1
|
Qiao W, Duan C, Ma J, Hu W, Xie Y, Yang L, Wang T, Wu S, Li X, Wang Y, Cheng K, Zhang Y, Zhang Y, Zhuang R. Costimulatory Molecule CD226 Regulates Atopic Dermatitis in a Mouse Model. J Invest Dermatol 2024; 144:1743-1753.e4. [PMID: 38325579 DOI: 10.1016/j.jid.2024.01.022] [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: 12/22/2022] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
This study investigated the role of CD226 in a 2,4-dinitrochlorobenzene (DNCB)-induced mouse model of atopic dermatitis. The results showed that the lack of CD226 (global and CD4+ T-cell specific) significantly increased ear thickness, reddening, swelling, and scaling of the skin as well as inflammatory cell and mast cell infiltration. RT-qPCR results demonstrated that the mRNA expressions of atopic dermatitis-related inflammatory cytokines and chemokines were markedly increased in the draining lymph nodes and lesioned ear skin tissues of global and CD4+ T-cell-specific CD226-deficient mice compared with that in control mice. In vitro assessment revealed that CD226 directly modulates TGFβ-mediated regulatory T (Treg) cell differentiation and proliferation. Notably, Treg cell-specific deletion of CD226 (Cd226fl/flFoxp3cre mice) resulted in more severe dermatitis and epidermal thickening than those observed in littermate mice upon DNCB treatment. Subsequent analysis showed that the infiltration of Treg cells in ear lesions and the number of Tregs in the spleen were significantly reduced in Cd226fl/flFoxp3cre mice after DNCB treatment. In addition, the lack of CD226 induced apoptosis of Treg cells through the activation of caspase 3. Therefore, these results suggest that CD226 has potential efficacy in atopic dermatitis, correlating with Treg cell inhibition.
Collapse
MESH Headings
- Animals
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/pathology
- Dermatitis, Atopic/genetics
- Mice
- Disease Models, Animal
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Mice, Knockout
- Dinitrochlorobenzene
- Apoptosis
- Cell Differentiation
- Mice, Inbred C57BL
- Skin/pathology
- Skin/immunology
Collapse
Affiliation(s)
- Wei Qiao
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China; Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Chujun Duan
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China; Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Wei Hu
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yang Xie
- Otolaryngological Department of Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lu Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Tingting Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuwen Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Xuemei Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yuan Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Liu Y, Zhou J, Chen B, Liu X, Cai Y, Liu W, Hao H, Li S. High-dimensional mass cytometry reveals systemic and local immune signatures in necrotizing enterocolitis. Front Immunol 2023; 14:1292987. [PMID: 38045686 PMCID: PMC10690805 DOI: 10.3389/fimmu.2023.1292987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
Objective Patients with necrotizing enterocolitis display severe gastrointestinal complications of prematurity, but the mechanism driving this clinical profile remains unknown. We used mass cytometry time-of-flight to characterize and compare immune cell populations in the blood and intestine tissue from patients with and without (controls) necrotizing enterocolitis at single-cell resolution. Methods We completed a deep mapping of the immune system of the peripheral blood mononuclear cells and intestinal mucosa tissue using mass cytometry to evaluate immune cell types, which revealed global immune dysregulation characteristics underlying necrotizing enterocolitis. Results Compared with controls, natural killer cells display signs of heightened activation and increased cytotoxic potential in the peripheral blood and mucosa of patients with necrotizing enterocolitis. Furthermore, CD4+ T effector memory cells, non-classical monocytes, active dendritic cells, and neutrophils were specifically enriched in the mucosa, suggesting trafficking from the periphery to areas of inflammation. Moreover, we mapped the systemic and local distinct immune signatures suggesting patterns of cell localization in necrotizing enterocolitis. Conclusion We used mass cytometry time-of-flight technology to identify immune cell populations specific to the peripheral blood and intestinal mucosa tissue from patients with necrotizing enterocolitis and controls. This information might be used to develop precise diagnosis and therapies that target specific cell populations in patients with necrotizing enterocolitis.
Collapse
Affiliation(s)
- Yufeng Liu
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, Guangzhou, China
| | - Jialiang Zhou
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Baozhu Chen
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao Liu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Liu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Massironi S, Mulinacci G, Gallo C, Elvevi A, Danese S, Invernizzi P, Vespa E. Mechanistic Insights into Eosinophilic Esophagitis: Therapies Targeting Pathophysiological Mechanisms. Cells 2023; 12:2473. [PMID: 37887317 PMCID: PMC10605530 DOI: 10.3390/cells12202473] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disease characterized by eosinophilic infiltration of the esophagus. It arises from a complex interplay of genetic predisposition (susceptibility loci), environmental triggers (allergens and dietary antigens), and a dysregulated immune response, mainly mediated by type 2 T helper cell (Th2)-released cytokines, such as interleukin (IL)-4, IL-5, and IL-13. These cytokines control eosinophil recruitment and activation as well as tissue remodeling, contributing to the characteristic features of EoE. The pathogenesis of EoE includes epithelial barrier dysfunction, mast cell activation, eosinophil degranulation, and fibrosis. Epithelial barrier dysfunction allows allergen penetration and promotes immune cell infiltration, thereby perpetuating the inflammatory response. Mast cells release proinflammatory mediators and promote eosinophil recruitment and the release of cytotoxic proteins and cytokines, causing tissue damage and remodeling. Prolonged inflammation can lead to fibrosis, resulting in long-term complications such as strictures and dysmotility. Current treatment options for EoE are limited and mainly focus on dietary changes, proton-pump inhibitors, and topical corticosteroids. Novel therapies targeting key inflammatory pathways, such as monoclonal antibodies against IL-4, IL-5, and IL-13, are emerging in clinical trials. A deeper understanding of the complex pathogenetic mechanisms behind EoE will contribute to the development of more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Giacomo Mulinacci
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Camilla Gallo
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Alessandra Elvevi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Edoardo Vespa
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
4
|
Zhovmer AS, Manning A, Smith C, Wang J, Ma X, Tsygankov D, Dokholyan NV, Cartagena-Rivera AX, Singh RK, Tabdanov ED. Septins Enable T Cell Contact Guidance via Amoeboid-Mesenchymal Switch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559597. [PMID: 37808814 PMCID: PMC10557721 DOI: 10.1101/2023.09.26.559597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Lymphocytes exit circulation and enter in-tissue guided migration toward sites of tissue pathologies, damage, infection, or inflammation. By continuously sensing and adapting to the guiding chemo-mechano-structural properties of the tissues, lymphocytes dynamically alternate and combine their amoeboid (non-adhesive) and mesenchymal (adhesive) migration modes. However, which mechanisms guide and balance different migration modes are largely unclear. Here we report that suppression of septins GTPase activity induces an abrupt amoeboid-to-mesenchymal transition of T cell migration mode, characterized by a distinct, highly deformable integrin-dependent immune cell contact guidance. Surprisingly, the T cell actomyosin cortex contractility becomes diminished, dispensable and antagonistic to mesenchymal-like migration mode. Instead, mesenchymal-like T cells rely on microtubule stabilization and their non-canonical dynein motor activity for high fidelity contact guidance. Our results establish septin's GTPase activity as an important on/off switch for integrin-dependent migration of T lymphocytes, enabling their dynein-driven fluid-like mesenchymal propulsion along the complex adhesion cues.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Alexis Manning
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jian Wang
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Xuefei Ma
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nikolay V Dokholyan
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, The Pennsylvania State University Hershey-Hummelstown, PA, USA
| | - Alexander X Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Rakesh K Singh
- Department of Obstetrics & Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Erdem D Tabdanov
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|
5
|
Yamashita K, Takebayashi S, Murata W, Hirai N, Ito Y, Mitsui M, Saito M, Sato K, Terada M, Niizeki N, Suzuki A, Ogitani K, Fujikawa T, Komori M, Inoue N, Arai N, Maekawa M. Analytical Performance of a Novel Latex Turbidimetric Immunoassay, "Nanopia TARC", for TARC/CCL17 Measurement: A Retrospective Observational Study. Diagnostics (Basel) 2023; 13:2935. [PMID: 37761302 PMCID: PMC10529481 DOI: 10.3390/diagnostics13182935] [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] [Received: 07/13/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Thymus- and activation-regulated chemokine (TARC, also known as CCL17) is used as a biomarker for atopic dermatitis. The methods currently used for its measurement are complex, time-consuming, and require large machinery, warranting the need for a method that is simple, has a quick turnaround time, and requires less complex machinery. We evaluated the analytical performance of a novel latex turbidimetric immunoassay method, "Nanopia TARC", on 174 residual serum samples from patients with skin or allergic diseases. This evaluation included the assessment of the limit of blank/detection/quantification (LOB/D/Q), precision, accuracy, linearity, interference, and commutability between Nanopia TARC and "HISCL TARC", based on the chemiluminescent enzyme immunoassay (CLEIA) method. The LOB/D/Q values were 13, 57, and 141 pg/mL, respectively. The coefficient of variation of the repeatability was 0.9-3.8%, and that of the intermediate precision was 2.1-5.4%. The total error of the accuracy was 1.9-13.4%. The linearity was 141 and 19,804 pg/mL for TARC. The correlation coefficient between Nanopia TARC and HISCL TARC determined using the Passing-Bablok regression analysis was 0.999. Furthermore, the concordance of diagnostic criteria with AD was 92%. Nanopia TARC was confirmed to have the same analytical performance for TARC measurement as the existing CLEIA method.
Collapse
Affiliation(s)
- Keita Yamashita
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Shiori Takebayashi
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Wataru Murata
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Nao Hirai
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Yui Ito
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Mayuka Mitsui
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Mina Saito
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Kei Sato
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Miyuki Terada
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Noriyasu Niizeki
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Akira Suzuki
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Kenya Ogitani
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| | - Toshihiko Fujikawa
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Marie Komori
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Nozomi Inoue
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Norimitsu Arai
- Diagnostic Products Development, Department Research & Development, Sekisui Medical Co., Ltd., Tokyo 103-0027, Japan; (T.F.); (M.K.); (N.I.); (N.A.)
| | - Masato Maekawa
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; (S.T.); (W.M.); (N.H.); (Y.I.); (M.M.); (M.S.); (K.S.); (M.T.); (N.N.); (A.S.); (K.O.)
| |
Collapse
|
6
|
Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, McDonald-Hyman C, Saha A, Aguilar EG, Koehn B, Osborn MJ, Panoskaltsis-Mortari A, Macdonald KPA, Hill GR, Murphy WJ, Serody JS, Maillard I, Kean LS, Kim SV, Littman DR, Blazar BR. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity. Am J Transplant 2023; 23:1102-1115. [PMID: 36878433 PMCID: PMC10475494 DOI: 10.1016/j.ajt.2023.01.030] [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: 12/19/2022] [Accepted: 01/31/2023] [Indexed: 03/07/2023]
Abstract
Damage to the gastrointestinal tract following allogeneic hematopoietic stem cell transplantation is a significant contributor to the severity and perpetuation of graft-versus-host disease. In preclinical models and clinical trials, we showed that infusing high numbers of regulatory T cells reduces graft-versus-host disease incidence. Despite no change in in vitro suppressive function, transfer of ex vivo expanded regulatory T cells transduced to overexpress G protein-coupled receptor 15 or C-C motif chemokine receptor 9, specific homing receptors for colon or small intestine, respectively, lessened graft-versus-host disease severity in mice. Increased regulatory T cell frequency and retention within the gastrointestinal tissues of mice that received gut homing T cells correlated with lower inflammation and gut damage early post-transplant, decreased graft-versus-host disease severity, and prolonged survival compared with those receiving control transduced regulatory T cells. These data provide evidence that enforced targeting of ex vivo expanded regulatory T cells to the gastrointestinal tract diminishes gut injury and is associated with decreased graft-versus-host disease severity.
