Jan Wahlström
- Respiratory Medicine Division, Department of Medicine, Solna, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | - Susanna Kullberg
- Respiratory Medicine Division, Department of Medicine, Solna, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | - Pernilla Darlington
- Respiratory Medicine Division, Department of Clinical Science and Education, Södersjukhuset and Karolinska Institutet, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Division, Department of Medicine, Solna, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Division, Department of Medicine, Solna, Karolinska Institutet, SE-171 76, Stockholm, Sweden; Respiratory Medicine Division, Department of Medicine Solna, And Center for Molecular Medicine (CMM), Karolinska Institutet; and Respiratory Medicine, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Significance of Mast Cell Formed Extracellular Traps in Microbial Defense. Clin Rev Allergy Immunol 2021; 62:160-179. [PMID: 34024033 PMCID: PMC8140557 DOI: 10.1007/s12016-021-08861-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Mast cells (MCs) are critically involved in microbial defense by releasing antimicrobial peptides (such as cathelicidin LL-37 and defensins) and phagocytosis of microbes. In past years, it has become evident that in addition MCs may eliminate invading pathogens by ejection of web-like structures of DNA strands embedded with proteins known together as extracellular traps (ETs). Upon stimulation of resting MCs with various microorganisms, their products (including superantigens and toxins), or synthetic chemicals, MCs become activated and enter into a multistage process that includes disintegration of the nuclear membrane, release of chromatin into the cytoplasm, adhesion of cytoplasmic granules on the emerging DNA web, and ejection of the complex into the extracellular space. This so-called ETosis is often associated with cell death of the producing MC, and the type of stimulus potentially determines the ratio of surviving vs. killed MCs. Comparison of different microorganisms with specific elimination characteristics such as S pyogenes (eliminated by MCs only through extracellular mechanisms), S aureus (removed by phagocytosis), fungi, and parasites has revealed important aspects of MC extracellular trap (MCET) biology. Molecular studies identified that the formation of MCET depends on NADPH oxidase-generated reactive oxygen species (ROS). In this review, we summarize the present state-of-the-art on the biological relevance of MCETosis, and its underlying molecular and cellular mechanisms. We also provide an overview over the techniques used to study the structure and function of MCETs, including electron microscopy and fluorescence microscopy using specific monoclonal antibodies (mAbs) to detect MCET-associated proteins such as tryptase and histones, and cell-impermeant DNA dyes for labeling of extracellular DNA. Comparing the type and biofunction of further MCET decorating proteins with ETs produced by other immune cells may help provide a better insight into MCET biology in the pathogenesis of autoimmune and inflammatory disorders as well as microbial defense.
Collapse
|
33
|
Korkmaz E, Balmert SC, Sumpter TL, Carey CD, Erdos G, Falo LD. Microarray patches enable the development of skin-targeted vaccines against COVID-19. Adv Drug Deliv Rev 2021; 171:164-186. [PMID: 33539853 PMCID: PMC8060128 DOI: 10.1016/j.addr.2021.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Stephen C Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
34
|
Berger AE, Durrieu C, Dzviga C, Perrot JL, Lambert C. Human peripheral basophils extended phenotype shows a high expression of CD244 immuno-regulatory receptor. J Immunol Methods 2021; 492:112951. [PMID: 33493550 DOI: 10.1016/j.jim.2020.112951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Basophils play a major physio-pathological role in hypersensitivity related diseases. Basophils express high affinity Immunoglobulin (Ig) E receptors (FcεRI), IgG and complement regulatory. Basophils also have immunoregulatory activity through interaction with T cells. The aim of this study was to look for the expression of markers reflecting the activation status of peripheral Basophil in healthy donors. METHOD the study was performed on 29 healthy donors, 62% females with a mean age of 50.1 + 17.0 years. Basophils were identified on their expression of CD123 without HLA-DR and/or CD193 in two 8 colors panels including CD46, CD55, CD59, CD203c, CD32 (FcγRII), CD64 (FcγRIII), CD163, CD137L (4-1BBL), CD252 (OX40L), CD244 (2B4) and CD3 on whole blood. Basophil activation with anti IgE was performed on 14 donors. RESULTS AND DISCUSSION Our results confirmed the Basophil expression of CD123, CD193 and CD203 (the latter is strongly increased under stimulation). Complement regulatory proteins (CD46, CD55, CD59) were expressed at the same levels as on other leukocytes; CD46, CD59 expression being slightly increased under stimulation. CD32 and CD163 scavenger were slightly higher than on lympho and not influenced by activation. CD252 or CD137L were expressed at low levels and significantly induced by stimulation. Most of all, CD244 was highly expressed on Basophils as compared to any other leukocytes in fresh peripheral blood. CONCLUSIONS Our study shows that human resting Basophils express IgE and IgG Fc receptors and check point receptor CD244 that could potentially play a role in their previously reported immunoregulatory activity in sensitization and even in tumor immune escape.
