251
|
Taylor S, Huang Y, Mallett G, Stathopoulou C, Felizardo TC, Sun MA, Martin EL, Zhu N, Woodward EL, Elias MS, Scott J, Reynolds NJ, Paul WE, Fowler DH, Amarnath S. PD-1 regulates KLRG1 + group 2 innate lymphoid cells. J Exp Med 2017; 214:1663-1678. [PMID: 28490441 PMCID: PMC5461001 DOI: 10.1084/jem.20161653] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/06/2017] [Accepted: 03/21/2017] [Indexed: 11/04/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC-2s) regulate immune responses to pathogens and maintain tissue homeostasis in response to cytokines. Positive regulation of ILC-2s through ICOS has been recently elucidated. We demonstrate here that PD-1 is an important negative regulator of KLRG1+ ILC-2 function in both mice and humans. Increase in KLRG1+ ILC-2 cell numbers was attributed to an intrinsic defect in PD-1 signaling, which resulted in enhanced STAT5 activation. During Nippostrongylus brasiliensis infection, a significant expansion of KLRG1+ ILC-2 subsets occurred in Pdcd1-/- mice and, upon adoptive transfer, Pdcd1-/- KLRG1+ ILC-2s significantly reduced worm burden. Furthermore, blocking PD-1 with an antibody increased KLRG1+ ILC-2 cell number and reduced disease burden. Therefore, PD-1 is required for maintaining the number, and hence function, of KLRG1+ ILC-2s.
Collapse
Affiliation(s)
- Samuel Taylor
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yuefeng Huang
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Grace Mallett
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Chaido Stathopoulou
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Tania C Felizardo
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ming-An Sun
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Evelyn L Martin
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Nathaniel Zhu
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Emma L Woodward
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Martina S Elias
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Jonathan Scott
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| | - Nick J Reynolds
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK.,Department of Dermatology, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, England, UK
| | - William E Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daniel H Fowler
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Shoba Amarnath
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, NE1 7RU, England, UK
| |
Collapse
|
252
|
Laffont S, Blanquart E, Savignac M, Cénac C, Laverny G, Metzger D, Girard JP, Belz GT, Pelletier L, Seillet C, Guéry JC. Androgen signaling negatively controls group 2 innate lymphoid cells. J Exp Med 2017; 214:1581-1592. [PMID: 28484078 PMCID: PMC5461006 DOI: 10.1084/jem.20161807] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/23/2017] [Accepted: 03/31/2017] [Indexed: 01/17/2023] Open
Abstract
At the onset of adolescence, asthma becomes less prevalent in males than in females, suggesting a protective role of male sex hormones. Here, Laffont et al. show that androgens negatively control ILC2 development and ILC2-driven lung inflammation in male mice. Prevalence of asthma is higher in women than in men, but the mechanisms underlying this sex bias are unknown. Group 2 innate lymphoid cells (ILC2s) are key regulators of type 2 inflammatory responses. Here, we show that ILC2 development is greatly influenced by male sex hormones. Male mice have reduced numbers of ILC2 progenitors (ILC2Ps) and mature ILC2s in peripheral tissues compared with females. In consequence, males exhibit reduced susceptibility to allergic airway inflammation in response to environmental allergens and less severe IL-33–driven lung inflammation, correlating with an impaired expansion of lung ILC2s. Importantly, orchiectomy, but not ovariectomy, abolishes the sex differences in ILC2 development and restores IL-33–mediated lung inflammation. ILC2Ps express the androgen receptor (AR), and AR signaling inhibits their differentiation into mature ILC2s. Finally, we show that hematopoietic AR expression limits IL-33–driven lung inflammation through a cell-intrinsic inhibition of ILC2 expansion. Thus, androgens play a crucial protective role in type 2 airway inflammation by negatively regulating ILC2 homeostasis, thereby limiting their capacity to expand locally in response to IL-33.
Collapse
Affiliation(s)
- Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| | - Eve Blanquart
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| | - Magali Savignac
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| | - Claire Cénac
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| | - Gilles Laverny
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Daniel Metzger
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Gabrielle T Belz
- The Walter and Elisa Hall Institute of Medical Research, University of Melbourne, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lucette Pelletier
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| | - Cyril Seillet
- The Walter and Elisa Hall Institute of Medical Research, University of Melbourne, Parkville, Victoria 3052, Australia .,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Institut National de la Santé et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UPS, 31300 Toulouse, France
| |
Collapse
|
253
|
Thiriou D, Morianos I, Xanthou G, Samitas K. Innate immunity as the orchestrator of allergic airway inflammation and resolution in asthma. Int Immunopharmacol 2017; 48:43-54. [PMID: 28463786 DOI: 10.1016/j.intimp.2017.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/15/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
The respiratory system is constantly in direct contact with the environment and, has therefore, developed strong innate and adaptive immune responses to combat pathogens. Unlike adaptive immunity which is mounted later in the course of the immune response and is naive at the outset, innate immunity provides the first line of defense against microbial agents, while also promoting resolution of inflammation. In the airways, innate immune effector cells mainly consist of eosinophils, neutrophils, mast cells, basophils, macrophages/monocytes, dendritic cells and innate lymphoid cells, which attack pathogens directly or indirectly through the release of inflammatory cytokines and antimicrobial peptides, and coordinate T and B cell-mediated adaptive immunity. Airway epithelial cells are also critically involved in shaping both the innate and adaptive arms of the immune response. Chronic allergic airway inflammation and linked asthmatic disease is often considered a result of aberrant activation of type 2 T helper cells (Th2) towards innocuous environmental allergens; however, innate immune cells are increasingly recognized as key players responsible for the initiation and the perpetuation of allergic responses. Moreover, innate cells participate in immune response regulation through the release of anti-inflammatory mediators, and guide tissue repair and the maintenance of airway homeostasis. The scope of this review is to outline existing knowledge on innate immune responses involved in allergic airway inflammation, highlight current gaps in our understanding of the underlying molecular and cellular mechanisms and discuss the potential use of innate effector cells in new therapeutic avenues.
Collapse
Affiliation(s)
- Despoina Thiriou
- 2(nd) Respiratory Medicine Dept., Athens Chest Hospital "Sotiria", Athens, Greece
| | - Ioannis Morianos
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Konstantinos Samitas
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece; 7(th) Respiratory Medicine Dept. and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece.
| |
Collapse
|
254
|
IL-33 induces both regulatory B cells and regulatory T cells in dextran sulfate sodium-induced colitis. Int Immunopharmacol 2017; 46:38-47. [DOI: 10.1016/j.intimp.2017.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/14/2017] [Accepted: 02/03/2017] [Indexed: 12/15/2022]
|
255
|
Griesenauer B, Paczesny S. The ST2/IL-33 Axis in Immune Cells during Inflammatory Diseases. Front Immunol 2017; 8:475. [PMID: 28484466 PMCID: PMC5402045 DOI: 10.3389/fimmu.2017.00475] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Il1rl1 (also known as ST2) is a member of the IL-1 superfamily, and its only known ligand is IL-33. ST2 exists in two forms as splice variants: a soluble form (sST2), which acts as a decoy receptor, sequesters free IL-33, and does not signal, and a membrane-bound form (ST2), which activates the MyD88/NF-κB signaling pathway to enhance mast cell, Th2, regulatory T cell (Treg), and innate lymphoid cell type 2 functions. sST2 levels are increased in patients with active inflammatory bowel disease, acute cardiac and small bowel transplant allograft rejection, colon and gastric cancers, gut mucosal damage during viral infection, pulmonary disease, heart disease, and graft-versus-host disease. Recently, sST2 has been shown to be secreted by intestinal pro-inflammatory T cells during gut inflammation; on the contrary, protective ST2-expressing Tregs are decreased, implicating that ST2/IL-33 signaling may play an important role in intestinal disease. This review will focus on what is known on its signaling during various inflammatory disease states and highlight potential avenues to intervene in ST2/IL-33 signaling as treatment options.
Collapse
Affiliation(s)
- Brad Griesenauer
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Department of Microbiology Immunology, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Department of Microbiology Immunology, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
256
|
Poposki JA, Klingler AI, Tan BK, Soroosh P, Banie H, Lewis G, Hulse KE, Stevens WW, Peters AT, Grammer LC, Schleimer RP, Welch KC, Smith SS, Conley DB, Raviv JR, Karras JG, Akbari O, Kern RC, Kato A. Group 2 innate lymphoid cells are elevated and activated in chronic rhinosinusitis with nasal polyps. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:233-243. [PMID: 28474861 PMCID: PMC5569375 DOI: 10.1002/iid3.161] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/20/2022]
Abstract
Background Chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) is characterized by type 2 inflammation with high levels of Th2 cytokines. Although T helper cytokines are released from T cells, innate lymphoid cells (ILC) are also known to produce high levels of the same cytokines. However, the presence of various types of ILC in CRS is poorly understood. Objective The objective of this study was to fully characterize the presence of all ILC subsets in CRS and to identify phenotypical differences of group 2 ILC (ILC2) in CRSwNP compared to ILC2 from non‐type 2 inflamed areas. Methods We investigated the presence of ILC subsets in peripheral blood mononuclear cells (PBMC) from healthy subjects, tonsil tissue, ethmoid tissue from control subjects and patients with non‐polypoid CRS (CRSsNP) and CRSwNP, as well as nasal polyp (NP) tissue from CRSwNP by flow cytometry. Sorted ILC2 were cultured in the presence and absence of IL‐33 and production of IL‐5 and IL‐13 was assessed by Luminex. Results We found that all ILC subsets were present in NP but ILC2 were dominant and significantly elevated compared to PBMC, tonsil, CRSsNP, and normal sinus tissue. We also found that inducible T‐cell co‐stimulator (ICOS) and side scatter were increased and CD127 was down‐regulated in ILC2 from NP compared to blood or tonsil ILC2. Thymic stromal lymphopoietin, IL‐7, and IL‐33 were able to down‐regulate expression of CD127 and increase side scatter in blood ILC2. Furthermore, sorted NP ILC2 but not blood ILC2 spontaneously released type 2 cytokines including IL‐5 and IL‐13. Conclusions and Clinical Relevance These results suggest that ILC2 are not only elevated but also activated in CRSwNP in vivo and that ILC2 may play important roles in the type 2 inflammation in CRSwNP.
