1
|
Zlotnikov ID, Kudryashova EV. Polymeric Infrared and Fluorescent Probes to Assess Macrophage Diversity in Bronchoalveolar Lavage Fluid of Asthma and Other Pulmonary Disease Patients. Polymers (Basel) 2024; 16:3427. [PMID: 39684172 DOI: 10.3390/polym16233427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Bronchial asthma remains a serious medical problem, as approximately 10% of patients fail to achieve adequate symptom control with available treatment options. Macrophages play a pivotal role in the pathophysiology of asthma, as well as in some other respiratory disorders. Typically, they are classified into two major classes, M1 and M2; however, recent findings have indicated that in fact there is a whole range of macrophage polarization and functional diversity beyond this bimodal division. The isolation of individual cell sub-populations and the identification of their role and diagnostic/therapeutic significance is still a challenge. Here, we have attempted to assess the differences between patient-derived macrophage populations from bronchoalveolar lavage fluid (BALF) samples in different pulmonary disease conditions, based on their capability to interact with a range of specific and relatively non-specific carbohydrate-based ligands (containing galactose (linear or cyclic form), mannose, trimannose, etc.). Obviously, the main target of these ligands was CD206; however, other minor receptors, able to bind carbohydrates, have also been reported for macrophages. Trimannose binds most specifically to CD206 macrophage receptors, while monomannose has intermediate affinity, and galactose has low affinity and may involve binding to other receptors. This clearly indicates the ligands were chosen based on their predicted binding strength and specificity for CD206, providing the rationale for the study. In some cases, the activated macrophage affinity to galactose base ligands was higher than that to mannose, indicating that complexes of CD206 or other carbohydrate-binding receptors may contribute substantially to macrophage functional features. In addition, variations in receptor clustering and distribution may substantially affect affinity to the same ligand. Interestingly, with a panel of 6-10 different carbohydrate-based ligands with FTIR or fluorescent marker, we were able not only to distinguish between healthy and disease states but also between closely related diseases such as purulent endobronchitis, obstructive bronchitis, pneumonia, and bronchial asthma. For further investigation, specific sub-populations of macrophages, seen as hallmarks to specific diseases, can be isolated and studied separately, likely giving new insights with diagnostic and therapeutic significance for hard-to-treat patients. The group of patients with resistant disease can also be identified with this approach as a fingerprint method to find a more targeted therapeutic strategy, improving their clinical outcomes. As expected, this will provide a large additional array of data for analysis, compared to the work going on in the world. The dataset used by other researchers mainly for known "antibody" ligands is semi-quantitative and insufficient for the purposes of typing as yet unknown and uncomplicated sub-populations. The analysis of the presented data in combination with personalized information from patients' medical records will be carried out using both traditional methods and machine learning methods.
Collapse
Affiliation(s)
- Igor D Zlotnikov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| | - Elena V Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| |
Collapse
|
2
|
Ulrich BJ, Zhang W, Kenworthy BT, Kharwadkar R, Olson MR, Kaplan MH. Activin A Promotes Differentiation of a Pathogenic Multicytokine IL-9-secreting CD4+ T Cell Population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:823-830. [PMID: 39058312 PMCID: PMC11371476 DOI: 10.4049/jimmunol.2300635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024]
Abstract
The development of Th subsets results from cellular and cytokine cues that are present in the inflammatory environment. The developing T cell integrates multiple signals from the environment that sculpt the cytokine-producing capacity of the effector T cell. Importantly, T cells can discriminate similar cytokine signals to generate distinct outcomes, and that discrimination is critical in Th subset development. IL-9-secreting Th9 cells regulate multiple immune responses, including immunity to pathogens and tumors, allergic inflammation, and autoimmunity. In combination with IL-4, TGF-β or activin A promotes IL-9 production; yet, it is not clear if both TGF-β family members generate Th9 cells with identical phenotype and function. We observed that in contrast to TGF-β that efficiently represses Th2 cytokines in murine Th9 cultures, differentiation with activin A produced a multicytokine T cell phenotype with secretion of IL-4, IL-5, IL-13, and IL-10 in addition to IL-9. Moreover, multicytokine secreting cells are more effective at promoting allergic inflammation. These observations suggest that although TGF-β and IL-4 were identified as cytokines that stimulate optimal IL-9 production, they might not be the only cytokines that generate optimal function from IL-9-producing T cells in immunity and disease.
