1
|
Huang YA, Wang X, Kim JC, Yao X, Sethi A, Strohm A, Doherty TA. PIP-seq identifies novel heterogeneous lung innate lymphocyte population activation after combustion product exposure. Sci Rep 2024; 14:20167. [PMID: 39215111 PMCID: PMC11364781 DOI: 10.1038/s41598-024-70880-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population that play diverse roles in airway inflammation after exposure to allergens and infections. However, how ILCs respond after exposure to environmental toxins is not well understood. Here we show a novel method for studying the heterogeneity of rare lung ILC populations by magnetic enrichment for lung ILCs followed by particle-templated instant partition sequencing (PIP-seq). Using this method, we were able to identify novel group 1 and group 2 ILC subsets that exist after exposure to both fungal allergen and burn pit-related constituents (BPC) that include dioxin, aromatic hydrocarbon, and particulate matter. Toxin exposure in combination with fungal allergen induced activation of specific ILC1/NK and ILC2 populations as well as promoted neutrophilic lung inflammation. Oxidative stress pathways and downregulation of specific ribosomal protein genes (Rpl41 and Rps19) implicated in anti-inflammatory responses were present after BPC exposure. Increased IFNγ expression and other pro-neutrophilic mediator transcripts were increased in BPC-stimulated lung innate lymphoid cells. Further, the addition of BPC induced Hspa8 (encodes HSC70) and aryl hydrocarbon transcription factor activity across multiple lung ILC subsets. Overall, using an airway disease model that develops after occupational and environmental exposures, we demonstrate an effective method to better understand heterogenous ILC subset activation.
Collapse
Affiliation(s)
- Yung-An Huang
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Xinyu Wang
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Jong-Chan Kim
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Xiang Yao
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Anshika Sethi
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Allyssa Strohm
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Taylor A Doherty
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA.
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA.
| |
Collapse
|
2
|
Schworer SA, Olbrich CL, Larsen LD, Howard E, Liu L, Koyama K, Spencer LA. Notch 2 signaling contributes to intestinal eosinophil adaptations in steady state and tissue burden following oral allergen challenge. J Leukoc Biol 2024; 116:379-391. [PMID: 38789100 PMCID: PMC11271981 DOI: 10.1093/jleuko/qiae122] [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: 11/10/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024] Open
Abstract
Eosinophils not only function as inflammatory effectors in allergic diseases, but also contribute to tissue homeostasis in steady state. Emerging data are revealing tissue eosinophils to be adaptive cells, imprinted by their local tissue microenvironment and exhibiting distinct functional phenotypes that may contribute to their homeostatic vs. inflammatory capacities. However, signaling pathways that regulate eosinophil tissue adaptations remain elusive. Notch signaling is an evolutionarily conserved pathway that mediates differential cell fate programming of both pre- and postmitotic immune cells. This study investigated a role for notch receptor 2 signaling in regulating eosinophil functions and tissue phenotype in both humans and mice. Notch 2 receptors were constitutively expressed and active in human blood eosinophils. Pharmacologic neutralization of notch 2 in ex vivo stimulated human eosinophils altered their activated transcriptome and prevented their cytokine-mediated survival. Genetic ablation of eosinophil-expressed notch 2 in mice diminished steady-state intestine-specific eosinophil adaptations and impaired their tissue retention in a food allergic response. In contrast, notch 2 had no effect on eosinophil phenotype or tissue inflammation within the context of allergic airways inflammation, suggesting that notch 2-dependent regulation of eosinophil phenotype and function is specific to the gut. These data reveal notch 2 signaling as a cell-intrinsic mechanism that contributes to eosinophil survival, function, and intestine-specific adaptations. The notch 2 pathway may represent a viable strategy to reprogram eosinophil functional phenotypes in gastrointestinal eosinophil-associated diseases.
Collapse
Affiliation(s)
- Stephen A Schworer
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Marsico Lung Institute, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Courtney L Olbrich
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, and Mucosal Inflammation Program, 12700 E. 19th Ave, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, 13123 E. 16th Ave, Children's Hospital Colorado, Aurora, CO 80045, United States
| | - Leigha D Larsen
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, and Mucosal Inflammation Program, 12700 E. 19th Ave, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, 13123 E. 16th Ave, Children's Hospital Colorado, Aurora, CO 80045, United States
| | - Emily Howard
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
| | - Linying Liu
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
| | - Kenya Koyama
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
- Department of Respiratory Medicine and Clinical Immunology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi 321-0293, Japan
| | - Lisa A Spencer
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, United States
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, and Mucosal Inflammation Program, 12700 E. 19th Ave, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, 13123 E. 16th Ave, Children's Hospital Colorado, Aurora, CO 80045, United States
| |
Collapse
|
3
|
Huang YA, Wang X, Kim JC, Yao X, Sethi A, Strohm A, Doherty TA. PIP-Seq identifies novel heterogeneous lung innate lymphocyte population activation after combustion product exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600420. [PMID: 38979234 PMCID: PMC11230265 DOI: 10.1101/2024.06.24.600420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population that play diverse roles in airway inflammation after exposure to allergens and infections. However, how ILCs respond after exposure to environmental toxins is not well understood. Here we show a novel method for studying the heterogeneity of rare lung ILC populations by magnetic enrichment for lung ILCs followed by particle-templated instant partition sequencing (PIP-seq). Using this method, we were able to identify novel group 1 and group 2 ILC subsets that exist after exposure to both fungal allergen and burn pit-related constituents (BPC) that include dioxin, aromatic hydrocarbon, and particulate matter. Toxin exposure in combination with fungal allergen induced activation of specific ILC1/NK and ILC2 populations as well as promoted neutrophilic lung inflammation. Oxidative stress pathways and downregulation of specific ribosomal protein genes ( Rpl41 and Rps19 ) implicated in anti-inflammatory responses were present after BPC exposure. Increased IFNγ expression and other pro-neutrophilic mediator transcripts were increased in BPC-stimulated lung innate lymphoid cells. Further, the addition of BPC induced Hspa8 (encodes HSC70) and aryl hydrocarbon transcription factor activity across multiple lung ILC subsets. Overall, using an airway disease model that develops after occupational and environmental exposures, we demonstrate an effective method to better understand heterogenous ILC subset activation.
Collapse
|
4
|
Ruhl A, Antão AV, Dietschmann A, Radtke D, Tenbusch M, Voehringer D. STAT6-induced production of mucus and resistin-like molecules in lung Club cells does not protect against helminth or influenza A virus infection. Eur J Immunol 2024; 54:e2350558. [PMID: 37855177 DOI: 10.1002/eji.202350558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
Airway epithelial cells contribute to a variety of lung diseases including allergic asthma, where IL-4 and IL-13 promote activation of the transcription factor STAT6. This leads to goblet cell hyperplasia and the secretion of effector molecules by epithelial cells. However, the specific effect of activated STAT6 in lung epithelial cells is only partially understood. Here, we created a mouse strain to selectively investigate the role of constitutively active STAT6 in Club cells, a subpopulation of airway epithelial cells. CCSP-Cre_STAT6vt mice and bronchiolar organoids derived from these show an enhanced expression of the chitinase-like protein Chil4 (Ym2) and resistin-like molecules (Relm-α, -β, -γ). In addition, goblet cells of these mice spontaneously secrete mucus into the bronchi. However, the activated epithelium resulted neither in impaired lung function nor conferred a protective effect against the migrating helminth Nippostrongylus brasiliensis. Moreover, CCSP-Cre_STAT6vt mice showed similar allergic airway inflammation induced by live conidia of the fungus Aspergillus fumigatus and similar recovery after influenza A virus infection compared to control mice. Together these results highlight that STAT6 signaling in Club cells induces the secretion of Relm proteins and mucus without impairing lung function, but this is not sufficient to confer protection against helminth or viral infections.
Collapse
Affiliation(s)
- Andreas Ruhl
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ana Vieira Antão
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Axel Dietschmann
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel Radtke
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - David Voehringer
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
5
|
Goode E, Marczylo E. A scoping review: What are the cellular mechanisms that drive the allergic inflammatory response to fungal allergens in the lung epithelium? Clin Transl Allergy 2023; 13:e12252. [PMID: 37357550 PMCID: PMC10234180 DOI: 10.1002/clt2.12252] [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: 02/15/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 06/27/2023] Open
Abstract
Allergic airway disease (AAD) is a collective term for respiratory disorders that can be exacerbated upon exposure to airborne allergens. The contribution of fungal allergens to AAD has become well established over recent years. We conducted a comprehensive review of the literature using Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to better understand the mechanisms involved in the allergic response to fungi in airway epithelia, identify knowledge gaps and make recommendations for future research. The search resulted in 61 studies for final analysis. Despite heterogeneity in the models and methods used, we identified major pathways involved in fungal allergy. These included the activation of protease-activated receptor 2, the EGFR pathway, adenosine triphosphate and purinergic receptor-dependent release of IL33, and oxidative stress, which drove mucin expression and goblet cell metaplasia, Th2 cytokine production, reduced barrier integrity, eosinophil recruitment, and airway hyperresponsiveness. However, there were several knowledge gaps and therefore we recommend future research should focus on the use of more physiologically relevant methods to directly compare key allergenic fungal species, clarify specific mechanisms of fungal allergy, and assess the fungal allergy in disease models. This will inform disease management and future interventions, ultimately reducing the burden of disease.
Collapse
Affiliation(s)
| | - Emma Marczylo
- Toxicology DepartmentUK Health Security AgencyChiltonUK
| |
Collapse
|
6
|
Shi Y, Zhu N, Qiu Y, Tan J, Wang F, Qin L, Dai A. Resistin-like molecules: a marker, mediator and therapeutic target for multiple diseases. Cell Commun Signal 2023; 21:18. [PMID: 36691020 PMCID: PMC9869618 DOI: 10.1186/s12964-022-01032-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/25/2023] Open
Abstract
Resistin-like molecules (RELMs) are highly cysteine-rich proteins, including RELMα, RELMβ, Resistin, and RELMγ. However, RELMs exhibit significant differences in structure, distribution, and function. The expression of RELMs is regulated by various signaling molecules, such as IL-4, IL-13, and their receptors. In addition, RELMs can mediate numerous signaling pathways, including HMGB1/RAGE, IL-4/IL-4Rα, PI3K/Akt/mTOR signaling pathways, and so on. RELMs proteins are involved in wide range of physiological and pathological processes, including inflammatory response, cell proliferation, glucose metabolism, barrier defense, etc., and participate in the progression of numerous diseases such as lung diseases, intestinal diseases, cardiovascular diseases, and cancers. Meanwhile, RELMs can serve as biomarkers, risk predictors, and therapeutic targets for these diseases. An in-depth understanding of the role of RELMs may provide novel targets or strategies for the treatment and prevention of related diseases. Video abstract.
Collapse
Affiliation(s)
- Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Yun Qiu
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Medicine, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China.
| |
Collapse
|
7
|
Moarbes V, Gaudreault V, Karkout R, Labrie L, Zhao H, Shan J, Fixman ED. STAT6-IP-Dependent Disruption of IL-33-Mediated ILC2 Expansion and Type 2 Innate Immunity in the Murine Lung. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2192-2202. [PMID: 36426982 DOI: 10.4049/jimmunol.2100688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/19/2022] [Indexed: 12/24/2022]
Abstract
Recent interest has focused on innate-type cytokines as promoters of type 2 immunity and targets for drug development in asthma. IL-33 induces production of IL-4 and/or IL-13, which is associated with STAT6-dependent responses in innate cells, including group 2 innate lymphoid cells (ILC2s), macrophages, and eosinophils. Our published data show that STAT6-immunomodulatory peptide (STAT6-IP), an immunomodulatory peptide designed to inhibit the STAT6 transcription factor, reduces induction of Th2 adaptive immunity in respiratory syncytial virus infection and asthma models. Nevertheless, the mechanism of STAT6-IP-dependent inhibition has remained obscure. In this study, we demonstrate that STAT6-IP reduced IL-33-induced type 2 innate lung inflammation. Specifically, our data show that STAT6-IP reduced recruitment and activation of eosinophils as well as polarization of alternatively activated macrophages. Decreases in these cells correlated with reduced levels of IL-5 and IL-13 as well as several type 2 chemokines in the bronchoalveolar lavage fluid. STAT6-IP effectively inhibited expansion of ILC2s as well as the number of IL-5- and IL-13-producing ILC2s. Our data suggest that STAT6-IP effectively disrupts IL-13-dependent positive feedback loops, initiated by ILC2 activation, to suppress IL-33-induced type 2 innate immunity in the murine lung.