Collapse
Affiliation(s)
- Julie A Poposki
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Aiko I Klingler
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bruce K Tan
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Pejman Soroosh
- Janssen Research and Development, San Diego, California, USA
| | - Homayon Banie
- Janssen Research and Development, San Diego, California, USA
| | - Gavin Lewis
- Janssen Research and Development, San Diego, California, USA
| | - Kathryn E Hulse
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Whitney W Stevens
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anju T Peters
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leslie C Grammer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert P Schleimer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kevin C Welch
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Stephanie S Smith
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - David B Conley
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Joseph R Raviv
- Division of Otolaryngology-Head and Neck Surgery, NorthShore University HealthSystem, The University of Chicago, Pritzker School of Medicine, Evanston, Illinois, USA
| | - James G Karras
- Janssen Research and Development, San Diego, California, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Robert C Kern
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Atsushi Kato
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
257
|
Furuhashi K, Chua YL, Wong KHS, Zhou Q, Lee DCP, Liong KH, Teo GH, Hutchinson PE, Kemeny DM. Priming with high and low respiratory allergen dose induces differential CD4 + T helper type 2 cells and IgE/IgG1 antibody responses in mice. Immunology 2017; 151:227-238. [PMID: 28190273 DOI: 10.1111/imm.12726] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 02/03/2017] [Accepted: 02/05/2017] [Indexed: 12/20/2022] Open
Abstract
Sensitization of allergic patients normally takes place over several years and is the result of repeated exposure to low levels of allergen. Most mouse asthma models use a high dose of allergen administered over a short period. We have investigated the role of dose in the immune response to an inhaled respiratory allergen (Blomia tropicalis). We observed the effect of priming dose on the allergic response in mice intranasally immunized with low (0·5 μg) and high (50 μg) doses of B. tropicalis extract and killed 1 day after the last challenge. For both doses of allergen, T helper type 2 (Th2) cells and Th2 cytokines were evident as well as eosinophilic inflammation accompanied by mucus hyper-secretion. By contrast, IgE and IgG1 antibody responses were normally only detected at high-dose priming. To investigate the mechanism for these effects, we found group 2 innate lymphoid cells (ILC2s) were increased 48 hr after challenge in the low-dose-treated but not the high-dose-treated mice. Furthermore, we determined whether repeated low-dose exposure with different priming protocols could induce an antibody response. Repeated low-dose exposure to 0·5 μg three times weekly for 4 weeks (cumulative 6 μg) had the same effect as a shorter high-dose exposure (cumulative 80 μg) and increasing cumulative dose induced antibody responses. These data indicate that low doses of allergen are sufficient to prime Th2 cells and ILC2s, but insufficient to induce antibody responses. Cumulative exposure to small amounts of allergen induces both Th2 and antibody responses and may better reflect natural sensitization.
Collapse
Affiliation(s)
- Kazuki Furuhashi
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Yen L Chua
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Kenneth H S Wong
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Qian Zhou
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Debbie C P Lee
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Ka H Liong
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Guo H Teo
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - Paul E Hutchinson
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| | - David M Kemeny
- Department of Microbiology, Immunology Programme, National University of Singapore, Singapore city, Singapore
| |
Collapse
|
258
|
Karta MR, Rosenthal PS, Beppu A, Vuong CY, Miller M, Das S, Kurten RC, Doherty TA, Broide DH. β 2 integrins rather than β 1 integrins mediate Alternaria-induced group 2 innate lymphoid cell trafficking to the lung. J Allergy Clin Immunol 2017; 141:329-338.e12. [PMID: 28366795 DOI: 10.1016/j.jaci.2017.03.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 01/23/2017] [Accepted: 03/15/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) expand in the lungs of mice during type 2 inflammation induced by the fungal allergen Alternaria alternata. The increase in ILC2 numbers in the lung has been largely attributed to local proliferation and whether ILC2s migrate from the circulation to the lung after Alternaria exposure is unknown. OBJECTIVE We examined whether human (lung, lymph node, and blood) and mouse lung ILC2s express β1 and β2 integrin adhesion molecules and whether these integrins are required for trafficking of ILC2s into the lungs of mice. METHODS Human and mouse ILC2s were assessed for surface expression of β1 and β2 integrin adhesion molecules by using flow cytometry. The role of β1 and β2 integrins in ILC2 trafficking to the lungs was assessed by in vivo blocking of these integrins before airway exposure to Alternaria in mice. RESULTS Both human and mouse lung ILC2s express high levels of β1 and β2 integrin adhesion receptors. Intranasal administration of Alternaria challenge reduced ILC2 numbers in the bone marrow and concurrently increased blood and lung ILC2 numbers. In vivo blocking of β2 integrins (CD18) significantly reduced ILC2 numbers in the lungs but did not alter ILC2 proliferation, apoptosis, and function. In contrast, in vivo blocking of β1 integrins or α4 integrins did not affect lung ILC2 numbers. CONCLUSION ILC2 numbers increase in the mouse lung not only through local proliferation but also through trafficking from the circulation into the lung using β2 rather than β1 or α4 integrins.
Collapse
Affiliation(s)
- Maya R Karta
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Peter S Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Andrew Beppu
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Christine Y Vuong
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Richard C Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital Research Institute, Little Rock, Ark
| | - Taylor A Doherty
- 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
|
259
|
Sato Y, Kato H, Ebina-Shibuya R, Itoh-Nakadai A, Okuyama R, Igarashi K. Bach2 Controls Homeostasis of Eosinophils by Restricting the Type-2 Helper Function of T Cells. TOHOKU J EXP MED 2017; 241:175-182. [PMID: 28216546 DOI: 10.1620/tjem.241.175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bach2 is a transcription factor which represses its target genes and plays important roles in the differentiation of B and T lymphoid cells. Bach2-deficient (KO) mice develop severe pulmonary alveolar proteinosis, which is associated with increased numbers of granulocytes and T cells. Bach2 is essential for the regulation of T cells, but its role in the regulation of granulocytes is not clear. Here, we observed increased numbers of eosinophils but not neutrophils in the bone marrow, spleen, peripheral blood, and bronchoalveolar lavage fluids of Bach2 KO mice compared with those of wild-type (WT) mice. Upon co-transplantation of the bone marrow cells from CD45.2 Bach2 KO and CD45.1/CD45.2 double-positive WT mice to irradiated WT CD45.1/CD45.2 mice, the reconstituted numbers of eosinophils were similar between Bach2 KO and WT cells. These results showed that the deficiency of Bach2 in eosinophils did not directly drive the differentiation of eosinophils. To investigate the effect of Bach2 KO CD4+ T cells upon eosinophils, we analyzed Rag2/Bach2-double deficient (dKO) mice which lack lymphocytes including CD4+ T cells. Rag2/Bach2 dKO mice did not show any increase in the numbers of eosinophils. Importantly, Bach2 KO mice showed an increase of interleukin-5 (Il-5) in the sera compared with WT mice. These results suggest that up-regulated functions of CD4+ T cells including secretion of Il-5 resulted in proliferation and/or migration to peripheral tissues of eosinophils in Bach2 KO mice. We propose that Bach2 controls homeostasis of eosinophils via restricting the production of Il-5 in CD4+ T cells.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Biochemistry, Tohoku University Graduate School of Medicine
| | | | | | | | | | | |
Collapse
|
260
|
Cheng H, Jin C, Wu J, Zhu S, Liu YJ, Chen J. Guards at the gate: physiological and pathological roles of tissue-resident innate lymphoid cells in the lung. Protein Cell 2017; 8:878-895. [PMID: 28271447 PMCID: PMC5712288 DOI: 10.1007/s13238-017-0379-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/02/2017] [Indexed: 01/03/2023] Open
Abstract
The lung is an important open organ and the primary site of respiration. Many life-threatening diseases develop in the lung, e.g., pneumonia, asthma, chronic obstructive pulmonary diseases (COPDs), pulmonary fibrosis, and lung cancer. In the lung, innate immunity serves as the frontline in both anti-irritant response and anti-tumor defense and is also critical for mucosal homeostasis; thus, it plays an important role in containing these pulmonary diseases. Innate lymphoid cells (ILCs), characterized by their strict tissue residence and distinct function in the mucosa, are attracting increased attention in innate immunity. Upon sensing the danger signals from damaged epithelium, ILCs activate, proliferate, and release numerous cytokines with specific local functions; they also participate in mucosal immune-surveillance, immune-regulation, and homeostasis. However, when their functions become uncontrolled, ILCs can enhance pathological states and induce diseases. In this review, we discuss the physiological and pathological functions of ILC subsets 1 to 3 in the lung, and how the pathogenic environment affects the function and plasticity of ILCs.
Collapse
Affiliation(s)
- Hang Cheng
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, 130061, China.,Department of Pediatrics, The First Hospital, Jilin University, Changchun, 130021, China
| | - Chengyan Jin
- Department of Thoracic Surgery, The Second Hospital, Jilin University, Changchun, 130041, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, 130061, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, 130061, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, 130061, China. .,Sanofi Research and Development, Cambridge, MA, 02139, USA.
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, 130061, China.
| |
Collapse
|
261
|
Group 2 innate lymphoid cells are recruited to the nasal mucosa in patients with aspirin-exacerbated respiratory disease. J Allergy Clin Immunol 2017; 140:101-108.e3. [PMID: 28279492 DOI: 10.1016/j.jaci.2016.11.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/26/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Aspirin-exacerbated respiratory disease (AERD) is characterized by tissue eosinophilia and mast cell activation, including abundant production of prostaglandin D2 (PGD2). Group 2 innate lymphoid cells (ILC2s), which promote tissue eosinophilia and mast cell responses, undergo chemotaxis and cytokine production in response to PGD2, but it is unknown whether ILC2s are active in patients with AERD. OBJECTIVE We sought to determine whether ILC2 numbers change in peripheral blood and the nasal mucosa during COX-1 inhibitor-induced reactions in patients with AERD. METHODS Blood and nasal scrapings were collected at baseline, during reactions, and after completion of ketorolac/aspirin challenge/desensitization in 12 patients with AERD. ILC2s and eosinophils were quantitated by means of flow cytometry. Urine was also collected, and quantification of PGD2 metabolite and leukotriene E4 levels was done by using ELISA. Baseline and nonsteroidal anti-inflammatory drug reaction clinical data were correlated with cell changes. RESULTS ILC2 numbers significantly increased in nasal mucosal samples and decreased in blood at the time of COX-1 inhibitor reactions in 12 patients with AERD. These changes were not observed in 2 patients without AERD. Furthermore, eosinophil numbers decreased in blood concurrently with significant increases in urinary PGD2 metabolite and leukotriene E4 levels. The magnitude of increases in nasal mucosal ILC2 numbers positively correlated with maximum symptom scores during challenges. Furthermore, blood ILC2 numbers during the reaction correlated with time for the reaction to resolve, possibly reflecting reaction severity. CONCLUSIONS ILC2s are recruited to the nasal mucosa during COX-1 inhibitor-induced reactions in patients with AERD, correlating with enhanced production of prostaglandins and leukotrienes.