Collapse
Affiliation(s)
- Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Wenwu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Blake T Kenworthy
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Rakshin Kharwadkar
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Matthew R Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
3
|
Yadav V, Pandey V, Gaglani P, Srivastava A, Soni, Subhashini. Inhibiting SIRT-2 by AK-7 restrains airway inflammation and oxidative damage promoting lung resurgence through NF-kB and MAP kinase signaling pathway. Front Immunol 2024; 15:1404122. [PMID: 38979411 PMCID: PMC11228164 DOI: 10.3389/fimmu.2024.1404122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/24/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a major global cause of mortality with limited effective treatments. Sirtuins (SIRT) are histone deacetylases that are involved in the regulation of redox and inflammatory homeostasis. Hence, the present study aims to investigate the role of SIRT-2 in modulating inflammation in a murine model of COPD. Methods COPD in mice was established by cigarette smoke (CS) exposure for 60 days, and AK-7 was used as the specific SIRT-2 inhibitor. AK-7 (100 µg/kg and 200 µg/kg body weight) was administered intranasally 1 h before CS exposure. Molecular docking was performed to analyze the binding affinity of different inflammatory proteins with AK-7. Results Immune cell analysis showed a significantly increased number of macrophages (F4/80), neutrophils (Gr-1), and lymphocytes (CD4+, CD8+, and CD19+) in the COPD, group and their population was declined by AK-7 administration. Total reactive oxygen species, total inducible nitric oxide synthase, inflammatory mediators such as neutrophil elastase, C-reactive protein, histamine, and cytokines as IL4, IL-6, IL-17, and TNF-α were elevated in COPD and declined in the AK-7 group. However, IL-10 showed reverse results representing anti-inflammatory potency. AK-7 administration by inhibiting SIRT-2 decreased the expression of p-NF-κB, p-P38, p-Erk, and p-JNK and increased the expression of Nrf-2. Furthermore, AK-7 also declined the lung injury by inhibiting inflammation, parenchymal destruction, emphysema, collagen, club cells, and Kohn pores. AK-7 also showed good binding affinity with inflammatory proteins. Discussion The current study reveals that SIRT-2 inhibition mitigates COPD severity and enhances pulmonary therapeutic interventions, suggesting AK-7 as a potential therapeutic molecule for COPD medication development.
Collapse
Affiliation(s)
- Vandana Yadav
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Vinita Pandey
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Pratikkumar Gaglani
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Atul Srivastava
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Soni
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Subhashini
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| |
Collapse
|
4
|
Adilis Maria Paiva Ferreira L, Karla Diega Paiva Ferreira L, Fragoso Pereira Cavalcanti R, Allysson de Assis Ferreira Gadelha F, Mangueira de Lima L, Francisco Alves A, Gabriel Lima Júnior C, Regina Piuvezam M. Morita-Baylis-Hillman adduct 2-(3-hydroxy-1-methyl-2-oxoindolin-3-il) acrylonitrile (CISACN) ameliorates the pulmonary allergic inflammation in CARAS model by increasing IFN-γ/IL-4 ratio towards the Th1 immune response. Int Immunopharmacol 2024; 130:111737. [PMID: 38401465 DOI: 10.1016/j.intimp.2024.111737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Combined allergic rhinitis and asthma syndrome (CARAS) is an airway-type 2 immune response with a profuse inflammatory process widely affecting the world population. Due to the compromise of quality of life and the lack of specific pharmacotherapy, the search for new molecules becomes relevant. This study aimed to evaluate the effectiveness of the Morita-Bailys-Hillman adduct (CISACN) treatment in the CARAS experimental model. Female BALB/c mice were ovalbumin (OVA) -sensitized and -challenged and treated with CISACN. The treatment decreased the eosinophil migration to the nasal and lung cavities and tissues and the goblet cell hyperplasia/hypertrophy, attenuated airway hyperactivity by reducing the hyperplasia/hypertrophy of the smooth muscle and the extracellular matrix's thickness. Also, the treatment reduced the clinical signs of rhinitis as nasal rubbing and sneezing in a histamine-induced nasal hyperreactivity assay. The immunomodulatory effect of CISACN was by reducing OVA-specific IgE serum level, and IL-33, IL-4, IL-13, and TGF-β production, dependent on IFN-γ increase. Furthermore, the effect of CISACN on lung granulocytes was by decreasing the p-p38MAPK/p65NF-κB signaling pathway. Indeed, CISACN reduced the p38MAPK and p65NF-κB activation. These data demonstrated the anti-inflammatory and immunomodulatory effects of the CISACN with scientific support to become a pharmacological tool to treat airway inflammatory diseases.