Collapse
Affiliation(s)
- Vanessa Moarbes
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Véronique Gaudreault
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rami Karkout
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Lydia Labrie
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Hedi Zhao
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jichuan Shan
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Elizabeth D Fixman
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Badrani JH, Strohm AN, Lacasa L, Civello B, Cavagnero K, Haung YA, Amadeo M, Naji LH, Lund SJ, Leng A, Kim H, Baum RE, Khorram N, Mondal M, Seumois G, Pilotte J, Vanderklish PW, McGee HM, Doherty TA. RNA-binding protein RBM3 intrinsically suppresses lung innate lymphoid cell activation and inflammation partially through CysLT1R. Nat Commun 2022; 13:4435. [PMID: 35908044 PMCID: PMC9338970 DOI: 10.1038/s41467-022-32176-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
Innate lymphoid cells (ILC) promote lung inflammation in asthma through cytokine production. RNA-binding proteins (RBPs) are critical post-transcriptional regulators, although less is known about RBPs in ILC biology. Here, we demonstrate that RNA-binding motif 3 (RBM3) is highly expressed in lung ILCs and is further induced by alarmins TSLP and IL-33. Rbm3-/- and Rbm3-/-Rag2-/- mice exposed to asthma-associated Alternaria allergen develop enhanced eosinophilic lung inflammation and ILC activation. IL-33 stimulation studies in vivo and in vitro show that RBM3 suppressed lung ILC responses. Further, Rbm3-/- ILCs from bone marrow chimeric mice display increased ILC cytokine production suggesting an ILC-intrinsic suppressive function of RBM3. RNA-sequencing of Rbm3-/- lung ILCs demonstrates increased expression of type 2/17 cytokines and cysteinyl leukotriene 1 receptor (CysLT1R). Finally, Rbm3-/-Cyslt1r-/- mice show dependence on CysLT1R for accumulation of ST2+IL-17+ ILCs. Thus, RBM3 intrinsically regulates lung ILCs during allergen-induced type 2 inflammation that is partially dependent on CysLT1R.
Collapse
Affiliation(s)
- Jana H. Badrani
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Allyssa N. Strohm
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,Veterans Affairs San Diego Health Care System, La Jolla, CA USA
| | - Lee Lacasa
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Blake Civello
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Kellen Cavagnero
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Yung-An Haung
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.145695.a0000 0004 1798 0922Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Michael Amadeo
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Luay H. Naji
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Sean J. Lund
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Anthea Leng
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Hyojoung Kim
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Rachel E. Baum
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Naseem Khorram
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Monalisa Mondal
- grid.185006.a0000 0004 0461 3162La Jolla Institute, La Jolla, CA USA
| | - Grégory Seumois
- grid.185006.a0000 0004 0461 3162La Jolla Institute, La Jolla, CA USA
| | - Julie Pilotte
- grid.214007.00000000122199231The Scripps Research Institute, La Jolla, CA USA
| | | | - Heather M. McGee
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.250671.70000 0001 0662 7144NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute, La Jolla, CA USA ,grid.410425.60000 0004 0421 8357Departments of Radiation Oncology and Immuno-Oncology, City of Hope, Duarte, CA USA ,Department of Molecular Medicine, La Jolla, CA USA
| | - Taylor A. Doherty
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,Veterans Affairs San Diego Health Care System, La Jolla, CA USA
| |
Collapse
|
9
|
Li Y, Zhang Q, Li L, Hao D, Cheng P, Li K, Li X, Wang J, Wang Q, Du Z, Ji H, Chen H. LKB1 deficiency upregulates RELM-α to drive airway goblet cell metaplasia. Cell Mol Life Sci 2021; 79:42. [PMID: 34921639 PMCID: PMC8738459 DOI: 10.1007/s00018-021-04044-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/08/2023]
Abstract
Targeting airway goblet cell metaplasia is a novel strategy that can potentially reduce the chronic obstructive pulmonary disease (COPD) symptoms. Tumor suppressor liver kinase B1 (LKB1) is an important regulator of the proliferation and differentiation of stem/progenitor cells. In this study, we report that LKB1 expression was downregulated in the lungs of patients with COPD and in those of cigarette smoke-exposed mice. Nkx2.1Cre; Lkb1f/f mice with conditional loss of Lkb1 in mouse lung epithelium displayed airway mucus hypersecretion and pulmonary macrophage infiltration. Single-cell transcriptomic analysis of the lung tissues from Nkx2.1Cre; Lkb1f/f mice further revealed that airway goblet cell differentiation was altered in the absence of LKB1. An organoid culture study demonstrated that Lkb1 deficiency in mouse airway (club) progenitor cells promoted the expression of FIZZ1/RELM-α, which drove airway goblet cell differentiation and pulmonary macrophage recruitment. Additionally, monocyte-derived macrophages in the lungs of Nkx2.1Cre; Lkb1f/f mice exhibited an alternatively activated M2 phenotype, while expressing RELM-α, which subsequently aggravated airway goblet cell metaplasia. Our findings suggest that the LKB1-mediated crosstalk between airway progenitor cells and macrophages regulates airway goblet cell metaplasia. Moreover, our data suggest that LKB1 agonists might serve as a potential therapeutic option to treat respiratory disorders associated with goblet cell metaplasia.
Collapse
Affiliation(s)
- Yu Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Qiuyang Zhang
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Li Li
- Department of Respiratory Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - De Hao
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
| | - Peiyong Cheng
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
| | - Kuan Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Xue Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Jianhai Wang
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Qi Wang
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Zhongchao Du
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China.
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China.
| |
Collapse
|
10
|
Pham AK, Miller M, Rosenthal P, Das S, Weng N, Jang S, Kurten RC, Badrani J, Doherty TA, Oliver B, Broide DH. ORMDL3 expression in ASM regulates hypertrophy, hyperplasia via TPM1 and TPM4, and contractility. JCI Insight 2021; 6:136911. [PMID: 33661765 PMCID: PMC8119187 DOI: 10.1172/jci.insight.136911] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
ORM1-like 3 (ORMDL3) has strong genetic linkage to childhood onset asthma. To determine whether ORMDL3 selective expression in airway smooth muscle (ASM) influences ASM function, we used Cre-loxP techniques to generate transgenic mice (hORMDL3Myh11eGFP-cre), which express human ORMDL3 selectively in smooth muscle cells. In vitro studies of ASM cells isolated from the bronchi of hORMDL3Myh11eGFP-cre mice demonstrated that they developed hypertrophy (quantitated by FACS and image analysis), developed hyperplasia (assessed by BrdU incorporation), and expressed increased levels of tropomysin proteins TPM1 and TPM4. siRNA knockdown of TPM1 or TPM4 demonstrated their importance to ORMDL3-mediated ASM proliferation but not hypertrophy. In addition, ASM derived from hORMDL3Myh11eGFP-cre mice had increased contractility to histamine in vitro, which was associated with increased levels of intracellular Ca2+; increased cell surface membrane Orai1 Ca2+ channels, which mediate influx of Ca2+ into the cytoplasm; and increased expression of ASM contractile genes sarco/endoplasmic reticulum Ca2+ ATPase 2b and smooth muscle 22. In vivo studies of hORMDL3Myh11eGFP-cre mice demonstrated that they had a spontaneous increase in ASM and airway hyperreactivity (AHR). ORMDL3 expression in ASM thus induces changes in ASM (hypertrophy, hyperplasia, increased contractility), which may explain the contribution of ORMDL3 to the development of AHR in childhood onset asthma, which is highly linked to ORMDL3 on chromosome 17q12-21.
Collapse
Affiliation(s)
- Alexa K. Pham
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Peter Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ning Weng
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sunghoon Jang
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard C. Kurten
- Department of Pediatrics, Arkansas Children’s Research Institute, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jana Badrani
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Taylor A. Doherty
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Veterans Affairs San Diego Health Care System, La Jolla, California, USA
| | - Brian Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - David H. Broide
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
11
|
Ihrie MD, Duke KS, Shipkowski KA, You DJ, Lee HY, Taylor-Just AJ, Bonner JC. STAT6-Dependent Exacerbation of House Dust Mite-Induced Allergic Airway Disease in Mice by Multi-Walled Carbon Nanotubes. NANOIMPACT 2021; 22:S2452-0748(21)00018-5. [PMID: 33860111 PMCID: PMC8043620 DOI: 10.1016/j.impact.2021.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
There is increasing evidence that inhaled multi-walled carbon nanotubes (MWCNTs) can have harmful effects on the respiratory system. Rodent studies suggest that individuals with asthma may be susceptible to the adverse pulmonary effects of MWCNTs. Asthma is an allergic lung disease characterized by a TH2 immune response that results in chronic airway disease characterized by eosinophilic lung inflammation, airway mucous cell metaplasia, and airway fibrosis. Signal transducer and activator of transcription 6 (STAT6) is a transcription factor with multiple roles in TH2 type inflammation. Herein we sought to examine the role of STAT6 in the exacerbation of house dust mite (HDM) allergen-induced allergic airway disease by MWCNTs. Male wild type (WT) and STAT6 knockout (Stat6 KO) mice were dosed via intranasal aspiration on days 0, 2, 4, 14, 16 and 18 with either vehicle, HDM extract, MWCNTs, or a combination of HDM and MWCNTs. Necropsy was performed on day 21 to collect bronchoalveolar lavage fluid (BALF), serum and lung tissue. MWCNTs exacerbated HDM-induced allergic endpoints, including eosinophilic lung inflammation, mucous cell metaplasia, and serum IgE levels. HDM-induced eosinophilic lung inflammation, mucous cell metaplasia, and serum IgE and exacerbation of these endpoints by MWCNTs were ablated in Stat6 KO mice. In addition, airway fibrosis was significantly increased by the combination of HDM and MWCNTs in WT mice but not in Stat6 KO mice. These findings provide new mechanistic insight by demonstrating a requirement for STAT6 in MWCNT-induced exacerbation of allergic respiratory disease.
Collapse
Affiliation(s)
- Mark D. Ihrie
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - Katherine S. Duke
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - Kelly A. Shipkowski
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - Dorothy J. You
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - Ho Young Lee
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - Alexia J. Taylor-Just
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| | - James C. Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695 U.S.A
| |
Collapse
|
12
|
Pai S, Njoku DB. The Role of Hypoxia-Induced Mitogenic Factor in Organ-Specific Inflammation in the Lung and Liver: Key Concepts and Gaps in Knowledge Regarding Molecular Mechanisms of Acute or Immune-Mediated Liver Injury. Int J Mol Sci 2021; 22:ijms22052717. [PMID: 33800244 PMCID: PMC7962531 DOI: 10.3390/ijms22052717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 01/15/2023] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), which is also known as resistin-like molecule α (RELM-α), found in inflammatory zone 1 (FIZZ1), or resistin-like alpha (retlna), is a cysteine-rich secretory protein and cytokine. HIMF has been investigated in the lung as a mediator of pulmonary fibrosis, inflammation and as a marker for alternatively activated macrophages. Although these macrophages have been found to have a role in acute liver injury and acetaminophen toxicity, few studies have investigated the role of HIMF in acute or immune-mediated liver injury. The aim of this focused review is to analyze the literature and examine the effects of HIMF and its human homolog in organ-specific inflammation in the lung and liver. We followed the guidelines set by PRISMA in constructing this review. The relevant checklist items from PRISMA were included. Items related to meta-analysis were excluded because there were no randomized controlled clinical trials. We found that HIMF was increased in most models of acute liver injury and reduced damage from acetaminophen-induced liver injury. We also found strong evidence for HIMF as a marker for alternatively activated macrophages. Our overall risk of bias assessment of all studies included revealed that 80% of manuscripts demonstrated some concerns in the randomization process. We also demonstrated some concerns (54.1%) and high risk (45.9%) of bias in the selection of the reported results. The need for randomization and reduction of bias in the reported results was similarly detected in the studies that focused on HIMF and the liver. In conclusion, we propose that HIMF could be utilized as a marker for M2 macrophages in immune-mediated liver injury. However, we also detected the need for randomized clinical trials and additional experimental and human prospective studies in order to fully comprehend the role of HIMF in acute or immune-mediated liver injury.