Collapse
|
262
|
Corren J. Inflammatory Disorders Associated with Allergy: Overview of Immunopathogenesis and Implications for Treatment. Immunol Allergy Clin North Am 2017; 37:233-246. [PMID: 28366474 DOI: 10.1016/j.iac.2017.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A number of chronic inflammatory diseases are associated with IgE-mediated immunologic hypersensitivity, including atopic dermatitis, chronic rhinosinusitis, and asthma. Pathogenetic studies of well-characterized patient groups has allowed investigators to more precisely define the molecular pathways involved in these diseases. Specific cytokines and chemokines, as well as other unique proteins, have now been identified in each of these common disorders and a number of medications are currently in development for inhibiting their actions. Continual refinement of our understanding of the pathogenesis of these diseases will undoubtedly yield increasingly precise, and potentially more effective, treatments.
Collapse
Affiliation(s)
- Jonathan Corren
- Department of Medicine, Section of Clinical Immunology and Allergy, David Geffen School of Medicine at UCLA, 10780 Santa Monica Boulevard, Suite 280, Los Angeles, CA 90025, USA.
| |
Collapse
|
263
|
Loghmani A, Raoofi R, Ownagh A, Delirezh N. Alternaria alternata acts on human Monocyte-derived Dendritic cells to mediate Th2/Th17 polarisation. Allergol Immunopathol (Madr) 2017; 45:175-182. [PMID: 27823917 DOI: 10.1016/j.aller.2016.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Although the mechanism of asthma is not precisely understood in humans, clinical and epidemiological studies have offered a potential relationship between exposure to environmental fungi, such as Alternaria alternata (A. alternata) and the development and exacerbation of asthma. The aim of this project is to investigate the mechanisms of Th2 responses by A. alternata as a clinically relevant model for the environmental exposure. MATERIALS AND METHODS Plastic adherent monocytes were cultured with granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4) to convert these cells into Monocyte-derived Dendritic cells (MoDc) and then matured in the presence of Monocyte-Conditioned Medium (MCM) as the control group and MCM+ A. alternata extract as the inductive groups. RESULTS The results indicated that the expression of CD14 decreased and CD83 and anti-human leukocyte antigen-DR (HLA-DR) increased in the inductive groups in comparison with the control group. More importantly, A. alternata inhibited IL-12 production by activated dendritic cells (DCs), and the DCs exposed to A. alternata enhanced the Th2 polarisation of CD4+ T cells. The production amount of IL-10 overcame IL-12 as well as Il-23 increased significantly, and hand in T cells the production of cytokines Interferon-γ (IFN-γ) decreased. However, both IL-17 and IL-4 increased (p<0.05). Phagocytic activity in the inductive groups decreased significantly compared with the control group. CONCLUSION The asthma-related environmental fungus A. alternata, with an effect on dendritic cells profile mediates TH2/TH17. Such immunodysregulation properties of causative environmental fungi may explain their strong relationship with human asthma and allergic diseases.
Collapse
Affiliation(s)
- A Loghmani
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - R Raoofi
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| | - A Ownagh
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| | - N Delirezh
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| |
Collapse
|
264
|
Johansson K, Malmhäll C, Ramos-Ramírez P, Rådinger M. MicroRNA-155 is a critical regulator of type 2 innate lymphoid cells and IL-33 signaling in experimental models of allergic airway inflammation. J Allergy Clin Immunol 2017; 139:1007-1016.e9. [DOI: 10.1016/j.jaci.2016.06.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/24/2016] [Accepted: 06/28/2016] [Indexed: 01/01/2023]
|
265
|
Wu J, Hu H, Xu L, Qi F, Bai S, Cui Y, Chai R, Wang D, Liu B. Natural helper cells are associated with the exacerbated airway inflammation seen during RSV reinfection of neonatally primed mice. Int Immunopharmacol 2017; 45:156-162. [PMID: 28219838 DOI: 10.1016/j.intimp.2017.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 11/26/2022]
Abstract
Infection with respiratory syncytial virus (RSV) in neonatal mice causes more aggressive airway disease when the mice are reinfected with the same virus as adults. However, the underlying mechanisms responsible for this phenomenon are not entirely defined. Natural helper (NH) cells are considered a key factor for virus-induced or exacerbated airway inflammation and airway hyper-responsiveness by producing type 2 cytokines. To confirm whether NH cells are involved in the aggravated lung pathology seen during reinfection, BALB/c mice were initially infected as neonates and reinfected in adulthood. We observed that neonatal RSV infection resulted in an enhanced infiltration of eosinophils and neutrophils in the lungs, in parallel with a significant increase in the levels of IL-5 and IL-13 in bronchoalveolar lavage fluids on day 2 after reinfection. It seems likely that pulmonary NH cells may play a role in the occurrence, since mice first infected at 1wk of age developed an additional increase in the number of NH cells as well as IL-5- and IL-13-producing NH cells in the lungs than those first infected as young adults. In fact, an elevated expression of mRNAs for IL-5 and IL-13 in pulmonary NH cells was detected in mice first infected as neonates. Furthermore, adoptive transfer of NH cells into neonatal mice was able to boost eosinophilic infiltration as well as the production of type 2 cytokines in the lungs after reinfection at adulthood. In contrast, the expression of mRNA for the type 1 cytokine IFN-γ was down-regulated markedly by adoptive transfer of NH cells. Thus, these results suggest that Th2-type NH cells may play a role in the exacerbated airway inflammation seen during RSV reinfection of neonatally primed mice.
Collapse
Affiliation(s)
- Jianqi Wu
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Haiyan Hu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Lei Xu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Feifei Qi
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Song Bai
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Yulin Cui
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Ruonan Chai
- The PLA Center of Respiratory and Allergic Disease Diagnosing Management, the General Hospital of Shenyang Military Command, Shenyang, China
| | - Dandan Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
266
|
Chen CC, Kobayashi T, Iijima K, Hsu FC, Kita H. IL-33 dysregulates regulatory T cells and impairs established immunologic tolerance in the lungs. J Allergy Clin Immunol 2017; 140:1351-1363.e7. [PMID: 28196763 DOI: 10.1016/j.jaci.2017.01.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/10/2016] [Accepted: 01/05/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Airway exposure to environmental antigens generally leads to immunologic tolerance. A fundamental question remains: Why is airway tolerance compromised in patients with allergic airway diseases? IL-33 promotes innate and adaptive type 2 immunity and might provide the answer to this question. OBJECTIVE The goal of this study was to investigate the roles played by IL-33 in altering regulatory T (Treg) cells in the lungs and in affecting previously established airway immunologic tolerance. METHODS We analyzed CD4+ forkhead box P3 (Foxp3)+ Treg cells that were isolated from the lungs of naive BALB/c mice and those treated with IL-33. Airway tolerance and allergen-induced airway inflammation models in mice were used to investigate how IL-33 affects established immunologic tolerance in vivo. RESULTS CD4+Foxp3+ Treg cells in the lungs expressed the IL-33 receptor ST2. When exposed to IL-33, Treg cells upregulated their expression of the canonical TH2 transcription factor GATA3, as well as ST2, and produced type 2 cytokines. Treg cells lost their ability to suppress effector T cells in the presence of IL-33. Airway administration of IL-33 with an antigen impaired immunologic tolerance in the lungs that had been established by prior exposure to the antigen. Dysregulated Foxp3+ Treg cells with distinct characteristics of TH2 cells increased in the lungs of mice undergoing IL-33-dependent allergen-driven airway inflammation. CONCLUSIONS IL-33 dysregulated lung Treg cells and impaired immunologic tolerance to inhaled antigens. Established airway tolerance might not be sustained in the presence of an innate immunologic stimulus, such as IL-33.
Collapse
Affiliation(s)
| | | | - Koji Iijima
- Department of Medicine, Mayo Clinic Rochester, Minn
| | - Fan-Chi Hsu
- Department of Immunology, Mayo Clinic Rochester, Minn
| | - Hirohito Kita
- Department of Immunology, Mayo Clinic Rochester, Minn; Department of Medicine, Mayo Clinic Rochester, Minn.
| |
Collapse
|
267
|
Abstract
IL-9 is a pro-inflammatory cytokine implicated in certain immune-mediated diseases where chronic or acute inflammation of the mucosa plays an important role. Although initially described as being produced by what was then thought to be Th2 cells, it was later described that specialized lymphocyte populations are involved in IL-9 production. In addition to the classical Th9 effector (subset of CD4+ T cells), IL-9 is also produced by nonconventional lymphocytes, namely invariant natural killer T (iNKT) cells and innate lymphoid cells (ILCs). The identification of IL-9-producing cells by flow cytometry and cytokine measurements are pivotal for assigning and defining functional cellular phenotypes. In this chapter we provide methods for the in vitro polarization of IL-9-producing nonconventional lymphocytes and the best conditions for the detection of IL-9 production by intracellular staining.
Collapse
Affiliation(s)
- Silvia C P Almeida
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-025, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, 2780-156, Oeiras, Portugal
| | - Luis Graca
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-025, Lisbon, Portugal.
- Instituto Gulbenkian de Ciencia, 2780-156, Oeiras, Portugal.
| |
Collapse
|
268
|
Abstract
Sepsis remains a major clinical problem with high morbidity and mortality. As new inflammatory mediators are characterized, it is important to understand their roles in sepsis. Interleukin 33 (IL-33) is a recently described member of the IL-1 family that is widely expressed in cells of barrier tissues. Upon tissue damage, IL-33 is released as an alarmin and activates various types of cells of both the innate and adaptive immune system through binding to the ST2/IL-1 receptor accessory protein complex. IL-33 has apparent pleiotropic functions in many disease models, with its actions strongly shaped by the local microenvironment. Recent studies have established a role for the IL-33-ST2 axis in the initiation and perpetuation of inflammation during endotoxemia, but its roles in sepsis appear to be organism and model dependent. In this review, we focus on the recent advances in understanding the role of the IL-33/ST2 axis in sepsis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213 USA.,State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 China
| | - Heth R Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213 USA.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Rosemary Hoffman
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213 USA.,State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 China
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213 USA
| |
Collapse
|
269
|
von Moltke J, O'Leary CE, Barrett NA, Kanaoka Y, Austen KF, Locksley RM. Leukotrienes provide an NFAT-dependent signal that synergizes with IL-33 to activate ILC2s. J Exp Med 2016; 214:27-37. [PMID: 28011865 PMCID: PMC5206504 DOI: 10.1084/jem.20161274] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/02/2016] [Accepted: 11/25/2016] [Indexed: 12/20/2022] Open
Abstract
von Moltke et al. demonstrate that optimal cytokine induction in group 2 innate lymphocytes results from synergy between NFAT-dependent leukotriene signaling and IL-33 signaling. This integration of signaling pathways may represent an innate substitute for the T cell receptor. Group 2 innate lymphoid cells (ILC2s) and type 2 helper T cells (Th2 cells) are the primary source of interleukin 5 (IL-5) and IL-13 during type 2 (allergic) inflammation in the lung. In Th2 cells, T cell receptor (TCR) signaling activates the transcription factors nuclear factor of activated T cells (NFAT), nuclear factor κB (NF-κB), and activator protein 1 (AP-1) to induce type 2 cytokines. ILC2s lack a TCR and respond instead to locally produced cytokines such as IL-33. Although IL-33 induces AP-1 and NF-κB, NFAT signaling has not been described in ILC2s. In this study, we report a nonredundant NFAT-dependent role for lipid-derived leukotrienes (LTs) in the activation of lung ILC2s. Using cytokine reporter and LT-deficient mice, we find that complete disruption of LT signaling markedly diminishes ILC2 activation and downstream responses during type 2 inflammation. Type 2 responses are equivalently attenuated in IL-33– and LT-deficient mice, and optimal ILC2 activation reflects potent synergy between these pathways. These findings expand our understanding of ILC2 regulation and may have important implications for the treatment of airways disease.
