1
|
Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a New Axis in the Genesis of Respiratory Diseases: Possible Therapeutic Implications. Int J Mol Sci 2023; 24:1783. [PMID: 36675299 PMCID: PMC9861898 DOI: 10.3390/ijms24021783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
It is well ascertained that airway inflammation has a key role in the genesis of numerous respiratory pathologies, including asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Pulmonary tissue inflammation and anti-inflammatory responses implicate an intricate relationship between local and infiltrating immune cells and structural pulmonary cells. Alarmins are endogenic proteins discharged after cell injury in the extracellular microenvironment. The purpose of our review is to highlight the alterations in respiratory diseases involving some alarmins, such as high mobility group box 1 (HMGB1) and interleukin (IL)-33, and their inter-relationships and relationships with genetic non-coding material, such as microRNAs. The role played by these alarmins in some pathophysiological processes confirms the existence of an axis composed of HMGB1 and IL-33. These alarmins have been implicated in ferroptosis, the onset of type 2 inflammation and airway alterations. Moreover, both factors can act on non-coding genetic material capable of modifying respiratory function. Finally, we present an outline of alarmins and RNA-based therapeutics that have been proposed to treat respiratory pathologies.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
2
|
Wang CJ, Cheng SL, Kuo SH. Asthma and COVID-19 Associations: Focus on IgE-Related Immune Pathology. Life (Basel) 2022; 12:life12020153. [PMID: 35207441 PMCID: PMC8874771 DOI: 10.3390/life12020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Management of patients with asthma during the coronavirus disease 2019 (COVID-19) pandemic is a concern, especially since asthma predisposes patients to respiratory problems. Interestingly, asthma characterized by type 2 inflammation, also known as T-helper type 2-high endotype, displays a cellular and molecular profile that may confer protective effects against COVID-19. The results of experimental and clinical studies have established the actions of immunoglobulin E (IgE) in inducing airway hyperreactivity and weakening an interferon-mediated antiviral response following respiratory viral infection. Robust evidence supports the beneficial effect of the anti-IgE biologic treatment omalizumab on reducing respiratory virus-induced asthma exacerbations and reducing the frequency, duration, and severity of respiratory viral illness in patients with asthma. Indeed, accumulating reports of patients with severe asthma treated with omalizumab during the pandemic have reassuringly shown that continuing omalizumab treatment during COVID-19 is safe, and in fact may help prevent the severe course of COVID-19. Accordingly, guidance issued by the Global Initiative for Asthma recommends that all patients with asthma continue taking their prescribed asthma medications, including biologic therapy, during the COVID-19 pandemic. The impact of biologic treatments on patients with asthma and COVID-19 will be better understood as more evidence emerges.
Collapse
Affiliation(s)
- Chung-Jen Wang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
| | - Shih-Lung Cheng
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
- Department of Chemical Engineering and Materials Science, Yuab Ze University, Taoyuan City 32003, Taiwan
| | - Sow-Hsong Kuo
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
- Correspondence:
| |
Collapse
|
3
|
Zheng C, Lu Z, Wu H, Cui L, Bi J, Wan X. Exogenous oxidative stress suppresses IL-33 -driven proliferation programming in group 2 innate lymphoid cells. Int Immunopharmacol 2021; 95:107541. [PMID: 33756232 DOI: 10.1016/j.intimp.2021.107541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 10/21/2022]
Abstract
Although exogenous oxidative stress has been suggested to promote the pathogenesis of airway inflammation, previous trials using conventional antioxidant therapy in asthma have been largely ineffective, suggesting the complex roles of oxidative stress in the regulation of airway inflammation and of its critical mediating lymphocyte populations. Group 2 innate lymphoid cells (ILC2s) proliferate and induce eosinophilia in response to tissue alarminsin the early phase of airway inflammation. We previously showed that IL-33 -induced endogenous reactive oxygen species is required for optimal metabolic activation of ILC2 functions, however, the role of exogenous oxidative stress in regulating ILC2 functions has not been investigated. Here, we found that exogenous oxidative stress induced by injection of ROS -generating reagent alleviated IL-33 -triggered ILC2 response and inflammation both in the airway and in the liver. Exogenous oxidative stress in ILC2s also compromised IL-33 -mediated accumulation of these cells, as well as subsequent recruitment of eosinophils, after adoptive transferring into ILC2 deficient hosts. Mechanistically, exogenous oxidative stress in ILC2s compromised the proliferation program, while preserving the expression levels of effector molecules in ILC2s. Impaired proliferation under exogenous oxidative stress led to reduced numbers of ILC2s, and an overall decrease in ILC2 response to IL-33 stimulation. Collectively, these data indicate that exogenous oxidative stress suppresses the proliferation program in ILC2s and alleviates IL-33 -triggered inflammation, suggesting that therapeutic induction of oxidative stress might alleviate ILC2 -mediated inflammation in the airway, and possibly also in other organs.