Collapse
Affiliation(s)
- Sananda Pai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Dolores B. Njoku
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
13
|
Cavagnero KJ, Badrani JH, Naji LH, Amadeo MB, Leng AS, Lacasa LD, Strohm AN, Renusch SR, Gasparian SS, Doherty TA. Cyclic-di-GMP Induces STING-Dependent ILC2 to ILC1 Shift During Innate Type 2 Lung Inflammation. Front Immunol 2021; 12:618807. [PMID: 33679760 PMCID: PMC7935536 DOI: 10.3389/fimmu.2021.618807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Type 2 inflammation is found in most forms of asthma, which may co-exist with recurrent viral infections, bacterial colonization, and host cell death. These processes drive the accumulation of intracellular cyclic-di-nucleotides such as cyclic-di-GMP (CDG). Group 2 innate lymphoid cells (ILC2s) are critical drivers of type 2 lung inflammation during fungal allergen exposure in mice; however, it is unclear how CDG regulates lung ILC responses during lung inflammation. Here, we show that intranasal CDG induced early airway type 1 interferon (IFN) production and dramatically suppressed CD127+ST2+ ILC2s and type 2 lung inflammation during Alternaria and IL-33 exposure. Further, CD127-ST2-Thy1.2+ lung ILCs, which showed a transcriptomic signature consistent with ILC1s, were expanded and activated by CDG combined with either Alternaria or IL-33. CDG-mediated suppression of type 2 inflammation occurred independent of IL-18R, IL-12, and STAT6 but required the stimulator of interferon genes (STING) and type 1 IFN signaling. Thus, CDG potently suppresses ILC2-driven lung inflammation and promotes ILC1 responses. These results suggest potential therapeutic modulation of STING to suppress type 2 inflammation and/or increase anti-viral responses during respiratory infections.
Collapse
Affiliation(s)
- Kellen J. Cavagnero
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Dermatology, University of California, San Diego, La Jolla, CA, United States
| | - Jana H. Badrani
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Luay H. Naji
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michael B. Amadeo
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Anthea S. Leng
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Lee Diego Lacasa
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Allyssa N. Strohm
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Samantha R. Renusch
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Suzanna S. Gasparian
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Taylor A. Doherty
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
- Veterans Affairs San Diego Health Care System, La Jolla, CA, United States
| |
Collapse
|
14
|
Zhou W, Zhang J, Toki S, Goleniewska K, Norlander AE, Newcomb DC, Wu P, Boyd KL, Kita H, Peebles RS. COX Inhibition Increases Alternaria-Induced Pulmonary Group 2 Innate Lymphoid Cell Responses and IL-33 Release in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 205:1157-1166. [PMID: 32690653 DOI: 10.4049/jimmunol.1901544] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023]
Abstract
The cyclooxygenase (COX) metabolic pathway regulates immune responses and inflammation. The effect of the COX pathway on innate pulmonary inflammation induced by protease-containing fungal allergens, such as Alternaria alternata, is not fully defined. In this study, we tested the hypothesis that COX inhibition augments Alternaria-induced pulmonary group 2 innate lymphoid cell (ILC2) responses and IL-33 release. Mice were treated with the COX inhibitors indomethacin, flurbiprofen, or vehicle and challenged intranasally with Alternaria extract for four consecutive days to induce innate lung inflammation. We found that indomethacin and flurbiprofen significantly increased the numbers of ILC2 and IL-5 and IL-13 expression by ILC2 in the lung. Indomethacin also increased ILC2 proliferation, the percentages of eosinophils, and mucus production in the lung. Both indomethacin and flurbiprofen augmented the release of IL-33 in bronchoalveolar lavage fluid after Alternaria challenge, suggesting that more IL-33 was available for ILC2 activation and that a COX product(s) inhibited IL-33 release. This is supported by the in vitro finding that the COX product PGE2 and the PGI2 analogs cicaprost decreased Alternaria extract-induced IL-33 release by human bronchial epithelial cells. Although contrasting effects of PGD2, PGE2, and PGI2 on ILC2 responses have been previously reported, the overall effect of the COX pathway on ILC2 function is inhibitory in Alternaria-induced innate airway inflammation.
Collapse
Affiliation(s)
- Weisong Zhou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232;
| | - Jian Zhang
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Shinji Toki
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Allison E Norlander
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Dawn C Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Pingsheng Wu
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
| | - R Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Nashville, TN 37232.,Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| |
Collapse
|
15
|
Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2020; 319:L422-L434. [PMID: 32692581 DOI: 10.1152/ajplung.00040.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The family of resistin-like molecules (RELMs) consists of four members in rodents (RELMα/FIZZ1/HIMF, RELMβ/FIZZ2, Resistin/FIZZ3, and RELMγ/FIZZ4) and two members in humans (Resistin and RELMβ), all of which exhibit inflammation-regulating, chemokine, and growth factor properties. The importance of these cytokines in many aspects of physiology and pathophysiology, especially in cardiothoracic diseases, is rapidly evolving in the literature. In this review article, we attempt to summarize the contribution of RELM signaling to the initiation and progression of lung diseases, such as pulmonary hypertension, asthma/allergic airway inflammation, chronic obstructive pulmonary disease, fibrosis, cancers, infection, and other acute lung injuries. The potential of RELMs to be used as biomarkers or risk predictors of these diseases also will be discussed. Better understanding of RELM signaling in the pathogenesis of pulmonary diseases may offer novel targets or approaches for the development of therapeutics to treat or prevent a variety of inflammation, tissue remodeling, and fibrosis-related disorders in respiratory, cardiovascular, and other systems.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Ho L, Yip A, Lao F, Botelho F, Richards CD. RELMα is Induced in Airway Epithelial Cells by Oncostatin M Without Requirement of STAT6 or IL-6 in Mouse Lungs In Vivo. Cells 2020; 9:cells9061338. [PMID: 32471168 PMCID: PMC7349350 DOI: 10.3390/cells9061338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/02/2023] Open
Abstract
Resistin-like molecule alpha (RELMα) and YM-1 are secreted proteins implicated in murine models of alternatively activated macrophage (AA/M2) accumulation and Th2-skewed inflammation. Since the gp130 cytokine Oncostatin M (OSM) induces a Th2-like cytokine and AA/M2 skewed inflammation in mouse lung, we here investigated regulation of RELMα and YM-1. Transient pulmonary overexpression of OSM by Adenovirus vector (AdOSM) markedly induced RELMα and YM-1 protein expression in total lung. In situ hybridization showed that RELMα mRNA was highly induced in airway epithelial cells (AEC) and was co-expressed with CD68 mRNA in some but not all CD68+ cells in parenchyma. IL-6 overexpression (a comparator gp130 cytokine) induced RELMα, but at significantly lower levels. IL-6 (assessing IL-6-/- mice) was not required, nor was STAT6 (IL-4/13 canonical signalling) for AdOSM-induction of RELMα in AEC. AEC responded directly to OSM in vitro as assessed by pSTAT3 activation. RELMα-deficient mice showed similar inflammatory cell infiltration and cytokine responses to wt in response to AdOSM, but showed less accumulation of CD206+ AA/M2 macrophages, reduced induction of extracellular matrix gene mRNAs for COL1A1, COL3A1, MMP13, and TIMP1, and reduced parenchymal alpha smooth muscle actin. Thus, RELMα is regulated by OSM in AEC and contributes to extracellular matrix remodelling in mouse lung.
Collapse
|
17
|
Ualiyeva S, Hallen N, Kanaoka Y, Ledderose C, Matsumoto I, Junger W, Barrett N, Bankova L. Airway brush cells generate cysteinyl leukotrienes through the ATP sensor P2Y2. Sci Immunol 2020; 5:5/43/eaax7224. [PMID: 31953256 PMCID: PMC7176051 DOI: 10.1126/sciimmunol.aax7224] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 12/19/2019] [Indexed: 01/12/2023]
Abstract
Chemosensory epithelial cells (EpCs) are specialized cells that promote innate type 2 immunity and protective neurally mediated reflexes in the airway. Their effector programs and modes of activation are not fully understood. Here, we define the transcriptional signature of two choline acetyltransferase-expressing nasal EpC populations. They are found in the respiratory and olfactory mucosa and express key chemosensory cell genes including the transcription factor Pou2f3, the cation channel Trpm5, and the cytokine Il25 Moreover, these cells share a core transcriptional signature with chemosensory cells from intestine, trachea and thymus, and cluster with tracheal brush cells (BrCs) independently from other respiratory EpCs, indicating that they are part of the brush/tuft cell family. Both nasal BrC subsets express high levels of transcripts encoding cysteinyl leukotriene (CysLT) biosynthetic enzymes. In response to ionophore, unfractionated nasal BrCs generate CysLTs at levels exceeding that of the adjacent hematopoietic cells isolated from naïve mucosa. Among activating receptors, BrCs express the purinergic receptor P2Y2. Accordingly, the epithelial stress signal ATP and aeroallergens that elicit ATP release trigger BrC CysLT generation, which is mediated by the P2Y2 receptor. ATP- and aeroallergen-elicited CysLT generation in the nasal lavage is reduced in mice lacking Pou2f3, a requisite transcription factor for BrC development. Last, aeroallergen-induced airway eosinophilia is reduced in BrC-deficient mice. These results identify a previously undescribed BrC sensor and effector pathway leading to generation of lipid mediators in response to luminal signals. Further, they suggest that BrC sensing of local damage may provide an important sentinel immune function.
Collapse
Affiliation(s)
- S. Ualiyeva
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - N. Hallen
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Y. Kanaoka
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - C. Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - W. Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - N.A. Barrett
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - L.G. Bankova
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Hadebe S, Brombacher F. Environment and Host-Genetic Determinants in Early Development of Allergic Asthma: Contribution of Fungi. Front Immunol 2019; 10:2696. [PMID: 31824491 PMCID: PMC6879655 DOI: 10.3389/fimmu.2019.02696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
Asthma is a chronic debilitating airway disease affecting millions of people worldwide. Although largely thought to be a disease of the first world, it is now clear that it is on the rise in many middle- and lower-income countries. The disease is complex, and its etiology is poorly understood, which explains failure of most treatment strategies. We know that in children, asthma is closely linked to poor lung function in the first 3-years of life, when the lung is still undergoing post-natal alveolarization phase. Epidemiological studies also suggest that environmental factors around that age do play a critical part in the establishment of early wheezing which persists until adulthood. Some of the factors that contribute to early development of asthma in children in Western world are clear, however, in low- to middle-income countries this is likely to differ significantly. The contribution of fungal species in the development of allergic diseases is known in adults and in experimental models. However, it is unclear whether early exposure during perinatal or post-natal lung development influences a protective or promotes allergic asthma. Host immune cells and responses will play a crucial part in early development of allergic asthma. How immune cells and their receptors may recognize fungi and promote allergic asthma or protect by tolerance among other immune mechanisms is not fully understood in this early lung development stage. The aim of this review is to discuss what fungal species are present during early exposure as well as their contribution to the development of allergic responses. We also discuss how the host has evolved to promote tolerance to limit hyper-responsiveness to innocuous fungi, and how host evasion by fungi during early development consequentially results in allergic diseases.
Collapse
Affiliation(s)
- Sabelo Hadebe
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
- Division of Immunology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| |
Collapse
|
19
|
Cavagnero KJ, Badrani JH, Naji LH, Amadeo MB, Shah VS, Gasparian S, Pham A, Wang AW, Seumois G, Croft M, Broide DH, Doherty TA. Unconventional ST2- and CD127-negative lung ILC2 populations are induced by the fungal allergen Alternaria alternata. J Allergy Clin Immunol 2019; 144:1432-1435.e9. [PMID: 31369800 DOI: 10.1016/j.jaci.2019.07.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/08/2019] [Accepted: 07/25/2019] [Indexed: 12/19/2022]
Affiliation(s)
| | - Jana H Badrani
- Department of Medicine, University of California, San Diego, Calif
| | - Luay H Naji
- Department of Medicine, University of California, San Diego, Calif
| | - Michael B Amadeo
- Department of Medicine, University of California, San Diego, Calif
| | - Veranca S Shah
- Department of Medicine, University of California, San Diego, Calif
| | | | - Alexa Pham
- Department of Medicine, University of California, San Diego, Calif
| | - Alice W Wang
- La Jolla Institute for Allergy and Immunology, La Jolla, Calif
| | - Grégory Seumois
- La Jolla Institute for Allergy and Immunology, La Jolla, Calif
| | - Michael Croft
- Department of Medicine, University of California, San Diego, Calif; La Jolla Institute for Allergy and Immunology, La Jolla, Calif
| | - David H Broide
- Department of Medicine, University of California, San Diego, Calif
| | - Taylor A Doherty
- Department of Medicine, University of California, San Diego, Calif.
| |
Collapse
|
20
|
Helfrich S, Mindt BC, Fritz JH, Duerr CU. Group 2 Innate Lymphoid Cells in Respiratory Allergic Inflammation. Front Immunol 2019; 10:930. [PMID: 31231357 PMCID: PMC6566538 DOI: 10.3389/fimmu.2019.00930] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Millions of people worldwide are suffering from allergic inflammatory airway disorders. These conditions are regarded as a consequence of multiple imbalanced immune events resulting in an inadequate response with the exact underlying mechanisms still being a subject of ongoing research. Several cell populations have been proposed to be involved but it is becoming increasingly evident that group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of respiratory allergic inflammation. ILC2s are important mediators of inflammation but also tissue remodeling by secreting large amounts of signature cytokines within a short time period. Thereby, ILC2s instruct innate but also adaptive immune responses. Here, we will discuss the recent literature on allergic inflammation of the respiratory tract with a focus on ILC2 biology. Furthermore, we will highlight different therapeutic strategies to treat pulmonary allergic inflammation and their potential influence on ILC2 function as well as discuss the perspective of using human ILC2s for diagnostic purposes.