Collapse
Affiliation(s)
- Jakob von Moltke
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143 .,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Claire E O'Leary
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Nora A Barrett
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115
| | - Yoshihide Kanaoka
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115
| | - K Frank Austen
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115
| | - Richard M Locksley
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143 .,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
270
|
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: 3.6] [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
|
271
|
Neumann K, Karimi K, Meiners J, Voetlause R, Steinmann S, Dammermann W, Lüth S, Asghari F, Wegscheid C, Horst AK, Tiegs G. A Proinflammatory Role of Type 2 Innate Lymphoid Cells in Murine Immune-Mediated Hepatitis. THE JOURNAL OF IMMUNOLOGY 2016; 198:128-137. [PMID: 27872212 DOI: 10.4049/jimmunol.1600418] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/20/2016] [Indexed: 12/24/2022]
Abstract
Type 2 innate lymphoid cells (ILC2) mediate inflammatory immune responses in the context of diseases triggered by the alarmin IL-33. In recent years, IL-33 has been implicated in the pathogenesis of immune-mediated liver diseases. However, the immunoregulatory function of ILC2s in the inflamed liver remains elusive. Using the murine model of Con A-induced immune-mediated hepatitis, we showed that selective expansion of ILC2s in the liver was associated with highly elevated hepatic IL-33 expression, severe liver inflammation, and infiltration of eosinophils. CD4+ T cell-mediated tissue damage and subsequent IL-33 release were responsible for the activation of hepatic ILC2s that produced the type 2 cytokines IL-5 and IL-13 during liver inflammation. Interestingly, ILC2 depletion correlated with less severe hepatitis and reduced accumulation of eosinophils in the liver, whereas adoptive transfer of hepatic ILC2s aggravated liver inflammation and tissue damage. We further showed that, despite expansion of hepatic ILC2s, 3-d IL-33 treatment before Con A challenge potently suppressed development of immune-mediated hepatitis. We found that IL-33 not only activated hepatic ILC2s but also expanded CD4+ Foxp3+ regulatory T cells (Treg) expressing the IL-33 receptor ST2 in the liver. This Treg subset also accumulated in the liver during resolution of immune-mediated hepatitis. In summary, hepatic ILC2s are poised to respond to the release of IL-33 upon liver tissue damage through expression of type 2 cytokines thereby participating in the pathogenesis of immune-mediated hepatitis. Inflammatory activity of ILC2s might be regulated by IL-33-elicited ST2+ Tregs that also arise in immune-mediated hepatitis.
Collapse
Affiliation(s)
- Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Khalil Karimi
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Jana Meiners
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Ruth Voetlause
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Silja Steinmann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Werner Dammermann
- 1. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany
| | - Stefan Lüth
- 1. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany
| | - Farahnaz Asghari
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Claudia Wegscheid
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Andrea K Horst
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany; and
| |
Collapse
|
272
|
Lee HS, Park DE, Lee JW, Chang Y, Kim HY, Song WJ, Kang HR, Park HW, Chang YS, Cho SH. IL-23 secreted by bronchial epithelial cells contributes to allergic sensitization in asthma model: role of IL-23 secreted by bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2016; 312:L13-L21. [PMID: 27864285 DOI: 10.1152/ajplung.00114.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 11/03/2016] [Indexed: 12/29/2022] Open
Abstract
IL-23 has been postulated to be a critical mediator contributing to various inflammatory diseases. Dermatophagoides pteronyssinus (Der p) is one of the most common inhalant allergens. However, the role of IL-23 in Der p-induced mouse asthma model is not well understood, particularly with regard to the development of allergic sensitization in the airways. The objective of this study was to evaluate roles of IL-23 in Der p sensitization and asthma development. BALB/c mice were repeatedly administered Der p intranasally to develop Der p allergic sensitization and asthma. After Der p local administration, changes in IL-23 expression were examined in lung tissues and primary epithelial cells. Anti-IL-23p19 antibody was given during the Der p sensitization period, and its effects were examined. Effects of anti-IL-23p19 antibody at bronchial epithelial levels were also examined in vitro. The expression of IL-23 at bronchial epithelial layers was increased after Der p local administration in mouse. In Der p-induced mouse models, anti-IL-23p19 antibody treatment during allergen sensitization significantly diminished Der p allergic sensitization and several features of allergic asthma including the production of Th2 cytokines and the population of type 2 innate lymphoid cells in lungs. The activation of dendritic cells in lung-draining lymph nodes was also reduced by anti-IL-23 treatment. In murine lung alveolar type II-like epithelial cell line (MLE-12) cells, IL-23 blockade prevented cytokine responses to Der p stimulation, such as IL-1α, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-33, and also bone marrow-derived dendritic cell activation. In conclusion, IL-23 is another important bronchial epithelial cell-driven cytokine which may contribute to the development of house dust mite allergic sensitization and asthma.
Collapse
Affiliation(s)
- Hyun Seung Lee
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Da-Eun Park
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Ji-Won Lee
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Yuna Chang
- Department of Medical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Young Kim
- Department of Medical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Jung Song
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; and
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; and
| | - Heung-Woo Park
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; and
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang-Heon Cho
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea; .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; and
| |
Collapse
|
273
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
274
|
Galle-Treger L, Suzuki Y, Patel N, Sankaranarayanan I, Aron JL, Maazi H, Chen L, Akbari O. Nicotinic acetylcholine receptor agonist attenuates ILC2-dependent airway hyperreactivity. Nat Commun 2016; 7:13202. [PMID: 27752043 PMCID: PMC5071851 DOI: 10.1038/ncomms13202] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
Allergic asthma is a complex and chronic inflammatory disorder that is associated with airway hyperreactivity (AHR) and driven by Th2 cytokine secretion. Type 2 innate lymphoid cells (ILC2s) produce large amounts of Th2 cytokines and contribute to the development of AHR. Here, we show that ILC2s express the α7-nicotinic acetylcholine receptor (α7nAChR), which is thought to have an anti-inflammatory role in several inflammatory diseases. We show that engagement of a specific agonist with α7nAChR on ILC2s reduces ILC2 effector function and represses ILC2-dependent AHR, while decreasing expression of ILC2 key transcription factor GATA-3 and critical inflammatory modulator NF-κB, and reducing phosphorylation of upstream kinase IKKα/β. Additionally, the specific α7nAChR agonist reduces cytokine production and AHR in a humanized ILC2 mouse model. Collectively, our data suggest that α7nAChR expressed by ILC2s is a potential therapeutic target for the treatment of ILC2-mediated asthma. Airway hyperreactivity is driven by type 2 cytokines produced by ILC2 and Th2 cells. Here the authors show that an α7-nicotinic receptor agonist (GTS-21) inhibits ILC2 responses and is therapeutic against Alternaria-induced airway hyperreactivity in a humanized mouse model.
Collapse
Affiliation(s)
- Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Yuzo Suzuki
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Nisheel Patel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Ishwarya Sankaranarayanan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Jennifer L Aron
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| | - Lin Chen
- Departments of Biological Science and Chemistry, University of Southern California, 1050 Childs Way RIH 201, Los Angeles, California 90089, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 1450 Biggy Street NRT 5509, Los Angeles, California 90033, USA
| |
Collapse
|
275
|
Rigas D, Lewis G, Aron JL, Wang B, Banie H, Sankaranarayanan I, Galle-Treger L, Maazi H, Lo R, Freeman GJ, Sharpe AH, Soroosh P, Akbari O. Type 2 innate lymphoid cell suppression by regulatory T cells attenuates airway hyperreactivity and requires inducible T-cell costimulator-inducible T-cell costimulator ligand interaction. J Allergy Clin Immunol 2016; 139:1468-1477.e2. [PMID: 27717665 DOI: 10.1016/j.jaci.2016.08.034] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/19/2016] [Accepted: 08/02/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND Atopic diseases, including asthma, exacerbate type 2 immune responses and involve a number of immune cell types, including regulatory T (Treg) cells and the emerging type 2 innate lymphoid cells (ILC2s). Although ILC2s are potent producers of type 2 cytokines, the regulation of ILC2 activation and function is not well understood. OBJECTIVE In the present study, for the first time, we evaluate how Treg cells interact with pulmonary ILC2s and control their function. METHODS ILC2s and Treg cells were evaluated by using in vitro suppression assays, cell-contact assays, and gene expression panels. Also, human ILC2s and Treg cells were adoptively transferred into NOD SCID γC-deficient mice, which were given isotype or anti-inducible T-cell costimulator ligand (ICOSL) antibodies and then challenged with IL-33 and assessed for airway hyperreactivity. RESULTS We show that induced Treg cells, but not natural Treg cells, effectively suppress the production of the ILC2-driven proinflammatory cytokines IL-5 and IL-13 both in vitro and in vivo. Mechanistically, our data reveal the necessity of inducible T-cell costimulator (ICOS)-ICOS ligand cell contact for Treg cell-mediated ILC2 suppression alongside the suppressive cytokines TGF-β and IL-10. Using a translational approach, we then demonstrate that human induced Treg cells suppress syngeneic human ILC2s through ICOSL to control airway inflammation in a humanized ILC2 mouse model. CONCLUSION These findings suggest that peripheral expansion of induced Treg cells can serve as a promising therapeutic target against ILC2-dependent asthma.