Collapse
Affiliation(s)
- Jemma H Larson
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sujeong Jin
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Loschi
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara Bolivar Wagers
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael C Zaiken
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cameron McDonald-Hyman
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ethan G Aguilar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brent Koehn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kelli P A Macdonald
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Immunology Department, Brisbane, Queensland, Australia
| | - Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA; Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Jonathan S Serody
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sangwon V Kim
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dan R Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
7
|
Brancaccio R, Murdaca G, Casella R, Loverre T, Bonzano L, Nettis E, Gangemi S. miRNAs' Cross-Involvement in Skin Allergies: A New Horizon for the Pathogenesis, Diagnosis and Therapy of Atopic Dermatitis, Allergic Contact Dermatitis and Chronic Spontaneous Urticaria. Biomedicines 2023; 11:1266. [PMID: 37238937 PMCID: PMC10216116 DOI: 10.3390/biomedicines11051266] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/29/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Skin inflammation is a common underlying feature of atopic dermatitis, allergic contact dermatitis and chronic spontaneous urticaria. The pathogenetic mechanisms have not been fully elucidated. The purpose of this study was to examine whether miRNA, by regulating inflammatory mechanisms through the modulation of innate and adaptive immune responses, could play a major role in the pathogenesis of these skin conditions. We conducted a narrative review using the Pubmed and Embase scientific databases and search engines to find the most relevant miRNAs related to the pathophysiology, severity and prognosis of skin conditions. The studies show that miRNAs are involved in the pathogenesis and regulation of atopic dermatitis and can reveal an atopic predisposition or indicate disease severity. In chronic spontaneous urticaria, different miRNAs which are over-expressed during urticaria exacerbations not only play a role in the possible response to therapy or remission, but also serve as a marker of chronic autoimmune urticaria and indicate associations with other autoimmune diseases. In allergic contact dermatitis, miRNAs are upregulated in inflammatory lesions and expressed during the sensitization phase of allergic response. Several miRNAs have been identified as potential biomarkers of these chronic skin conditions, but they are also possible therapeutic targets.
Collapse
Affiliation(s)
- Raffaele Brancaccio
- Dermatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio nell’Emilia, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Rossella Casella
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70124 Bari, Italy
| | - Teresa Loverre
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70124 Bari, Italy
| | - Laura Bonzano
- Dermatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio nell’Emilia, Italy
| | - Eustachio Nettis
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70124 Bari, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Division of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
8
|
Nakamura T, Tachibana Y, Murata T. 12-HETE promotes late-phase responses in a murine model of allergic rhinitis. Allergy 2023; 78:574-577. [PMID: 36208108 DOI: 10.1111/all.15547] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Tatsuro Nakamura
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuri Tachibana
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Hulme J. Staphylococcus Infection: Relapsing Atopic Dermatitis and Microbial Restoration. Antibiotics (Basel) 2023; 12:antibiotics12020222. [PMID: 36830133 PMCID: PMC9952585 DOI: 10.3390/antibiotics12020222] [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: 12/15/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Atopic Dermatitis (AD) skin is susceptible to Staphylococcus aureus (SA) infection, potentially exposing it to a plethora of toxins and virulent determinants, including Panton-Valentine leukocidin (PVL) (α-hemolysin (Hla) and phenol-soluble modulins (PSMs)), and superantigens. Depending on the degree of infection (superficial or invasive), clinical treatments may encompass permanganate (aq) and bleach solutions coupled with intravenous/oral antibiotics such as amoxicillin, vancomycin, doxycycline, clindamycin, daptomycin, telavancin, linezolid, or tigecycline. However, when the skin is significantly traumatized (sheathing of epidermal sections), an SA infection can rapidly ensue, impairing the immune system, and inducing local and systemic AD presentations in susceptible areas. Furthermore, when AD presents systemically, desensitization can be long (years) and intertwined with periods of relapse. In such circumstances, the identification of triggers (stress or infection) and severity of the flare need careful monitoring (preferably in real-time) so that tailored treatments targeting the underlying pathological mechanisms (SA toxins, elevated immunoglobulins, impaired healing) can be modified, permitting rapid resolution of symptoms.
Collapse
Affiliation(s)
- John Hulme
- Gachon Bio-Nano Institute, Gachon University, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
10
|
Sztuk TKS, Rigby NM, Nørskov-Nielsen L, Koppelman SJ, Sancho AI, Knudsen NPH, Marsh J, Johnson P, Gupta S, Mackie AR, Larsen JM, Bøgh KL. Dose and route of administration determine the efficacy of prophylactic immunotherapy for peanut allergy in a Brown Norway rat model. Front Immunol 2023; 14:1121497. [PMID: 36911669 PMCID: PMC9996042 DOI: 10.3389/fimmu.2023.1121497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Allergen-specific immunotherapy (IT) is emerging as a viable option for treatment of peanut allergy. Yet, prophylactic IT remains unexplored despite early introduction of peanut in infancy was shown to prevent allergy. There is a need to understand how allergens interact with the immune system depending on the route of administration, and how different dosages of allergen may protect from sensitisation and a clinical active allergy. Here we compared peanut allergen delivery via the oral, sublingual (SL), intragastric (IG) and subcutaneous (SC) routes for the prevention of peanut allergy in Brown Norway (BN) rats. Methods BN rats were administered PBS or three different doses of peanut protein extract (PPE) via either oral IT (OIT), SLIT, IGIT or SCIT followed by intraperitoneal (IP) injections of PPE to assess the protection from peanut sensitisation. The development of IgE and IgG1 responses to PPE and the major peanut allergens were evaluated by ELISAs. The clinical response to PPE was assessed by an ear swelling test (EST) and proliferation was assessed by stimulating splenocytes with PPE. Results Low and medium dose OIT (1 and 10 mg) and all doses of SCIT (1, 10, 100 µg) induced sensitisation to PPE, whereas high dose OIT (100 mg), SLIT (10, 100 or 1000 µg) or IGIT (1, 10 and 100 mg) did not. High dose OIT and SLIT as well as high and medium dose IGIT prevented sensitisation from the following IP injections of PPE and suppressed PPE-specific IgE levels in a dose-dependent manner. Hence, administration of peanut protein via different routes confers different risks for sensitisation and protection from peanut allergy development. Overall, the IgE levels toward the individual major peanut allergens followed the PPE-specific IgE levels. Discussion Collectively, this study showed that the preventive effect of allergen-specific IT is determined by the interplay between the specific site of PPE delivery for presentation to the immune system, and the allergen quantity, and that targeting and modulating tolerance mechanisms at specific mucosal sites may be a prophylactic strategy for prevention of peanut allergy.
Collapse
Affiliation(s)
| | - Neil Marcus Rigby
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | - Stef J Koppelman
- Institute of Agriculture and Natural Resources, Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Ana Isabel Sancho
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Justin Marsh
- Institute of Agriculture and Natural Resources, Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Philip Johnson
- Institute of Agriculture and Natural Resources, Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - Alan Robert Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | - Jeppe Madura Larsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | |
Collapse
|
11
|
Hippen KL, Hefazi M, Larson JH, Blazar BR. Emerging translational strategies and challenges for enhancing regulatory T cell therapy for graft-versus-host disease. Front Immunol 2022; 13:926550. [PMID: 35967386 PMCID: PMC9366169 DOI: 10.3389/fimmu.2022.926550] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 02/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for many types of cancer. Genetic disparities between donor and host can result in immune-mediated attack of host tissues, known as graft versus host disease (GVHD), a major cause of morbidity and mortality following HSCT. Regulatory CD4+ T cells (Tregs) are a rare cell type crucial for immune system homeostasis, limiting the activation and differentiation of effector T cells (Teff) that are self-reactive or stimulated by foreign antigen exposure. Adoptive cell therapy (ACT) with Treg has demonstrated, first in murine models and now in patients, that prophylactic Treg infusion can also suppress GVHD. While clinical trials have demonstrated Treg reduce severe GVHD occurrence, several impediments remain, including Treg variability and practical need for individualized Treg production for each patient. Additionally, there are challenges in the use of in vitro expansion techniques and in achieving in vivo Treg persistence in context of both immune suppressive drugs and in lymphoreplete patients being treated for GVHD. This review will focus on 3 main translational approaches taken to improve the efficacy of tTreg ACT in GVHD prophylaxis and development of treatment options, following HSCT: genetic modification, manipulating TCR and cytokine signaling, and Treg production protocols. In vitro expansion for Treg ACT presents a multitude of approaches for gene modification to improve efficacy, including: antigen specificity, tissue targeting, deletion of negative regulators/exhaustion markers, resistance to immunosuppressive drugs common in GVHD treatment. Such expansion is particularly important in patients without significant lymphopenia that can drive Treg expansion, enabling a favorable Treg:Teff ratio in vivo. Several potential therapeutics have also been identified that enhance tTreg stability or persistence/expansion following ACT that target specific pathways, including: DNA/histone methylation status, TCR/co-stimulation signaling, and IL-2/STAT5 signaling. Finally, this review will discuss improvements in Treg production related to tissue source, Treg subsets, therapeutic approaches to increase Treg suppression and stability during tTreg expansion, and potential for storing large numbers of Treg from a single production run to be used as an off-the-shelf infusion product capable of treating multiple recipients.
Collapse
Affiliation(s)
- Keli L. Hippen
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Jemma H. Larson
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Bruce R. Blazar
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| |
Collapse
|
12
|
Jin R, Luo L, Zheng J. The Trinity of Skin: Skin Homeostasis as a Neuro-Endocrine-Immune Organ. Life (Basel) 2022; 12:725. [PMID: 35629392 PMCID: PMC9144330 DOI: 10.3390/life12050725] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 12/15/2022] Open
Abstract
For a long time, skin was thought to be no more than the barrier of our body. However, in the last few decades, studies into the idea of skin as an independent functional organ have gradually deepened our understanding of skin and its functions. In this review, we gathered evidence that presented skin as a "trinity" of neuro-endocrine-immune function. From a neuro perspective, skin communicates through nerves and receptors, releasing neurotrophins and neuropeptides; from an endocrine perspective, skin is able to receive and secrete most hormones and has the cutaneous equivalent of the hypothalamic-pituitary-adrenal (HPA) axis; from an immune perspective, skin is protected not only by its physical barrier, but also immune cells and molecules, which can also cause inflammation. Together as an organ, skin works bidirectionally by operating peripheral neuro-endocrine-immune function and being regulated by the central nervous system, endocrine system and immune system at the same time, maintaining homeostasis. Additionally, to further explain the "trinity" of cutaneous neuro-endocrine-immune function and how it works in disease pathophysiology, a disease model of rosacea is presented.
Collapse
Affiliation(s)
- Rong Jin
- Department of Dermatology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Lan Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Jie Zheng
- Department of Dermatology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
13
|
Humeau M, Boniface K, Bodet C. Cytokine-Mediated Crosstalk Between Keratinocytes and T Cells in Atopic Dermatitis. Front Immunol 2022; 13:801579. [PMID: 35464457 PMCID: PMC9022745 DOI: 10.3389/fimmu.2022.801579] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/07/2022] [Indexed: 01/22/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by barrier dysfunction, dysregulated immune response, and dysbiosis with increased Staphylococcus aureus colonization. Infiltration of various T helper cell subsets into lesional skin and subsequent cytokine release are a hallmark of AD. Release of cytokines by both T cells and keratinocytes plays a key role in skin inflammation and drives many AD features. This review aims to discuss cytokine-mediated crosstalk between T cells and keratinocytes in AD pathogenesis and the potential impact of virulence factors produced by Staphylococcus aureus on these interactions.