Collapse
Affiliation(s)
- Anne-Emmanuelle Berger
- Immunology laboratory, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France
| | - Coralie Durrieu
- Immunology laboratory, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France
| | - Charles Dzviga
- Allergology unit, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France
| | - Jean-Luc Perrot
- Dermatology department, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France
| | - Claude Lambert
- Immunology laboratory, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France; Allergology unit, University Hospital Saint-Etienne, 42055 Saint-Etienne, Cedex 2, France.
| |
Collapse
|
35
|
Pius-Sadowska E, Machaliński B. Pleiotropic activity of nerve growth factor in regulating cardiac functions and counteracting pathogenesis. ESC Heart Fail 2021; 8:974-987. [PMID: 33465292 PMCID: PMC8006610 DOI: 10.1002/ehf2.13138] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiac innervation density generally reflects the levels of nerve growth factor (NGF) produced by the heart—changes in NGF expression within the heart and vasculature contribute to neuronal remodelling (e.g. sympathetic hyperinnervation or denervation). Its synthesis and release are altered under different pathological conditions. Although NGF is well known for its survival effects on neurons, it is clear that these effects are more wide ranging. Recent studies reported both in vitro and in vivo evidence for beneficial actions of NGF on cardiomyocytes in normal and pathological hearts, including prosurvival and antiapoptotic effects. NGF also plays an important role in the crosstalk between the nervous and cardiovascular systems. It was the first neurotrophin to be implicated in postnatal angiogenesis and vasculogenesis by autocrine and paracrine mechanisms. In connection with these unique cardiovascular properties of NGF, we have provided comprehensive insight into its function and potential effect of NGF underlying heart sustainable/failure conditions. This review aims to summarize the recent data on the effects of NGF on various cardiovascular neuronal and non‐neuronal functions. Understanding these mechanisms with respect to the diversity of NGF functions may be crucial for developing novel therapeutic strategies, including NGF action mechanism‐guided therapies.
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, Szczecin, 70111, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, Szczecin, 70111, Poland
| |
Collapse
|
36
|
Wan J, Wu Y, Huang L, Tian Y, Ji X, Abdelaziz MH, Cai W, Dineshkumar K, Lei Y, Yao S, Sun C, Su Z, Wang S, Xu H. ILC2-derived IL-9 inhibits colorectal cancer progression by activating CD8 + T cells. Cancer Lett 2021; 502:34-43. [PMID: 33429004 DOI: 10.1016/j.canlet.2021.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s), characterized by secretion of type 2 cytokines, regulate multiple immune responses. ILC2s are found in different tumor tissues, and ILC2-derived interleukin (IL)-4, IL-5, and IL-13 act on the cells in tumor microenvironment to participate in tumor progression. ILC2s are abundant in colorectal cancer (CRC) tissue, but the role of ILC2s in CRC remains unclear. In this study, we found that the percentage of ILC2s was higher in CRC tissue than in the adjacent normal tissue and that these ILC2s were the dominant IL-9-secreting cell-subsets in CRC tissue, as shown by flow cytometry analysis. ILC2s-derived IL-9 could activate CD8+ T cells to inhibit tumor growth, while anti-IL-9 reversed this effect. In vivo experiments showed that neutralizing ILC2s promoted tumor growth, while tumor inhibition occurred by intravenous injection of IL-9. In conclusion, our results demonstrated that ILC2-derived IL-9 could activate CD8+ T cells to promote anti-tumor effects in CRC.