Collapse
Affiliation(s)
- Diamanda Rigas
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Gavin Lewis
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif; Janssen Research and Development, San Diego, Calif
| | - Jennifer L Aron
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Bowen Wang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | | | - Ishwarya Sankaranarayanan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Richard Lo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Mass
| | - Arlene H Sharpe
- Department of Microbiology and Immunology, Harvard Medical School, Boston, Mass
| | | | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
| |
Collapse
|
276
|
Abstract
Interleukin-33 (IL-33) - a member of the IL-1 family - was originally described as an inducer of type 2 immune responses, activating T helper 2 (TH2) cells and mast cells. Now, evidence is accumulating that IL-33 also potently stimulates group 2 innate lymphoid cells (ILC2s), regulatory T (Treg) cells, TH1 cells, CD8+ T cells and natural killer (NK) cells. This pleiotropic nature is reflected in the role of IL-33 in tissue and metabolic homeostasis, infection, inflammation, cancer and diseases of the central nervous system. In this Review, we highlight the molecular and cellular characteristics of IL-33, together with its major role in health and disease and the potential therapeutic implications of these findings in humans.
Collapse
|
277
|
Gordon ED, Palandra J, Wesolowska-Andersen A, Ringel L, Rios CL, Lachowicz-Scroggins ME, Sharp LZ, Everman JL, MacLeod HJ, Lee JW, Mason RJ, Matthay MA, Sheldon RT, Peters MC, Nocka KH, Fahy JV, Seibold MA. IL1RL1 asthma risk variants regulate airway type 2 inflammation. JCI Insight 2016; 1:e87871. [PMID: 27699235 PMCID: PMC5033813 DOI: 10.1172/jci.insight.87871] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/21/2016] [Indexed: 01/19/2023] Open
Abstract
Genome-wide association studies of asthma have identified genetic variants in the IL1RL1 gene, but the molecular mechanisms conferring risk are unknown. IL1RL1 encodes the ST2 receptor (ST2L) for IL-33 and an inhibitory decoy receptor (sST2). IL-33 promotes type 2 inflammation, which is present in some but not all asthmatics. We find that two single nucleotide polymorphisms (SNPs) in IL1RL1 - rs1420101 and rs11685480 - are strongly associated with plasma sST2 levels, though neither is an expression quantitative trait locus (eQTL) in whole blood. Rather, rs1420101 and rs11685480 mark eQTLs in airway epithelial cells and distal lung parenchyma, respectively. We find that the genetically determined plasma sST2 reservoir, derived from the lung, neutralizes IL-33 activity, and these eQTL SNPs additively increase the risk of airway type 2 inflammation among asthmatics. These risk variants define a population of asthmatics at risk of IL-33-driven type 2 inflammation.
Collapse
Affiliation(s)
- Erin D. Gordon
- Department of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Joe Palandra
- Pfizer Inc., Pharmacodynamics and Metabolism, Andover, Massachusetts, USA
| | | | - Lando Ringel
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Cydney L. Rios
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | | | - Louis Z. Sharp
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Jamie L. Everman
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Hannah J. MacLeod
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Jae W. Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, UCSF, San Francisco, California, USA
| | - Robert J. Mason
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Michael A. Matthay
- Department of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, UCSF, San Francisco, California, USA
| | | | - Michael C. Peters
- Department of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Karl H. Nocka
- Pfizer Inc., Inflammation and Immunology, Cambridge, Massachusetts, USA
| | - John V. Fahy
- Department of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Max A. Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-Denver, Denver, Colorado, USA
| |
Collapse
|
278
|
Stier MT, Bloodworth MH, Toki S, Newcomb DC, Goleniewska K, Boyd KL, Quitalig M, Hotard AL, Moore ML, Hartert TV, Zhou B, McKenzie AN, Peebles RS. Respiratory syncytial virus infection activates IL-13-producing group 2 innate lymphoid cells through thymic stromal lymphopoietin. J Allergy Clin Immunol 2016; 138:814-824.e11. [PMID: 27156176 PMCID: PMC5014571 DOI: 10.1016/j.jaci.2016.01.050] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 01/12/2016] [Accepted: 01/29/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a major health care burden with a particularly high worldwide morbidity and mortality rate among infants. Data suggest that severe RSV-associated illness is in part caused by immunopathology associated with a robust type 2 response. OBJECTIVE We sought to determine the capacity of RSV infection to stimulate group 2 innate lymphoid cells (ILC2s) and the associated mechanism in a murine model. METHODS Wild-type (WT) BALB/c, thymic stromal lymphopoietin receptor (TSLPR) knockout (KO), or WT mice receiving an anti-TSLP neutralizing antibody were infected with the RSV strain 01/2-20. During the first 4 to 6 days of infection, lungs were collected for evaluation of viral load, protein concentration, airway mucus, airway reactivity, or ILC2 numbers. Results were confirmed with 2 additional RSV clinical isolates, 12/11-19 and 12/12-6, with known human pathogenic potential. RESULTS RSV induced a 3-fold increase in the number of IL-13-producing ILC2s at day 4 after infection, with a concurrent increase in total lung IL-13 levels. Both thymic stromal lymphopoietin (TSLP) and IL-33 levels were increased 12 hours after infection. TSLPR KO mice did not mount an IL-13-producing ILC2 response to RSV infection. Additionally, neutralization of TSLP significantly attenuated the RSV-induced IL-13-producing ILC2 response. TSLPR KO mice displayed reduced lung IL-13 protein levels, decreased airway mucus and reactivity, attenuated weight loss, and similar viral loads as WT mice. Both 12/11-19 and 12/12-6 similarly induced IL-13-producing ILC2s through a TSLP-dependent mechanism. CONCLUSION These data demonstrate that multiple pathogenic strains of RSV induce IL-13-producing ILC2 proliferation and activation through a TSLP-dependent mechanism in a murine model and suggest the potential therapeutic targeting of TSLP during severe RSV infection.
Collapse
Affiliation(s)
- Matthew T Stier
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Melissa H Bloodworth
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Shinji Toki
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Dawn C Newcomb
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn; Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kelli L Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Marc Quitalig
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Anne L Hotard
- Division of Infectious Disease, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, Ga
| | - Martin L Moore
- Division of Infectious Disease, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, Ga
| | - Tina V Hartert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Baohua Zhou
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind
| | - Andrew N McKenzie
- MRC Laboratory of Molecular Biology, Cambridge University, Cambridge, United Kingdom
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn; Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn.
| |
Collapse
|
279
|
Fat-associated lymphoid clusters control local IgM secretion during pleural infection and lung inflammation. Nat Commun 2016; 7:12651. [PMID: 27582256 PMCID: PMC5025788 DOI: 10.1038/ncomms12651] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/18/2016] [Indexed: 12/25/2022] Open
Abstract
Fat-associated lymphoid clusters (FALC) are inducible structures that support rapid innate-like B-cell immune responses in the serous cavities. Little is known about the physiological cues that activate FALCs in the pleural cavity and more generally the mechanisms controlling B-cell activation in FALCs. Here we show, using separate models of pleural nematode infection with Litomosoides sigmodontis and Altenaria alternata induced acute lung inflammation, that inflammation of the pleural cavity rapidly activates mediastinal and pericardial FALCs. IL-33 produced by FALC stroma is crucial for pleural B1-cell activation and local IgM secretion. However, B1 cells are not the direct target of IL-33, which instead requires IL-5 for activation. Moreover, lung inflammation leads to increased IL-5 production by type 2 cytokine-producing innate lymphoid cells (ILC2) in the FALC. These findings reveal a link between inflammation, IL-33 release by FALC stromal cells, ILC2 activation and pleural B-cell activation in FALCs, resulting in local and antigen-specific IgM production. Fat-associated lymphoid clusters (FALC) in the serous cavities house rapid IgM-producing B1 cells, but how the clusters are activated to respond to infection is unclear. Here the authors show that in response to lung inflammation or pleural nematode infection adipose stromal cell-derived IL-33 activates ILC2s to produce IL-5, thus driving the B1 response in the FALCs.
Collapse
|
280
|
Almeida FF, Belz GT. Innate lymphoid cells: models of plasticity for immune homeostasis and rapid responsiveness in protection. Mucosal Immunol 2016; 9:1103-12. [PMID: 27484190 DOI: 10.1038/mi.2016.64] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/16/2016] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILCs) have stormed onto the immune landscape as "newly discovered" cell types. These tissue-resident sentinels are enriched at mucosal surfaces and engage in complex cross talk with elements of the adaptive immune system and microenvironment to orchestrate immune homeostasis. Many parallels exist between innate cells and T cells leading to the initial partitioning of ILCs into rather rigid subsets that reflect their "adaptive-like" effector cytokines profiles. ILCs themselves, however, have unique attributes that are only just beginning to be elucidated. These features result in complementarity with, rather than complete duplication of, functions of the adaptive immune system. Key transcription factors determine the pathway of differentiation of progenitors towards an ILC1, ILC2, or ILC3 subset. Once formed, flexibility in the responses of these subsets to stimuli unexpectedly allows transdifferentation between the different subsets and the acquisition of altered phenotypes and function. This provides a mechanism for rapid innate immune responsiveness. Here, we discuss the models of differentiation for maintenance and activation of tissue-resident ILCs in maintaining immune homeostasis and protection.
Collapse
Affiliation(s)
- F F Almeida
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - G T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
281
|
Abstract
Innate lymphoid cells (ILCs), a newly identified member of the lymphoid population, play a critical role in the transition from innate to adaptive immunity in host defense. ILCs are important in mucosal barrier immunity, tissue homeostasis, and immune regulation throughout the body. Significant alterations in ILC responses in lung diseases have been observed and reported. Emerging evidence has shown that ILCs are importantly involved in the pathogenesis and development of a variety of lung diseases, i.e., helminth infections, allergic airway inflammation, and airway hyper-responsiveness. However, as a tissue-resident cell population, the role of ILCs in the lung remains poorly characterized. In this review, we discuss the role of ILCs in lung diseases, the mechanisms underlying the ILC-mediated regulation of immunity, and the therapeutic potential of modulating ILC responses.