Collapse
Affiliation(s)
- Mélanie Humeau
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines LITEC UR 15560, Université de Poitiers, Poitiers, France
| | - Katia Boniface
- ImmunoConcEpT, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5164, University of Bordeaux, Bordeaux, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines LITEC UR 15560, Université de Poitiers, Poitiers, France
| |
Collapse
|
14
|
Abbott J, Mukherjee A, Wu W, Ye T, Jung HS, Cheung KM, Gertner RS, Basan M, Ham D, Park H. Multi-parametric functional imaging of cell cultures and tissues with a CMOS microelectrode array. LAB ON A CHIP 2022; 22:1286-1296. [PMID: 35266462 PMCID: PMC8963257 DOI: 10.1039/d1lc00878a] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 06/01/2023]
Abstract
Electrode-based impedance and electrochemical measurements can provide cell-biology information that is difficult to obtain using optical-microscopy techniques. Such electrical methods are non-invasive, label-free, and continuous, eliminating the need for fluorescence reporters and overcoming optical imaging's throughput/temporal resolution limitations. Nonetheless, electrode-based techniques have not been heavily employed because devices typically contain few electrodes per well, resulting in noisy aggregate readouts. Complementary metal-oxide-semiconductor (CMOS) microelectrode arrays (MEAs) have sometimes been used for electrophysiological measurements with thousands of electrodes per well at sub-cellular pitches, but only basic impedance mappings of cell attachment have been performed outside of electrophysiology. Here, we report on new field-based impedance mapping and electrochemical mapping/patterning techniques to expand CMOS-MEA cell-biology applications. The methods enable accurate measurement of cell attachment, growth/wound healing, cell-cell adhesion, metabolic state, and redox properties with single-cell spatial resolution (20 μm electrode pitch). These measurements allow the quantification of adhesion and metabolic differences of cells expressing oncogenes versus wild-type controls. The multi-parametric, cell-population statistics captured by the chip-scale integrated device opens up new avenues for fully electronic high-throughput live-cell assays for phenotypic screening and drug discovery applications.
Collapse
Affiliation(s)
- Jeffrey Abbott
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
- Department of Physics, Harvard University, Cambridge, Massachusetts, USA
| | - Avik Mukherjee
- Department of System Biology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Wenxuan Wu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Tianyang Ye
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Han Sae Jung
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Kevin M Cheung
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Rona S Gertner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Markus Basan
- Department of System Biology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Donhee Ham
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Hongkun Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
- Department of Physics, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Shannon JL, Corcoran DL, Murray JC, Ziegler SF, MacLeod AS, Zhang JY. Thymic stromal lymphopoietin controls hair growth. Stem Cell Reports 2022; 17:649-663. [PMID: 35216683 PMCID: PMC9039851 DOI: 10.1016/j.stemcr.2022.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/17/2022] Open
Abstract
Skin tissue regeneration after injury involves the production and integration of signals by stem cells residing in hair follicles (HFSCs). Much remains unknown about how specific wound-derived factors modulate stem cell contribution to hair growth. We demonstrate that thymic stromal lymphopoietin (TSLP) is produced in response to skin injury and during the anagen phase of the hair cycle. Intradermal injection of TSLP promoted wound-induced hair growth (WIHG), whereas neutralizing TSLP receptor (TSLPR) inhibited WIHG. Using flow cytometry and fluorescent immunostaining, we found that TSLP promoted proliferation of transit-amplifying cells. Lgr5CreER-mediated deletion of Tslpr in HFSCs inhibited both wound-induced and exogenous TSLP-induced hair growth. Our data highlight a novel function for TSLP in regulation of hair follicle activity during homeostasis and wound healing.
Collapse
Affiliation(s)
- Jessica L Shannon
- Department of Dermatology, Duke University, P.O. Box 103052, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA
| | - David L Corcoran
- Genomic and Computational Biology, Duke University, Durham, NC 27705, USA
| | - John C Murray
- Department of Dermatology, Duke University, P.O. Box 103052, Durham, NC 27710, USA
| | - Steven F Ziegler
- Benaroya Research Institute, Seattle, WA 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, P.O. Box 103052, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Jennifer Y Zhang
- Department of Dermatology, Duke University, P.O. Box 103052, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
16
|
Abstract
The microbiome plays a significant role in human health, homeostasis, immune system, and disease pathogenesis. Disrupted communication between the microbiome and host has been extensively studied in gastrointestinal diseases. To a lesser extent, there is emerging research on the skin microbiome and its connection with the gut, referred to as the gut-skin axis and its effects on dermatologic conditions. A basic overview will be provided of the gut and skin microbiome with a focus on the impact of this connection on cutaneous diseases, such as psoriasis, atopic dermatitis, rosacea, acne vulgaris, photoaging, and cutaneous wounds. In addition, we shall discuss nutrition-based approaches mediated through the gut-skin axis and topical treatments that could serve as potential adjunctive management by manipulation of the microbiome. In particular, there is a growing body of research on oral probiotics, prebiotics, and dietary modifications that may help improve symptoms for a variety of dermatologic conditions in select demographic groups.
Collapse
Affiliation(s)
- Shivani Sinha
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut, USA
| | - Gloria Lin
- Department of Dermatology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Katalin Ferenczi
- Department of Dermatology, University of Connecticut School of Medicine, Farmington, Connecticut, USA.
| |
Collapse
|
17
|
Chemokines and Innate Lymphoid Cells in Skin Inflammation. Cells 2021; 10:cells10113074. [PMID: 34831296 PMCID: PMC8621478 DOI: 10.3390/cells10113074] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
As the outermost barrier, skin plays an important role in protecting our bodies against outside invasion. Under stable conditions or during inflammation, leukocytes migration is essential for restoring homeostasis in the skin. Immune cells trafficking is orchestrated by chemokines; leukocytes express receptors that bind to chemokines and trigger migration. The homeostasis of the immune ecosystem is an extremely complicated dynamic process that requires the cooperation of innate and adaptive immune cells. Emerging studies have been shedding a light on the unique characteristics of skin-resident innate lymphoid cells (ILCs). In this review, we discuss how chemokines orchestrate skin ILCs trafficking and contribute to tissue homeostasis and how abnormal chemokine–chemokine receptor interactions contribute to and augment skin inflammation, as seen in conditions such as contact hypersensitivity, atopic dermatitis, and psoriasis.
Collapse
|
18
|
Yoshie O. CCR4 as a Therapeutic Target for Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13215542. [PMID: 34771703 PMCID: PMC8583476 DOI: 10.3390/cancers13215542] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CCR4 is a chemokine receptor selectively expressed on normal T cell subsets such as type 2 helper T cells, skin-homing T cells and regulatory T cells, and on skin-associated T cell malignancies such as adult T cell leukemia/lymphoma (ATLL), which is etiologically associated with human T lymphocyte virus type 1 (HTLV-1), and cutaneous T cell lymphomas (CTCLs). Mogamulizumab is a fully humanized and glyco-engineered monoclonal anti-CCR4 antibody used for the treatment of refractory/relapsed ATLL and CTCLs, often resulting in complete remission. The clinical applications of Mogamulizumab are now being extended to solid tumors, exploring the therapeutic effect of regulatory T cell depletion. This review overviews the expression of CCR4 in various T cell subsets, HTLV-1-infected T cells, ATLL and CTCLs, and the clinical applications of Mogamulizumab. Abstract CCR4 is a chemokine receptor mainly expressed by T cells. It is the receptor for two CC chemokine ligands, CCL17 and CCL22. Originally, the expression of CCR4 was described as highly selective for helper T type 2 (Th2) cells. Later, its expression was extended to other T cell subsets such as regulatory T (Treg) cells and Th17 cells. CCR4 has long been regarded as a potential therapeutic target for allergic diseases such as atopic dermatitis and bronchial asthma. Furthermore, the findings showing that CCR4 is strongly expressed by T cell malignancies such as adult T cell leukemia/lymphoma (ATLL) and cutaneous T cell lymphomas (CTCLs) have led to the development and clinical application of the fully humanized and glyco-engineered monoclonal anti-CCR4 Mogamulizumab in refractory/relapsed ATLL and CTCLs with remarkable successes. However, Mogamulizumab often induces severe adverse events in the skin possibly because of its efficient depletion of Treg cells. In particular, treatment with Mogamulizumab prior to allogenic hematopoietic stem cell transplantation (allo-HSCT), the only curative option of these T cell malignancies, often leads to severe glucocorticoid-refractory graft-versus-host diseases. The efficient depletion of Treg cells by Mogamulizumab has also led to its clinical trials in advanced solid tumors singly or in combination with immune checkpoint inhibitors. The main focus of this review is CCR4; its expression on normal and malignant T cells and its significance as a therapeutic target in cancer immunotherapy.
Collapse
Affiliation(s)
- Osamu Yoshie
- Health and Kampo Institute, Sendai 981-3205, Japan;
- Kindai University, Osaka 577-8502, Japan
- Aoinosono-Sendai Izumi Long-Term Health Care Facility, Sendai 981-3126, Japan
| |
Collapse
|
19
|
Inoue T, Omori-Miyake M, Maruyama S, Okabe M, Kuwahara M, Honda H, Miura H, Yamashita M. The Loss of H3K27 Histone Demethylase Utx in T Cells Aggravates Allergic Contact Dermatitis. THE JOURNAL OF IMMUNOLOGY 2021; 207:2223-2234. [PMID: 34588217 DOI: 10.4049/jimmunol.2001160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 08/24/2021] [Indexed: 11/19/2022]
Abstract
The pathogenesis of allergic contact dermatitis (ACD) requires the activation of Ag-specific T cells, including effector and regulatory T cells. The differentiation and function of these T cells is epigenetically regulated through DNA methylation and histone modifications. However, the roles of altered histone H3K27 methylation in T cells in the development of ACD remain unknown. Two types of histone H3K27 demethylases, Utx and Jmjd3, have been reported in mammals. To determine the role of the histone H3K27 demethylase expression of T cells in the development of ACD, we generated T cell-specific, Utx-deficient (Utx KO) mice or Jmjd3-deficient (Jmjd3 KO) mice. Unlike control mice, Utx KO mice had severer symptoms of ACD, whereas Jmjd3 KO mice showed symptoms identical to those in control mice. In Utx KO mice with ACD, the massive infiltration of myeloid cells, including neutrophils and dendritic cells, has been observed. In addition, the expression of proinflammatory cytokines in CD4+ T cells of the draining lymph nodes (LNs) and in CD8+ T cells of the skin was increased in Utx KO mice, whereas the ratio of Foxp3+ regulatory CD4+ T cells to Foxp3- conventional CD4+ T cells was decreased in both the draining LNs and the skin of Utx KO mice with ACD. Furthermore, Foxp3+ regulatory CD4+ T cells of Utx KO mice with ACD expressed a decreased level of CCR4 (a skin-tropic chemokine receptor) in comparison with control. Thus, in CD4+ T cells, Utx could potentially be involved in the regulation of the pathogenesis of ACD.