Collapse
Affiliation(s)
- Jie Wan
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yinqiu Wu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lan Huang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Tian
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiaoyun Ji
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | | | - Wei Cai
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Kesavan Dineshkumar
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuqing Lei
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Shun Yao
- Center for Pituitary Tumor Surgery, Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Caixia Sun
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China; Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212001, China
| | - Huaxi Xu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
37
|
Elieh Ali Komi D, Shafaghat F, Kovanen PT, Meri S. Mast cells and complement system: Ancient interactions between components of innate immunity. Allergy 2020; 75:2818-2828. [PMID: 32446274 DOI: 10.1111/all.14413] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/09/2020] [Accepted: 04/26/2020] [Indexed: 12/23/2022]
Abstract
The emergence and evolution of the complement system and mast cells (MCs) can be traced back to sea urchins and the ascidian Styela plicata, respectively. Acting as a cascade of enzymatic reactions, complement is activated through the classical (CP), the alternative (AP), and the lectin pathway (LP) based on the recognized molecules. The system's main biological functions include lysis, opsonization, and recruitment of phagocytes. MCs, beyond their classic role as master cells of allergic reactions, play a role in other settings, as well. Thus, MCs are considered as extrahepatic producers of complement proteins. They express various complement receptors, including those for C3a and C5a. C3a and C5a not only activate the C3aR and C5aR expressing MCs but also act as chemoattractants for MCs derived from different anatomic sites, such as from the bone marrow, human umbilical cord blood, or skin in vitro. Cross talk between MCs and complement is facilitated by the production of complement proteins by MCs and their activation by the MC tryptase. The coordinated activity between MCs and the complement system plays a key role, for example, in a number of allergic, cutaneous, and vascular diseases. At a molecular level, MCs and complement system interactions are based on the production of several complement zymogens by MCs and their activation by MC-released proteases. Additionally, at a cellular level, MCs act as potent effector cells of complement activation by expressing receptors for C3a and C5a through which their chemoattraction and activation are mediated by anaphylatoxins in a paracrine and autocrine fashion.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | - Farzaneh Shafaghat
- Immunology Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | | | - Seppo Meri
- Department of Bacteriology and Immunology Immunobiology Research Program University of Helsinki Helsinki Finland
- HUSLAB Helsinki University Central Hospital Helsinki Finland
| |
Collapse
|
38
|
Zhao YB, Yang SH, Shen J, Deng K, Li Q, Wang Y, Cui W, Ye H. Interaction between regulatory T cells and mast cells via IL-9 and TGF-β production. Oncol Lett 2020; 20:360. [PMID: 33133260 PMCID: PMC7590434 DOI: 10.3892/ol.2020.12224] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Research on the immunosuppression of cancer cells has attracted much attention in recent years. The present study sought to provide a new strategy for tumor immunotherapy targeting mast cells by studying the mechanisms underlying mast cell function in cancer immunosuppression. Between January 2015 and December 2017, the tumor tissues of 40 patients with gastric cancer (GC) were collected and grouped in Lihuili Hospital of Ningbo City, China. Pathological sections were prepared and an immunofluorescence assay was performed to analyze the expression of forkhead Box Protein P3 (FOXP3), tryptase, TGFβ1, TGF-βR, IL-9, IL-9R and Oxford 40 ligand (OX40L). Then, the correlations between FOXP3 and tryptase, TGFβ1 and tryptase expression, and the expression of OX40L in patients with GC with different stages were analyzed. The results revealed that high levels of mast cells were present in patients GC, and tryptase and FOXP3 expressions were positively correlated. Mast cells regulate T regulatory (reg) cells in the gastric tumor microenvironment by secreting TGFβ1. Tregs, in turn, promote the survival of mast cells in the tumor microenvironment by producing IL-9. Furthermore, OX40L expression in mast cells was significantly associated with Tumor-Node-Metastasis staging of GC. Overall, the present study reported a positive feedback system that functions through TGFβ1 and IL-9 to allow cross-talk between Tregs and mast cells. Moreover, OX40L may be a potential target for the diagnosis and treatment of GC. These results may provide a new strategy for tumor immunotherapy targeting mast cells.