Collapse
Affiliation(s)
- Deng-Ming Lai
- Department of Cardiovascular Surgery, the Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052 China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Qiang Shu
- Department of Cardiovascular Surgery, the Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052 China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240 USA
| |
Collapse
|
282
|
Percopo CM, Brenner TA, Ma M, Kraemer LS, Hakeem RMA, Lee JJ, Rosenberg HF. SiglecF+Gr1hi eosinophils are a distinct subpopulation within the lungs of allergen-challenged mice. J Leukoc Biol 2016; 101:321-328. [PMID: 27531929 DOI: 10.1189/jlb.3a0416-166r] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 01/06/2023] Open
Abstract
Although eosinophils as a group are readily identified by their unique morphology and staining properties, flow cytometry provides an important means for identification of subgroups based on differential expression of distinct surface Ags. Here, we characterize an eosinophil subpopulation defined by high levels of expression of the neutrophil Ag Gr1 (CD45+CD11c-SiglecF+Gr1hi). SiglecF+Gr1hi eosinophils, distinct from the canonical SiglecF+Gr1- eosinophil population, were detected in allergen-challenged wild-type and granule protein-deficient (EPX-/- and MBP-1-/-) mice, but not in the eosinophil-deficient ΔdblGATA strain. In contrast to Gr1+ neutrophils, which express both cross-reacting Ags Ly6C and Ly6G, SiglecF+Gr1hi eosinophils from allergen-challenged lung tissue are uniquely Ly6G+ Although indistinguishable from the more-numerous SiglecF+Gr1- eosinophils under light microscopy, FACS-isolated populations revealed prominent differences in cytokine contents. The lymphocyte-targeting cytokines CXCL13 and IL-27 were identified only in the SiglecF+Gr1hi eosinophil population (at 3.9 and 4.8 pg/106 cells, respectively), as was the prominent proinflammatory mediator IL-13 (72 pg/106 cells). Interestingly, bone marrow-derived (SiglecF+), cultured eosinophils include a more substantial Gr1+ subpopulation (∼50%); Gr1+ bmEos includes primarily a single Ly6C+ and a smaller, double-positive (Ly6C+Ly6G+) population. Taken together, our findings characterize a distinct SiglecF+Gr1hi eosinophil subset in lungs of allergen-challenged, wild-type and granule protein-deficient mice. SiglecF+Gr1hi eosinophils from wild-type mice maintain a distinct subset of cytokines, including those active on B and T lymphocytes. These cytokines may facilitate eosinophil-mediated immunomodulatory responses in the allergen-challenged lung as well as in other distinct microenvironments.
Collapse
Affiliation(s)
- Caroline M Percopo
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Todd A Brenner
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michelle Ma
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura S Kraemer
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Reem M A Hakeem
- Molecular Signal Transduction Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA; and
| | | | - Helene F Rosenberg
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
283
|
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: 1.7] [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
|
284
|
Alternative splicing of interleukin-33 and type 2 inflammation in asthma. Proc Natl Acad Sci U S A 2016; 113:8765-70. [PMID: 27432971 DOI: 10.1073/pnas.1601914113] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Type 2 inflammation occurs in a large subgroup of asthmatics, and novel cytokine-directed therapies are being developed to treat this population. In mouse models, interleukin-33 (IL-33) activates lung resident innate lymphoid type 2 cells (ILC2s) to initiate airway type 2 inflammation. In human asthma, which is chronic and difficult to model, the role of IL-33 and the target cells responsible for persistent type 2 inflammation remain undefined. Full-length IL-33 is a nuclear protein and may function as an "alarmin" during cell death, a process that is uncommon in chronic stable asthma. We demonstrate a previously unidentified mechanism of IL-33 activity that involves alternative transcript splicing, which may operate in stable asthma. In human airway epithelial cells, alternative splicing of the IL-33 transcript is consistently present, and the deletion of exons 3 and 4 (Δ exon 3,4) confers cytoplasmic localization and facilitates extracellular secretion, while retaining signaling capacity. In nonexacerbating asthmatics, the expression of Δ exon 3,4 is strongly associated with airway type 2 inflammation, whereas full-length IL-33 is not. To further define the extracellular role of IL-33 in stable asthma, we sought to determine the cellular targets of its activity. Comprehensive flow cytometry and RNA sequencing of sputum cells suggest basophils and mast cells, not ILC2s, are the cellular sources of type 2 cytokines in chronic asthma. We conclude that IL-33 isoforms activate basophils and mast cells to drive type 2 inflammation in chronic stable asthma, and novel IL-33 inhibitors will need to block all biologically active isoforms.
Collapse
|
285
|
Drake LY, Iijima K, Bartemes K, Kita H. Group 2 Innate Lymphoid Cells Promote an Early Antibody Response to a Respiratory Antigen in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 197:1335-42. [PMID: 27421480 DOI: 10.4049/jimmunol.1502669] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/13/2016] [Indexed: 01/31/2023]
Abstract
Innate lymphoid cells (ILCs) are a new family of immune cells that play important roles in innate immunity in mucosal tissues, and in the maintenance of tissue and metabolic homeostasis. Recently, group 2 ILCs (ILC2s) were found to promote the development and effector functions of Th2-type CD4(+) T cells by interacting directly with T cells or by activating dendritic cells, suggesting a role for ILC2s in regulating adaptive immunity. However, our current knowledge on the role of ILCs in humoral immunity is limited. In this study, we found that ILC2s isolated from the lungs of naive BALB/c mice enhanced the proliferation of B1- as well as B2-type B cells and promoted the production of IgM, IgG1, IgA, and IgE by these cells in vitro. Soluble factors secreted by ILC2s were sufficient to enhance B cell Ig production. By using blocking Abs and ILC2s isolated from IL-5-deficient mice, we found that ILC2-derived IL-5 is critically involved in the enhanced production of IgM. Furthermore, when adoptively transferred to Il7r(-/-) mice, which lack ILC2s and mature T cells, lung ILC2s promoted the production of IgM Abs to a polysaccharide Ag, 4-hydroxy-3-nitrophenylacetyl Ficoll, within 7 d of airway exposure in vivo. These findings add to the growing body of literature regarding the regulatory functions of ILCs in adaptive immunity, and suggest that lung ILC2s promote B cell production of early Abs to a respiratory Ag even in the absence of T cells.
Collapse
Affiliation(s)
- Li Yin Drake
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Koji Iijima
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Kathleen Bartemes
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905; and Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
286
|
Lipid mediators as regulators of human ILC2 function in allergic diseases. Immunol Lett 2016; 179:36-42. [PMID: 27396531 DOI: 10.1016/j.imlet.2016.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/06/2016] [Accepted: 07/06/2016] [Indexed: 12/29/2022]
Abstract
Group 2 innate lymphoid cells (ILC2) are specialized in type 2 immunity. ILC2 are activated early in immune responses and, despite their low abundance, are able to initiate and amplify allergic inflammation by orchestrating other type 2 immune cells. Based on recent discoveries, the spectrum of ILC2 regulating factors has been extended. It is now well established that not only epithelial cell-derived innate cytokines, but also bioactive lipids can regulate ILC2 activity and accumulation. Additionally, ILC2 appear to be susceptible to changes in the cytokine milieu and can acquire an ILC1-like phenotype due to a high degree of cellular plasticity. As ILC2 are fundamentally involved in the pathogenesis of type 2 diseases, they represent a promising therapeutic target for allergic airway and skin diseases. In this review we summarize the current knowledge about ILC2 biology in the allergy context, with a particular focus on the emerging role of lipid mediators in regulating ILC2 function.
Collapse
|
287
|
The IL-33 receptor (ST2) regulates early IL-13 production in fungus-induced allergic airway inflammation. Mucosal Immunol 2016; 9:937-49. [PMID: 26555705 DOI: 10.1038/mi.2015.106] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/22/2015] [Indexed: 02/04/2023]
Abstract
Allergic airway inflammation (AAI) in response to environmental antigens is an increasing medical problem, especially in the Western world. Type 2 interleukins (IL) are central in the pathological response but their importance and cellular source(s) often rely on the particular allergen. Here, we highlight the cellular sources and regulation of the prototypic type 2 cytokine, IL-13, during the establishment of AAI in a fungal infection model using Cryptococcus neoformans. IL-13 reporter mice revealed a rapid onset of IL-13 competence within innate lymphoid cells type 2 (ILC2) and IL-33R(+) T helper (Th) cells. ILC2 showed IL-33-dependent proliferation upon infection and significant IL-13 production. Th cells essentially required IL-33 to become either GATA3(+) or GATA3(+)/Foxp3(+) hybrids. GATA3(+) Th cells almost exclusively contributed to IL-13 production but hybrid GATA3(+)/Foxp3(+) Th cells did not. In addition, alveolar macrophages upregulated the IL-33R and subsequently acquired a phenotype of alternative activation (Ym1(+), FIZZ1(+), and arginase-1(+)) linked to type 2 immunity. Absence of adaptive immunity in rag2(-/-) mice resulted in attenuated AAI, revealing the need for Th2 cells for full AAI development. Taken together, in pulmonary cryptococcosis ILC2 and GATA3(+) Th2 cells produce early IL-13 largely IL-33R-dependent, thereby promoting goblet cell metaplasia, pulmonary eosinophilia, and alternative activation of alveolar macrophages.
Collapse
|
288
|
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: 40] [Impact Index Per Article: 4.4] [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
|
289
|
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: 5.4] [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
|
290
|
Zhou W, Toki S, Zhang J, Goleniewksa K, Newcomb DC, Cephus JY, Dulek DE, Bloodworth MH, Stier MT, Polosuhkin V, Gangula RD, Mallal SA, Broide DH, Peebles RS. Prostaglandin I2 Signaling and Inhibition of Group 2 Innate Lymphoid Cell Responses. Am J Respir Crit Care Med 2016; 193:31-42. [PMID: 26378386 DOI: 10.1164/rccm.201410-1793oc] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RATIONALE Group 2 innate lymphoid cells (ILC2s) robustly produce IL-5 and IL-13, cytokines central to the asthma phenotype; however, the effect of prostaglandin (PG) I2 on ILC2 function is unknown. OBJECTIVES To determine the effect of PGI2 on mouse and human ILC2 cytokine expression in vitro and the effect of endogenous PGI2 and the PGI2 analog cicaprost on lung ILC2s in vivo. METHODS Flow-sorted bone marrow ILC2s of wild-type (WT) and PGI2 receptor-deficient (IP(-/-)) mice were cultured with IL-33 and treated with the PGI2 analog cicaprost. WT and IP(-/-) mice were challenged intranasally with Alternaria alternata extract for 4 consecutive days to induce ILC2 responses, and these were quantified. Prior to A. alternata extract, challenged WT mice were treated with cicaprost. Human flow-sorted peripheral blood ILC2s were cultured with IL-33 and IL-2 and treated with the PGI2 analog cicaprost. MEASUREMENT AND MAIN RESULTS We demonstrate that PGI2 inhibits IL-5 and IL-13 protein expression by IL-33-stimulated ILC2s purified from mouse bone marrow in a manner that was dependent on signaling through the PGI2 receptor IP. In a mouse model of 4 consecutive days of airway challenge with an extract of A. alternata, a fungal aeroallergen associated with severe asthma exacerbations, endogenous PGI2 signaling significantly inhibited lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s, as well as the mean fluorescence intensity of IL-5 and IL-13 staining. In addition, exogenous administration of a PGI2 analog inhibited Alternaria extract-induced lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s and the mean fluorescence intensity of IL-5 and IL-13 staining. Finally, a PGI2 analog inhibited IL-5 and IL-13 expression by human ILC2s that were stimulated with IL-2 and IL-33. CONCLUSIONS These results suggest that PGI2 may be a potential therapy to reduce the ILC2 response to protease-containing aeroallergens, such as Alternaria.