Collapse
Affiliation(s)
- Chaoyue Zheng
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China
| | - Zhen Lu
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Haisi Wu
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Lulu Cui
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China
| | - Jiacheng Bi
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; University of Chinese Academy of Sciences, Beijing, People's Republic of China.
| | - Xiaochun Wan
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People's Republic of China; University of Chinese Academy of Sciences, Beijing, People's Republic of China.
| |
Collapse
|
4
|
Zhang J, Feng X, Fan Y, Zhu G, Bai C. Molecular hydrogen alleviates asthma through inhibiting IL-33/ILC2 axis. Inflamm Res 2021; 70:569-579. [PMID: 33852061 DOI: 10.1007/s00011-021-01459-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Asthma is one of the most common noninfectious chronic diseases characterized by type II inflammation. This study aimed to investigate the effects of molecular hydrogen on the pathogenesis of asthma. METHODS OVA sensitized asthma mouse model and house dust mite treated 16HBE cellular model were established and hydrogen/oxygen mixture was used to treat asthmatic mice and 16HBE cells. Serum and BALF cytokines were measured with specific ELISA assays. E-cadherin and ZO-1 were detected by immunohistochemical staining and expression of caspase 3 and 9, NF-κB, IL-33 and ST2 was assessed by quantitative real-time PCR, western blot and/or immunofluorescence. IL-33 promoter activity was analyzed by dual-luciferase assay. ILC2 population was assayed by flow cytometry and differentially expressed miRNAs were detected using miRNA array. RESULTS Serum and BALF levels of IL-33 and other alarmin and type II cytokines were greatly increased by OVA and inhibited by H2 in asthmatic mice. The expression of NF-κB (p65) and ST2 was upregulated by OVA and suppressed by H2. ILC2 population was markedly increased in OVA-induced asthmatic mice, and such increase was inhibited by H2. E-cadherin and ZO-1 levels in airway tissues of asthmatic mice were significantly lower than that of control mice, and the reduction was recovered by H2 treatment. H2 alleviated HDM induced apoptosis of 16HBE cells, upregulation of IL-33 and ST2, and elevation of IL-33 promoter activity. A group of miRNAs differentially expressed in HDM and HDM + H2 treated 16HBE cells were identified. CONCLUSIONS These data demonstrated that H2 is efficient in suppressing allergen-induced asthma and could be developed as a therapeutics for asthma and other conditions of type II inflammation.
Collapse
Affiliation(s)
- Jingxi Zhang
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China.
| | - Xiumin Feng
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Yunxin Fan
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Guanglin Zhu
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Chong Bai
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| |
Collapse
|
5
|
Yang D, Guo X, Huang T, Liu C. The Role of Group 3 Innate Lymphoid Cells in Lung Infection and Immunity. Front Cell Infect Microbiol 2021; 11:586471. [PMID: 33718260 PMCID: PMC7947361 DOI: 10.3389/fcimb.2021.586471] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/19/2021] [Indexed: 02/05/2023] Open
Abstract
The lung is constantly exposed to environmental particulates such as aeroallergens, pollutants, or microorganisms and is protected by a poised immune response. Innate lymphoid cells (ILCs) are a population of immune cells found in a variety of tissue sites, particularly barrier surfaces such as the lung and the intestine. ILCs play a crucial role in the innate immune system, and they are involved in the maintenance of mucosal homeostasis, inflammation regulation, tissue remodeling, and pathogen clearance. In recent years, group 3 innate lymphoid cells (ILC3s) have emerged as key mediators of mucosal protection and repair during infection, mainly through IL-17 and IL-22 production. Although research on ILC3s has become focused on the intestinal immunity, the biology and function of pulmonary ILC3s in the pathogenesis of respiratory infections and in the development of chronic pulmonary inflammatory diseases remain elusive. In this review, we will mainly discuss how pulmonary ILC3s act on protection against pathogen challenge and pulmonary inflammation, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Dan Yang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Xinning Guo
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Tingxuan Huang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Chuntao Liu