Collapse
Affiliation(s)
- Takashi Inoue
- Department of Bone and Joint Surgery, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Miyuki Omori-Miyake
- Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Saho Maruyama
- Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan; and
| | - Makoto Kuwahara
- Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Honda
- Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiromasa Miura
- Department of Bone and Joint Surgery, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan; .,Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
20
|
Wang S, Zissler UM, Buettner M, Heine S, Heldner A, Kotz S, Pechtold L, Kau J, Plaschke M, Ullmann JT, Guerth F, Oelsner M, Alessandrini F, Blank S, Chaker AM, Schmidt‐Weber CB, Jakwerth CA. An exhausted phenotype of T H 2 cells is primed by allergen exposure, but not reinforced by allergen-specific immunotherapy. Allergy 2021; 76:2827-2839. [PMID: 33969495 DOI: 10.1111/all.14896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Studies show that proallergic TH 2 cells decrease after successful allergen-specific immunotherapy (AIT). It is likely that iatrogenic administration of allergens drives these cells to exhaustion due to chronic T-cell receptor stimulation. This study aimed to investigate the exhaustion of T cells in connection with allergen exposure during AIT in mice and two independent patient cohorts. METHODS OVA-sensitized C57BL/6J mice were challenged and treated with OVA, and the development of exhaustion in local and systemic TH 2 cells was analyzed. In patients, the expression of exhaustion-associated surface markers on TH 2 cells was evaluated using flow cytometry in a cross-sectional grass pollen allergy cohort with and without AIT. The treatment effect was further studied in PBMC collected from a prospective long-term AIT cohort. RESULTS The exhaustion-associated surface markers CTLA-4 and PD-1 were significantly upregulated on TH 2 cells upon OVA aerosol exposure in OVA-allergic compared to non-allergic mice. CTLA-4 and PD-1 decreased after AIT, in particular on the surface of local lung TH 2 cells. Similarly, CTLA-4 and PD-1 expression was enhanced on TH 2 cells from patients with allergic rhinitis with an even stronger effect in those with concomitant asthma. Using an unbiased Louvain clustering analysis, we discovered a late-differentiated TH 2 population expressing both markers that decreased during up-dosing but persisted long term during the maintenance phase. CONCLUSIONS This study shows that allergen exposure promotes CTLA-4 and PD-1 expression on TH 2 cells and that the dynamic change in frequencies of exhausted TH 2 cells exhibits a differential pattern during the up-dosing versus the maintenance phases of AIT.
Collapse
|
21
|
Wirtz S, Schulz-Kuhnt A, Neurath MF, Atreya I. Functional Contribution and Targeted Migration of Group-2 Innate Lymphoid Cells in Inflammatory Lung Diseases: Being at the Right Place at the Right Time. Front Immunol 2021; 12:688879. [PMID: 34177944 PMCID: PMC8222800 DOI: 10.3389/fimmu.2021.688879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
During the last decade, group-2 innate lymphoid cells (ILC2s) have been discovered and successfully established as crucial mediators of lung allergy, airway inflammation and fibrosis, thus affecting the pathogenesis and clinical course of many respiratory diseases, like for instance asthma, cystic fibrosis and chronic rhinosinusitis. As an important regulatory component in this context, the local pulmonary milieu at inflammatory tissue sites does not only determine the activation status of lung-infiltrating ILC2s, but also influences their motility and migratory behavior. In general, many data collected in recent murine and human studies argued against the former concept of a very strict tissue residency of innate lymphoid cells (ILCs) and instead pointed to a context-dependent homing capacity of peripheral blood ILC precursors and the inflammation-dependent capacity of specific ILC subsets for interorgan trafficking. In this review article, we provide a comprehensive overview of the so far described molecular mechanisms underlying the pulmonary migration of ILC2s and thereby the numeric regulation of local ILC2 pools at inflamed or fibrotic pulmonary tissue sites and discuss their potential to serve as innovative therapeutic targets in the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Stefan Wirtz
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Anja Schulz-Kuhnt
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
22
|
Koenig JFE, Bruton K, Phelps A, Grydziuszko E, Jiménez-Saiz R, Jordana M. Memory Generation and Re-Activation in Food Allergy. Immunotargets Ther 2021; 10:171-184. [PMID: 34136419 PMCID: PMC8200165 DOI: 10.2147/itt.s284823] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Recent evidence has highlighted the critical role of memory cells in maintaining lifelong food allergies, thereby identifying these cells as therapeutic targets. IgG+ memory B cells replenish pools of IgE-secreting cells upon allergen exposure, which contract thereafter due to the short lifespan of tightly regulated IgE-expressing cells. Advances in the detection and highly dimensional analysis of allergen-specific B and T cells from allergic patients have provided insight on their phenotype and function. The newly identified Th2A and Tfh13 populations represent a leap in our understanding of allergen-specific T cell phenotypes, although how these populations contribute to IgE memory responses remains poorly understood. Within, we discuss the mechanisms by which memory B and T cells are activated, integrating knowledge from human systems and fundamental research. We then focus on memory reactivation, specifically, on the pathways of secondary IgE responses. Throughout, we identify areas of future research which will help identify immunotargets for a transformative therapy for food allergy.
Collapse
Affiliation(s)
- Joshua F E Koenig
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kelly Bruton
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Allyssa Phelps
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Emily Grydziuszko
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Rodrigo Jiménez-Saiz
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IIS-IP), Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - Manel Jordana
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
23
|
Catherine J, Roufosse F. What does elevated TARC/CCL17 expression tell us about eosinophilic disorders? Semin Immunopathol 2021; 43:439-458. [PMID: 34009399 PMCID: PMC8132044 DOI: 10.1007/s00281-021-00857-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/14/2021] [Indexed: 12/19/2022]
Abstract
Eosinophilic disorders encompass a large spectrum of heterogeneous diseases sharing the presence of elevated numbers of eosinophils in blood and/or tissues. Among these disorders, the role of eosinophils can vary widely, ranging from a modest participation in the disease process to the predominant perpetrator of tissue damage. In many cases, eosinophilic expansion is polyclonal, driven by enhanced production of interleukin-5, mainly by type 2 helper cells (Th2 cells) with a possible contribution of type 2 innate lymphoid cells (ILC2s). Among the key steps implicated in the establishment of type 2 immune responses, leukocyte recruitment toward inflamed tissues is particularly relevant. Herein, the contribution of the chemo-attractant molecule thymus and activation-regulated chemokine (TARC/CCL17) to type 2 immunity will be reviewed. The clinical relevance of this chemokine and its target, C-C chemokine receptor 4 (CCR4), will be illustrated in the setting of various eosinophilic disorders. Special emphasis will be put on the potential diagnostic, prognostic, and therapeutic implications related to activation of the TARC/CCL17-CCR4 axis.
Collapse
Affiliation(s)
- Julien Catherine
- Department of Internal Medicine, Hôpital Erasme, 808 Route de Lennik, 1070, Brussels, Belgium. .,Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Gosselies, Brussels, Belgium.
| | - Florence Roufosse
- Department of Internal Medicine, Hôpital Erasme, 808 Route de Lennik, 1070, Brussels, Belgium.,Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Gosselies, Brussels, Belgium
| |
Collapse
|
24
|
Ahamad N, Kar A, Mehta S, Dewani M, Ravichandran V, Bhardwaj P, Sharma S, Banerjee R. Immunomodulatory nanosystems for treating inflammatory diseases. Biomaterials 2021; 274:120875. [PMID: 34010755 DOI: 10.1016/j.biomaterials.2021.120875] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/26/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023]
Abstract
Inflammatory disease (ID) is an umbrella term encompassing all illnesses involving chronic inflammation as the central manifestation of pathogenesis. These include, inflammatory bowel diseases, hepatitis, pulmonary disorders, atherosclerosis, myocardial infarction, pancreatitis, arthritis, periodontitis, psoriasis. The IDs create a severe burden on healthcare and significantly impact the global socio-economic balance. Unfortunately, the standard therapies that rely on a combination of anti-inflammatory and immunosuppressive agents are palliative and provide only short-term relief. In contrast, the emerging concept of immunomodulatory nanosystems (IMNs) has the potential to address the underlying causes and prevent reoccurrence, thereby, creating new opportunities for treating IDs. The IMNs offer exquisite ability to precisely modulate the immune system for a therapeutic advantage. The nano-sized dimension of IMNs allows them to efficiently infiltrate lymphatic drainage, interact with immune cells, and subsequently to undergo rapid endocytosis by hyperactive immune cells (HICs) at inflamed sites. Thus, IMNs serve to restore dysfunctional or HICs and alleviate the inflammation. We identified that different IMNs exert their immunomodulatory action via either of the seven mechanisms to modulate; cytokine production, cytokine neutralization, cellular infiltration, macrophage polarization, HICs growth inhibition, stimulating T-reg mediated tolerance and modulating oxidative-stress. In this article, we discussed representative examples of IMNs by highlighting their rationalization, design principle, and mechanism of action in context of treating various IDs. Lastly, we highlighted technical challenges in the application of IMNs and explored the future direction of research, which could potentially help to overcome those challenges.
Collapse
Affiliation(s)
- Nadim Ahamad
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Abhinanda Kar
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Sourabh Mehta
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India; IITB-Monash Research Academy IIT Bombay, Powai, Mumbai, 400076, India
| | - Mahima Dewani
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vasanthan Ravichandran
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Prateek Bhardwaj
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shivam Sharma
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rinti Banerjee
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
25
|
Scadding GK, Scadding GW. Innate and Adaptive Immunity: ILC2 and Th2 Cells in Upper and Lower Airway Allergic Diseases. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:1851-1857. [PMID: 33618052 DOI: 10.1016/j.jaip.2021.02.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
Advances in our understanding of the immune system, with the recent discovery of a parallel set of innate T lymphocytes, the innate lymphocytes (ILCs), have led to a reassessment of the pathogenesis of allergic and eosinophilic airway disorders, including allergic rhinitis (AR), asthma, and chronic rhinosinusitis with nasal polyps. We review current understanding of both elements of type-2 inflammatory responses and their relative influence in these common conditions and consider possible impacts of this on treatment selection.
Collapse
|
26
|
Shibaguchi H, Yasutaka Y, Futagami K. Novel method to analyze cell kinetics for the rapid diagnosis and determination of the causative agent in allergy. PLoS One 2021; 16:e0246125. [PMID: 33606707 PMCID: PMC7895410 DOI: 10.1371/journal.pone.0246125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/23/2020] [Indexed: 11/28/2022] Open
Abstract
Drug-induced allergy (DIA), an unexpectedly triggered side effect of drugs used for therapeutic purposes, is a serious clinical issue that needs to be resolved because it interrupts the treatment of the primary disease. Since conventional allergy testing is insufficient to accurately predict the occurrence of DIA or to determine the drugs causing it, the development of diagnostic and predictive tools for allergic reactions is important. We demonstrated a novel method, termed high-sensitive allergy test (HiSAT), for the rapid diagnosis of allergy (within 1 hr; with true-positive diagnosis rates of 89% and 9% for patients with and without allergy-like symptoms, respectively). HiSAT analyzes the cell kinetics as an index against chemotactic factors in a patient’s serum, as different from the diagnosis using conventional methods. Once allergy has occurred, HiSAT can be used to determine the causative medicine using culture supernatants incubated with the subject’s lymphocytes and the test allergen. This test is more efficient (60%) than the lymphocyte transformation test (20%). Furthermore, in HiSAT, cell mobility significantly increases in a dose-dependent manner against supernatant incubated with lymphocytes from a subject with pollinosis collected at a time when the subject is without allergic symptoms and the antigen. The result demonstraed that HiSAT might be a promising method to rapidly diagnose DIA or to determine with high accuracy the antigen causing allergy.