Collapse
Affiliation(s)
- Yi-Bin Zhao
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shao-Hui Yang
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jie Shen
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Ke Deng
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qi Li
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Yu Wang
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Wei Cui
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Hua Ye
- Department of Gastroenterology, Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
39
|
Eissmann MF, Buchert M, Ernst M. IL33 and Mast Cells-The Key Regulators of Immune Responses in Gastrointestinal Cancers? Front Immunol 2020; 11:1389. [PMID: 32719677 PMCID: PMC7350537 DOI: 10.3389/fimmu.2020.01389] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
The Interleukin (IL-)1 family IL33 is best known for eliciting type 2 immune responses by stimulating mast cells (MCs), regulatory T-cells (Tregs), innate lymphoid cells (ILCs) and other immune cells. MCs and IL33 provide critical control of immunological and epithelial homeostasis in the gastrointestinal (GI) tract. Meanwhile, the role of MCs in solid malignancies appears tissue-specific with both pro and anti-tumorigenic activities. Likewise, IL33 signaling significantly shapes immune responses in the tumor microenvironment, but these effects remain often dichotomous when assessed in experimental models of cancer. Thus, the balance between tumor suppressing and tumor promoting activities of IL33 are highly context dependent, and most likely dictated by the mixture of cell types responding to IL33. Adding to this complexity is the promiscuous nature by which MCs respond to cytokines other than IL33 and release chemotactic factors that recruit immune cells into the tumor microenvironment. In this review, we integrate the outcomes of recent studies on the role of MCs and IL33 in cancer with our own observations in the GI tract. We propose a working model where the most abundant IL33 responsive immune cell type is likely to dictate an overall tumor-supporting or tumor suppressing outcome in vivo. We discuss how these opposing responses affect the therapeutic potential of targeting MC and IL33, and highlight the caveats and challenges facing our ability to effectively harness MCs and IL33 biology for anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| |
Collapse
|
40
|
Komi DEA, Khomtchouk K, Santa Maria PL. A Review of the Contribution of Mast Cells in Wound Healing: Involved Molecular and Cellular Mechanisms. Clin Rev Allergy Immunol 2020; 58:298-312. [PMID: 30729428 DOI: 10.1007/s12016-019-08729-w] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells (MCs), apart from their classic role in allergy, contribute to a number of biologic processes including wound healing. In particular, two aspects of their histologic distribution within the skin have attracted the attention of researchers to study their wound healing role; they represent up to 8% of the total number of cells within the dermis and their cutaneous versions are localized adjacent to the epidermis and the subdermal vasculature and nerves. At the onset of a cutaneous injury, the accumulation of MCs and release of proinflammatory and immunomodulatory mediators have been well documented. The role of MC-derived mediators has been investigated through the stages of wound healing including inflammation, proliferation, and remodeling. They contribute to hemostasis and clot formation by enhancing the expression of factor XIIIa in dermal dendrocytes through release of TNF-α, and contribute to clot stabilization. Keratinocytes, by secreting stem cell factor (SCF), recruit MCs to the site. MCs in return release inflammatory mediators, including predominantly histamine, VEGF, interleukin (IL)-6, and IL-8, that contribute to increase of endothelial permeability and vasodilation, and facilitate migration of inflammatory cells, mainly monocytes and neutrophils to the site of injury. MCs are capable of activating the fibroblasts and keratinocytes, the predominant cells involved in wound healing. MCs stimulate fibroblast proliferation during the proliferative phase via IL-4, vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF) to produce a new extracellular matrix (ECM). MC-derived mediators including fibroblast growth factor-2, VEGF, platelet-derived growth factor (PDGF), TGF-β, nerve growth factor (NGF), IL-4, and IL-8 contribute to neoangiogenesis, fibrinogenesis, or reepithelialization during the repair process. MC activation inhibition and targeting the MC-derived mediators are potential therapeutic strategies to improve wound healing through reduced inflammatory responses and scar formation.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kelly Khomtchouk
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Stanford University, 801 Welch Rd, Stanford, CA, 94305, USA
| | - Peter Luke Santa Maria
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Stanford University, 801 Welch Rd, Stanford, CA, 94305, USA.