Collapse
Affiliation(s)
- Sofia Helfrich
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara C Mindt
- Department of Microbiology & Immunology, McGill University, Montréal, QC, Canada.,McGill University Research Centre on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada.,FOCiS Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, QC, Canada
| | - Jörg H Fritz
- Department of Microbiology & Immunology, McGill University, Montréal, QC, Canada.,McGill University Research Centre on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada.,FOCiS Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, QC, Canada.,Department of Physiology, McGill University, Montréal, QC, Canada
| | - Claudia U Duerr
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
21
|
Zhao H, Moarbes V, Gaudreault V, Shan J, Aldossary H, Cyr L, Fixman ED. Sex Differences in IL-33-Induced STAT6-Dependent Type 2 Airway Inflammation. Front Immunol 2019; 10:859. [PMID: 31118931 PMCID: PMC6504808 DOI: 10.3389/fimmu.2019.00859] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 04/03/2019] [Indexed: 12/02/2022] Open
Abstract
Sex differences in asthma prevalence are well-documented but poorly understood. Murine models have contributed to our understanding of mechanisms that could regulate this sex disparity, though the majority of these studies have examined responses present after Th2 adaptive immunity is established. We have now investigated how sex influences acute activation of innate cell populations in the lung upon initial exposure to the model antigen, ovalbumin (OVA), in the presence of IL-33 (OVA+IL-33), to prime the lungs for type 2 immunity. We also examined how inflammatory responses induced by OVA+IL-33 were altered in mice lacking the STAT6 transcription factor, which is activated by IL-13, an effector cytokine of IL-33. Our data demonstrate that type 2 inflammation induced by OVA+IL-33 was more severe in female mice compared to males. Females exhibited greater cytokine and chemokine production, eosinophil influx and activation, macrophage polarization to the alternatively activated phenotype, and expansion of group 2 innate lymphoid cells (ILC2s). While increases in ILC2s and eosinophils were largely independent of STAT6 in both males and females, many other responses were STAT6-dependent only in female mice. Our findings indicate that a subset of type 2 inflammatory responses induced by OVA+IL-33 require STAT6 in both males and females and that enhanced type 2 inflammation in females, compared to males, is associated with greater IL-13 protein production. Our findings suggest blunted IL-13 production in males may protect against type 2 inflammation initiated by OVA+IL-33 delivery to the lung.
Collapse
Affiliation(s)
- Hedi Zhao
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Vanessa Moarbes
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Véronique Gaudreault
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Jichuan Shan
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Haya Aldossary
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Louis Cyr
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Elizabeth D Fixman
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
22
|
Peng X, Maltz MR, Botthoff JK, Aronson EL, Nordgren TM, Lo DD, Cocker DR. Establishment and characterization of a multi-purpose large animal exposure chamber for investigating health effects. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2019; 90:035115. [PMID: 30927824 PMCID: PMC6910591 DOI: 10.1063/1.5042097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Air pollution poses a significant threat to the environment and human health. Most in vivo health studies conducted regarding air pollutants, including particulate matter (PM) and gas phase pollutants, have been either through traditional medical intranasal treatment or using a tiny chamber, which limit animal activities. In this study, we designed and tested a large, whole-body, multiple animal exposure chamber with uniform dispersion and exposure stability for animal studies. The chamber simultaneously controls particle size distribution and PM mass concentration. Two different methods were used to generate aerosol suspension through either soluble material (Alternaria extract), liquid particle suspension (nanosilica solution), or dry powder (silica powder). We demonstrate that the chamber system provides well controlled and characterized whole animal exposures, where dosage is by inhalation of particulate matter.
Collapse
Affiliation(s)
| | | | | | | | | | - David D. Lo
- Author to whom correspondence should be addressed:
| | | |
Collapse
|
23
|
Miller M, Vuong C, Garcia MF, Rosenthal P, Das S, Weng N, Pham A, Kim YJ, Broide DH. Does reduced zona pellucida binding protein 2 (ZPBP2) expression on chromosome 17q21 protect against asthma? J Allergy Clin Immunol 2018; 142:706-709.e4. [PMID: 29709669 PMCID: PMC6078789 DOI: 10.1016/j.jaci.2018.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/09/2018] [Accepted: 04/20/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Christine Vuong
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | | | - Peter Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Ning Weng
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Alexa Pham
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Yu Jin Kim
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - David H Broide
- Department of Medicine, University of California San Diego, La Jolla, Calif.
| |
Collapse
|
24
|
Bartemes KR, Kita H. Innate and adaptive immune responses to fungi in the airway. J Allergy Clin Immunol 2018; 142:353-363. [PMID: 30080527 PMCID: PMC6083885 DOI: 10.1016/j.jaci.2018.06.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022]
Abstract
Fungi are ubiquitous outdoors and indoors. Exposure, sensitization, or both to fungi are strongly associated with development of asthma and allergic airway diseases. Furthermore, global climate change will likely increase the prevalence of fungi and enhance their antigenicity. Major progress has been made during the past several years regarding our understanding of antifungal immunity. Fungi contain cell-wall molecules, such as β-glucan and chitin, and secrete biologically active proteases and glycosidases. Airway epithelial cells and innate immune cells, such as dendritic cells, are equipped with cell-surface molecules that react to these fungal products, resulting in production of cytokines and proinflammatory mediators. As a result, the adaptive arm of antifungal immunity, including TH1-, TH2-, and TH17-type CD4+ T cells, is established, reinforcing protection against fungal infection and causing detrimental immunopathology in certain subjects. We are only in the beginning stages of understanding the complex biology of fungi and detailed mechanisms of how they activate the immune response that can protect against or drive diseases in human subjects. Here we describe our current understanding with an emphasis on airway allergic immune responses. The gaps in our knowledge and desirable future directions are also discussed.
Collapse
Affiliation(s)
- Kathleen R Bartemes
- Division of Allergic Diseases, Department of Internal Medicine, and the Department of Immunology, Mayo Clinic, Rochester, Minn
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, and the Department of Immunology, Mayo Clinic, Rochester, Minn.
| |
Collapse
|
25
|
Li BWS, de Bruijn MJW, Lukkes M, van Nimwegen M, Bergen IM, KleinJan A, GeurtsvanKessel CH, Andeweg A, Rimmelzwaan GF, Hendriks RW. T cells and ILC2s are major effector cells in influenza-induced exacerbation of allergic airway inflammation in mice. Eur J Immunol 2018; 49:144-156. [PMID: 29762870 PMCID: PMC6585726 DOI: 10.1002/eji.201747421] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/14/2018] [Accepted: 05/09/2018] [Indexed: 12/17/2022]
Abstract
Influenza virus infection is an important cause of severe asthma exacerbations, but it remains unclear how a Th1‐mediated antiviral response triggers a prototypical Th2 disease. We investigated CD4+ T cells and group 2 innate lymphoid cells (ILC2s) in influenza virus‐infected mice. We found that ILC2s accumulated in the lung rapidly after influenza virus infection, but the induction of IL‐5 and IL‐13 secretion was delayed and concomitant with T cell activation. In an influenza‐induced exacerbation of allergic airway inflammation model we noticed an initial reduction of ILC2 numbers and cytokine production in broncho‐alveolar lavage compared to chronic house dust mite (HDM)‐mediated airway inflammation alone. ILC2s phenotype was characterized by low T1/ST2, ICOS, KLRG1, and CD25 expression, resembling naïve ILC2s. The contribution of ILC2s to type 2 cytokine production in the early stage of the influenza‐induced exacerbation was limited. In contrast, T cells showed increased IL‐4 and IL‐5 production when exposed to both HDM and influenza virus. Upon virus clearance, ILC2s regained an activated T1/ST2highICOShighKLRG1highCD25high phenotype paired with cytokine production and were major contributors to the type 2 cytokine milieu. Collectively, our data indicate that both T cells and ILC2s contribute to influenza‐induced exacerbation of allergic airway inflammation, but with different kinetics.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | | | - Melanie Lukkes
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Ingrid M Bergen
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | | | - Arno Andeweg
- Department of Viroscience, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
26
|
Pine GM, Batugedara HM, Nair MG. Here, there and everywhere: Resistin-like molecules in infection, inflammation, and metabolic disorders. Cytokine 2018; 110:442-451. [PMID: 29866514 DOI: 10.1016/j.cyto.2018.05.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/13/2018] [Accepted: 05/15/2018] [Indexed: 02/07/2023]
Abstract
The Resistin-Like Molecules (RELM) α, β, and γ and their namesake, resistin, share structural and sequence homology but exhibit significant diversity in expression and function within their mammalian host. RELM proteins are expressed in a wide range of diseases, such as: microbial infections (eg. bacterial and helminth), inflammatory diseases (eg. asthma, fibrosis) and metabolic disorders (eg. diabetes). While the expression pattern and molecular regulation of RELM proteins are well characterized, much controversy remains over their proposed functions, with evidence of host-protective and pathogenic roles. Moreover, the receptors for RELM proteins are unclear, although three receptors for resistin, decorin, adenylyl cyclase-associated protein 1 (CAP1), and Toll-like Receptor 4 (TLR4) have recently been proposed. In this review, we will first summarize the molecular regulation of the RELM gene family, including transcription regulation and tissue expression in humans and mouse disease models. Second, we will outline the function and receptor-mediated signaling associated with RELM proteins. Finally, we will discuss recent studies suggesting that, despite early misconceptions that these proteins are pathogenic, RELM proteins have a more nuanced and potentially beneficial role for the host in certain disease settings.
Collapse
Affiliation(s)
- Gabrielle M Pine
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Hashini M Batugedara
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States.
| |
Collapse
|
27
|
Yamaguchi M, Samuchiwal SK, Quehenberger O, Boyce JA, Balestrieri B. Macrophages regulate lung ILC2 activation via Pla2g5-dependent mechanisms. Mucosal Immunol 2018; 11:615-626. [PMID: 29346348 PMCID: PMC5976507 DOI: 10.1038/mi.2017.99] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/17/2017] [Indexed: 02/04/2023]
Abstract
Group V phospholipase A2 (Pla2g5) is a lipid-generating enzyme necessary for macrophage effector functions in pulmonary inflammation. However, the lipid mediators involved and their cellular targets have not been identified. Mice lacking Pla2g5 showed markedly reduced lung ILC2 activation and eosinophilia following repetitive Alternaria Alternata inhalation. While Pla2g5-null mice had Wt levels of immediate IL-33 release after one Alternaria dose, they failed to upregulate IL-33 in macrophages following repeated Alternaria administration. Unexpectedly, while adoptive transfer of bone marrow-derived (BM)-macrophages restored ILC2 activation and eosinophilia in Alternaria-exposed Pla2g5-null mice, exogenous IL-33 did not. Conversely, transfers of Pla2g5-null BM-macrophages reduced inflammation in Alternaria-exposed Wt mice. Mass spectrometry analysis of free fatty acids (FFAs) demonstrated significantly reduced FFAs (including linoleic acid (LA) and oleic acid (OA)) in lung and BM-macrophages lacking Pla2g5. Exogenous administration of LA or LA+OA to Wt mice sharply potentiated IL-33-induced lung eosinophilia and ILC2 expansion in vitro and in vivo. In contrast, OA potentiated IL-33-induced inflammation and ILC2 expansion in Pla2g5-null mice, but LA was inactive both in vivo and in vitro. Notably, Pla2g5-null ILC2s showed significantly reduced expression of the FFA-receptor-1 compared to Wt ILC2s. Thus, macrophage-associated Pla2g5 contributes significantly to type-2 immunity through regulation of IL-33 induction and FFA-driven ILC2 activation.