Collapse
Affiliation(s)
- Larissa Adilis Maria Paiva Ferreira
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Raquel Fragoso Pereira Cavalcanti
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Louise Mangueira de Lima
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Adriano Francisco Alves
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Marcia Regina Piuvezam
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil; Drug Research Institute of the Federal University of Paraíba, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil.
| |
Collapse
|
5
|
Qian G, Jiang W, Sun D, Sun Z, Chen A, Fang H, Wang J, Liu Y, Yin Z, Wei H, Fang H, Zhang X. B-cell-derived IL-10 promotes allergic sensitization in asthma regulated by Bcl-3. Cell Mol Immunol 2023; 20:1313-1327. [PMID: 37653127 PMCID: PMC10616210 DOI: 10.1038/s41423-023-01079-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/15/2023] [Indexed: 09/02/2023] Open
Abstract
Aeroallergen sensitization, mainly mediated by lung epithelium and dendritic cells (DCs), is integral to allergic asthma pathogenesis and progression. IL-10 has a dual role in immune responses, as it inhibits myeloid cell activation but promotes B-cell responses and epithelial cell proliferation. Here, we report a proinflammatory function of B-cell-derived IL-10 modulated by Bcl-3 in allergic asthma. Specifically, Bcl-3-/- mice showed elevated IL-10 levels and were found to be highly vulnerable to allergic asthma induced by house dust mites (HDMs). IL-10 had a positive correlation with the levels of the DC chemoattractant CCL-20 in HDM-sensitized mice and in patients with asthma and induced a selective increase in CCL-20 production by mouse lung epithelial cells. Blockade of IL-10 or IL-10 receptors during sensitization dampened both HDM-induced sensitization and asthma development. IL-10 levels peaked 4 h post sensitization with HDM and IL-10 was primarily produced by B cells under Bcl-3-Blimp-1-Bcl-6 regulation. Mice lacking B-cell-derived IL-10 displayed decreased lung epithelial CCL-20 production and diminished DC recruitment to the lungs upon HDM sensitization, thereby demonstrating resistance to HDM-induced asthma. Moreover, responses to HDM stimulation in Bcl-3-/- mice lacking B-cell-derived IL-10 were comparable to those in Bcl-3+/+ mice. The results revealed an unexpected role of B-cell-derived IL-10 in promoting allergic sensitization and demonstrated that Bcl-3 prevents HDM-induced asthma by inhibiting B-cell-derived IL-10 production. Thus, targeting the Bcl-3/IL-10 axis to inhibit allergic sensitization is a promising approach for treating allergic asthma. IL-10 is released rapidly from lung plasma cells under Bcl-3-Blimp-1-Bcl-6 regulation upon house dust mite exposure and amplifies lung epithelial cell (EC)-derived CCL-20 production and subsequent dendritic cell (DC) recruitment to promote allergic sensitization in asthma.