| |
Collapse
|
41
|
Kechida M. Update on Autoimmune Diseases Pathogenesis. Curr Pharm Des 2020; 25:2947-2952. [PMID: 31686634 DOI: 10.2174/1381612825666190709205421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 06/30/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autoimmune diseases result from the interplay of cellular effectors like T and B cells, regulatory cells in addition to molecular factors like cytokines and regulatory molecules. METHODS Different electronic databases were searched in a non-systematic way to find out the literature of interest. RESULTS Pathogenesis of autoimmune diseases involves typical factors such as genetic background including HLA and non HLA system genes, environmental factors such as infectious agents and inflammatory cells mainly T and B lymphocytes abnormally activated leading to immune dysfunction. Other recently reported less typical factors such as micro-RNAs, circular RNAs, myeloperoxidase, vimentine and microbiome dysbiosis seem to be potential target therapies. CONCLUSION We aimed in this manuscript to review common factors in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Melek Kechida
- Internal Medicine and Endocrinology Department of Fattouma Bourguiba University Hospital, University of Monastir, BP 56 Avenue Taher Haddad, Monastir 5000, Tunisia
| |
Collapse
|
42
|
Zhou Z, Zeiter S, Schmid T, Sakai D, Iatridis JC, Zhou G, Richards RG, Alini M, Grad S, Li Z. Effect of the CCL5-Releasing Fibrin Gel for Intervertebral Disc Regeneration. Cartilage 2020; 11:169-180. [PMID: 29582673 PMCID: PMC7097979 DOI: 10.1177/1947603518764263] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE To explore if chemokine (C-C motif) ligand 5 (CCL5) delivery could recruit annulus fibrosus (AF) cells to the injury sites and facilitate the repair of ruptured AF. DESIGN The effects of CCL5 on bovine AF cells in vitro were tested by transwell assay and quantitative real-time polymerase chain reaction. Fibrin gel containing CCL5 was used to treat annulotomized bovine caudal discs cultured under dynamic loading conditions. After 14 days of loading, the samples were collected for histological examination. A pilot animal study was performed using sheep cervical discs to investigate the effect of fibrin gel encapsulated with CCL5 for the treatment of ruptured AF. After 14 weeks, the animals were sacrificed, and the discs were scanned with magnetic resonance imaging before histopathological examination. RESULTS CCL5 showed a chemotactic effect on AF cells in a dose-dependent manner. AF cells cultured with CCL5 in vitro did not show any change of the gene expression of CCL5 receptors, catabolic and proinflammatory markers. In vitro release study showed that CCL5 exhibited sustained release from the fibrin gel into the culture media; however, in the organ culture study CCL5 did not stimulate homing of AF cells toward the defect sites. The pilot animal study did not show any repair effect of CCL5. CONCLUSIONS CCL5 has a chemotactic effect on AF cells in vitro, but no ex vivo or in vivo regenerative effect when delivered within fibrin gel. Further study with a stronger chemotactic agent and/or an alternate biomaterial that is more conductive of cell migration is warranted.
Collapse
Affiliation(s)
- Zhiyu Zhou
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- AO Research Institute Davos, Davos, Switzerland
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Stephan Zeiter
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
| | - Tanja Schmid
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
| | - Daisuke Sakai
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
- Department of Orthopaedic Surgery, Surgical Science and Research Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - James C. Iatridis
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guangqian Zhou
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - R. Geoff Richards
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
| | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
- Collaborative Research Partner Annulus Fibrosus Repair Program, AO Foundation, Davos, Switzerland
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
43
|
Gu L, Wang F, Lin Z, Xu T, Lin D, Xing M, Yang S, Chao Z, Ye B, Lin P, Hui C, Lu L, Hou S. Genetic characteristics of Jiaji Duck by whole genome re-sequencing. PLoS One 2020; 15:e0228964. [PMID: 32049997 PMCID: PMC7015413 DOI: 10.1371/journal.pone.0228964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
Jiaji Duck (JJ) is a Muscovy duck species that possesses many superior characteristics, and it has become an important genetic resource in China. However, to date, its genetic characteristics and genetic relationship with other duck breeds have not been explored yet, which greatly limits the utilization of JJ. In the present study, we investigated the genome sequences of 15 individual ducks representing five different duck populations, including JJ, French Muscovy duck (FF), mallard (YD), hong duck (HD) and Beijing duck (BD). Moreover, we investigated the characteristics of JJ-specific single nucleotide polymorphisms (SNPs) and compared the genome sequences of JJ vs. YD and JJ vs. BD using integrated strategies, including mutation detection, selective screening, and Gene Ontology (GO) analysis. More than 40 Gb of clean data were obtained for each population (mean coverage of 13.46 Gb per individual). A total number of 22,481,367 SNPs and 4,156,829 small insertion-deletions (Indels) were identified for the five duck populations, which could be used as molecular markers in breeding and utilization of JJ. Moreover, we identified 1,447,932 JJ-specific SNPs, and found that genes covering at least one JJ-specific SNP mainly involved in protein phosphorylation and dephosphorylation, as well as DNA modification. Phylogenetic tree and principal components analysis (PCA) revealed that the genetic relationship of JJ was closest to FF, while it was farthest to BD. A total of 120 and 111 genes were identified as positive selection genes for JJ vs. BD and JJ vs. YD, respectively. GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that the positive selection genes for JJ vs. BD ducks mainly involved in pigmentation, muscle contraction and stretch, gland secretion, and immunology, while the positive selection genes obtained from JJ vs. YD ducks mainly involved in embryo development, muscle contraction and stretch, and gland secretion. Taken together, our findings enabled us to better understand the characteristics of JJ and provided a molecular basis for the breeding and hybrid utilization of JJ in the future.