Collapse
Affiliation(s)
- Munehiro Yamaguchi
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Sachin K. Samuchiwal
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Oswald Quehenberger
- Department of Medicine, Department of Pharmacology, University of California, San Diego, CA 92093, USA
| | - Joshua A. Boyce
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Barbara Balestrieri
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
28
|
Li BWS, Beerens DMJM, Brem MD, Hendriks RW. Characterization of Group 2 Innate Lymphoid Cells in Allergic Airway Inflammation Models in the Mouse. Methods Mol Biol 2018; 1559:169-183. [PMID: 28063044 DOI: 10.1007/978-1-4939-6786-5_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Allergic asthma is a chronic inflammatory lung disease mediated by type 2 cytokines produced by T helper 2 (Th2) cells as well as the recently discovered group 2 innate lymphoid cells (ILC2). Due to a lack of unique markers, the accurate phenotypic characterization and quantification of ILC2 requires a comprehensive panel of fluorescently labeled antibodies. The markers that are currently used to characterize ILC2 have not been standardized and often vary between research groups, which poses significant challenges when comparing data. Intranasal administration of the pro-inflammatory cytokine IL-33 in mice is associated with strong, Th2 cell-independent ILC2 activation. ILC2 are also activated in mouse models of allergic asthma based on the physiologically relevant house dust mite (HDM) allergen, which parallel eosinophilic airway inflammation observed in asthma patients. Here, we describe the analysis of ILC2 by flow cytometry in these two commonly used allergic airway inflammation models in the mouse.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Dior M J M Beerens
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Maarten D Brem
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, 2040, 3000, CA, Rotterdam, The Netherlands.
| |
Collapse
|
29
|
Peng X, Madany AM, Jang JC, Valdez JM, Rivas Z, Burr AC, Grinberg YY, Nordgren TM, Nair MG, Cocker D, Carson MJ, Lo DD. Continuous Inhalation Exposure to Fungal Allergen Particulates Induces Lung Inflammation While Reducing Innate Immune Molecule Expression in the Brainstem. ASN Neuro 2018; 10:1759091418782304. [PMID: 30016877 PMCID: PMC6053578 DOI: 10.1177/1759091418782304] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/17/2018] [Accepted: 05/20/2018] [Indexed: 12/13/2022] Open
Abstract
Continuous exposure to aerosolized fine (particle size ≤2.5 µm) and ultrafine (particle size ≤0.1 µm) particulates can trigger innate inflammatory responses in the lung and brain depending on particle composition. Most studies of manmade toxicants use inhalation exposure routes, whereas most studies of allergens use soluble solutions administered via intranasal or injection routes. Here, we tested whether continuous inhalation exposure to aerosolized Alternaria alternata particulates (a common fungal allergen associated with asthma) would induce innate inflammatory responses in the lung and brain. By designing a new environmental chamber able to control particle size distribution and mass concentration, we continuously exposed adult mice to aerosolized ultrafine Alternaria particulates for 96 hr. Despite induction of innate immune responses in the lung, induction of innate immune responses in whole brain samples was not detected by quantitative polymerase chain reaction or flow cytometry. However, exposure did trigger decreases in Arginase 1, inducible nitric oxide synthase, and tumor necrosis factor alpha mRNA in the brainstem samples containing the central nervous system respiratory circuit (the dorsal respiratory group, ventral respiratory group, and the pre-Bötzinger and Bötzinger complexes). In addition, a significant decrease in the percentage of Toll-like receptor 2-expressing brainstem microglia was detected by flow cytometry. Histologic analysis revealed a significant decrease in Iba1 but not glial fibrillary acidic protein immunoreactivity in both the brainstem and the hippocampus. Together these data indicate that inhalation exposure to a natural fungal allergen under conditions sufficient to induce lung inflammation surprisingly causes reductions in baseline expression of select innate immune molecules (similar to that observed during endotoxin tolerance) in the region of the central nervous system controlling respiration.
Collapse
Affiliation(s)
- Xinze Peng
- BREATHE Center, University of California, Riverside, CA,
USA
- Department of Chemical and Environmental Engineering, Bourns
College of Engineering, Center for Environmental Research and Technology
(Ce-Cert),
University
of California, Riverside, CA, USA
| | - Abdullah M. Madany
- BREATHE Center, University of California, Riverside, CA,
USA
- Center for Glial-Neuronal Interactions,
University
of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
| | - Jessica C. Jang
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Microbiology Graduate Program,
University
of California, Riverside, CA, USA
| | - Joseph M. Valdez
- BREATHE Center, University of California, Riverside, CA,
USA
- Center for Glial-Neuronal Interactions,
University
of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Neuroscience Graduate Program,
University
of California, Riverside, CA, USA
| | - Zuivanna Rivas
- BREATHE Center, University of California, Riverside, CA,
USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
| | - Abigail C. Burr
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
| | - Yelena Y. Grinberg
- Center for Glial-Neuronal Interactions,
University
of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
| | - Tara M. Nordgren
- BREATHE Center, University of California, Riverside, CA,
USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Biomedical Sciences Graduate Program,
University
of California, Riverside, CA, USA
| | - Meera G. Nair
- BREATHE Center, University of California, Riverside, CA,
USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Microbiology Graduate Program,
University
of California, Riverside, CA, USA
- Biomedical Sciences Graduate Program,
University
of California, Riverside, CA, USA
| | - David Cocker
- BREATHE Center, University of California, Riverside, CA,
USA
- Department of Chemical and Environmental Engineering, Bourns
College of Engineering, Center for Environmental Research and Technology
(Ce-Cert),
University
of California, Riverside, CA, USA
| | - Monica J. Carson
- BREATHE Center, University of California, Riverside, CA,
USA
- Center for Glial-Neuronal Interactions,
University
of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Neuroscience Graduate Program,
University
of California, Riverside, CA, USA
- Biomedical Sciences Graduate Program,
University
of California, Riverside, CA, USA
| | - David D. Lo
- BREATHE Center, University of California, Riverside, CA,
USA
- Center for Glial-Neuronal Interactions,
University
of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine,
University
of California, Riverside, CA, USA
- Microbiology Graduate Program,
University
of California, Riverside, CA, USA
- Biomedical Sciences Graduate Program,
University
of California, Riverside, CA, USA
| |
Collapse
|
30
|
Niu C, Liu N, Liu J, Zhang M, Ying L, Wang L, Tian D, Dai J, Luo Z, Liu E, Zou L, Fu Z. Vitamin A maintains the airway epithelium in a murine model of asthma by suppressing glucocorticoid-induced leucine zipper. Clin Exp Allergy 2017; 46:848-60. [PMID: 26399569 DOI: 10.1111/cea.12646] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 08/05/2015] [Accepted: 08/12/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND The effects of glucocorticoids (GCs) on the repair of the airway epithelium in asthma are controversial, and we previously reported that the GC dexamethasone (Dex) inhibits the repair of human airway epithelial cells and that this process is mediated by glucocorticoid-induced leucine zipper (GILZ) through MAPK-ERK signaling in vitro. Vitamin A (VA) is involved in the regulation of the MAPK-ERK pathway but has not been widely supplied during asthma treatment. It is unclear whether VA attenuates the negative regulation of GILZ on the MAPK-ERK pathway and maintains airway epithelium integrity during asthma treatment. METHODS Female BALB/c mice were sensitized and challenged with ovalbumin (OVA) and subsequently treated with Dex, VA or intranasal inhalation of adenovirus sh-GILZ vectors. Indexes of airway epithelium integrity, including pathological alterations, pulmonary EGFR expression and airway hyperresponsiveness (AHR), were then measured. The expression of GILZ and key components of activated MAPK-ERK signals (p-Raf-1, p-MEK, and p-Erk1/2) were also detected. RESULTS Dex failed to relieve OVA-induced asthma airway epithelium injury, as assessed through H&E staining, EGFR expression and AHR. Moreover, in the OVA-challenged mice treated with Dex, GLIZ expression was increased, whereas the ratios of p-Raf-1/Raf-1, p-MEK/MEK and p-Erk1/2/Erk1/2 were significantly decreased. Further study indicated that GILZ expression was decreased and that the ratios of p-Raf-1/Raf-1, p-MEK/MEK and p-Erk1/2/Erk1/2 were up-regulated in the GILZ-silenced OVA-challenged mice and VA-fed OVA-challenged mice, independent of Dex treatment. The airway epithelium integrity of the OVA-challenged mice was maintained by treatment with both VA and Dex. CONCLUSIONS Vitamin A maintained the Dex-treated asthma airway epithelium via the down-regulation of GILZ expression and the activation MAPK-ERK signaling, and these effects might contribute to improving the effects of GC therapeutics on asthma.
Collapse
Affiliation(s)
- C Niu
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - N Liu
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - J Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - M Zhang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - L Ying
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - L Wang
- Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - D Tian
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - J Dai
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Z Luo
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - E Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - L Zou
- Center for Clinical Molecular Medicine, Chongqing Stem Cell Therapy Technology Research Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Z Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Nikota J, Banville A, Goodwin LR, Wu D, Williams A, Yauk CL, Wallin H, Vogel U, Halappanavar S. Stat-6 signaling pathway and not Interleukin-1 mediates multi-walled carbon nanotube-induced lung fibrosis in mice: insights from an adverse outcome pathway framework. Part Fibre Toxicol 2017; 14:37. [PMID: 28903780 PMCID: PMC5598059 DOI: 10.1186/s12989-017-0218-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/05/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The accumulation of MWCNTs in the lung environment leads to inflammation and the development of disease similar to pulmonary fibrosis in rodents. Adverse Outcome Pathways (AOPs) are a framework for defining and organizing the key events that comprise the biological changes leading to undesirable events. A putative AOP has been developed describing MWCNT-induced pulmonary fibrosis; inflammation and the subsequent healing response induced by inflammatory mechanisms have been implicated in disease progression. The objective of the present study was to address a key data gap in this AOP: empirical data supporting the essentiality of pulmonary inflammation as a key event prior to fibrosis. Specifically, Interleukin-1 Receptor1 (IL-1R1) and Signal Transducer and Activator of Transcription 6 (STAT6) knock-out (KO) mice were employed to target inflammation and the subsequent healing response using MWCNTs as a model pro-fibrotic stressor to determine whether this altered the development of fibrosis. RESULTS Wild type (WT) C57BL/6, IL-1R1 (KO) or STAT6 KO mice were exposed to a high dose of Mitsui-7 MWCNT by intratracheal administration. Inflammation was assessed 24 h and 28 days post MWCNT administration, and fibrotic lesion development was assessed 28 days post MWCNT administration. MWCNT-induced acute inflammation was suppressed in IL-1R1 KO mice at the 24 h time point relative to WT mice, but this suppression was not observed 28 days post exposure, and IL-1R1 KO did not alter fibrotic disease development. In contrast, STAT6 KO mice exhibited suppressed acute inflammation and attenuated fibrotic disease in response to MWCNT administration compared to STAT6 WT mice. Whole genome analysis of all post-exposure time points identified a subset of differentially expressed genes associated with fibrosis in both KO mice compared to WT mice. CONCLUSION The findings support the essentiality of STAT6-mediated signaling in the development of MWCNT-induced fibrotic disease. The IL-1R1 KO results also highlight the nature of the inflammatory response associated with MWCNT exposure, and indicate a system with multiple redundancies. These data add to the evidence supporting an existing AOP, and will be useful in designing screening strategies that could be used by regulatory agencies to distinguish between MWCNTs of varying toxicity.