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Cephus J, Gandhi VD, Shah R, Brooke Davis J, Fuseini H, Yung JA, Zhang J, Kita H, Polosukhin VV, Zhou W, Newcomb DC. Estrogen receptor-α signaling increases allergen-induced IL-33 release and airway inflammation. Allergy 2021; 76:255-268. [PMID: 32648964 DOI: 10.1111/all.14491] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/04/2020] [Accepted: 06/20/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2) are stimulated by IL-33 to increase IL-5 and IL-13 production and airway inflammation. While sex hormones regulate airway inflammation, it remained unclear whether estrogen signaling through estrogen receptor-α (ER-α, Esr1) or ER-β (Esr2) increased ILC2-mediated airway inflammation. We hypothesize that estrogen signaling increases allergen-induced IL-33 release, ILC2 cytokine production, and airway inflammation. METHODS Female Esr1-/- , Esr2-/- , wild-type (WT), and IL33fl/fl eGFP mice were challenged with Alternaria extract (Alt Ext) or vehicle for 4 days. In select experiments, mice were administered tamoxifen or vehicle pellets for 21 days prior to challenge. Lung ILC2, IL-5 and IL-13 production, and BAL inflammatory cells were measured on day 5 of Alt Ext challenge model. Bone marrow from WT and Esr1-/- female mice was transferred (1:1 ratio) into WT female recipients for 6 weeks followed by Alt Ext challenge. hBE33 cells and normal human bronchial epithelial cells (NHBE) were pretreated with 17β-estradiol (E2), propyl-pyrazole-triol (PPT, ER-α agonist), or diarylpropionitrile (DPN, ER-β agonist) before allergen challenge to determine IL-33 gene expression and release, extracellular ATP release, DUOX-1 production, and necrosis. RESULTS Alt Ext challenged Esr1-/- , but not Esr2-/- , mice had decreased IL-5 and IL-13 production, BAL eosinophils, and IL-33 release compared to WT mice. Tamoxifen decreased IL-5 and IL-13 production and BAL eosinophils. IL-33eGFP + epithelial cells were decreased in Alt Ext challenged Esr1-/- mice compared to WT mice. 17β-E2 or PPT, but not DPN, increased IL-33 gene expression, release, and DUOX-1 production in hBE33 or NHBE cells. CONCLUSION Estrogen receptor -α signaling increased IL-33 release and ILC2-mediated airway inflammation.
Collapse
Affiliation(s)
- Jacqueline‐Yvonne Cephus
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Vivek D. Gandhi
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Ruchi Shah
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Jordan Brooke Davis
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Hubaida Fuseini
- Department of Pathology, Microbiology, and Immunology Vanderbilt University Nashville Tennessee USA
| | - Jeffrey A. Yung
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Jian Zhang
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Hirohito Kita
- Allergic Diseases Research Laboratory Mayo Clinic Phoenix Arizona USA
| | - Vasiliy V. Polosukhin
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Weisong Zhou
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
| | - Dawn C. Newcomb
- Department of Medicine Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center Nashville Tennessee USA
- Department of Pathology, Microbiology, and Immunology Vanderbilt University Nashville Tennessee USA
| |
Collapse
|
7
|
Causton B, Pardo-Saganta A, Gillis J, Discipio K, Kooistra T, Rajagopal J, Xavier RJ, Cho JL, Medoff BD. CARMA3 Mediates Allergic Lung Inflammation in Response to Alternaria alternata. Am J Respir Cell Mol Biol 2019; 59:684-694. [PMID: 29958012 DOI: 10.1165/rcmb.2017-0181oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The airway epithelial cell (AEC) response to allergens helps initiate and propagate allergic inflammation in asthma. CARMA3 is a scaffold protein that mediates G protein-coupled receptor-induced NF-κB activation in airway epithelium. In this study, we demonstrate that mice with CARMA3-deficient AECs have reduced airway inflammation, as well as reduced type 2 cytokine levels in response to Alternaria alternata. These mice also have reduced production of IL-33 and IL-25, and reduced numbers of innate lymphoid cells in the lung. We also show that CARMA3-deficient human AECs have decreased production of proasthmatic mediators in response to A. alternata. Finally, we show that CARMA3 interacts with inositol 1,4,5-trisphosphate receptors in AECs, and that inhibition of CARMA3 signaling reduces A. alternata-induced intracellular calcium release. In conclusion, we show that CARMA3 signaling in AECs helps mediate A. alternata-induced allergic airway inflammation, and that CARMA3 is an important signaling molecule for type 2 immune responses in the lung.