Collapse
Affiliation(s)
- Lihong Gu
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Feng Wang
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Zhemin Lin
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Tieshan Xu
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, P. R. China
| | - Dajie Lin
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Manping Xing
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Shaoxiong Yang
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Zhe Chao
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Baoguo Ye
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Peng Lin
- Hainan Chuanwei Muscovy Duck Breeding Co., Ltd, Wenchang, P. R. China
| | - Chunhui Hui
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Sciences, Haikou, P. R. China
| | - Lizhi Lu
- Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, P. R. China
| | - Shuisheng Hou
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China
| |
Collapse
|
44
|
Mustafa AS. Vaccine Potential of Mycobacterial Antigens against Asthma. Med Princ Pract 2020; 29:404-411. [PMID: 32422630 PMCID: PMC7511680 DOI: 10.1159/000508719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/17/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is a cause of substantial burden of disease in the world, including both premature deaths and reduced quality of life. A leading hypothesis to explain the worldwide increase of asthma is the "hygiene hypothesis," which suggests that the increase in the prevalence of asthma is due to the reduction in exposure to infections/microbial antigens. In allergic asthma, the most common type of asthma, antigen-specific T helper (Th)2 and Th17 cells and their cytokines are primary mediators of the pathological consequences. In contrast, Th1 and T regulatory (Treg) cells and their cytokines play a protective role. This article aims to review the information on the effect of mycobacteria and their antigens in modulating Th2/Th17 responses towards Th1/Treg responses and protection against asthma in humans and animal models.
Collapse
Affiliation(s)
- Abu Salim Mustafa
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait,
| |
Collapse
|
45
|
Kaesler S, Wölbing F, Kempf WE, Skabytska Y, Köberle M, Volz T, Sinnberg T, Amaral T, Möckel S, Yazdi A, Metzler G, Schaller M, Hartmann K, Weide B, Garbe C, Rammensee HG, Röcken M, Biedermann T. Targeting tumor-resident mast cells for effective anti-melanoma immune responses. JCI Insight 2019; 4:125057. [PMID: 31578309 DOI: 10.1172/jci.insight.125057] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 08/27/2019] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint blockade has revolutionized cancer treatment. Patients developing immune mediated adverse events, such as colitis, appear to particularly benefit from immune checkpoint inhibition. Yet, the contributing mechanisms are largely unknown. We identified a systemic LPS signature in melanoma patients with colitis following anti-cytotoxic T lymphocyte-associated antigen 4 (anti-CTLA-4) checkpoint inhibitor treatment and hypothesized that intestinal microbiota-derived LPS contributes to therapeutic efficacy. Because activation of immune cells within the tumor microenvironment is considered most promising to effectively control cancer, we analyzed human and murine melanoma for known sentinels of LPS. We identified mast cells (MCs) accumulating in and around melanomas and showed that effective melanoma immune control was dependent on LPS-activated MCs recruiting tumor-infiltrating effector T cells by secretion of CXCL10. Importantly, CXCL10 was also upregulated in human melanomas with immune regression and in patients with colitis induced by anti-CTLA-4 antibody. Furthermore, we demonstrate that CXCL10 upregulation and an MC signature at the site of melanomas are biomarkers for better patient survival. These findings provide conclusive evidence for a "Trojan horse treatment strategy" in which the plasticity of cancer-resident immune cells, such as MCs, is used as a target to boost tumor immune defense.