Collapse
Affiliation(s)
- Jake Nikota
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Allyson Banville
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Laura Rose Goodwin
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Dongmei Wu
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Carole Lynn Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| | - Håkan Wallin
- Department of Biological and Chemical Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Ulla Vogel
- National Research Centre for the Working Environment, Lerso Parkallé 105, DK-2100 Copenhagen, Denmark
- Department of Micro- and Nanotechnology, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9 Canada
| |
Collapse
|
32
|
Identification of trans Protein QTL for Secreted Airway Mucins in Mice and a Causal Role for Bpifb1. Genetics 2017; 207:801-812. [PMID: 28851744 DOI: 10.1534/genetics.117.300211] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/22/2017] [Indexed: 12/14/2022] Open
Abstract
Mucus hyper-secretion is a hallmark feature of asthma and other muco-obstructive airway diseases. The mucin proteins MUC5AC and MUC5B are the major glycoprotein components of mucus and have critical roles in airway defense. Despite the biomedical importance of these two proteins, the loci that regulate them in the context of natural genetic variation have not been studied. To identify genes that underlie variation in airway mucin levels, we performed genetic analyses in founder strains and incipient lines of the Collaborative Cross (CC) in a house dust mite mouse model of asthma. CC founder strains exhibited significant differences in MUC5AC and MUC5B, providing evidence of heritability. Analysis of gene and protein expression of Muc5ac and Muc5b in incipient CC lines (n = 154) suggested that post-transcriptional events were important regulators of mucin protein content in the airways. Quantitative trait locus (QTL) mapping identified distinct, trans protein QTL for MUC5AC (chromosome 13) and MUC5B (chromosome 2). These two QTL explained 18 and 20% of phenotypic variance, respectively. Examination of the MUC5B QTL allele effects and subsequent phylogenetic analysis allowed us to narrow the MUC5B QTL and identify Bpifb1 as a candidate gene. Bpifb1 mRNA and protein expression were upregulated in parallel to MUC5B after allergen challenge, and Bpifb1 knockout mice exhibited higher MUC5B expression. Thus, BPIFB1 is a novel regulator of MUC5B.
Collapse
|
33
|
Song DJ, Miller M, Beppu A, Rosenthal P, Das S, Karta M, Vuong C, Mehta AK, Croft M, Broide DH. Rhinovirus Infection of ORMDL3 Transgenic Mice Is Associated with Reduced Rhinovirus Viral Load and Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2017; 199:2215-2224. [PMID: 28827284 DOI: 10.4049/jimmunol.1601412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 07/27/2017] [Indexed: 01/29/2023]
Abstract
Orosomucoid like 3 (ORMDL3), a gene localized to chromosome 17q21, has been linked in epidemiologic studies to childhood asthma and rhinovirus (RV) infections. As the single nucleotide polymorphisms linking ORMDL3 to asthma are associated with increased expression of ORMDL3, we have used hORMDL3zp3-Cre mice (which have universal increased expression of human ORMDL3) to determine whether infection of these transgenic mice with RV influences levels of airway inflammation or RV viral load. RV infection of hORMDL3zp3-Cre mice resulted in reduced RV viral load assessed by quantitative real-time PCR (lung and airway epithelium), as well as reduced airway inflammation (total bronchoalveolar lavage cells, neutrophils, macrophages, and lymphocytes) compared with RV-infected wild-type mice. Levels of the antiviral pathways including IFNs (IFN-α, IFN-β, IFN-λ) and RNAse L were significantly increased in the lungs of RV-infected hORMDL3zp3-Cre mice. Levels of the antiviral mouse oligoadenylate synthetase (mOas)1g pathway and RNAse L were upregulated in the lungs of unchallenged hORMDL3zp3-Cre mice. In addition, levels of mOas2, but not mOas1 (mOas1a, mOas1b, mOas1g), or mOas3 pathways were significantly more upregulated by IFNs (IFN-α, IFN-β, IFN-λ) in epithelial cells from hORMDL3zp3-Cre mice compared with RV-infected wild-type mouse epithelial cells. RNAse L-deficient mice infected with RV had increased RV viral load. Overall, these studies suggest that increased levels of ORMDL3 contribute to antiviral defense to RV infection in mice through pathways that may include IFNs (IFN-α, IFN-β, IFN-λ), OAS, and RNAse L.
Collapse
Affiliation(s)
- Dae Jin Song
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093.,Department of Pediatrics, Korea University College of Medicine, Seoul 03080, Korea; and
| | - Marina Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Andrew Beppu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter Rosenthal
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sudipta Das
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Maya Karta
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Christine Vuong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Amit Kumar Mehta
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - David H Broide
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
| |
Collapse
|
34
|
Wang Y, Zhu J, Zhang L, Zhang Z, He L, Mou Y, Deng Y, Cao Y, Yang P, Su Y, Zhao J, Zhang S, Yu Q, Hu J, Chen Z, Ning Q, Xiang X, Xu Y, Wang CY, Xiong W. Role of C/EBP homologous protein and endoplasmic reticulum stress in asthma exacerbation by regulating the IL-4/signal transducer and activator of transcription 6/transcription factor EC/IL-4 receptor α positive feedback loop in M2 macrophages. J Allergy Clin Immunol 2017; 140:1550-1561.e8. [PMID: 28238747 DOI: 10.1016/j.jaci.2017.01.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 12/30/2016] [Accepted: 01/23/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND C/EBP homologous protein (Chop), a marker of endoplasmic reticulum (ER) stress, exhibits aberrant expression patterns during asthma development. However, its exact role in asthma pathogenesis is not fully understood. OBJECTIVES We aimed to determine the function and mechanism of Chop in the pathogenesis of allergic asthma in patients and animals. METHODS Studies were conducted in asthmatic patients and Chop-/- mice to dissect the role of Chop and ER stress in asthma pathogenesis. An ovalbumin (OVA)-induced allergic airway inflammation model was used to address the effect of Chop deficiency on asthma development. Next, the effect of Chop deficiency on macrophage polarization and related signaling pathways was investigated to demonstrate the underlying mechanisms. RESULTS Asthmatic patients and mice after OVA induction exhibited aberrant Chop expression along with ER stress. Specifically, Chop was noted to be specifically overexpressed in macrophages, and mice deficient in Chop were protected from OVA-induced allergic airway inflammation, as manifested by attenuated airway inflammation, remodeling, and hyperresponsiveness. Chop was found to exacerbate allergic airway inflammation by enhancing M2 programming in macrophages. Mechanistic studies characterized an IL-4/signal transducer and activator of transcription 6/transcription factor EC (Tfec)/IL-4 receptor α positive feedback regulatory loop, in which IL-4 induces Chop expression, which then promotes signal transducer and activator of transcription 6 signaling to transcribe Tfec expression. Finally, Tfec transcribes IL-4 receptor α expression to promote M2 programming in macrophages. CONCLUSIONS Chop and ER stress are implicated in asthma pathogenesis, which involves regulation of M2 programming in macrophages.
Collapse
Affiliation(s)
- Yi Wang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jianghui Zhu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Lei Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Zhijun Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Long He
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Yong Mou
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yanhan Deng
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yong Cao
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Ping Yang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Ga
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Shu Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Qilin Yu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Jifa Hu
- Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Zhishui Chen
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xudong Xiang
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongjian Xu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Cong-Yi Wang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
| | - Weining Xiong
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China.
| |
Collapse
|
35
|
Movassagh H, Shan L, Mohammed A, Halayko AJ, Gounni AS. Semaphorin 3E Deficiency Exacerbates Airway Inflammation, Hyperresponsiveness, and Remodeling in a Mouse Model of Allergic Asthma. THE JOURNAL OF IMMUNOLOGY 2017; 198:1805-1814. [PMID: 28108561 DOI: 10.4049/jimmunol.1601514] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/19/2016] [Indexed: 01/17/2023]
Abstract
Semaphorin 3E (Sema3E) plays a crucial role in axon guidance, vascular patterning, and immune regulation. Nevertheless, the role of Sema3E in asthma is still elusive. In this study, we show that genetic ablation of Sema3E in mice results in increased lung granulocytosis, airway hyperresponsiveness, mucus overproduction, collagen deposition, and Th2/Th17 inflammation. Transfer of Sema3e-/- bone marrow progenitor cells to irradiated wild-type (WT) recipients exacerbates airway hyperresponsiveness and inflammation, whereas transfer of WT bone marrow progenitor cells ameliorates asthma pathology in Sema3e-/- recipients. Sema3e-/- mice display a higher frequency of CD11b+ pulmonary dendritic cells than their WT controls at the baseline and after sensitization with house dust mite. Adoptive transfer of CD11b+ pulmonary dendritic cells from Sema3e-/- mice into WT recipients increases house dust mite-induced Th2/Th17 inflammation in the airway. Together, these findings identify Sema3E as a novel regulatory molecule in allergic asthma that acts upstream of proallergic events and suggest that targeting this molecule could be a novel approach to treat allergic asthma.
Collapse
Affiliation(s)
- Hesam Movassagh
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| | - Lianyu Shan
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| | - Ashfaque Mohammed
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| | - Andrew J Halayko
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and.,Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5 Canada
| | - Abdelilah S Gounni
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| |
Collapse
|
36
|
Valladao AC, Frevert CW, Koch LK, Campbell DJ, Ziegler SF. STAT6 Regulates the Development of Eosinophilic versus Neutrophilic Asthma in Response to Alternaria alternata. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:4541-4551. [PMID: 27815425 PMCID: PMC5136320 DOI: 10.4049/jimmunol.1600007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 10/11/2016] [Indexed: 11/19/2022]
Abstract
Human asthma is a heterogeneous disease characterized by the expression of both Th2 and Th17 cytokines. In vitro and in vivo studies have shown a reciprocal regulation between Th2 and Th17 pathways, suggesting a potential induction of neutrophil-promoting Th17 inflammation in the absence of a Th2 response. Alternaria alternata is a clinically relevant allergen that is associated with severe and fatal asthma exacerbations. Exposure to A. alternata is characterized by a predominant Th2 response, but can also induce the production of factors associated with Th17 responses (e.g., CXCL8) from epithelial cells. Using a mouse model, we found that wild-type mice develop an eosinophilic Th2 airway disease in response to A. alternata exposure, whereas IL-4-, IL-13-, and STAT6-deficient mice exhibit a primarily neutrophilic response. Neutrophilic asthma in STAT6-/- mice was accompanied by elevated lung levels of TNF-α, CXCL1, CXCL2, and CXCL5, and was steroid resistant. Neutralization of Th17 signaling only partially reduced neutrophil numbers and total airway inflammation. Airway neutrophilia developed in RAG-deficient and CD4-depleted BALB/c mice, suggesting that the suppression of neutrophil responses is dependent on Th2 cytokine production by T cells and that airway neutrophilia is primarily an innate response to allergen. These results highlight the importance of combination therapies for treatment of asthma and establish a role for factors other than IL-17 as targets for neutrophilic asthma.