Collapse
Affiliation(s)
- Hirotomo Shibaguchi
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Department of Hospital Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- * E-mail:
| | - Yuki Yasutaka
- Department of Hospital Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- Department of Health Care Management, Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka, Japan
| | - Koujiro Futagami
- Department of Hospital Pharmacy, Fukuoka University Hospital, Fukuoka, Japan
- Department of Health Care Management, Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
27
|
Lee YG, Lee SH, Hong J, Lee PH, Jang AS. Titanium dioxide particles modulate epithelial barrier protein, Claudin 7 in asthma. Mol Immunol 2021; 132:209-216. [PMID: 33483086 DOI: 10.1016/j.molimm.2021.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/15/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Epithelial barrier dysfunction is involved in allergic inflammation and asthma, due to increased exposure of sub-epithelial tissues to inhaled allergens and air pollutants. The tight junction proteins claudins (CLDNs) are important regulators of paracellular permeability. CLDN7 is expressed in the alveolar epithelium; however, its contribution to airway barrier function remains unclear. The aim of this study was to assess the effects of TiO2 on epithelial barrier function in asthma. Mice were sensitized and challenged with OVA or exposed to TiO2 on days 21-23. The effect of TiO2 on CLDN7 was assessed by ELISA, immunoblotting, and immunohistochemical analysis. The levels of CLDN7 in the plasma of patients with asthma and healthy individuals were also examined. CLDN7 levels were lower in plasma from patients with asthma compared with healthy individuals. CLDN7 levels were associated with FEV1/FVC and the blood eosinophils (%) in patients with asthma. Although CLDN7 expression was elevated in the lungs of mice with asthma and in NHBE cells treated with HDM extracts, its expression was suppressed by exposure to TiO2. p-AKT and p-ERK was increased in asthmatic mice and decreased in mice with TiO2 treatment. p-AKT and p-ERK was decreased in NHBE cells treated with TiO2 and HDM extracts. Trans-epithelial electrical resistance (TEER) was higher in NHBE cells treated with TiO2 or HDM extracts; however, this was decreased by concurrent TiO2 and HDM extracts treatment. Our data suggest that particulate matter contributes to airway epithelial barrier dysfunction and results in airway inflammation and responsiveness.
Collapse
Affiliation(s)
- Yun-Gi Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Sun-Hye Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Jisu Hong
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Pureun-Haneul Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - An-Soo Jang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea.
| |
Collapse
|
28
|
Inhibition of miR-99a-5p prevents allergen-driven airway exacerbations without compromising type-2 memory responses in the intestine following helminth infection. Mucosal Immunol 2021; 14:912-922. [PMID: 33846533 PMCID: PMC8222002 DOI: 10.1038/s41385-021-00401-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/16/2021] [Accepted: 03/14/2021] [Indexed: 02/04/2023]
Abstract
Acute exacerbations (AE) of asthma, remain one of the biggest concerns for patients living with asthma. As such, identifying the causes, the molecular mechanisms involved and new therapeutic interventions to prevent AE is a high priority. Immunity to intestinal helminths involves the reactivation of type-2 immune responses leading to smooth muscle contraction and mucus hypersecretion-physiological processes very similar to acute exacerbations in the airways following allergen exposure. In this study, we employed a murine model of intestinal helminth infection, using Heligmosomoides polygyrus, to identify miRNAs during active expulsion, as a system for the identification of miRNAs that may contribute to AE in the airways. Concomitant with type-2 immunity and expulsion of H. polygyrus, we identified miR-99a-5p, miR-148a-3p and miR-155-5p that were differentially regulated. Systemic inhibition of these miRNAs, alone or in combination, had minimal impact on expulsion of H. polygyrus, but inhibition of miR-99a-5p or miR-155-5p significantly reduced house dust mite (HDM)-driven acute inflammation, modelling human acute exacerbations. Immunological, pathological and transcriptional analysis identified that miR-155-5p or miR-99a-5p contribute significantly to HDM-driven AE and that transient inhibition of these miRNAs may provide relief from allergen-driven AE, without compromising anti-helminth immunity in the gut.
Collapse
|
29
|
Korde A, Ahangari F, Haslip M, Zhang X, Liu Q, Cohn L, Gomez JL, Chupp G, Pober JS, Gonzalez A, Takyar SS. An endothelial microRNA-1-regulated network controls eosinophil trafficking in asthma and chronic rhinosinusitis. J Allergy Clin Immunol 2020; 145:550-562. [PMID: 32035607 DOI: 10.1016/j.jaci.2019.10.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/25/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Airway eosinophilia is a prominent feature of asthma and chronic rhinosinusitis (CRS), and the endothelium plays a key role in eosinophil trafficking. To date, microRNA-1 (miR-1) is the only microRNA known to be regulated in the lung endothelium in asthma models. OBJECTIVE We sought to determine the role of endothelial miR-1 in allergic airway inflammation. METHODS We measured microRNA and mRNA expression using quantitative RT-PCR. We used ovalbumin and house dust mite models of asthma. Endothelium-specific overexpression of miR-1 was achieved through lentiviral vector delivery or induction of a transgene. Tissue eosinophilia was quantified by using Congo red and anti-eosinophil peroxidase staining. We measured eosinophil binding with a Sykes-Moore adhesion chamber. Target recruitment to RNA-induced silencing complex was assessed by using anti-Argonaute2 RNA immunoprecipitation. Surface P-selectin levels were measured by using flow cytometry. RESULTS Serum miR-1 levels had inverse correlations with sputum eosinophilia, airway obstruction, and number of hospitalizations in asthmatic patients and sinonasal tissue eosinophilia in patients with CRS. IL-13 stimulation decreased miR-1 levels in human lung endothelium. Endothelium-specific overexpression of miR-1 reduced airway eosinophilia and asthma phenotypes in murine models and inhibited IL-13-induced eosinophil binding to endothelial cells. miR-1 recruited P-selectin, thymic stromal lymphopoietin, eotaxin-3, and thrombopoietin receptor to the RNA-induced silencing complex; downregulated these genes in the lung endothelium; and reduced surface P-selectin levels in IL-13-stimulated endothelial cells. In our asthma and CRS cohorts, miR-1 levels correlated inversely with its target genes. CONCLUSION Endothelial miR-1 regulates eosinophil trafficking in the setting of allergic airway inflammation. miR-1 has therapeutic potential in asthmatic patients and patients with CRS.
Collapse
Affiliation(s)
- Asawari Korde
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Farida Ahangari
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Maria Haslip
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn; Yale School of Nursing, Orange, Conn
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Conn
| | - Qing Liu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Jordan S Pober
- Department of Immunobiology, Yale School of Medicine, New Haven, Conn
| | | | - Shervin S Takyar
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
30
|
Wu G, Zhang X, Chen X, Wang J, Yang J, Wang L, Sun S, Qi Y, Wang H, Yin Y, Xu W. Streptococcus pneumoniae aminopeptidase N regulates dendritic cells that attenuates type-2 airway inflammation in murine allergic asthma. Br J Pharmacol 2020; 177:5063-5077. [PMID: 32726465 DOI: 10.1111/bph.15216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/27/2020] [Accepted: 07/13/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Epidemiological and experimental studies suggest that microbial exposure in early childhood is linked with reduced risk to suffer asthma. Thus microbial components with immunoregulatory capabilities might serve as a preventive strategy for allergic asthma. Recently, it was identified that Streptococcus pneumoniae aminopeptidase N (PepN) could suppress T cell effector function. We sought to investigate the effect of PepN on murine allergic asthma and elucidate the underlying mechanism. EXPERIMENTAL APPROACH The effects of intranasal administration of PepN during or before sensitization were examined in ovalbumin (OVA)-induced murine allergic asthma. The roles of CD11b+ dendritic cells in PepN treated OVA-induced allergic asthma were evaluated by flow cytometry, cytokines detection and adoptive transfer. Moreover, the numbers of lung type 2 innate lymphoid cells (ILC2s) were also detected. KEY RESULTS Administration of PepN during or before sensitization attenuated type-2 airway inflammation (eosinophilia, mucus hypersecretion, Th2 cytokines production and IgE production) in allergic asthma mice. PepN reduced lung accumulation of CD11b+ dendritic cells, which was accompanied by diminished dendritic cell-attracting chemokine CCL20 production as well as CCL17 and CCL22, which are Th2-cell chemokines predominantly produced by CD11b+ dendritic cells. Adoptive transfer of BM-derived CD11b+ dendritic cells abolished the inhibitory effect of PepN on OVA-induced type-2 airway inflammation. The numbers of lung ILC2s were decreased in asthmatic mice receiving PepN. CONCLUSION AND IMPLICATIONS PepN alleviated type-2 inflammation in OVA-induced allergic asthma mice, which was mediated by regulation of lung CD11b+ dendritic cells. Our study provides a novel strategy for the prevention of allergic asthma.
Collapse
Affiliation(s)
- Guangying Wu
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xu Chen
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jian Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Yang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Si Sun
- The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Yuhong Qi
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Wenchun Xu
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Kim TK, Lee JC, Im SH, Lee MS. Amelioration of Autoimmune Diabetes of NOD Mice by Immunomodulating Probiotics. Front Immunol 2020; 11:1832. [PMID: 33013834 PMCID: PMC7496355 DOI: 10.3389/fimmu.2020.01832] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/08/2020] [Indexed: 01/01/2023] Open
Abstract
Type 1 autoimmune diabetes is an autoimmune disease characterized by specific destruction of pancreatic β-cells producing insulin. Recent studies have shown that gut microbiota and immunity are closely linked to systemic immunity, affecting the balance between pro-inflammatory and regulatory immune responses. Altered gut microbiota may be causally related to the development of immune-mediated diseases, and probiotics have been suggested to have modulatory effects on inflammatory diseases and immune disorders. We studied whether a probiotic combination that has immunomodulatory effects on several inflammatory diseases can reduce the incidence of diabetes in non-obese diabetic (NOD) mice, a classical animal model of human T1D. When Immune Regulation and Tolerance 5 (IRT5), a probiotic combination comprising Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus reuteri, Bifidobacterium bifidium, and Streptococcus thermophiles, was administered 6 times a week for 36 weeks to NOD mice, beginning at 4 weeks of age, the incidence of diabetes was significantly reduced. Insulitis score was also significantly reduced, and β-cell mass was conversely increased by IRT5 administration. IRT5 administration significantly reduced gut permeability in NOD mice. The proportion of total regulatory T cells was not changed by IRT5 administration; however, the proportion of CCR9+ regulatory T (Treg) cells expressing gut-homing receptor was significantly increased in pancreatic lymph nodes (PLNs) and lamina propria of the small intestine (SI-LP). Type 1 T helper (Th1) skewing was reduced in PLNs by IRT5 administration. IRT5 could be a candidate for an effective probiotic combination, which can be safely administered to inhibit or prevent type 1 diabetes (T1D).
Collapse
Affiliation(s)
- Tae Kang Kim
- Department of Internal Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Sin-Hyeog Im
- ImmunoBiome. Inc., Pohang, South Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Myung-Shik Lee
- Department of Internal Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Lee J, Kim B, Chu H, Zhang K, Kim H, Kim JH, Kim SH, Pan Y, Noh JY, Sun Z, Lee J, Jeong KY, Park KH, Park JW, Kupper TS, Park CO, Lee KH. FABP5 as a possible biomarker in atopic march: FABP5-induced Th17 polarization, both in mouse model and human samples. EBioMedicine 2020; 58:102879. [PMID: 32711257 PMCID: PMC7387782 DOI: 10.1016/j.ebiom.2020.102879] [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: 03/18/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 01/09/2023] Open
Abstract
Background While the incidence of patients with atopic dermatitis (AD) with atopic march (AM) showing respiratory allergy is steadily rising, the pathomechanism is still unknown. There are currently no biomarkers to predict progression of AM. Methods To explore the mechanism of AM, patients with AD and AM and healthy controls were recruited and RNA microarray, flow cytometry, quantitative real-time polymerase chain reaction, and immunofluorescence staining were performed. We also co-cultured dendritic cells and CD4+T cells with various Dermatophagoides farinae allergen fractions. Cytokine levels were evaluated using enzyme-linked immunosorbent assay. Findings Both fatty-acid-binding protein 5 (FABP5) and Th17-related genes were more highly expressed in AM. FABP5 knockdown significantly decreased Th17-inducing cytokines in keratinocytes and IL-17A in T cells from AM patients. Further confirmation was obtained using an AM mice model compared to mice without AM. Der f 1, a major D. farinae allergen, increased FABP5 and IL-17A expression in T cells from AM patients. Higher serum FABP5 levels from AM patients were positively correlated with serum IL-17A levels. Interpretation FABP5 expression, possibly enhanced by higher epicutaneous and respiratory sensitization to Der f 1, may directly promote Th17 responses in AD patients with AM. Thus, AM progression can be explained by Th17 reaction induced by FABP5. FABP5 was identified as a potential biomarker in AM. Funding This study was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (Ministry of Science and ICT; No. NRF-2017R1A2B4009568), grants of the Korean Health Technology R&D Project, Ministry for Health, Welfare & Family Affairs, and the Republic of Korea (HI13C0010, HI14C1324, HI14C1799).