Collapse
Affiliation(s)
- Guojun Qian
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, 200001, Shanghai, China.
| | - Wenxia Jiang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Donglin Sun
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China
| | - Zhun Sun
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China
| | - Anning Chen
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China
| | - Hongwei Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jingyao Wang
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China
| | - Yongzhong Liu
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200032, Shanghai, China
| | - Zhinan Yin
- Zhuhai People's Hospital, Biomedical Translational Research Institute, Jinan University, 510632, Guangzhou, China
| | - Haiming Wei
- Institute of Immunology, University of Science and Technology of China, 230000, Hefei, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Department of Anesthesiology, Minhang Hospital, Fudan University, 201100, Shanghai, China.
| | - Xiaoren Zhang
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, 511436, Guangzhou, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
6
|
Schneble D, El-Gazzar A, Kargarpour Z, Kramer M, Metekol S, Stoshikj S, Idzko M. Cell-type-specific role of P2Y2 receptor in HDM-driven model of allergic airway inflammation. Front Immunol 2023; 14:1209097. [PMID: 37790940 PMCID: PMC10543084 DOI: 10.3389/fimmu.2023.1209097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Allergic airway inflammation (AAI) is a chronic respiratory disease that is considered a severe restriction in daily life and is accompanied by a constant risk of acute aggravation. It is characterized by IgE-dependent activation of mast cells, infiltration of eosinophils, and activated T-helper cell type 2 (Th2) lymphocytes into airway mucosa. Purinergic receptor signaling is known to play a crucial role in inducing and maintaining allergic airway inflammation. Previous studies in an ovalbumin (OVA)-alum mouse model demonstrated a contribution of the P2Y2 purinergic receptor subtype (P2RY2) in allergic airway inflammation. However, conflicting data concerning the mechanism by which P2RY2 triggers AAI has been reported. Thus, we aimed at elucidating the cell-type-specific role of P2RY2 signaling in house dust mite (HDM)-driven model of allergic airway inflammation. Thereupon, HDM-driven AAI was induced in conditional knockout mice, deficient or intact for P2ry2 in either alveolar epithelial cells, hematopoietic cells, myeloid cells, helper T cells, or dendritic cells. To analyze the functional role of P2RY2 in these mice models, flow cytometry of bronchoalveolar lavage fluid (BALF), cytokine measurement of BALF, invasive lung function measurement, HDM re-stimulation of mediastinal lymph node (MLN) cells, and lung histology were performed. Mice that were subjected to an HDM-based model of allergic airway inflammation resulted in reduced signs of acute airway inflammation including eosinophilia in BALF, peribronchial inflammation, Th2 cytokine production, and bronchial hyperresponsiveness in mice deficient for P2ry2 in alveolar epithelial cells, hematopoietic cells, myeloid cells, or dendritic cells. Furthermore, the migration of bone-marrow-derived dendritic cells and bone-marrow-derived monocytes, both deficient in P2ry2, towards ATP was impaired. Additionally, we found reduced levels of MCP-1/CCL2 and IL-8 homologues in the BALF of mice deficient in P2ry2 in myeloid cells and lower concentrations of IL-33 in the lung tissue of mice deficient in P2ry2 in alveolar epithelial cells. In summary, our results show that P2RY2 contributes to HDM-induced airway inflammation by mediating proinflammatory cytokine production in airway epithelial cells, monocytes, and dendritic cells and drives the recruitment of lung dendritic cells and monocytes.