Collapse
Affiliation(s)
- Benjamin Causton
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Ana Pardo-Saganta
- 1 Division of Pulmonary and Critical Care Medicine.,4 Center for Computational and Integrative Biology, and
| | - Jacob Gillis
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Katherine Discipio
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Tristan Kooistra
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Jayaraj Rajagopal
- 1 Division of Pulmonary and Critical Care Medicine.,4 Center for Computational and Integrative Biology, and
| | - Ramnik J Xavier
- 5 Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; and.,2 Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Josalyn L Cho
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Benjamin D Medoff
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| |
Collapse
|
8
|
Critical role of interleukin-23 in development of asthma promoted by cigarette smoke. J Mol Med (Berl) 2019; 97:937-949. [PMID: 31020341 DOI: 10.1007/s00109-019-01768-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/12/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
It has been recently reported that cigarette smoke exposure during allergen sensitization facilitates the development of allergic asthma; however, the underlying mechanisms remain elusive. We evaluated the role of interleukin (IL-23) in a cigarette smoke extract (CSE)-induced Dermatophagoides pteronyssinus (Dp)-allergic asthma mouse model. BALB/c mice were exposed to CSE during allergen sensitization period. Anti-IL-23p19 or IL-23R antibody was administered during the sensitization period. And we evaluated several immunological responses. The expression of IL-23 and IL-23 receptor (IL-23R) was examined in lung tissue. IL-23 and IL-23R expression was increased in the airway epithelium of Dp/CSE co-administered mice. CSE administration during the sensitization promoted Dp-allergic sensitization and the development of asthma phenotypes. Additionally, the proportion of innate lymphoid type 2 cells (ILC2) was also increased by CSE and Dp co-instillation. Anti-IL-23 or IL-23R antibody treatment during allergen sensitization significantly diminished phenotypes of allergic asthma and the ILC2 population. The levels of IL-33 and thymic stromal lymphopoietin (TSLP) were also significantly reduced by anti-IL-23 or IL-23R antibody treatment. IL-23 may thus play a significant role in cigarette smoke-induced allergic sensitization and asthma development. Clinically, the increase in allergen sensitization due to cigarette exposure causes onset of asthma, and IL-23 may be important in this mechanism. KEY MESSAGES: IL-23 and IL-23R expression was increased in the lung epithelium of Dp and CSE co-exposed mice during sensitization period. The population of ILC2s was increased in Dp and CSE co-exposed mice during sensitization period. Anti-IL23 or IL-23R antibody treatment with co-administration of CSE and HDM during sensitization period significantly suppresses ILC2. In vitro, IL-23 blockade in Dp and CSE-stimulated epithelial cells suppressed IL-13 expression in ILC2.
Collapse
|
9
|
Abukabda AB, McBride CR, Batchelor TP, Goldsmith WT, Bowdridge EC, Garner KL, Friend S, Nurkiewicz TR. Group II innate lymphoid cells and microvascular dysfunction from pulmonary titanium dioxide nanoparticle exposure. Part Fibre Toxicol 2018; 15:43. [PMID: 30413212 PMCID: PMC6230229 DOI: 10.1186/s12989-018-0280-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Background The cardiovascular effects of pulmonary exposure to engineered nanomaterials (ENM) are poorly understood, and the reproductive consequences are even less understood. Inflammation remains the most frequently explored mechanism of ENM toxicity. However, the key mediators and steps between lung exposure and uterine health remain to be fully defined. The purpose of this study was to determine the uterine inflammatory and vascular effects of pulmonary exposure to titanium dioxide nanoparticles (nano-TiO2). We hypothesized that pulmonary nano-TiO2 exposure initiates a Th2 inflammatory response mediated by Group II innate lymphoid cells (ILC2), which may be associated with an impairment in uterine microvascular reactivity. Methods Female, virgin, Sprague-Dawley rats (8–12 weeks) were exposed to 100 μg of nano-TiO2 via intratracheal instillation 24 h prior to microvascular assessments. Serial blood samples were obtained at 0, 1, 2 and 4 h post-exposure for multiplex cytokine analysis. ILC2 numbers in the lungs were determined. ILC2s were isolated and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) levels were measured. Pressure myography was used to assess vascular reactivity of isolated radial arterioles. Results Pulmonary nano-TiO2 exposure was associated with an increase in IL-1ß, 4, 5 and 13 and TNF- α 4 h post-exposure, indicative of an innate Th2 inflammatory response. ILC2 numbers were significantly increased in lungs from exposed animals (1.66 ± 0.19%) compared to controls (0.19 ± 0.22%). Phosphorylation of the transactivation domain (Ser-468) of NF-κB in isolated ILC2 and IL-33 in lung epithelial cells were significantly increased (126.8 ± 4.3% and 137 ± 11% of controls respectively) by nano-TiO2 exposure. Lastly, radial endothelium-dependent arteriolar reactivity was significantly impaired (27 ± 12%), while endothelium-independent dilation (7 ± 14%) and α-adrenergic sensitivity (8 ± 2%) were not altered compared to control levels. Treatment with an anti- IL-33 antibody (1 mg/kg) 30 min prior to nano-TiO2 exposure resulted in a significant improvement in endothelium-dependent dilation and a decreased level of IL-33 in both plasma and bronchoalveolar lavage fluid. Conclusions These results provide evidence that the uterine microvascular dysfunction that follows pulmonary ENM exposure may be initiated via activation of lung-resident ILC2 and subsequent systemic Th2-dependent inflammation. Electronic supplementary material The online version of this article (10.1186/s12989-018-0280-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alaeddin Bashir Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll Rolland McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas Paul Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William Travis Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Compton Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Lee Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Robert Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
10
|
Chirumbolo S, Bjørklund G, Sboarina A, Vella A. The role of basophils as innate immune regulatory cells in allergy and immunotherapy. Hum Vaccin Immunother 2018; 14:815-831. [PMID: 29257936 DOI: 10.1080/21645515.2017.1417711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Basophils are circulating cells that are associated quite exclusively with allergy response and hypersensitivity reactions but their role in the immune network might be much more intriguing and complex than previously expected. The feasibility of testing their biology in vitro for allergy research and diagnosis, due fundamentally to their quite easy availability in the peripheral blood, made them the major source for assessing allergy in the laboratory assay, when yet many further cells such as mast cells and eosinophils are much more involved as effector cells in allergy than circulating basophils. Interestingly, basophil numbers change rarely in peripheral blood during an atopic response, while we might yet observe an increase in eosinophils and modification in the biology of mast cells in the tissue during an hypersensitivity response. Furthermore, the fact that basophils are very scanty in numbers suggests that they should mainly serve as regulatory cells in immunity, rather than effector leukocytes, as still believed by the majority of physicians. In this review we will try to describe and elucidate the possible role of these cells, known as "innate IL4-producing cells" in the immune regulation of allergy and their function in allergen immunotherapy.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- a Department of Neurological and Movement Sciences , University of Verona , Verona , Italy
| | - Geir Bjørklund
- b Council for Nutritional and Environmental Medicine (CONEM) , Mo i Rana , Norway
| | - Andrea Sboarina
- c Department of Surgery , Dentistry, Paediatrics and Gynaecology-University of Verona , Verona , Italy
| | - Antonio Vella
- d Unit of Immunology-Azienda Ospedaliera Universitaria Integrata (AOUI) , Verona , Italy
| |
Collapse
|
11
|
Interleukin 33 exacerbates antigen driven airway hyperresponsiveness, inflammation and remodeling in a mouse model of asthma. Sci Rep 2017; 7:4219. [PMID: 28652606 PMCID: PMC5484710 DOI: 10.1038/s41598-017-03674-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/04/2017] [Indexed: 12/14/2022] Open
Abstract
Interleukin 33 (IL-33) represents a potential link between the airway epithelium and induction of Th2-type inflammatory responses associated with the development of asthma. This study investigated the potential of IL-33 to exacerbate antigen driven asthma responses. An ovalbumin (OVA) asthma model was used in which sensitized C57BL/6 mice were exposed to IL-33 before each OVA challenge. IL-33 given to sensitized mice acted synergistically with antigen and aggravated airway inflammation, hyperresponsiveness and remodeling compared with mice that were only OVA sensitized and challenged and mice that were only exposed to IL-33. Elevated levels of local and systemic mast cell protease mMCP-1, as well as antigen-specific IgE production, were observed following IL-33 administration to sensitized mice. Similarly, exposing OVA-sensitized mice to IL-33 increased the Th2 cytokine levels, including IL-4, IL-5 and IL-13. Furthermore, IL-33 and OVA administration to OVA-sensitized mice increased ILC2s in the lung, suggesting a role for ILC2s in IL-33-mediated exacerbation of OVA-induced airway responses. Collectively, these findings show that IL-33 aggravates important features of antigen-driven asthma, which may have implications for asthma exacerbations.