Collapse
Affiliation(s)
- Jungsoo Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Department of Dermatology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Republic of Korea
| | - Bomi Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Howard Chu
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - KeLun Zhang
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeran Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ji Hye Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seo Hyeong Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youdong Pan
- Department of Dermatology & Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Harvard Medical School, Boston, MA, USA
| | - Ji Yeon Noh
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - ZhengWang Sun
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jongsun Lee
- Division of Allergy and Immunology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Yong Jeong
- Division of Allergy and Immunology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hee Park
- Division of Allergy and Immunology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung-Won Park
- Division of Allergy and Immunology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Thomas S Kupper
- Department of Dermatology & Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, Harvard Medical School, Boston, MA, USA
| | - Chang Ook Park
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Kwang Hoon Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Berghi NO, Dumitru M, Vrinceanu D, Ciuluvica RC, Simioniuc-Petrescu A, Caragheorgheopol R, Tucureanu C, Cornateanu RS, Giurcaneanu C. Relationship between chemokines and T lymphocytes in the context of respiratory allergies (Review). Exp Ther Med 2020; 20:2352-2360. [PMID: 32765714 PMCID: PMC7401840 DOI: 10.3892/etm.2020.8961] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Allergic diseases have been classified in the last decades using various theories. The main classes of the newest classification in allergic respiratory diseases focus on the characterization of the endotype (which takes into account biomarkers related to determinant pathophysiological mechanisms) and of the phenotype (based on the description of the disease). Th2, Th1 and Th17 lymphocytes and the type of inflammatory response mediated by them represent the basis for Th2 and non-Th2 endotype classification. In addition, new lymphocytes were also used to characterize allergic diseases: Th9 lymphocytes, Th22 lymphocytes, T follicular helper cells (TFH) lymphocytes and invariant natural killer T (iNKT) lymphocytes. In the last decade, a growing body of evidence focused on chemokines, chemoattractant cytokines, which seems to have an important contribution to the pathogenesis of this pathology. This review presents the interactions between chemokines and Th lymphocytes in the context of Th2/non-Th2 endotype classification of respiratory allergies.
Collapse
Affiliation(s)
- Nicolae Ovidiu Berghi
- Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 011461 Bucharest, Romania
| | - Mihai Dumitru
- Anatomy Department, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daniela Vrinceanu
- ENT Department, 'Carol Davila' University of Medicine and Pharmacy, 010271 Bucharest, Romania
| | | | - Anca Simioniuc-Petrescu
- ENT Department, 'Carol Davila' University of Medicine and Pharmacy, 010271 Bucharest, Romania
| | - Ramona Caragheorgheopol
- Immunology Laboratory, 'Cantacuzino' National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Catalin Tucureanu
- Immunology Laboratory, 'Cantacuzino' National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Roxana Sfrent Cornateanu
- Department of Physiopathology and Immunology, 'Carol Davila' University of Medicine and Pharmacy, 041914 Bucharest, Romania
| | - Calin Giurcaneanu
- Department of Oncologic Dermatology, 'Elias' Emergency University Hospital, 'Carol Davila' University of Medicine and Pharmacy, 011461 Bucharest, Romania
| |
Collapse
|
34
|
Pietka W, Sundnes O, Hammarström C, Zucknick M, Khnykin D, Haraldsen G. Lack of interleukin-33 and its receptor does not prevent calcipotriol-induced atopic dermatitis-like inflammation in mice. Sci Rep 2020; 10:6451. [PMID: 32296080 PMCID: PMC7160114 DOI: 10.1038/s41598-020-63410-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/20/2020] [Indexed: 01/01/2023] Open
Abstract
Current studies addressing the influence of interleukin-33 or its receptor (IL-33R/ST2) on development of atopic dermatitis-like inflammation in mice have reported conflicting results. We compared the response in single- and double-deficient IL-33−/−/ST2−/− C57BL/6J BomTac mice in the well-established calcipotriol-induced model of atopic dermatitis. All genotypes (groups of up to 14 mice) developed atopic dermatitis-like inflammation yet we observed no biologically relevant difference between groups in gross anatomy or ear thickness. Moreover, histological examination of skin revealed no differences in mononuclear leukocyte and granulocyte infiltration nor Th2 cytokine levels (IL-4 and IL-13). Finally, skin CD45+ cells and CD3+ cells were found at similar densities across all groups. Our findings indicate that lack of interleukin-33 and its receptor ST2 does not prevent the development of AD-like skin inflammation.
Collapse
Affiliation(s)
- Wojciech Pietka
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Olav Sundnes
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Rheumatology, Dermatology and Infectious Diseases, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Clara Hammarström
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Manuela Zucknick
- Oslo Center for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Denis Khnykin
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Oslo, Norway. .,Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
35
|
Lai JF, Thompson LJ, Ziegler SF. TSLP drives acute T H2-cell differentiation in lungs. J Allergy Clin Immunol 2020; 146:1406-1418.e7. [PMID: 32304753 DOI: 10.1016/j.jaci.2020.03.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/14/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is an epithelial-derived cytokine that is important for the development of type 2 inflammatory responses at mucosal surfaces. OBJECTIVE In humans, TSLP level has been found to be elevated in the lungs of patients with asthma, and in mouse models, TSLP can promote type 2 airway inflammation, primarily through the activation of dendritic cells. However, the mechanisms underlying its role remain unclear. The objective of this study was to provide a mechanistic analysis of TSLP-mediated type 2 airway inflammation METHODS: To dissect the mechanisms of TSLP-mediated type 2 responses, mice were treated with TSLP and antigen to evaluate cellular immune responses. Flow cytometric analyses were used to follow responses in the airways, and conditional deletion of TSLP receptor and adoptive transfer were used to identify the cellular subsets involved in this inflammatory response. RESULTS We showed that TSLP can directly promote TH2-cell differentiation in the lung, independent of the draining lymph nodes. We also identified a population of patrolling monocytes/interstitial macrophages (IMs) (CD11c-expressing IMs) that are both necessary and sufficient for TSLP-mediated TH2-cell differentiation and airway inflammation. TH2-cell-driven airway eosinophilia is attenuated by ablation of CD11c-expressing IMs or by selective deficiency of TSLP receptor signaling in these cells. More importantly, CD11c-expressing IMs are sufficient for the induction of acute TH2-cell responses in the lungs that is independent of dendritic cells and T-cell priming in the draining lymph nodes. CONCLUSION These findings indicate a novel mechanistic role for TSLP and CD11c-expressing IMs in the development of acute TH2-cell-dependent allergic airway inflammation. This work also demonstrates a new role for TSLP in promoting type 2 responses directly in the lung.
Collapse
Affiliation(s)
- Jen-Feng Lai
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | | | | |
Collapse
|
36
|
Ogita T, Yamamoto Y, Mikami A, Shigemori S, Sato T, Shimosato T. Oral Administration of Flavonifractor plautii Strongly Suppresses Th2 Immune Responses in Mice. Front Immunol 2020; 11:379. [PMID: 32184789 PMCID: PMC7058663 DOI: 10.3389/fimmu.2020.00379] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
The bacterium Flavonifractor plautii (FP), which is found in human feces, has been reported to participate in catechin metabolism in the gut, but this bacterium's effects on immune function are unclear. We assessed the effect of oral administration of FP on the immune response in ovalbumin (OVA) -sensitized mice. We demonstrated that the FP treatment suppressed interleukin (IL)-4 in splenocytes and OVA-specific IgE production in serum from OVA-sensitized mice. Moreover, oral administration of FP augmented CD4+CD25+ T cells and CD103+CD11c+ DCs. In animals of the FP group, the proportion of FP was increased in the mesenteric lymph nodes (MLNs), as was the proportion of Deferribacteres in the cecum. Oral administration of FP may inhibit the Th2 immune response by incorporation into the MLNs and/or by inducing changes in the gut microbiota. Thus, FP may be useful in alleviating antigen-induced Th2 immune responses.
Collapse
Affiliation(s)
- Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Yoshinari Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ayane Mikami
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Suguru Shigemori
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takashi Sato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
37
|
Hossain FMA, Park SO, Kim HJ, Eo JC, Choi JY, Uyangaa E, Kim B, Kim K, Eo SK. CCR5 attenuates neutrophilic airway inflammation exacerbated by infection with rhinovirus. Cell Immunol 2020; 351:104066. [PMID: 32089258 DOI: 10.1016/j.cellimm.2020.104066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 01/13/2020] [Accepted: 02/14/2020] [Indexed: 12/15/2022]
Abstract
Human rhinovirus (hRV) is the most common cause of asthma exacerbation characterized by clinical and pathophysiological heterogeneity. Steroid-sensitive, Th2 type-eosinophilic asthma has been somewhat studied, but hRV-induced neutrophilic asthma exacerbation is poorly understood. Here, CCR5 was found to play a role in attenuating neutrophilic airway inflammation in hRV-induced asthma exacerbation using chicken ovalbumin (OVA)-based model. CCR5 deficiency resulted in exacerbated neutrophilic asthmatic responses in airways following hRV infection. CCR5-deficient mice showed enhanced mucus expression and altered expression of tight junction proteins in lung tissues. CCR5-deficient mice were also manifested with influx of CD45+CD11b+Siglec-F+Gr-1+ neutrophils, along with enhanced production of IL-17A, IFN-γ, IL-6, IL-1β cytokines in inflamed tissues. In contrast, CCR5-deficient mice elicited down-regulation of Th2-related cytokine proteins following hRV infection. More interestingly, the lack of CCR5 altered the equilibrium of CD4+FoxP3+ Tregs and IL-17+CD4+ Th17 in inflamed tissues. CCR5-deficient mice showed increased frequency and absolute number of IL-17-producing CD4+ Th17 cells in lung tissues compared to wild-type mice, whereas the reduced infiltration of CD4+FoxP3+ Treg cells was observed. CCR5 deficiency resulted in the skewed production of Th17 and Th1 cytokines in lymph nodes and lungs upon OVA stimulation. Likewise, CCR5-deficient mice showed enhanced expression of Th17-inducing cytokines (IL-1β, IL-6, and TNF-α) in lung tissues. These results imply that CCR5 deficiency facilitates Th17 airway inflammation during hRV-induced asthma exacerbation, along with suppressing Th2 responses. Furthermore, our results suggest that CCR5 attenuates hRV-induced neutrophilic airway inflammation through conserving the equilibrium of CD4+Foxp3+ Treg cells and IL-17+CD4+ Th17 cells in hRV-induced asthma exacerbation.