Collapse
Affiliation(s)
- Dominik Schneble
- Department of Pneumology, Medical Center – University of Freiburg, Freiburg, Germany
| | - Ahmed El-Gazzar
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Zahra Kargarpour
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Markus Kramer
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Seda Metekol
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Slagjana Stoshikj
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Department of Pneumology, Medical Center – University of Freiburg, Freiburg, Germany
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Live attenuated pertussis vaccine for prevention and treatment of allergic airway inflammation in mice. NPJ Vaccines 2022; 7:66. [PMID: 35739108 PMCID: PMC9226346 DOI: 10.1038/s41541-022-00494-w] [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: 01/10/2022] [Accepted: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Live attenuated vaccines often have beneficial non-specific effects, protecting against heterologous infectious and non-infectious diseases. We have developed a live attenuated pertussis vaccine, named BPZE1, currently in advanced clinical development. Here, we examined the prophylactic and therapeutic potential of its pertactin-deficient derivative BPZE1P in a mouse model of house dust mite (HDM)-induced allergic airway inflammation (AAI). BPZE1P was given nasally either before or after sensitization with HDM, followed by HDM challenge, or between two challenge episodes. Vaccination prior to sensitization reduced resistance in the airways, the numbers of infiltrating eosinophils and the concentrations of proinflammatory cytokines, such as IL-1α, IL-1β and IL-33, in the lungs but had no effect on Th2 cytokine levels. BPZE1P also protected when delivered after sensitization or between two challenge episodes. However, in this case the levels of Th2 cytokines in the lung were decreased without significant effects on IL-1α, IL-1β and IL-33 production. The vaccine restored lung function and decreased eosinophil influx in the lungs of HDM-treated mice. BPZE1P has a better take than BPZE1 in hosts vaccinated with acellular pertussis vaccines. Therefore, it has interesting potential as a preventive and therapeutic agent against AAI, even in acellular pertussis-vaccinated populations.
Collapse
|
8
|
Misiukiewicz-Stepien P, Paplinska-Goryca M. Biological effect of PM 10 on airway epithelium-focus on obstructive lung diseases. Clin Immunol 2021; 227:108754. [PMID: 33964432 DOI: 10.1016/j.clim.2021.108754] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
Recently, a continuous increase in environmental pollution has been observed. Despite wide-scale efforts to reduce air pollutant emissions, the problem is still relevant. Exposure to elevated levels of airborne particles increased the incidence of respiratory diseases. PM10 constitute the largest fraction of air pollutants, containing particles with a diameter of less than 10 μm, metals, pollens, mineral dust and remnant material from anthropogenic activity. The natural airway defensive mechanisms against inhaled material, such as mucus layer, ciliary clearance and macrophage phagocytic activity, may be insufficient for proper respiratory function. The epithelium layer can be disrupted by ongoing oxidative stress and inflammatory processes induced by exposure to large amounts of inhaled particles as well as promote the development and exacerbation of obstructive lung diseases. This review draws attention to the current state of knowledge about the physical features of PM10 and its impact on airway epithelial cells, and obstructive pulmonary diseases.
Collapse
Affiliation(s)
- Paulina Misiukiewicz-Stepien
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland; Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland.
| | | |
Collapse
|
9
|
Paiva Ferreira LKD, Paiva Ferreira LAM, Bezerra Barros GC, Mozzini Monteiro T, de Araújo Silva LA, Pereira RDA, Figueiredo PTR, Alves AF, Rodrigues LC, Piuvezam MR. MHTP, a synthetic alkaloid, attenuates combined allergic rhinitis and asthma syndrome through downregulation of the p38/ERK1/2 MAPK signaling pathway in mice. Int Immunopharmacol 2021; 96:107590. [PMID: 33857802 DOI: 10.1016/j.intimp.2021.107590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/24/2021] [Accepted: 03/11/2021] [Indexed: 01/16/2023]
Abstract
The combined allergic rhinitis and asthma syndrome (CARAS) is a chronic airway inflammation of allergic individuals, with a type 2 immune response. Pharmacotherapy is based on drugs with relevant side effects. Thus, the goal of this study was to evaluate the synthetic alkaloid, MHTP in the experimental model of CARAS. Therefore, BALB/c mice were ovalbumin (OVA) -sensitized and -challenged and treated with MHTP by intranasal or oral routes. Treated animals showed a decrease (p < 0.05) of sneezing, nasal rubbings, and histamine nasal hyperactivity. Besides, MHTP presented binding energy and favorable interaction for adequate anchoring in the histamine H1 receptor. MHTP treatment inhibited the eosinophil migration into the nasal (NALF) and the bronchoalveolar (BALF) fluids. Histological analysis showed that the alkaloid decreased the inflammatory cells in the subepithelial and perivascular regions of nasal tissue and in the peribronchiolar and perivascular regions of lung tissue. The MHTP treatment also reduced the pulmonary hyperactivity by decreasing the smooth muscle layer hypertrophy and the collagen fiber deposition in the extracellular matrix. The immunomodulatory effect of the alkaloid was due to the decrease of cytokines like IL-5 and IL-17A (type 2 and 3), TSLP (epithelial), and the immunoregulatory cytokine, TGF-β. These MHTP effects on granulocytes were dependent on the p38/ERK1/2 MAP kinase signaling pathway axis. Indeed, the synthetic alkaloid reduced the frequency of activation of both kinases independent of the NF-κB (p65) pathway indicating that the molecule shut down the intracellular transduction signals underlie the cytokine gene transcription.