Collapse
Affiliation(s)
- Andrea C Valladao
- Department of Immunology, University of Washington, Seattle, WA 98101
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; and
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, WA 98101
| | - Lisa K Koch
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; and
| | - Daniel J Campbell
- Department of Immunology, University of Washington, Seattle, WA 98101
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; and
| | - Steven F Ziegler
- Department of Immunology, University of Washington, Seattle, WA 98101;
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; and
| |
Collapse
|
37
|
Im J, Kim K, Yhee JY, O'Grady SM, Nho RS. Desensitization of idiopathic pulmonary fibrosis fibroblasts to Alternaria alternata extract-mediated necrotic cell death. Physiol Rep 2016; 4:4/21/e13020. [PMID: 27905295 PMCID: PMC5112498 DOI: 10.14814/phy2.13020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/06/2016] [Accepted: 10/08/2016] [Indexed: 12/31/2022] Open
Abstract
Alternaria alternata is an allergenic fungus and known to cause an upper respiratory tract infection and asthma in humans with compromised immunity. Although A. alternata's effect on airway epithelial cells has previously been examined, the potential role of A. alternata on lung fibroblast viability is not understood. Since lung fibroblasts derived from patients with idiopathic pulmonary fibrosis (IPF) display a distinct phenotype that is resistant to stress and cell death inducing conditions, the investigation of the role of Alternaria on pathological IPF fibroblasts provides a better understanding of the fibrotic process induced by an allergenic fungus. Therefore, we examined cell viability of control and IPF fibroblasts (n = 8 each) in response to A. alternata extract. Control fibroblast cell death was increased while IPF fibroblasts were resistant when exposed to 50-100 μg/mL of A. alternata extract. However, there was no significant difference in kinetics or magnitude of Ca2+ responses from control lung and IPF fibroblasts. In contrast, unlike control fibroblasts, intracellular reactive oxygen species (ROS) levels remained low when IPF cells were treated with A. alternata extracts as a function of time. Caspase 3/7 and TUNEL assay revealed that enhanced cell death caused by A. alternata extract was likely due to necrosis, and 7-AAD assay and the use of sodium pyruvate for ATP generation further supported our findings that IPF fibroblasts become resistant to A. alternata extract-induced necrotic cell death. Our results suggest that exposure to A. alternata potentially worsens the fibrotic process by promoting normal lung fibroblast cell death in patients with IPF.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kyutae Kim
- College of Biological Science, University of Minnesota, St. Paul, Minnesota
| | - Ji Young Yhee
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Scott M O'Grady
- Department of Animal Science, Integrative Biology and Physiology, University of Minnesota, St. Paul, Minnesota
| | - Richard S Nho
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
38
|
Eddens T, Campfield BT, Serody K, Manni ML, Horne W, Elsegeiny W, McHugh KJ, Pociask D, Chen K, Zheng M, Alcorn JF, Wenzel S, Kolls JK. A Novel CD4 + T Cell-Dependent Murine Model of Pneumocystis-driven Asthma-like Pathology. Am J Respir Crit Care Med 2016; 194:807-820. [PMID: 27007260 DOI: 10.1164/rccm.201511-2205oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Infection with Pneumocystis, an opportunistic fungal pathogen, can result in fulminant pneumonia in the clinical setting of patients with immunosuppression. In murine models, Pneumocystis has previously been shown to induce a CD4+ T cell-dependent eosinophilic response in the lung capable of providing protection. OBJECTIVES We sought to explore the role of Pneumocystis in generating asthma-like lung pathology, given the natural eosinophilic response to infection. METHODS Pneumocystis infection or antigen treatment was used to induce asthma-like pathology in wild-type mice. The roles of CD4+ T cells and eosinophils were examined using antibody depletion and knockout mice, respectively. The presence of anti-Pneumocystis antibodies in human serum samples was detected by ELISA and Western blotting. MEASUREMENTS AND MAIN RESULTS Pneumocystis infection generates a strong type II response in the lung that requires CD4+ T cells. Pneumocystis infection was capable of priming a Th2 response similar to that of a commonly studied airway allergen, the house dust mite. Pneumocystis antigen treatment was also capable of inducing allergic inflammation in the lung, resulting in anti-Pneumocystis IgE production, goblet cell hyperplasia, and increased airway resistance. In the human population, patients with severe asthma had increased levels of anti-Pneumocystis IgG and IgE compared with healthy control subjects. Patients with severe asthma with elevated anti-Pneumocystis IgG levels had worsened symptom scores and lung parameters such as decreased forced expiratory volume and increased residual volume compared with patients with severe asthma who had low anti-Pneumocystis IgG. CONCLUSIONS The present study demonstrates for the first time, to our knowledge, that Pneumocystis is an airway allergen capable of inducing asthma-like lung pathology.
Collapse
Affiliation(s)
- Taylor Eddens
- 1 Richard King Mellon Foundation Institute for Pediatric Research and.,2 Department of Immunology
| | - Brian T Campfield
- 1 Richard King Mellon Foundation Institute for Pediatric Research and.,3 Division of Pediatric Infectious Diseases, Department of Pediatrics, and
| | - Katelin Serody
- 1 Richard King Mellon Foundation Institute for Pediatric Research and
| | - Michelle L Manni
- 4 Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - William Horne
- 1 Richard King Mellon Foundation Institute for Pediatric Research and
| | - Waleed Elsegeiny
- 1 Richard King Mellon Foundation Institute for Pediatric Research and.,2 Department of Immunology
| | - Kevin J McHugh
- 4 Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Derek Pociask
- 5 Department of Pulmonary Diseases, Critical Care, and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Kong Chen
- 1 Richard King Mellon Foundation Institute for Pediatric Research and
| | - Mingquan Zheng
- 1 Richard King Mellon Foundation Institute for Pediatric Research and
| | - John F Alcorn
- 4 Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Sally Wenzel
- 6 Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Jay K Kolls
- 1 Richard King Mellon Foundation Institute for Pediatric Research and
| |
Collapse
|
39
|
Caniga M, Cabal A, Mehta K, Ross DS, Gil MA, Woodhouse JD, Eckman J, Naber JR, Callahan MK, Goncalves L, Hill SE, Mcleod RL, McIntosh F, Freke MC, Visser SA, Johnson N, Salmon M, Cicmil M. Preclinical Experimental and Mathematical Approaches for Assessing Effective Doses of Inhaled Drugs, Using Mometasone to Support Human Dose Predictions. J Aerosol Med Pulm Drug Deliv 2016; 29:362-77. [PMID: 26859446 DOI: 10.1089/jamp.2015.1253] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Michael Caniga
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Antonio Cabal
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Khamir Mehta
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - David S. Ross
- Center for Applied and Computational Mathematics, Rochester Institute of Technology, Rochester, New York
| | - Malgorzata A. Gil
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Janice D. Woodhouse
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Joseph Eckman
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - John R. Naber
- Department of Discovery Pharmaceutical Sciences, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Marissa K. Callahan
- Department of Discovery Pharmaceutical Sciences, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Luciano Goncalves
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Susan E. Hill
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Robbie L. Mcleod
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Fraser McIntosh
- Discovery Research Services Charles River Laboratories, Senneville, Quebec, Canada
| | - Mark C. Freke
- Discovery Research Services Charles River Laboratories, Senneville, Quebec, Canada
| | - Sandra A.G. Visser
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Neil Johnson
- Department of Safety Assessment and Laboratory Animal Sciences, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Michael Salmon
- Department of Biology Discovery, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| | - Milenko Cicmil
- Department of Pharmacology, Merck Research Laboratories, Boston, Massachusetts, and West Point, Pennsylvania
| |
Collapse
|
40
|
Anderson EL, Kobayashi T, Iijima K, Bartemes KR, Chen CC, Kita H. IL-33 mediates reactive eosinophilopoiesis in response to airborne allergen exposure. Allergy 2016; 71:977-88. [PMID: 26864308 DOI: 10.1111/all.12861] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Exposure to aeroallergens induces eosinophilic airway inflammation in patients with asthma and allergic airway diseases. The circulating number of eosinophils in peripheral blood is relatively small, leading us to hypothesize that bone marrow needs to be engaged quickly to meet the demands of the tissues. METHODS To investigate the communication between the lungs and bone marrow, we used acute allergen exposure and airway inflammation models in mice. Gene-deficient mice and cytokine reporter mice as well as in vitro cell culture models were used to dissect the mechanisms. RESULTS Naïve BALB/c mice produced increased numbers of eosinophil precursors and mature eosinophils in the bone marrow when their airways were exposed to a common fungal allergen, Alternaria alternata. Expression of IL-5 and IL-33 increased rapidly in the lungs, but not in the bone marrow. Sera from allergen-exposed mice promoted eosinophilopoiesis in bone marrow cells from naïve mice, which was blocked by anti-IL-5 antibody. Mice deficient in the IL-33 receptor ST2 (i.e., Il1rl1(-/-) mice) were unable to increase their serum levels of IL-5 and allergen-induced eosinophilopoiesis in the bone marrow after allergen exposure. Finally, group 2 innate lymphoid cells (ILC2s) in the lungs showed robust expression of IL-5 after Alternaria exposure. CONCLUSIONS These finding suggests that lung IL-33, through innate activation of ILC2s and their production of IL-5, plays a key role in promoting acute reactive eosinophilopoiesis in the bone marrow when naïve animals are exposed to airborne allergens. Therefore, bone marrow eosinophilopoiesis may be affected by atmospheric environmental conditions.
Collapse
Affiliation(s)
| | - T. Kobayashi
- Division of Allergic Diseases; Department of Internal Medicine; Mayo Clinic; Rochester MN USA
| | - K. Iijima
- Division of Allergic Diseases; Department of Internal Medicine; Mayo Clinic; Rochester MN USA
| | | | - C.-C. Chen
- Department of Immunology; Mayo Clinic; Rochester MN USA
| | - H. Kita
- Department of Immunology; Mayo Clinic; Rochester MN USA
- Division of Allergic Diseases; Department of Internal Medicine; Mayo Clinic; Rochester MN USA
| |
Collapse
|
41
|
Li BWS, de Bruijn MJW, Tindemans I, Lukkes M, KleinJan A, Hoogsteden HC, Hendriks RW. T cells are necessary for ILC2 activation in house dust mite-induced allergic airway inflammation in mice. Eur J Immunol 2016; 46:1392-403. [PMID: 27062360 DOI: 10.1002/eji.201546119] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/14/2016] [Accepted: 03/31/2016] [Indexed: 01/29/2023]
Abstract
Allergic asthma is a chronic inflammation of the airways mediated by an adaptive type 2 immune response. Upon allergen exposure, group 2 innate lymphoid cells (ILC2s) can be rapidly activated and represent an early innate source of IL-5 and IL-13. Here, we used a house dust mite (HDM)-driven asthma mouse model to study the induction of ILC2s in allergic airway inflammation. In BALF, lungs, and lymph nodes, ILC2 activation is critically dependent on prior sensitization with HDM. Importantly, T cells are required for ILC2 induction, whereby T-cell activation precedes ILC2 induction. During HDM-driven allergic airway inflammation the accumulation of ILC2s in BALF is IL-33 independent, although infiltrating ILC2s produce less cytokines in Il33(-/-) mice. Transfer of in vitro polarized OVA-specific OT-II Th2 cells alone or in combination with Th17 cells followed by OVA and HDM challenge is not sufficient to induce ILC2, despite significant eosinophilic inflammation and T-cell activation. In this asthma model, ILC2s are therefore not an early source of Th2 cytokines, but rather contribute to type 2 inflammation in which Th2 cells play a key role. Taken together, ILC2 induction in HDM-mediated allergic airway inflammation in mice critically depends on activation of T cells.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | | | - Irma Tindemans
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - Melanie Lukkes
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - Henk C Hoogsteden
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
42
|
Abstract
Recent discoveries have led to the identification of a novel group of immune cells, the innate lymphoid cells (ILCs). The members of this group are divided into three subpopulations: ILC1s, ILC2s, and ILC3s. ILC2s produce Th2 cytokines, IL-4, IL-5, and IL-13, upon activation by epithelial cell-derived cytokines, lipid mediators (cysteinyl leukotrienes and prostaglandin D2), and TNF family member TL1A and promote structural and immune cell responses in the airways after antigen exposure. In addition, ILC2 function is also influenced by inducible T cell costimulator (ICOS)/ICOS-ligand (ICOS-L) interactions via direct contact between immune cells. The most common airway antigens are allergens and viruses which are highly linked to the induction of airway diseases with underlying type 2 inflammation including asthma and allergic rhinitis. Based on recent findings linking ILC2s and airway Th2 responses, there is intensive investigation into the role of ILC2s in human disease with the hope of a better understanding of the pathophysiology and the discovery of novel potential therapeutic targets. This review summarizes the recent advances made in elucidating ILC2 involvement in human Th2 airway disease.
Collapse
|
43
|
Cianferoni A, Spergel J. The importance of TSLP in allergic disease and its role as a potential therapeutic target. Expert Rev Clin Immunol 2015; 10:1463-74. [PMID: 25340427 DOI: 10.1586/1744666x.2014.967684] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thymic stromal lymphopoietin (TSLP) is an epithelial-derived cytokine similar to IL- 7, whose gene is located on chromosome 5q22.1 and it exerts its biological function through the TSLP-Receptor (TSLP-R). TSLP is expressed primarily by epithelial cells at barrier surfaces such as the skin, gut and lung in response to danger signals. Since it was cloned in 1994, there has been accumulating evidence that TSLP is crucial for the maturation of antigen presenting cells and hematopoietic cells. TSLP genetic variants and its dysregulated expression have been linked to atopic diseases such as atopic dermatitis, asthma, allergic rhinitis and eosinophilic esophagitis.