Collapse
Affiliation(s)
- Weisong Zhou
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Shinji Toki
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Jian Zhang
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kasia Goleniewksa
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Dawn C Newcomb
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Jacqueline Y Cephus
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Daniel E Dulek
- 2 Division of Infectious Diseases, Department of Pediatrics, and
| | - Melissa H Bloodworth
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Matthew T Stier
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Vasiliy Polosuhkin
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Rama D Gangula
- 3 Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Simon A Mallal
- 3 Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - David H Broide
- 4 Department of Medicine, University of California San Diego, La Jolla, California
| | - R Stokes Peebles
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
291
|
Zuo L, Pannell BK, Liu Z. Characterization and redox mechanism of asthma in the elderly. Oncotarget 2016; 7:25010-21. [PMID: 26843624 PMCID: PMC5041886 DOI: 10.18632/oncotarget.7075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 01/17/2016] [Indexed: 12/15/2022] Open
Abstract
Asthma is a chronic disease characterized by reversible airflow limitation, coughing, bronchial constriction, and an inflammatory immune response. While asthma has frequently been categorized as emerging in childhood, evidence has begun to reveal that the elderly population is certainly susceptible to late-onset, or even long-standing asthma. Non-atopic asthma, most commonly found in elderly patients is associated with elevated levels of serum and sputum neutrophils and may be more detrimental than atopic asthma. The mortality of asthma is high in the elderly since these patients often possess more severe symptoms than younger populations. The redox mechanisms that mediate inflammatory reactions during asthma have not been thoroughly interpreted in the context of aging. Thus, we review the asthmatic symptoms related to reactive oxygen species (ROS) and reactive nitrogen species (RNS) in seniors. Moreover, immune status in the elderly is weakened in part by immunosenescence, which is broadly defined as the decline in functionality of the immune system that corresponds with increasing age. The effects of immunosenescence on the expression of biomarkers potentially utilized in the clinical diagnosis of asthma remain unclear. It has also been shown that existing asthma treatments are less effective in the elderly. Thus, it is necessary that clinicians approach the diagnosis and treatment of asthmatic senior patients using innovative methods. Asthma in the elderly demands more intentional diagnostic and therapeutic research since it is potentially one of the few causes of mortality and morbidity in the elderly that is largely reversible.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH, USA
- The Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Benjamin K. Pannell
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Zewen Liu
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Anesthesiology, Affiliated Ezhou Central Hospital, Renmin Hospital of Wuhan University Medical School, Hubei, China
| |
Collapse
|
292
|
Nayak AP, Green BJ, Lemons AR, Marshall NB, Goldsmith WT, Kashon ML, Anderson SE, Germolec DR, Beezhold DH. Subchronic exposures to fungal bioaerosols promotes allergic pulmonary inflammation in naïve mice. Clin Exp Allergy 2016; 46:861-70. [PMID: 26892490 DOI: 10.1111/cea.12724] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/05/2016] [Accepted: 02/07/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Epidemiological surveys indicate that occupants of mold contaminated environments are at increased risk of respiratory symptoms. The immunological mechanisms associated with these responses require further characterization. OBJECTIVE The aim of this study was to characterize the immunotoxicological outcomes following repeated inhalation of dry Aspergillus fumigatus spores aerosolized at concentrations potentially encountered in contaminated indoor environments. METHODS Aspergillus fumigatus spores were delivered to the lungs of naïve BALB/cJ mice housed in a multi-animal nose-only chamber twice a week for a period of 13 weeks. Mice were evaluated at 24 and 48 h post-exposure for histopathological changes in lung architecture, recruitment of specific immune cells to the airways, and serum antibody responses. RESULT Germinating A. fumigatus spores were observed in lungs along with persistent fungal debris in the perivascular regions of the lungs. Repeated exposures promoted pleocellular infiltration with concomitant epithelial mucus hypersecretion, goblet cell metaplasia, subepithelial fibrosis and enhanced airway hyperreactivity. Cellular infiltration in airways was predominated by CD4(+) T cells expressing the pro-allergic cytokine IL-13. Furthermore, our studies show that antifungal T cell responses (IFN-γ(+) or IL-17A(+) ) co-expressed IL-13, revealing a novel mechanism for the dysregulated immune response to inhaled fungi. Total IgE production was augmented in animals repeatedly exposed to A. fumigatus. CONCLUSIONS & CLINICAL RELEVANCE Repeated inhalation of fungal aerosols resulted in significant pulmonary pathology mediated by dynamic shifts in specific immune populations and their cytokines. These studies provide novel insights into the immunological mechanisms and targets that govern the health outcomes that result from repeated inhalation of fungal bioaerosols in contaminated environments.
Collapse
Affiliation(s)
- A P Nayak
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - B J Green
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - A R Lemons
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - N B Marshall
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - W T Goldsmith
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - M L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - S E Anderson
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - D R Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - D H Beezhold
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| |
Collapse
|
293
|
Hamilos DL. Drivers of chronic rhinosinusitis: Inflammation versus infection. J Allergy Clin Immunol 2016; 136:1454-1459. [PMID: 26654194 DOI: 10.1016/j.jaci.2015.10.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/21/2015] [Indexed: 12/15/2022]
Abstract
Studies of the underlying cause or causes of chronic rhinosinusitis (CRS) over the past 20 or more years have expanded from a focus on systemic immune and allergic mechanisms to an intense search for the underlying drivers of mucosal inflammation. These drivers involve mucosal inflammatory pathways that become activated by allergens, microbial stimuli, or poorly understood exogenous or endogenous stimuli. The holy grail in the study of CRS is to identify specific drivers of mucosal inflammation and translate these into more effective treatment for CRS. Certain deficiencies in local innate immunity have been described in patients with CRS that predispose to increased sinus mucosal bacterial colonization/infection, including deficient local production of antimicrobial lactoferrin and deficient functioning of the bitter taste receptor TAS2R38. Conversely, certain innate factors, namely IL-25, IL-33, and thymic stromal lymphopoietin (TSLP), are elaborated by sinus epithelial cells in response to microbial stimulation or airway injury and promote local TH2 inflammation. The precise physiologic role of these factors in innate or adaptive immunity is unclear, although IL-33 might function as an alarmin triggered by damage-associated molecular patterns. The cytokines IL-25 and TSLP, similarly promote proinflammatory tissue responses. Another feature of epithelial dysregulation in patients with CRS is overproduction of eosinophil-promoting C-C chemokines by sinus epithelium, perhaps driven in part through innate stimuli, as well as TH2 cytokines, such as IL-13. Strategies to reduce the microbial stimulation of maladaptive TH2 inflammation or to suppress the local elaboration of TH2-promoting epithelial factors, such as IL-33, have potential therapeutic benefit in patients with CRS, although the extent to which this is realized in patient care remains limited at present. This rostrum will summarize my views on the major microbial drivers of mucosal inflammation and dysregulation of innate TH2-promoting factors in patients with CRS based on recent experimental data.
Collapse
Affiliation(s)
- Daniel L Hamilos
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, Mass.
| |
Collapse
|
294
|
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
|
295
|
Tada H, Matsuyama T, Nishioka T, Hagiwara M, Kiyoura Y, Shimauchi H, Matsushita K. Porphyromonas gingivalis Gingipain-Dependently Enhances IL-33 Production in Human Gingival Epithelial Cells. PLoS One 2016; 11:e0152794. [PMID: 27058037 PMCID: PMC4825981 DOI: 10.1371/journal.pone.0152794] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/19/2016] [Indexed: 01/21/2023] Open
Abstract
The cytokine IL-33 is constitutively expressed in epithelial cells and it augments Th2 cytokine-mediated inflammatory responses by regulating innate immune cells. We aimed to determine the role of the periodontal pathogen, Porphyromonas gingivalis, in the enhanced expression of IL-33 in human gingival epithelial cells. We detected IL-33 in inflamed gingival epithelium from patients with chronic periodontitis, and found that P. gingivalis increased IL-33 expression in the cytoplasm of human gingival epithelial cells in vitro. In contrast, lipopolysaccharide, lipopeptide, and fimbriae derived from P. gingivalis did not increase IL-33 expression. Specific inhibitors of P. gingivalis proteases (gingipains) suppressed IL-33 mRNA induction by P. gingivalis and the P. gingivalis gingipain-null mutant KDP136 did not induce IL-33 expression. A small interfering RNA for protease-activated receptor-2 (PAR-2) as well as inhibitors of phospholipase C, p38 and NF-κB inhibited the expression of IL-33 induced by P. gingivalis. These results indicate that the PAR-2/IL-33 axis is promoted by P. gingivalis infection in human gingival epithelial cells through a gingipain-dependent mechanism.
Collapse
Affiliation(s)
- Hiroyuki Tada
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Division of Oral Microbiology, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
- * E-mail:
| | - Takashi Matsuyama
- Department of Periodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Sakuragaoka, Kagoshima, Japan
| | - Takashi Nishioka
- Division of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Makoto Hagiwara
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, Koriyama, Fukushima, Japan
| | - Hidetoshi Shimauchi
- Division of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
296
|
Innate lymphoid cells contribute to allergic airway disease exacerbation by obesity. J Allergy Clin Immunol 2016; 138:1309-1318.e11. [PMID: 27177781 DOI: 10.1016/j.jaci.2016.03.019] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 03/06/2016] [Accepted: 03/21/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Epidemiologic and clinical observations identify obesity as an important risk factor for asthma exacerbation, but the underlying mechanisms remain poorly understood. Type 2 innate lymphoid cells (ILC2s) and type 3 innate lymphoid cells (ILC3s) have been implicated, respectively, in asthma and adipose tissue homeostasis and in obesity-associated airway hyperresponsiveness (AHR). OBJECTIVE We sought to determine the potential involvement of innate lymphoid cells (ILCs) in allergic airway disease exacerbation caused by high-fat diet (HFD)-induced obesity. METHODS Obesity was induced by means of HFD feeding, and allergic airway inflammation was subsequently induced by means of intranasal administration of house dust mite (HDM) extract. AHR, lung and visceral adipose tissue inflammation, humoral response, cytokines, and innate and adaptive lymphoid populations were analyzed in the presence or absence of ILCs. RESULTS HFD feeding exacerbated allergic airway disease features, including humoral response, airway and tissue eosinophilia, AHR, and TH2 and TH17 pulmonary profiles. Notably, nonsensitized obese mice already exhibited increased lung ILC counts and tissue eosinophil infiltration compared with values in lean mice in the absence of AHR. The numbers of total and cytokine-expressing lung ILC2s and ILC3s further increased in HDM-challenged obese mice compared with those in HDM-challenged lean mice, and this was accompanied by high IL-33 and IL-1β levels and decreased ILC markers in visceral adipose tissue. Furthermore, depletion of ILCs with an anti-CD90 antibody, followed by T-cell reconstitution, led to a profound decrease in allergic airway inflammatory features in obese mice, including TH2 and TH17 infiltration. CONCLUSION These results indicate that HFD-induced obesity might exacerbate allergic airway inflammation through mechanisms involving ILC2s and ILC3s.