Collapse
Affiliation(s)
- Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea; Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Seong Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Hyo Jin Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Jun Cheol Eo
- Division of Biotechnology, College of Environmental & Biosource Science, Jeonbuk National University, Iksan 54596, South Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea.
| |
Collapse
|
38
|
Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García FJ. Mitochondria: An Integrative Hub Coordinating Circadian Rhythms, Metabolism, the Microbiome, and Immunity. Front Cell Dev Biol 2020; 8:51. [PMID: 32117978 PMCID: PMC7025554 DOI: 10.3389/fcell.2020.00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
There is currently some understanding of the mechanisms that underpin the interactions between circadian rhythmicity and immunity, metabolism and immune response, and circadian rhythmicity and metabolism. In addition, a wealth of studies have led to the conclusion that the commensal microbiota (mainly bacteria) within the intestine contributes to host homeostasis by regulating circadian rhythmicity, metabolism, and the immune system. Experimental studies on how these four biological domains interact with each other have mainly focused on any two of those domains at a time and only occasionally on three. However, a systematic analysis of how these four domains concurrently interact with each other seems to be missing. We have analyzed current evidence that signposts a role for mitochondria as a key hub that supports and integrates activity across all four domains, circadian clocks, metabolic pathways, the intestinal microbiota, and the immune system, coordinating their integration and crosstalk. This work will hopefully provide a new perspective for both hypothesis-building and more systematic experimental approaches.
Collapse
Affiliation(s)
- Bruno A Aguilar-López
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Hazel M Dockrell
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Francisco Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
39
|
Tindemans I, Joosse ME, Samsom JN. Dissecting the Heterogeneity in T-Cell Mediated Inflammation in IBD. Cells 2020; 9:E110. [PMID: 31906479 PMCID: PMC7016883 DOI: 10.3390/cells9010110] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022] Open
Abstract
Infiltration of the lamina propria by inflammatory CD4+ T-cell populations is a key characteristic of chronic intestinal inflammation. Memory-phenotype CD4+ T-cell frequencies are increased in inflamed intestinal tissue of IBD patients compared to tissue of healthy controls and are associated with disease flares and a more complicated disease course. Therefore, a tightly controlled balance between regulatory and inflammatory CD4+ T-cell populations is crucial to prevent uncontrolled CD4+ T-cell responses and subsequent intestinal tissue damage. While at steady state, T-cells display mainly a regulatory phenotype, increased in Th1, Th2, Th9, Th17, and Th17.1 responses, and reduced Treg and Tr1 responses have all been suggested to play a role in IBD pathophysiology. However, it is highly unlikely that all these responses are altered in each individual patient. With the rapidly expanding plethora of therapeutic options to inhibit inflammatory T-cell responses and stimulate regulatory T-cell responses, a crucial need is emerging for a robust set of immunological assays to predict and monitor therapeutic success at an individual level. Consequently, it is crucial to differentiate dominant inflammatory and regulatory CD4+ T helper responses in patients and relate these to disease course and therapy response. In this review, we provide an overview of how intestinal CD4+ T-cell responses arise, discuss the main phenotypes of CD4+ T helper responses, and review how they are implicated in IBD.
Collapse
Affiliation(s)
| | | | - Janneke N. Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
40
|
Lee PH, Kim BG, Park MK, Hong J, Lee YG, Jang AS. The Impact of Diesel Exhaust Particles on Tight Junctional Proteins on Nose and Lung in a Mouse Model. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 13:350-352. [PMID: 33474868 PMCID: PMC7840873 DOI: 10.4168/aair.2021.13.2.350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Pureun Haneul Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Byeong Gon Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Moo Kyun Park
- Department of Otolaryngology, Seoul National University, Seoul, Korea
| | - Jisu Hong
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Yun Gi Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - An Soo Jang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| |
Collapse
|
41
|
Plundrich NJ, Smith AR, Borst LB, Snider DB, Käser T, Dellon ES, Blikslager AT, Odle J, Lila MA, Laster SM. Oesophageal eosinophilia accompanies food allergy to hen egg white protein in young pigs. Clin Exp Allergy 2020; 50:95-104. [PMID: 31702085 PMCID: PMC6930966 DOI: 10.1111/cea.13527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Esophagitis with eosinophilia, inflammation, and fibrosis represent a chronic condition in humans with food allergies. OBJECTIVE In this investigation, we asked whether esophagitis with an eosinophilic component is observed in young pigs rendered allergic to hen egg white protein (HEWP). METHODS Food allergy was induced in young pigs using two protocols. In one protocol, sensitized pigs were challenged by gavage with a single dose of HEWP. Clinical signs were monitored for 24 hours, and then, gastrointestinal (GI) tissues were collected for histological examination. The phenotype of circulating, ovalbumin (OVA)-specific T cells also was examined in HEWP challenged animals. In the second protocol, sensitized animals were fed HEWP for 28 days. Animals were then examined by endoscopy and gastrointestinal tissues collected for histological examination. RESULTS In pigs challenged by gavage with HEWP, clinical signs were noted in 5/6 pigs including diarrhoea, emesis, and skin rash. Clinical signs were not seen in any control group. Histological analysis revealed significant levels of oesophageal eosinophilic infiltration (P < .05) in 4/6 of these animals, with two also displaying eosinophilic infiltration in the stomach. Eosinophils were not increased in ileum or colon samples. Increased numbers of circulating, OVA-specific CD4+ T cells also were observed in pigs that received HEWP by gavage. In the group of animals fed HEWP, endoscopy revealed clinical signs of esophagitis including oedema, granularity, white spots, and furrowing, while histology revealed oedema, immune cell infiltration, and basal zone hyperplasia. CONCLUSIONS AND CLINICAL RELEVANCE Food allergy in the pig can be associated with esophagitis based on histological and endoscopic findings, including eosinophilic infiltration. The young pig may, therefore, be a useful large animal model for the study of eosinophilic esophagitis in humans.
Collapse
Affiliation(s)
- Nathalie J. Plundrich
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Andrew R. Smith
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Luke B. Borst
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Douglas B. Snider
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine and Translational Research Training Program, Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Tobias Käser
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Evan S. Dellon
- Center for Esophageal Diseases and Swallowing, Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anthony T. Blikslager
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Jack Odle
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Mary Ann Lila
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Scott M. Laster
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
42
|
Kikuchi T, Nakae J, Kawano Y, Watanabe N, Onodera M, Itoh H. Foxo in T Cells Regulates Thermogenic Program through Ccr4/Ccl22 Axis. iScience 2019; 22:81-96. [PMID: 31756626 PMCID: PMC6880116 DOI: 10.1016/j.isci.2019.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 09/21/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
Crosstalk between immunity and the thermogenic program has provided insight into metabolic energy regulation. Here, we generated thermogenic program-accelerating mice (T-QKO), in which Foxo1 is knockout and Foxo3 is hetero-knockout in CD4+ T cells. T-QKO exhibit lean phenotype under HFD due to increased energy expenditure. Cold exposure significantly increased expression of the thermogenic genes (Ppargc1a and Ucp1), Th2 cytokines (Il4 and Il13), and Th2 marker gene (Gata3) in subcutaneous adipose tissue (SC) of T-QKO. Furthermore, Ccr4 expression was significantly increased in Th2 cells of T-QKO, and cold exposure induced Ccl22 expression in SC, leading to increased accumulation of Th2 cell population in SC of T-QKO. These data reveal a mechanism by which cold exposure induces selective recruitment of Th2 cells into SC, leading to regulation of energy expenditure by generating beige adipocyte and suggest that inhibition of Foxo in T cells may support a strategy to prevent and treat obesity. T-QKO increases Gata3 and Ccr4 expression in CD4+ T cells Cold exposure increases Ccl22 expression in subcutaneous adipose tissue Cold exposure increases SC-specific recruitment of Th2 cells in T-QKO Recruited Th2 cells secrete IL-4 and IL-13 and increase beiging of adipocytes
Collapse
Affiliation(s)
- Tetsuhiro Kikuchi
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jun Nakae
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Physiology, International University of Health and Welfare School of Medicine, Narita 286-8686, Japan.
| | - Yoshinaga Kawano
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Hiroshi Itoh
- Navigation Medicine of Kidney and Metabolism, Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
43
|
Rudulier CD, Tonti E, James E, Kwok WW, Larché M. Modulation of CRTh2 expression on allergen-specific T cells following peptide immunotherapy. Allergy 2019; 74:2157-2166. [PMID: 31077596 PMCID: PMC6817377 DOI: 10.1111/all.13867] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/15/2019] [Accepted: 03/01/2019] [Indexed: 12/28/2022]
Abstract
Background Allergen immunotherapy using synthetic peptide T‐cell epitopes (Cat‐PAD) from the major cat allergen Fel d 1 has been shown, in allergen exposure studies, to significantly reduce symptoms of allergic rhinoconjunctivitis in cat‐allergic subjects. However, the immunological mechanisms underlying clinical benefit remain only partially understood. Since previous studies of whole allergen immunotherapy demonstrated a reduction in the frequency of allergen‐specific (MHC II tetramer+) CD4+ T cells expressing the chemokine receptor CRTh2, we assessed the impact of Cat‐PAD on the frequency and functional phenotype of Fel d 1‐specific CD4+ T cells. Methods Using before and after treatment samples from subjects enrolled in a randomized, double‐blind, placebo‐controlled trial of Cat‐PAD, we employed Fel d 1 MHC II tetramers and flow cytometry to analyze the expression of chemokine receptors CCR3, CCR4, CCR5, CXCR3, and CRTh2, together with markers of memory phenotype (CD27 and CCR7) on Fel d 1‐specific CD4+ T cells. Results No statistically significant change in the frequency of Fel d 1‐specific CD4+ T cells, nor in their expression of chemokine receptors or memory phenotype, was observed. However, a significant reduction in cell surface expression of CRTh2 was observed between the placebo and active groups (P = 0.047). Conclusions Peptide immunotherapy with Cat‐PAD does not significantly alter the frequency or phenotype of Fel d 1‐CD4+ T cells, but may decrease their expression of CRTh2.
Collapse
Affiliation(s)
- Christopher D. Rudulier
- Department of Medicine, Division of Clinical Immunology & Allergy McMaster University Hamilton Ontario Canada
| | - Elena Tonti
- Department of Medicine, Division of Clinical Immunology & Allergy McMaster University Hamilton Ontario Canada
| | - Eddie James
- Benaroya Research Institute at Virginia Mason Seattle Washington
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason Seattle Washington
| | - Mark Larché
- Department of Medicine, Division of Clinical Immunology & Allergy McMaster University Hamilton Ontario Canada
- Department of Medicine, Division of Respirology Firestone Institute for Respiratory Health, The Research Institute at St. Joe’s Hamilton Ontario Canada
| |
Collapse
|
44
|
Chen M, Reed RR, Lane AP. Chronic Inflammation Directs an Olfactory Stem Cell Functional Switch from Neuroregeneration to Immune Defense. Cell Stem Cell 2019; 25:501-513.e5. [PMID: 31523027 DOI: 10.1016/j.stem.2019.08.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/08/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Although olfactory mucosa possesses long-lived horizontal basal stem cells (HBCs) and remarkable regenerative capacity, the function of human olfactory neuroepithelium is significantly impaired in chronic inflammatory rhinosinusitis. Here, we show that, while inflammation initially damages olfactory neurons and activates HBC-mediated regeneration, continued inflammation locks HBCs in an undifferentiated state. Global gene expression in mouse HBCs reveals broad upregulation of NF-κB-regulated cytokines and chemokines including CCL19, CCL20, and CXCL10, accompanied by enhancement of "stemness"-related transcription factors. Loss-of-function studies identify an NF-κB-dependent role of HBCs in amplifying inflammatory signaling, contributing to macrophage and T cell local proliferation. Chronically activated HBCs signal macrophages to maintain immune defense and prevent Treg development. In diseased human olfactory tissue, activated HBCs in a P63+ undifferentiated state similarly contribute to inflammation through chemokine production. These observations establish a mechanism of chronic rhinosinusitis-associated olfactory loss, caused by a functional switch of neuroepithelial stem cells from regeneration to immune defense.