Collapse
Affiliation(s)
- Laércia K D Paiva Ferreira
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Larissa A M Paiva Ferreira
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Grasiela C Bezerra Barros
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Luiz A de Araújo Silva
- Biotechnology Center, Federal University of Paraíba, Laboratory of Organic Synthesis, Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Ramon de A Pereira
- Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, MG, Brazil
| | - Pedro T R Figueiredo
- Drug Research Institute of the Federal University of Paraíba, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Adriano Francisco Alves
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Luís Cezar Rodrigues
- Biotechnology Center, Federal University of Paraíba, Laboratory of Organic Synthesis, Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Marcia Regina Piuvezam
- Laboratory of Immunopharmacology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil; Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa, PB, Brazil.
| |
Collapse
|
10
|
Liu KY, Wang LT, Wang HC, Wang SN, Tseng LW, Chai CY, Chiou SS, Huang SK, Hsu SH. Aryl Hydrocarbon Receptor is Essential in the Control of Lung Club Cell Homeostasis. J Inflamm Res 2021; 14:299-311. [PMID: 33574691 PMCID: PMC7872937 DOI: 10.2147/jir.s284800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Club cells play an important role in maintaining lung homeostasis and aryl hydrocarbon receptor (AhR) is known to be important in xenobiotic metabolism, but its role in regulating club cells is currently unknown. METHODS To this end, mice with club cell-specific AhR deficiency were generated and evaluated in a model of antigen (ovalbumin, OVA)-induced airway inflammation for the number of infiltrating inflammatory cells, the levels of cytokines and CC10 and Notch signaling by standard methods. RESULTS After OVA sensitization and challenge, Scgb1a1-Cre; Ahrflox/flox mice showed aggravated levels of pulmonary inflammation with increased levels of inflammatory cells and cytokines 1 day after challenge as compared to those seen in their littermate controls, but in contrast to the littermate controls, no significant change in the levels of CC10 and SP-D was noted in Scgb1a1-Cre; Ahrflox/flox mice. Surprisingly, 7 days after the challenge, while, as expected, wild-type mice recovered from acute inflammation, significantly increased lymphocytic infiltration was noted in Scgb1a1-Cre; Ahrflox/flox mice, suggesting their defective mechanism of recovery. Mechanistically, this was due, in part, to the decreased Notch1 signaling and expression of its downstream gene, HES5, while AhR was shown to positively regulate Notch1 expression via its transactivating activity targeting the xenobiotic response element in the promoter region of Notch1 gene. CONCLUSION Under the condition of pulmonary inflammation, AhR is critical in controlling lung club cell homeostasis via targeting Notch1 signaling and the generation of anti-inflammatory mediators.