Collapse
|
297
|
Anti-Inflammatory Effect of Quercetin on RAW 264.7 Mouse Macrophages Induced with Polyinosinic-Polycytidylic Acid. Molecules 2016; 21:450. [PMID: 27049378 PMCID: PMC6273652 DOI: 10.3390/molecules21040450] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/22/2016] [Accepted: 03/29/2016] [Indexed: 12/20/2022] Open
Abstract
Quercetin (3,3′,4′,5,6-pentahydroxyflavone) is a well-known antioxidant and a flavonol found in many fruits, leaves, and vegetables. Quercetin also has known anti-inflammatory effects on lipopolysaccharide-induced macrophages. However, the effects of quercetin on virus-induced macrophages have not been fully reported. In this study, the anti-inflammatory effect of quercetin on double-stranded RNA (dsRNA)-induced macrophages was examined. Quercetin at concentrations up to 50 μM significantly inhibited the production of NO, IL-6, MCP-1, IP-10, RANTES, GM-CSF, G-CSF, TNF-α, LIF, LIX, and VEGF as well as calcium release in dsRNA (50 µg/mL of polyinosinic-polycytidylic acid)-induced RAW 264.7 mouse macrophages (p < 0.05). Quercetin at concentrations up to 50 μM also significantly inhibited mRNA expression of signal transducer and activated transcription 1 (STAT1) and STAT3 in dsRNA-induced RAW 264.7 cells (p < 0.05). In conclusion, quercetin had alleviating effects on viral inflammation based on inhibition of NO, cytokines, chemokines, and growth factors in dsRNA-induced macrophages via the calcium-STAT pathway.
Collapse
|
298
|
Shan S, Li Y, Wang J, Lv Z, Yi D, Huang Q, Corrigan CJ, Wang W, Quangeng Z, Ying S. Nasal administration of interleukin-33 induces airways angiogenesis and expression of multiple angiogenic factors in a murine asthma surrogate. Immunology 2016; 148:83-91. [PMID: 27035894 DOI: 10.1111/imm.12589] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/14/2016] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Abstract
The T-helper cell type 2-promoting cytokine interleukin-33 (IL-33) has been implicated in asthma pathogenesis. Angiogenesis is a feature of airways remodelling in asthma. We hypothesized that IL-33 induces airways angiogenesis and expression of angiogenic factors in an established murine surrogate of asthma. In the present study, BALB/c mice were subjected to serial intranasal challenge with IL-33 alone for up to 70 days. In parallel, ovalbumin (OVA) -sensitized mice were subjected to serial intranasal challenge with OVA or normal saline to serve as positive and negative controls, respectively. Immunohistochemical analysis of expression of von Willebrand factor and erythroblast transformation-specific-related gene, both blood vessel markers, and angiogenic factors angiogenin, insulin-like growth factor-1, endothelin-1, epidermal growth factor and amphiregulin was performed in lung sections ex vivo. An established in-house assay was used to test whether IL-33 was able to induce microvessel formation by human vascular endothelial cells. Results showed that serial intranasal challenge of mice with IL-33 or OVA resulted in proliferation of peribronchial von Willebrand factor-positive blood vessels to a degree closely related to the total expression of the angiogenic factors amphiregulin, angiogenin, endothelin-1, epidermal growth factor and insulin-like growth factor-1. IL-33 also induced microvessel formation by human endothelial cells in a concentration-dependent fashion in vitro. Our data are consistent with the hypothesis that IL-33 has the capacity to induce angiogenesis at least partly by increasing local expression of multiple angiogenic factors in an allergen-independent murine asthma surrogate, and consequently that IL-33 or its receptor is a potential novel molecular target for asthma therapy.
Collapse
Affiliation(s)
- Shan Shan
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Li
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Wang
- The Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Zhe Lv
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dawei Yi
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qiong Huang
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Division of Asthma, Allergy & Lung Biology, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Wei Wang
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhang Quangeng
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sun Ying
- The Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Division of Asthma, Allergy & Lung Biology, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| |
Collapse
|
299
|
Shelite TR, Liang Y, Wang H, Mendell NL, Trent BJ, Sun J, Gong B, Xu G, Hu H, Bouyer DH, Soong L. IL-33-Dependent Endothelial Activation Contributes to Apoptosis and Renal Injury in Orientia tsutsugamushi-Infected Mice. PLoS Negl Trop Dis 2016; 10:e0004467. [PMID: 26943125 PMCID: PMC4778942 DOI: 10.1371/journal.pntd.0004467] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/27/2016] [Indexed: 01/23/2023] Open
Abstract
Endothelial cells (EC) are the main target for Orientia tsutsugamushi infection and EC dysfunction is a hallmark of severe scrub typhus in patients. However, the molecular basis of EC dysfunction and its impact on infection outcome are poorly understood. We found that C57BL/6 mice that received a lethal dose of O. tsutsugamushi Karp strain had a significant increase in the expression of IL-33 and its receptor ST2L in the kidneys and liver, but a rapid reduction of IL-33 in the lungs. We also found exacerbated EC stress and activation in the kidneys of infected mice, as evidenced by elevated angiopoietin (Ang) 2/Ang1 ratio, increased endothelin 1 (ET-1) and endothelial nitric oxide synthase (eNOS) expression. Such responses were significantly attenuated in the IL-33-/- mice. Importantly, IL-33-/- mice also had markedly attenuated disease due to reduced EC stress and cellular apoptosis. To confirm the biological role of IL-33, we challenged wild-type (WT) mice with a sub-lethal dose of O. tsutsugamushi and gave mice recombinant IL-33 (rIL-33) every 2 days for 10 days. Exogenous IL-33 significantly increased disease severity and lethality, which correlated with increased EC stress and activation, increased CXCL1 and CXCL2 chemokines, but decreased anti-apoptotic gene BCL-2 in the kidneys. To further examine the role of EC stress, we infected human umbilical vein endothelial cells (HUVEC) in vitro. We found an infection dose-dependent increase in the expression of IL-33, ST2L soluble ST2 (sST2), and the Ang2/Ang1 ratio at 24 and 48 hours post-infection. This study indicates a pathogenic role of alarmin IL-33 in a murine model of scrub typhus and highlights infection-triggered EC damage and IL-33-mediated pathological changes during the course of Orientia infection.
Collapse
Affiliation(s)
- Thomas R Shelite
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hui Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nicole L Mendell
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Brandon J Trent
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Guang Xu
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H Bouyer
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America.,Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
300
|
Yang Q, Ge MQ, Kokalari B, Redai IG, Wang X, Kemeny DM, Bhandoola A, Haczku A. Group 2 innate lymphoid cells mediate ozone-induced airway inflammation and hyperresponsiveness in mice. J Allergy Clin Immunol 2016; 137:571-8. [PMID: 26282284 PMCID: PMC4747855 DOI: 10.1016/j.jaci.2015.06.037] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 06/09/2015] [Accepted: 06/29/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Asthmatic patients are highly susceptible to air pollution and in particular to the effects of ozone (O3) inhalation, but the underlying mechanisms remain unclear. OBJECTIVE Using mouse models of O3-induced airway inflammation and airway hyperresponsiveness (AHR), we sought to investigate the role of the recently discovered group 2 innate lymphoid cells (ILC2s). METHODS C57BL/6 and BALB/c mice were exposed to Aspergillus fumigatus, O3, or both (3 ppm for 2 hours). ILC2s were isolated by means of fluorescence-activated cell sorting and studied for Il5 and Il13 mRNA expression. ILC2s were depleted with anti-Thy1.2 mAb and replaced by means of intratracheal transfer of ex vivo expanded Thy1.1 ILC2s. Cytokine levels (ELISA and quantitative PCR), inflammatory cell profile, and AHR (flexiVent) were assessed in the mice. RESULTS In addition to neutrophil influx, O3 inhalation elicited the appearance of eosinophils and IL-5 in the airways of BALB/c but not C57BL/6 mice. Although O3-induced expression of IL-33, a known activator of ILC2s, in the lung was similar between these strains, isolated pulmonary ILC2s from O3-exposed BALB/c mice had significantly greater Il5 and Il13 mRNA expression than C57BL/6 mice. This suggested that an altered ILC2 function in BALB/c mice might mediate the increased O3 responsiveness. Indeed, anti-Thy1.2 treatment abolished but ILC2s added back dramatically enhanced O3-induced AHR. CONCLUSIONS O3-induced activation of pulmonary ILC2s was necessary and sufficient to mediate asthma-like changes in BALB/c mice. This previously unrecognized role of ILC2s might help explain the heightened susceptibility of human asthmatic airways to O3 exposure.
Collapse
Affiliation(s)
- Qi Yang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pa; T-Cell Biology and Development Section, Laboratory of Genome Integrity, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Moyar Q Ge
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pa; Translational Lung Biology Center, Pulmonary, Critical Care & Sleep Medicine, University of California, Davis, Calif; Department of Microbiology, National University of Singapore, Singapore
| | - Blerina Kokalari
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Imre G Redai
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Xinxin Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pa
| | - David M Kemeny
- Department of Microbiology, National University of Singapore, Singapore
| | - Avinash Bhandoola
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pa; T-Cell Biology and Development Section, Laboratory of Genome Integrity, National Cancer Institute, National Institutes of Health, Bethesda, Md.
| | - Angela Haczku
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pa; Translational Lung Biology Center, Pulmonary, Critical Care & Sleep Medicine, University of California, Davis, Calif.
| |
Collapse
|