Collapse
|
12
|
Vinton CL, Ortiz AM, Calantone N, Mudd JC, Deleage C, Morcock DR, Whitted S, Estes JD, Hirsch VM, Brenchley JM. Cytotoxic T Cell Functions Accumulate When CD4 Is Downregulated by CD4 + T Cells in African Green Monkeys. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:4403-4412. [PMID: 28438898 PMCID: PMC5502537 DOI: 10.4049/jimmunol.1700136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/29/2017] [Indexed: 01/06/2023]
Abstract
African green monkeys (AGMs) are a natural host of SIV that do not develop simian AIDS. Adult AGMs naturally have low numbers of CD4+ T cells and a large population of MHC class II-restricted CD8αα T cells that are generated through CD4 downregulation in CD4+ T cells. In this article, we study the functional profiles and SIV infection status in vivo of CD4+ T cells, CD8αα T cells, and CD8αβ T cells in lymph nodes, peripheral blood, and bronchoalveolar lavage fluid of AGMs and rhesus macaques (in which CD4 downregulation is not observed). We show that, although CD8αα T cells in AGMs maintain functions associated with CD4+ T cells (including Th follicular functionality in lymphoid tissues and Th2 responses in bronchoalveolar lavage fluid), they also accumulate functions normally attributed to canonical CD8+ T cells. These hyperfunctional CD8αα T cells are found to circulate peripherally, as well as reside within the lymphoid tissue. Due to their unique combination of CD4 and CD8 T cell effector functions, these CD4- CD8αα T cells are likely able to serve as an immunophenotype capable of Th1, follicular Th, and CTL functionalities, yet they are unable to be infected by SIV. These data demonstrate the ambiguity of CD4/CD8 expression in dictating the functional capacities of T cells and suggest that accumulation of hyperfunctional CD8αα T cells in AGMs may lead to tissue-specific antiviral immune responses in lymphoid follicles that limit SIV replication in this particular anatomical niche.
Collapse
Affiliation(s)
- Carol L Vinton
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexandra M Ortiz
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Nina Calantone
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Joseph C Mudd
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Claire Deleage
- Retroviral Immunopathology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702; and
| | - David R Morcock
- Retroviral Immunopathology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702; and
| | - Sonya Whitted
- Nonhuman Primate Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jacob D Estes
- Retroviral Immunopathology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702; and
| | - Vanessa M Hirsch
- Nonhuman Primate Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
13
|
|
14
|
Abstract
Type-2 innate lymphoid cells (ILC2) belong to an expanding family of innate lymphocytes that provide a potent source of immune effector cytokines at the initiation of immune responses. ILC2 arise, under the control of the transcription factors RORα and GATA3, from lymphoid progenitors in the bone marrow, to secrete type-2 cytokines including IL-5 and IL-13. Using experimental models, ILC2 have been implicated in allergic diseases, such as asthma and atopic dermatitis, but also in metabolic homeostasis. Furthermore, recent reports have indicated that ILC2 not only play roles at the initiation of type-2 immunity but can also contribute to chronic pathology, such as fibrosis, and can impact on the priming of the adaptive T-cell response. The identification of ILC2 in patients with allergic dermatitis and allergic rhinitis indicates that these cells may represent new therapeutic targets.
Collapse
|
15
|
Affiliation(s)
- S. Scheurer
- Molecular Allergology; Paul-Ehrlich-Institut; Langen Germany
| | - M. Toda
- Molecular Allergology; Paul-Ehrlich-Institut; Langen Germany
| | - S. Vieths
- Molecular Allergology; Paul-Ehrlich-Institut; Langen Germany
| |
Collapse
|
16
|
Russi AE, Walker-Caulfield ME, Ebel ME, Brown MA. Cutting edge: c-Kit signaling differentially regulates type 2 innate lymphoid cell accumulation and susceptibility to central nervous system demyelination in male and female SJL mice. THE JOURNAL OF IMMUNOLOGY 2015; 194:5609-13. [PMID: 25972476 DOI: 10.4049/jimmunol.1500068] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/10/2015] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis preferentially affects women, and this sexual dimorphism is recapitulated in the SJL mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). In this study, we demonstrate that signaling through c-Kit exerts distinct effects on EAE susceptibility in male and female SJL mice. Previous studies in females show that Kit mutant (W/W(v)) mice are less susceptible to EAE than are wild-type mice. However, male W/W(v) mice exhibit exacerbated disease, a phenotype independent of mast cells and corresponding to a shift from a Th2- to a Th17-dominated T cell response. We demonstrate a previously undescribed deficit in c-Kit(+) type 2 innate lymphoid cells (ILC2s) in W/W(v) mice. ILC2s are also significantly reduced in EAE-susceptible wild-type females, indicating that both c-Kit signals and undefined male-specific factors are required for ILC2 function. We propose that deficiencies in Th2-promoting ILC2s remove an attenuating influence on the encephalitogenic T cell response and therefore increases disease susceptibility.