Collapse
Affiliation(s)
- Kwei-Yan Liu
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, 518020, People’s Republic of China
| | - Li-Ting Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsueh-Chun Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan
| | - Shen-Nien Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital
- Department of Surgery, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Li-Wen Tseng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shyh-Shin Chiou
- Division of Hematology-Oncology Department of Pediatrics, Kaohsiung Medical University Hospital Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shau-Ku Huang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, 518020, People’s Republic of China
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
11
|
Chu KH, Lin SY, Chiang BL. STAT6 Pathway Is Critical for the Induction and Function of Regulatory T Cells Induced by Mucosal B Cells. Front Immunol 2021; 11:615868. [PMID: 33584704 PMCID: PMC7878545 DOI: 10.3389/fimmu.2020.615868] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
B cells could convert naïve T cells into regulatory T cells (so-called Treg-of-B cells) which have the ability to treat animal models of inflammatory diseases, including allergic asthma, collagen-induced arthritis and colitis; however, the mechanisms of Treg-of-B cell generation remain unclear. In this study, we investigated the role of STAT6 in the generation of Treg-of-B (P) cells, which Treg cells were generated by Peyer’s patch B cells (P stands for Peyer’s patch). CD4+CD25- T cells from wild type, STAT6 knockout and IL-4 knockout mice were cocultured with wild type Peyer’s patch B cells for Treg-of-B (P) cell generation. A murine asthmatic model was used to analyze the in vivo regulatory function of Treg-of-B (P) cells. The data demonstrated that STAT6 played a critical role in the generation of Treg-of-B (P) cells, which confirmed with STAT6-deficient T cells and the STAT6 inhibitor AS1517499. When STAT6 was lacking, Treg-of-B (P) cells exerted impaired suppressive ability with decreased LAG3 expression. Furthermore, Peyer’s patch B cells played an essential role in regulatory T cell generation. In the absence of Peyer’s patch B cells, T cells expressed decreased phosphorylated STAT6, which was followed by decreased LAG3 expression and impaired suppressive ability, suggesting that Peyer’s patch B cells provided the critical signal to activate STAT6 phosphorylation in T cells. Moreover, STAT6 deficient Treg-of-B (P) cells could not alleviate inflammation in an animal model of asthma in vivo. IL-4 was downstream of phosphorylated STAT6 and maintained Treg-of-B (P) cell survival with increased expression of Bcl-2 and BclXL. We reported a novel finding that the STAT6-LAG3 signaling axis is important for the induction and function of Treg-of-B (P) cells.
Collapse
Affiliation(s)
- Kuan-Hua Chu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Yu Lin
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.,Allergy Center, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Suen JL, Wu TT, Li YH, Lee CL, Kuo FC, Yan PS, Wu CF, Tran M, Wang CJ, Hung CH, Wu MT, Chan MWY, Huang SK. Environmental Factor-Mediated Transgenerational Inheritance of Igf2r Hypomethylation and Pulmonary Allergic Response via Targeting Dendritic Cells. Front Immunol 2020; 11:603831. [PMID: 33424850 PMCID: PMC7786300 DOI: 10.3389/fimmu.2020.603831] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/23/2020] [Indexed: 11/13/2022] Open
Abstract
The developmental origin of allergic diseases has been suggested, but the molecular basis remains enigmatic. Exposure to environmental factors, such as di-(2-ethylhexyl) phthalate (DEHP; a common plasticizer), is suggested to be associated with increased childhood allergic asthma, but the causal relationship and its underlying mechanism remain unknown. This study explored the transgenerational mechanism of DEHP on allergic asthma and dendritic cell (DC) homeostasis through epigenetic modification. In a murine model, ancestral exposure of C57BL/6 mice to low-dose DEHP led to trans-generational promoter hypomethylation of the insulin-like growth factor 2 receptor (Igf2r), concomitant with enhanced Igf2r expression and increased apoptosis prominently in CD8α+ DCs upon ligand stimulation, with consequent reduction in their IL-12 secretion and subsequent T cell-derived IFN-γ, thereby promoting a default Th2-associated pulmonary allergic response. Increased apoptosis was also noted in circulating IGF2Rhigh human DCs. Further, in human placenta, the methylation level at the orthologous IGF2R promoter region was shown to be inversely correlated with the level of maternal DEHP intake. These results support the importance of ancestral phthalate exposure in conferring the trans-generational risk of allergic phenotypes, featuring hypo-methylation of the IGF2R gene and dysregulated DC homeostasis.