Collapse
Affiliation(s)
- Antonella Cianferoni
- The Children's Hospital of Philadelphia - Allergy, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
44
|
Lee DCP, Walker SA, Byrne AJ, Gregory LG, Buckley J, Bush A, Shaheen SO, Saglani S, Lloyd CM. Perinatal paracetamol exposure in mice does not affect the development of allergic airways disease in early life. Thorax 2015; 70:528-36. [PMID: 25841236 PMCID: PMC4453715 DOI: 10.1136/thoraxjnl-2014-205280] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/24/2015] [Indexed: 01/15/2023]
Abstract
Background Current data concerning maternal paracetamol intake during pregnancy, or intake during infancy and risk of wheezing or asthma in childhood is inconclusive based on epidemiological studies. We have investigated whether there is a causal link between maternal paracetamol intake during pregnancy and lactation and the development of house dust mite (HDM) induced allergic airways disease (AAD) in offspring using a neonatal mouse model. Methods Pregnant mice were administered paracetamol or saline by oral gavage from the day of mating throughout pregnancy and/or lactation. Subsequently, their pups were exposed to intranasal HDM or saline from day 3 of life for up to 6 weeks. Assessments of airway hyper-responsiveness, inflammation and remodelling were made at weaning (3 weeks) and 6 weeks of age. Results Maternal paracetamol exposure either during pregnancy and/or lactation did not affect development of AAD in offspring at weaning or at 6 weeks. There were no effects of maternal paracetamol at any time point on airway remodelling or IgE levels. Conclusions Maternal paracetamol did not enhance HDM induced AAD in offspring. Our mechanistic data do not support the hypothesis that prenatal paracetamol exposure increases the risk of childhood asthma.
Collapse
Affiliation(s)
- Debbie C P Lee
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK Immunology Programme, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Simone A Walker
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam J Byrne
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa G Gregory
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - James Buckley
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrew Bush
- Department of Respiratory Paediatrics, Royal Brompton Hospital, and National Heart and Lung Institute, Imperial College London, London, UK
| | - Seif O Shaheen
- Centre for Primary Care and Public Health, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Sejal Saglani
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK Department of Respiratory Paediatrics, Royal Brompton Hospital, and National Heart and Lung Institute, Imperial College London, London, UK
| | - Clare M Lloyd
- Leukocyte Biology Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
45
|
Doherty TA. At the bench: understanding group 2 innate lymphoid cells in disease. J Leukoc Biol 2015; 97:455-67. [PMID: 25473099 PMCID: PMC4338843 DOI: 10.1189/jlb.5bt0814-374r] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/21/2014] [Accepted: 10/29/2014] [Indexed: 12/18/2022] Open
Abstract
The conventional paradigm of type 2 inflammatory responses is characterized by activation of CD4(+) Th2 cells that produce IL-4, IL-5, and IL-13, resulting in tissue eosinophil infiltration, mucus metaplasia, AHR, and IgE production. However, the recent discovery of ILC2s in mice and humans has brought forth a novel pathway in type 2 immunity that may work independent of, or in concert with, adaptive Th2 responses. ILC2s were described initially as lineage-negative lymphocytes that produce high levels of Th2 cytokines IL-5 and IL-13 in response to IL-25 and IL-33 and promote protection against helminth infections. More recent investigations have identified novel upstream regulators, as well as novel ILC2 products. ILC2s are found in mucosal surfaces, including respiratory tract and skin, and studies from experimental asthma and atopic dermatitis models support a role for ILC2s in promoting type 2 inflammatory responses. There are many unanswered questions about the role of ILC2s in chronic allergic diseases, including how ILC2s or upstream pathways can be targeted for therapy. As ILC2s are not antigen specific and may be activated after exposures to a variety of infectious agents and irritants thought to contribute to respiratory and skin diseases, future strategies to target ILC2 function in human disease may be promising. Our intent is to identify priority areas for ILC2 translational research based on basic research insights.
Collapse
Affiliation(s)
- Taylor A Doherty
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
46
|
Doherty TA, Broide DH. Group 2 innate lymphoid cells: new players in human allergic diseases. J Investig Allergol Clin Immunol 2015; 25:1-11. [PMID: 25898689 PMCID: PMC4545833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023] Open
Abstract
Allergic diseases are characterized by tissue eosinophilia, mucus secretion, IgE production, and activation of mast cells and TH2 cells. Production of TH2 cytokines including IL-4, IL-5, IL-9, and IL-13 has mainly been attributed to CD4+T(H)2 cells. However, the recent discovery of group 2 innate lymphoid cells (ILC2s) in humans and findings from experimental disease models have challenged conventional concepts associated with the contribution of specific cells to type 2 inflammation in allergic diseases. ILC2s produce high levels of T(H)2 cytokines and have been detected in human lung tissue, peripheral blood, the gastrointestinal tract, skin, and sinonasal tissue, suggesting that ILC2s could contribute to chronic rhinosinusitis, asthma, atopic dermatitis, and gastrointestinal allergic disease. Moreover, depletion of ILC2s in animal models suggests a role for these cells in atopic dermatitis and asthma. This review will focus on the role of ILC2s in human allergy and asthma and provide a mechanistic insight from animal models.
Collapse
|
47
|
Jiménez-Garcia L, Herránz S, Luque A, Hortelano S. Critical role of p38 MAPK in IL-4-induced alternative activation of peritoneal macrophages. Eur J Immunol 2014; 45:273-86. [DOI: 10.1002/eji.201444806] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/28/2014] [Accepted: 10/13/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Lidia Jiménez-Garcia
- Unidad de Terapias Farmacológicas. Área de Genética Humana. Instituto de Investigación de Enfermedades Raras (IIER); Instituto de Salud Carlos III; Madrid Spain
| | - Sandra Herránz
- Unidad de Terapias Farmacológicas. Área de Genética Humana. Instituto de Investigación de Enfermedades Raras (IIER); Instituto de Salud Carlos III; Madrid Spain
| | - Alfonso Luque
- Unidad de Terapias Farmacológicas. Área de Genética Humana. Instituto de Investigación de Enfermedades Raras (IIER); Instituto de Salud Carlos III; Madrid Spain
| | - Sonsoles Hortelano
- Unidad de Terapias Farmacológicas. Área de Genética Humana. Instituto de Investigación de Enfermedades Raras (IIER); Instituto de Salud Carlos III; Madrid Spain
| |
Collapse
|
48
|
Lee MR, Shim D, Yoon J, Jang HS, Oh SW, Suh SH, Choi JH, Oh GT. Retnla overexpression attenuates allergic inflammation of the airway. PLoS One 2014; 9:e112666. [PMID: 25415454 PMCID: PMC4240542 DOI: 10.1371/journal.pone.0112666] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023] Open
Abstract
Resistin-like molecule alpha (Retnla), also known as ‘Found in inflammatory zone 1’, is a secreted protein that has been found in bronchoalveolar lavage (BAL) fluid of ovalbumin (OVA)-induced asthmatic mice and plays a role as a regulator of T helper (Th)2-driven inflammation. However, the role of Retnla in the progress of Th2-driven airway inflammation is not yet clear. To better understand the function of Retnla in Th2-driven airway inflammation, we generated Retnla-overexpressing (Retnla-Tg) mice. Retnla-Tg mice showed increased expression of Retnla protein in BAL fluid and airway epithelial cells. Retnla overexpression itself did not induce any alteration in lung histology or lung function compared to non-Tg controls. However, OVA-sensitized/challenged Retnla-Tg mice had decreased numbers of cells in BAL and inflammatory cells accumulating in the lung. They also showed a reduction in mucus production in the airway epithelium, concomitant with a decreased Muc5ac level. These results were accompanied by reduced levels of Th2 cytokines, including interleukin (IL)-4, IL-5, and IL-13, with no effect on levels of OVA-specific immunoglobulin isotypes. Furthermore, phosphorylation of ERK was markedly reduced in the lungs of OVA-challenged Retnla-Tg mice. Taken together, these results indicates that Retnla protects against Th2-mediated inflammation in an experimental mouse model of asthma, suggesting that therapeutic approaches to enhance the production of Retnla or Retnla-like molecules could be valuable for preventing allergic lung inflammation.
Collapse
Affiliation(s)
- Mi-Ran Lee
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Dahee Shim
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jihye Yoon
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Hyung Seok Jang
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Se-Woong Oh
- Yuhan Research Institute, Yuhan Corporation, Gongse-Dong, Giheung-Gu, Yongin-Si, Gyeonggi-Do, Republic of Korea
| | - Suk Hyo Suh
- Department of Physiology Medical School, Ewha Womans University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- * E-mail: (JHC); (GTO)
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
- * E-mail: (JHC); (GTO)
| |
Collapse
|
49
|
Drake LY, Iijima K, Kita H. Group 2 innate lymphoid cells and CD4+ T cells cooperate to mediate type 2 immune response in mice. Allergy 2014; 69:1300-7. [PMID: 24939388 DOI: 10.1111/all.12446] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Innate lymphoid cells (ILCs) play important roles in innate immunity and tissue remodeling via production of various cytokines and growth factors. Group 2 ILCs (ILC2s) were recently shown to mediate the immune pathology of asthma even without adaptive immunity. However, little is known about possible interactions between ILC2s and other immune cells. We sought to investigate the capacity of ILC2s to regulate effector functions of T cells. METHODS We isolated ILC2s from the lungs of naïve mice. We cultured CD4(+) T cells with ILC2s in vitro and examined the functions of these cell types. The mechanisms were investigated using blocking antibodies and cells isolated from cytokine-deficient mice. For the in vivo study, we adoptively transferred ILC2s and CD4(+) T cells into Il7ra(-/-) mice and subsequently exposed the mice to ovalbumin and a cysteine protease. RESULTS Lung ILC2s enhanced CD4(+) T-cell proliferation and promoted production of type 2 cytokines in vitro. The interaction between ILC2s and CD4(+) T cells involved costimulatory molecule OX40L and cytokine IL-4, which was mainly derived from ILC2s. Adoptive transfer of both ILC2 and CD4(+) T-cell populations, but not each population alone, into Il7ra(-/-) mice resulted in induction of a robust antigen-specific type 2 cytokine response and airway inflammation. CONCLUSION Lung ILC2s function to promote adaptive immunity in addition to their established roles in innate immunity. This novel function of ILC2s needs to be taken into account when considering the pathophysiology of asthma and other allergic airway diseases.
Collapse
Affiliation(s)
- L. Y. Drake
- Division of Allergic Diseases and Department of Medicine; Mayo Clinic; Rochester MN USA
| | - K. Iijima
- Division of Allergic Diseases and Department of Medicine; Mayo Clinic; Rochester MN USA
| | - H. Kita
- Division of Allergic Diseases; Departments of Medicine and Department of Immunology; Mayo Clinic; Rochester MN USA
| |
Collapse
|
50
|
Walford HH, Lund SJ, Baum RE, White AA, Bergeron CM, Husseman J, Bethel KJ, Scott DR, Khorram N, Miller M, Broide DH, Doherty TA. Increased ILC2s in the eosinophilic nasal polyp endotype are associated with corticosteroid responsiveness. Clin Immunol 2014; 155:126-135. [PMID: 25236785 DOI: 10.1016/j.clim.2014.09.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/21/2014] [Accepted: 09/09/2014] [Indexed: 01/08/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) have recently been identified in human nasal polyps, but whether numbers of ILC2s differ by polyp endotype or are influenced by corticosteroid use is unknown. Here, we show that eosinophilic nasal polyps contained double the number of ILC2s vs. non-eosinophilic polyps. Polyp ILC2s were also reduced by 50% in patients treated with systemic corticosteroids. Further, using a fungal allergen challenge mouse model, we detected greatly reduced Th2 cytokine-producing and Ki-67+ proliferating lung ILC2s in mice receiving dexamethasone. Finally, ILC2 Annexin V staining revealed extensive apoptosis after corticosteroid treatment in vivo and in vitro. Thus, ILC2s are elevated in the eosinophilic nasal polyp endotype and systemic corticosteroid treatment correlated with reduced polyp ILC2s. Finally, allergen-challenged mice showed reduced ILC2s and increased ILC2 apoptosis after corticosteroid treatment suggesting that ILC2 may be responsive to corticosteroids in eosinophilic respiratory disease.
Collapse
Affiliation(s)
- Hannah H Walford
- Department of Medicine, University of California-San Diego, San Diego, CA, USA; Rady's Children's Hospital of San Diego, Division of Rheumatology, Allergy and Immunology, San Diego, CA, USA
| | - Sean J Lund
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Rachel E Baum
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Andrew A White
- Division of Allergy, Asthma and Immunology, Scripps Clinic, La Jolla, CA, USA
| | | | - Jacob Husseman
- Department of Surgery, Division of Otolaryngology, University of California-San Diego, San Diego, CA, USA
| | - Kelly J Bethel
- Division of Surgical Pathology, Scripps Green Hospital, La Jolla, CA, USA
| | - David R Scott
- Division of Allergy, Asthma and Immunology, Scripps Clinic, La Jolla, CA, USA
| | - Naseem Khorram
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Marina Miller
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - David H Broide
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Taylor A Doherty
- Department of Medicine, University of California-San Diego, San Diego, CA, USA.
| |
Collapse
|