Collapse
Affiliation(s)
- Mengfei Chen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Randall R Reed
- Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew P Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
45
|
Mousavinasab F, Babaie D, Nilipour Y, Mansouri M, Imanzadeh F, Dara N, Rohani P, Khatami K, Sayyari A, Khoddami M, Kazemiaghdam M, Mesdaghi M. Increased number of regulatory T cells in esophageal tissue of patients with eosinophilic esophagitis in comparison to gastro esophageal reflux disease and control groups. Allergol Immunopathol (Madr) 2019; 47:431-436. [PMID: 31178311 DOI: 10.1016/j.aller.2019.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a primarily polygenic allergic disorder. Although most patients have IgE sensitization, it seems that non-IgE mediated responses mainly contribute to the pathogenesis of EoE. Regulatory T cells (Tregs) may have an important role in allergies. There are limited data on the association of Tregs and EoE. In this study, we enumerated and compared T lymphocytes and Tregs in esophageal tissue of patients with EoE, gastroesophageal reflux disease (GERD) and normal controls. METHODS Ten patients with EoE, ten patients with GERD and eight normal controls were included. Immunohistochemistry staining was used to enumerate T lymphocytes and Tregs. CD3+ cells were considered as T cells and FOXP3+, CD3+ cells were considered as Tregs. T cells and Tregs were counted in 10 high power fields (HPF) (×400) for each patient and the average of 10 HPFs was recorded. RESULTS The mean±SEM of Tregs in esophageal tissue of patients with EoE (10.90±2.14cells/HPF) was significantly higher than the GERD (2.77±0.66cells/HPF) and control groups (0.37±0.08cells/HPF) (P<0.001). Additionally, the mean±SEM of T lymphocytes in esophageal tissue of patients with EoE (24.39±3.86cells/HPF) were increased in comparison to the GERD (10.07±2.65cells/HPF) and control groups (3.17±0.93cells/HPF) (P<0.001). CONCLUSION There is an increase in the number of esophageal T lymphocytes and regulatory T cells in patients with EoE compared to the GERD and control groups.
Collapse
|
46
|
Rasmark Roepke E, Bruno V, Nedstrand E, Boij R, Strid CP, Piccione E, Berg G, Svensson-Arvelund J, Jenmalm MC, Rubér M, Ernerudh J. Low-molecular-weight-heparin increases Th1- and Th17-associated chemokine levels during pregnancy in women with unexplained recurrent pregnancy loss: a randomised controlled trial. Sci Rep 2019; 9:12314. [PMID: 31444404 PMCID: PMC6707182 DOI: 10.1038/s41598-019-48799-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/13/2019] [Indexed: 11/29/2022] Open
Abstract
Low-molecular-weight heparin (LMWH) is widely used to treat recurrent pregnancy loss (RPL) because of its anti-coagulant effects. Although in vitro studies have suggested additional immunological effects, these are debated. We therefore investigated whether LMWH could modulate immune responses in vivo during pregnancy of women with unexplained RPL. A Swedish open multi-centre randomised controlled trial included 45 women treated with tinzaparin and 42 untreated women. Longitudinally collected plasma samples were obtained at gestational weeks (gw) 6, 18, 28 and 34 and analysed by multiplex bead technology for levels of 11 cytokines and chemokines, chosen to represent inflammation and T-helper subset-associated immunity. Mixed linear models test on LMWH-treated and untreated women showed differences during pregnancy of the Th1-associated chemokines CXCL10 (p = 0.01), CXCL11 (p < 0.001) and the Th17-associated chemokine CCL20 (p = 0.04), while CCL2, CCL17, CCL22, CXCL1, CXCL8, CXCL12, CXCL13 and IL-6 did not differ. Subsequent Student's t-test showed significantly higher plasma levels of CXCL10 and CXCL11 in treated than untreated women at gw 28 and 34. The consistent increase in the two Th1-associated chemokines suggests a potential proinflammatory and unfavourable effect of LMWH treatment during later stages of pregnancy, when Th1 immunity is known to disrupt immunological tolerance.
Collapse
Affiliation(s)
- E Rasmark Roepke
- Department of Obstetrics and Gynecology, Skåne University Hospital, Malmö and Lund University, Lund, Sweden.
| | - V Bruno
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Section of Gynecology and Obstetrics, Academic Department of Biomedicine and Prevention, and Clinical Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| | - E Nedstrand
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - R Boij
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C Petersson Strid
- Departmen of Obstetrics and Gynecology, Kalmar Hospital, Kalmar, Sweden
| | - E Piccione
- Section of Gynecology and Obstetrics, Academic Department of Biomedicine and Prevention, and Clinical Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| | - G Berg
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - J Svensson-Arvelund
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M C Jenmalm
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M Rubér
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - J Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
47
|
Gaylo-Moynihan A, Prizant H, Popović M, Fernandes NRJ, Anderson CS, Chiou KK, Bell H, Schrock DC, Schumacher J, Capece T, Walling BL, Topham DJ, Miller J, Smrcka AV, Kim M, Hughson A, Fowell DJ. Programming of Distinct Chemokine-Dependent and -Independent Search Strategies for Th1 and Th2 Cells Optimizes Function at Inflamed Sites. Immunity 2019; 51:298-309.e6. [PMID: 31399281 PMCID: PMC6904228 DOI: 10.1016/j.immuni.2019.06.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/18/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
T-helper (Th) cell differentiation drives specialized gene programs that dictate effector T cell function at sites of infection. Here, we have shown Th cell differentiation also imposes discrete motility gene programs that shape Th1 and Th2 cell navigation of the inflamed dermis. Th1 cells scanned a smaller tissue area in a G protein-coupled receptor (GPCR) and chemokine-dependent fashion, while Th2 cells scanned a larger tissue area independent of GPCR signals. Differential chemokine reliance for interstitial migration was linked to STAT6 transcription-factor-dependent programming of integrin αVβ3 expression: Th2 cell differentiation led to high αVβ3 expression relative to Th1 cells. Th1 and Th2 cell modes of motility could be switched simply by manipulating the amount of αVβ3 on the cell surface. Deviating motility modes from those established during differentiation impaired effector function. Thus, programmed expression of αVβ3 tunes effector T cell reliance on environmental cues for optimal exploration of inflamed tissues.
Collapse
Affiliation(s)
- Alison Gaylo-Moynihan
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hen Prizant
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Milan Popović
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ninoshka R J Fernandes
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Christopher S Anderson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin K Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah Bell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dillon C Schrock
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Justin Schumacher
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Tara Capece
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brandon L Walling
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jim Miller
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Angela Hughson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah J Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
48
|
Da Rocha S, Bigot J, Onodi F, Cosette J, Corre G, Poupiot J, Fenard D, Gjata B, Galy A, Neildez-Nguyen TMA. Temporary Reduction of Membrane CD4 with the Antioxidant MnTBAP Is Sufficient to Prevent Immune Responses Induced by Gene Transfer. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:285-299. [PMID: 31497619 PMCID: PMC6718808 DOI: 10.1016/j.omtm.2019.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/29/2019] [Indexed: 12/14/2022]
Abstract
Unexpectedly, the synthetic antioxidant MnTBAP was found to cause a rapid and reversible downregulation of CD4 on T cells in vitro and in vivo. This effect resulted from the internalization of membrane CD4 T cell molecules into clathrin-coated pits and involved disruption of the CD4/p56Lck complex. The CD4 deprivation induced by MnTBAP had functional consequences on CD4-dependent infectious processes or immunological responses as shown in various models, including gene therapy. In cultured human T cells, MnTBAP-induced downregulation of CD4 functionally suppressed gp120- mediated lentiviral transduction in a model relevant for HIV infection. The injection of MnTBAP in mice reduced membrane CD4 on lymphocytes in vivo within 5 days of treatment, preventing OVA peptide T cell immunization while allowing subsequent immunization once treatment was stopped. In a mouse gene therapy model, MnTBAP treatment at the time of adenovirus-associated virus (AAV) vector administration, successfully controlled the induction of anti-transgene and anti-capsid immune responses mediated by CD4+ T cells, enabling the redosing mice with the same vector. These functional data provide new avenues to develop alternative therapeutic immunomodulatory strategies based on temporary regulation of CD4. These could be particularly useful for AAV gene therapy in which novel strategies for redosing are needed.
Collapse
Affiliation(s)
- Sylvie Da Rocha
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | - Jérémy Bigot
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | - Fanny Onodi
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | | | - Guillaume Corre
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | - Jérôme Poupiot
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | - David Fenard
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | | | - Anne Galy
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| | - Thi My Anh Neildez-Nguyen
- Ecole Pratique des Hautes Etudes, PSL Research University, INTEGRARE UMR_S951, INSERM, Généthon, Univ-Evry, 91002 Evry, France
| |
Collapse
|
49
|
Nieto-Fontarigo JJ, González-Barcala FJ, San José E, Arias P, Nogueira M, Salgado FJ. CD26 and Asthma: a Comprehensive Review. Clin Rev Allergy Immunol 2019; 56:139-160. [PMID: 27561663 PMCID: PMC7090975 DOI: 10.1007/s12016-016-8578-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asthma is a heterogeneous and chronic inflammatory family of disorders of the airways with increasing prevalence that results in recurrent and reversible bronchial obstruction and expiratory airflow limitation. These diseases arise from the interaction between environmental and genetic factors, which collaborate to cause increased susceptibility and severity. Many asthma susceptibility genes are linked to the immune system or encode enzymes like metalloproteases (e.g., ADAM-33) or serine proteases. The S9 family of serine proteases (prolyl oligopeptidases) is capable to process peptide bonds adjacent to proline, a kind of cleavage-resistant peptide bonds present in many growth factors, chemokines or cytokines that are important for asthma. Curiously, two serine proteases within the S9 family encoded by genes located on chromosome 2 appear to have a role in asthma: CD26/dipeptidyl peptidase 4 (DPP4) and DPP10. The aim of this review is to summarize the current knowledge about CD26 and to provide a structured overview of the numerous functions and implications that this versatile enzyme could have in this disease, especially after the detection of some secondary effects (e.g., viral nasopharyngitis) in type II diabetes mellitus patients (a subset with a certain risk of developing obesity-related asthma) upon CD26 inhibitory therapy.
Collapse
Affiliation(s)
- Juan J Nieto-Fontarigo
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Francisco J González-Barcala
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Respiratory Department, Clinic University Hospital (CHUS), Santiago de Compostela, Spain
| | - Esther San José
- Clinical Analysis Service, Clinic University Hospital (CHUS), Santiago de Compostela, Spain
| | - Pilar Arias
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Montserrat Nogueira
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Francisco J Salgado
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain.
| |
Collapse
|
50
|
Abstract
The laboratory mouse Mus musculus has long been used as a model organism to test hypotheses and treatments related to understanding the mechanisms of disease in humans; however, for these experiments to be relevant, it is important to know the complex ways in which mice are similar to humans and, crucially, the ways in which they differ. In this chapter, an in-depth analysis of these similarities and differences is provided to allow researchers to use mouse models of human disease and primary cells derived from these animal models under the most appropriate and meaningful conditions. Although there are considerable differences between mice and humans, particularly regarding genetics, physiology, and immunology, a more thorough understanding of these differences and their effects on the function of the whole organism will provide deeper insights into relevant disease mechanisms and potential drug targets for further clinical investigation. Using specific examples of mouse models of human lung disease, i.e., asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis, this chapter explores the most salient features of mouse models of human disease and provides a full assessment of the advantages and limitations of these models, focusing on the relevance of disease induction and their ability to replicate critical features of human disease pathophysiology and response to treatment. The chapter concludes with a discussion on the future of using mice in medical research with regard to ethical and technological considerations.
Collapse
|