Collapse
Affiliation(s)
- Abigail E Russi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | | | - Mark E Ebel
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| |
Collapse
|
17
|
Liu B, Lee JB, Chen CY, Hershey GKK, Wang YH. Collaborative interactions between type 2 innate lymphoid cells and antigen-specific CD4+ Th2 cells exacerbate murine allergic airway diseases with prominent eosinophilia. THE JOURNAL OF IMMUNOLOGY 2015; 194:3583-93. [PMID: 25780046 DOI: 10.4049/jimmunol.1400951] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 02/16/2015] [Indexed: 01/21/2023]
Abstract
Type-2 innate lymphoid cells (ILC2s) and the acquired CD4(+) Th2 and Th17 cells contribute to the pathogenesis of experimental asthma; however, their roles in Ag-driven exacerbation of chronic murine allergic airway diseases remain elusive. In this study, we report that repeated intranasal rechallenges with only OVA Ag were sufficient to trigger airway hyperresponsiveness, prominent eosinophilic inflammation, and significantly increased serum OVA-specific IgG1 and IgE in rested mice that previously developed murine allergic airway diseases. The recall response to repeated OVA inoculation preferentially triggered a further increase of lung OVA-specific CD4(+) Th2 cells, whereas CD4(+) Th17 and ILC2 cell numbers remained constant. Furthermore, the acquired CD4(+) Th17 cells in Stat6(-/-)/IL-17-GFP mice, or innate ILC2s in CD4(+) T cell-ablated mice, failed to mount an allergic recall response to OVA Ag. After repeated OVA rechallenge or CD4(+) T cell ablation, the increase or loss of CD4(+) Th2 cells resulted in an enhanced or reduced IL-13 production by lung ILC2s in response to IL-25 and IL-33 stimulation, respectively. In return, ILC2s enhanced Ag-mediated proliferation of cocultured CD4(+) Th2 cells and their cytokine production, and promoted eosinophilic airway inflammation and goblet cell hyperplasia driven by adoptively transferred Ag-specific CD4(+) Th2 cells. Thus, these results suggest that an allergic recall response to recurring Ag exposures preferentially triggers an increase of Ag-specific CD4(+) Th2 cells, which facilitates the collaborative interactions between acquired CD4(+) Th2 cells and innate ILC2s to drive the exacerbation of a murine allergic airway diseases with an eosinophilic phenotype.
Collapse
Affiliation(s)
- Bo Liu
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45220
| | - Jee-Boong Lee
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45220
| | - Chun-Yu Chen
- Division of Hematology, Oncology, and Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH 43205; and
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45220
| | - Yui-Hsi Wang
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45220;
| |
Collapse
|
18
|
Lung ILC2s link innate and adaptive responses in allergic inflammation. Trends Immunol 2015; 36:189-95. [PMID: 25704560 DOI: 10.1016/j.it.2015.01.005] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/19/2014] [Accepted: 01/21/2015] [Indexed: 12/24/2022]
Abstract
How allergens trigger the T helper 2 (Th2) response that characterizes allergic lung inflammation is not well understood. Epithelium-derived alarmins released after an allergen encounter activate the innate immune system, including group 2 innate lymphoid cells (ILC2s) which produce the type 2 interleukins IL-5 and IL-13. It has been recently shown that ILC2-derived cytokines are responsible not only for the innate responses underlying allergic inflammation but also for the initiation of the adaptive Th2 response. We review the role of lung ILC2s in the development of allergic inflammation and, in the context of recent findings, propose a common pathway wherein ILC2s, activated by the epithelium-derived cytokine IL-33, link the innate and the adaptive responses after allergen encounter in the lung.
Collapse
|
19
|
McKenzie ANJ, Spits H, Eberl G. Innate lymphoid cells in inflammation and immunity. Immunity 2014; 41:366-374. [PMID: 25238094 DOI: 10.1016/j.immuni.2014.09.006] [Citation(s) in RCA: 276] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs) were first described as playing important roles in the development of lymphoid tissues and more recently in the initiation of inflammation at barrier surfaces in response to infection or tissue damage. It has now become apparent that ILCs play more complex roles throughout the duration of immune responses, participating in the transition from innate to adaptive immunity and contributing to chronic inflammation. The proximity of ILCs to epithelial surfaces and their constitutive strategic positioning in other tissues throughout the body ensures that, in spite of their rarity, ILCs are able to regulate immune homeostasis effectively. Dysregulation of ILC function might result in chronic pathologies such as allergies, autoimmunity, and inflammation. A new role for ILCs in the maintenance of metabolic homeostasis has started to emerge, underlining their importance in fundamental physiological processes beyond infection and immunity.