Collapse
Affiliation(s)
- Susanne Kaesler
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Florian Wölbing
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Wolfgang Eberhard Kempf
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Yuliya Skabytska
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Dermatology, Eberhard Karls University, Tübingen, Germany.,Clinical Unit Allergology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Sinnberg
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Sigrid Möckel
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Amir Yazdi
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Gisela Metzler
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Schaller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Karin Hartmann
- Division of Allergy, Department of Dermatology, University of Basel, Basel, Switzerland
| | - Benjamin Weide
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.,Department of Immunology, Institute of Cell Biology, and German Cancer Consortium, German Cancer Research Center partner site Tübingen, Eberhard Karls University, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute of Cell Biology, and German Cancer Consortium, German Cancer Research Center partner site Tübingen, Eberhard Karls University, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Dermatology, Eberhard Karls University, Tübingen, Germany.,Clinical Unit Allergology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
46
|
Mast cell-mediated splanchnic cholestatic inflammation. Clin Res Hepatol Gastroenterol 2019; 43:561-574. [PMID: 30853494 DOI: 10.1016/j.clinre.2019.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/21/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Splanchnic mast cells increase in chronic liver and in acute-on-chronic liver diseases. We administered Ketotifen, a mast cell stabilizer, and measured the mast cells in the splanchnic organs of cholestatic rats. MATERIAL AND METHODS These groups were studied: sham-operated rats (S; n = 15), untreated microsurgical cholestasic rats (C; n = 20) and rats treated with Ketotifen: early (SK-e; n = 20 and CKe; n = 18), and late (SK-l; n = 15 and CK-l; n = 14). RESULTS The cholestatic rats showed systemic and splanchnic impairments, such as ascites, portal hypertension, and biliary proliferation and fibrosis. The rats also showed a splanchnic increase of TNF-α, IL-1β and MCP-1, and a reduction of IL-4, IL-10 and antioxidants. An increase of VEGF in the ileum and mesenteric lymphatic complex was associated with a liver reduction of TGF-β1. Ketotifen reduces the degree of hepatic insufficiency and the splanchnic inflammatory mediators, as well as VEGF and TGF-ß1 levels. Ketotifen also reduces the connective tissue mast cells in the mesenteric lymphatic complex of cholestatic rats, while increases the hepatic mucosal mast cells. CONCLUSIONS In cholestatic rats, Ketotifen improves liver function and ascites, and also reduces pro-inflammatory mediators in the splanchnic area. The decrease in connective tissue mast cells in the mesenteric lymphatic complex due to the administration of Ketotifen would lead to the improvement of the inflammatory splanchnic response, and consequently the abovementioned complications.
Collapse
|
47
|
Elieh Ali Komi D, Bjermer L. Mast Cell-Mediated Orchestration of the Immune Responses in Human Allergic Asthma: Current Insights. Clin Rev Allergy Immunol 2019; 56:234-247. [PMID: 30506113 DOI: 10.1007/s12016-018-8720-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Improving the lung function after experimental allergen challenge by blocking of mast cell (MC) mediators and the capability of MC mediators (including histamine, prostaglandin (PG) D2, and leukotriene (LT) C4) in induction of mucosal edema, bronchoconstriction, and mucus secretion provide evidence that MCs play a key role in pathophysiology of asthma. In asthma, the number of MCs increases in the airways and infiltration of MCs in a variety of anatomical sites including the epithelium, the submucosal glands, and the smooth muscle bundles occurs. MC localization within the ASM is accompanied with the hypertrophy and hyperplasia of the layer, and smooth muscle dysfunction that is mainly observed in forms of bronchial hyperresponsiveness, and variable airflow obstruction. Owing to the expression of a wide range of surface receptors and releasing various cytoplasmic mediators, MCs orchestrate the pathologic events of the disease. MC-released preformed mediators including chymase, tryptase, and histamine and de novo synthesized mediators such as PGD2, LTC4, and LTE4 in addition of cytokines mainly TGFβ1, TSLP, IL-33, IL-4, and IL-13 participate in pathogenesis of asthma. The release of MC mediators and MC/airway cell interactions during remodeling phase of asthma results in persistent cellular and structural changes in the airway wall mainly epithelial cell shedding, goblet cell hyperplasia, hypertrophy of ASM bundles, fibrosis in subepithelial region, abnormal deposition of extracellular matrix (ECM), increased tissue vascularity, and basement membrane thickening. We will review the current knowledge regarding the participation of MCs in each stage of asthma pathophysiology including the releasing mediators and their mechanism of action, expression of receptors by which they respond to stimuli, and finally the pharmaceutical products designed based on the strategy of blocking MC activation and mediator release.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leif Bjermer
- Department of Respiratory Medicine & Allergology, Inst for Clinical Science, Lund University, Lund, Sweden.