Collapse
Affiliation(s)
- Andrew N J McKenzie
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| | - Hergen Spits
- Academic Medical Center, University of Amsterdam, Department of Cell Biology & Histology, 1105BK Amsterdam, the Netherlands
| | - Gerard Eberl
- Institut Pasteur, Lymphoid Tissue Development Unit, Paris 75724, France
| |
Collapse
|
20
|
Vatrella A, Fabozzi I, Calabrese C, Maselli R, Pelaia G. Dupilumab: a novel treatment for asthma. J Asthma Allergy 2014; 7:123-30. [PMID: 25214796 PMCID: PMC4159398 DOI: 10.2147/jaa.s52387] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Simultaneously with the steady progress towards a better knowledge of the pathobiology of asthma, the potential usefulness of anticytokine therapies is emerging as one of the key concepts in the newly developing treatments of this widespread airway disease. In particular, given the key role played by interleukin (IL)-4 and IL-13 in the pathophysiology of the most typical aspects of asthma, such as chronic airway inflammation, tissue remodeling, and bronchial hyperresponsiveness, these pleiotropic cytokines are now considered as suitable therapeutic targets. Among the recently developed antiasthma biologic drugs, the monoclonal antibody dupilumab is very promising because of its ability to inhibit the biological effects of both IL-4 and IL-13. Indeed, dupilumab prevents IL-4/13 interactions with the α-subunit of the IL-4 receptor complex. A recent trial showed that in patients with difficult-to-control asthma, dupilumab can markedly decrease asthma exacerbations and improve respiratory symptoms and lung function; these effects were paralleled by significant reductions in T-helper 2-associated inflammatory biomarkers. However, further larger and longer trials are required to extend and validate these preliminary results, and also to carefully study the safety and tolerability profile of dupilumab.
Collapse
Affiliation(s)
| | - Immacolata Fabozzi
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Cecilia Calabrese
- Department of Cardiothoracic and Respiratory Sciences, Second University of Naples, Naples, Italy
| | - Rosario Maselli
- Department of Medical and Surgical Sciences, University Magna Græcia, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Medical and Surgical Sciences, University Magna Græcia, Catanzaro, Italy
| |
Collapse
|
21
|
Key mediators in the immunopathogenesis of allergic asthma. Int Immunopharmacol 2014; 23:316-29. [PMID: 24933589 DOI: 10.1016/j.intimp.2014.05.034] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 12/20/2022]
Abstract
Asthma is described as a chronic inflammatory disorder of the conducting airways. It is characterized by reversible airway obstruction, eosinophil and Th2 infiltration, airway hyper-responsiveness and airway remodeling. Our findings to date have largely been dependent on work done using animal models, which have been instrumental in broadening our understanding of the mechanism of the disease. However, using animals to model a uniquely human disease is not without its drawbacks. This review aims to examine some of the key mediators and cells of allergic asthma learned from animal models and shed some light on emerging mediators in the pathogenesis allergic airway inflammation in acute and chronic asthma.
Collapse
|
22
|
Nau D, Altmayer N, Mattner J. Mechanisms of innate lymphoid cell and natural killer T cell activation during mucosal inflammation. J Immunol Res 2014; 2014:546596. [PMID: 24987710 PMCID: PMC4058452 DOI: 10.1155/2014/546596] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 04/28/2014] [Indexed: 02/07/2023] Open
Abstract
Mucosal surfaces in the airways and the gastrointestinal tract are critical for the interactions of the host with its environment. Due to their abundance at mucosal tissue sites and their powerful immunomodulatory capacities, the role of innate lymphoid cells (ILCs) and natural killer T (NKT) cells in the maintenance of mucosal tolerance has recently moved into the focus of attention. While NKT cells as well as ILCs utilize distinct transcription factors for their development and lineage diversification, both cell populations can be further divided into three polarized subpopulations reflecting the distinction into Th1, Th2, and Th17 cells in the adaptive immune system. While bystander activation through cytokines mediates the induction of ILC and NKT cell responses, NKT cells become activated also through the engagement of their canonical T cell receptors (TCRs) by (glyco)lipid antigens (cognate recognition) presented by the atypical MHC I like molecule CD1d on antigen presenting cells (APCs). As both innate lymphocyte populations influence inflammatory responses due to the explosive release of copious amounts of different cytokines, they might represent interesting targets for clinical intervention. Thus, we will provide an outlook on pathways that might be interesting to evaluate in this context.
Collapse
Affiliation(s)
- David Nau
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, 91054 Erlangen, Germany
| | - Nora Altmayer
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, 91054 Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, 91054 Erlangen, Germany
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|