- Lung and Allergy Research, Skane University Hospital, Lasarettsgatan 7, 22185, Lund, Sweden.
| |
Collapse
|
48
|
Elieh Ali Komi D, Ribatti D. Mast cell-mediated mechanistic pathways in organ transplantation. Eur J Pharmacol 2019; 857:172458. [PMID: 31202799 DOI: 10.1016/j.ejphar.2019.172458] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 02/02/2023]
Abstract
Adaptive immunity has gained importance in transplant immunology for years, based on models in which T-cells orchestrate the immune responses during rejection. Most recently, researches revealed that innate immune cells, including mast cells (MCs) also play a pivotal role in allograft rejection. MC mediated immunoregulatory responses influence the innate and adaptive immune responses. Their capability to produce an array of both pro-inflammatory and anti-inflammatory mediators, expressing a wide range of costimulatory molecules in addition to acting as antigen-presenting cells (APCs), make them effective immune cells far beyond their classical role as primary orchestrator cells of allergy. Activated regulatory Tcells (Treg) cells contribute to MC recruitment into grafts by releasing interleukin (IL)-9. Tregs are capable of stabilizing MCs and suppressing IgE mediated degranulation through interaction of Treg expressing OX40 with MCs expressing OX40L. MCs in turn release transforming growth factor (TGF)-β and IL-10 which possess suppressive properties. Thus, these cells can suppress the proliferation of T-cells and support the generation of Tregs. MCs in addition to orchestrating immune responses in grafts by cell-to-cell interactions with variety of immune cells, cause histologic changes, mainly fibrosis by releasing mediators such as histamine, fibroblast growth factor-2 (FGF-2), TGF-β, chymase, and cathepsin G. The role of MCs in transplant rejection remains controversial. The accumulation of MCs in rejected grafts suggests that they play a role in preventing graft tolerance, and contribute to the progression of chronic rejection of allografts. However, high expression of MC-related gene products in tolerant grafts and their known interaction with Tregs on the other hand, support the notion that they are an integral component in achieving peripheral tolerance.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
49
|
Kritikou E, van der Heijden T, Swart M, van Duijn J, Slütter B, Wezel A, Smeets HJ, Maffia P, Kuiper J, Bot I. Hypercholesterolemia Induces a Mast Cell-CD4 + T Cell Interaction in Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 202:1531-1539. [PMID: 30683705 DOI: 10.4049/jimmunol.1800648] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/20/2018] [Indexed: 11/19/2022]
Abstract
Mast cells (MCs) are potent innate immune cells that aggravate atherosclerosis through the release of proinflammatory mediators inside atherosclerotic plaques. Similarly, CD4+ T cells are constituents of the adaptive immune response and accumulate within the plaques following lipid-specific activation by APCs. Recently it has been proposed that these two cell types can interact in a direct manner. However, no indication of such an interaction has been investigated in the context of atherosclerosis. In our study, we aimed to examine whether MCs can act as APCs in atherosclerosis, thereby modulating CD4+ T cell responses. We observed that MCs increased their MHC class II expression under hyperlipidemic conditions both in vivo and in vitro. Furthermore, we showed that MCs can present Ags in vivo via MHC class II molecules. Serum from high-fat diet-fed mice also enhanced the expression of the costimulatory molecule CD86 on cultured MCs, whereas OVA peptide-loaded MCs increased OT-II CD4+ T cell proliferation in vitro. The aortic CD4+ and TH1 cell content of atherosclerotic mice that lack MCs was reduced as compared with their wild-type counterparts. Importantly, we identified MCs that express HLA-DR in advanced human atheromata, indicating that these cells are capable of Ag presentation within human atherosclerotic plaques. Therefore, in this artice, we show that MCs may directly modulate adaptive immunity by acting as APCs in atherosclerosis.
Collapse
Affiliation(s)
- Eva Kritikou
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands;
| | - Thomas van der Heijden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Maarten Swart
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom; and.,Department of Pharmacy, University of Naples Federico II, 80138 Naples, Italy
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| |
Collapse
|
50
|
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
|