Collapse
Affiliation(s)
- Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tai-Ting Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yue-Hyuan Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Lai Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu-Chen Kuo
- Department of Gynecology and Obstetrics, E-Da Hospital, Kaohsiung, Taiwan
- Graduate Institute of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Pearlly S. Yan
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Chia-Fang Wu
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mita Tran
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chien-Jen Wang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Hsing Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Ming-Tsang Wu
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Community Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Taiwan
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Bao H, Zhou Q, Li Q, Niu M, Chen S, Yang P, Liu Z, Xia L. Differentially expressed circular RNAs in a murine asthma model. Mol Med Rep 2020; 22:5412-5422. [PMID: 33173985 PMCID: PMC7647044 DOI: 10.3892/mmr.2020.11617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/25/2020] [Indexed: 01/14/2023] Open
Abstract
Allergic asthma is one of the most common allergic diseases; however, the mechanisms underlying its development have yet to be fully elucidated. Although allergic diseases are inheritable, genetic variance alone cannot explain the notable increase in the prevalence of allergic diseases over a short period of time in recent decades. Recently, research focus has been shifting to epigenetic factors, such as non-coding RNAs. Circular RNAs (circRNAs) are involved in the pathogenesis of various diseases. The aim of the present study was to further elucidate the etiology of allergic asthma by analyzing aberrantly expressed circRNAs in a murine asthma model. A mouse model of house dust mite allergen-induced asthma was established, and the qualified libraries were sequenced using next-generation sequencing. The expression levels of circRNAs were validated by reverse transcription-quantitative PCR (RT-qPCR) analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed for biological pathway classification and enrichment analysis of the aberrantly expressed circRNAs. In addition, the interaction network of the differentially expressed circRNAs and microRNAs (miRNAs) was constructed using Cytoscape. By next-generation sequencing, a total of 150 circRNAs were revealed to be upregulated and 130 were downregulated in the murine asthma model group compared with in the control group. GO and KEGG analyses demonstrated that the differentially expressed circRNAs were mainly involved in processes such as ‘autoimmune disease’, ‘cell adhesion molecules (CAMs)’ and ‘endocytosis’, among others. The expression levels of six circRNAs, namely three upregulated (circ_0000909, circ_0000629 and circ_0000455) and three downregulated (circ_0001454, circ_0000723 and circ_0001389) circRNAs, were validated by RT-qPCR. In conclusion, the analyses suggested that circRNAs performed critical functions via endocytosis (such as macrophage endocytosis), cell adhesion molecules and lipid metabolism in allergic asthma. The interaction network revealed that certain miRNAs that may serve a role in asthma could be regulated by the differentially expressed circRNAs.
Collapse
Affiliation(s)
- Hui Bao
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Qiuyan Zhou
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Qiuju Li
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Mengmeng Niu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Sanfeng Chen
- Department of Internal Medicine, Aged Care Hospital of Hangzhou, Hangzhou, Zhejiang 310015, P.R. China
| | - Pingchang Yang
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Zhigang Liu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Lixin Xia
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
14
|
The Potential of IgG to Induce Murine and Human Thymic Maturation of IL-10+ B Cells (B10) Revealed in a Pilot Study. Cells 2020; 9:cells9102239. [PMID: 33027887 PMCID: PMC7600151 DOI: 10.3390/cells9102239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/13/2022] Open
Abstract
Regulatory B (B10) cells can control several inflammatory diseases, including allergies; however, the origin of peripheral B10 cells is not fully understood, and the involvement of primary lymphoid organs (PLOs) as a primary site of maturation is not known. Here, using a murine model of allergy inhibition mediated by maternal immunization with ovalbumin (OVA), we aimed to evaluate whether B10 cells can mature in the thymus and whether IgG can mediate this process. Female mice were immunized with OVA, and offspring thymus, bone marrow, spleen, lung, and serum samples were evaluated at different times and after passive transfer of purified IgG or thymocytes. A translational approach was implemented using human nonatopic thymus samples, nonatopic peripheral blood mononuclear cells (PBMCs), and IgG from atopic or nonatopic individuals. Based on the expression of CD1d on B cells during maturation stages, we suggest that B10 cells can also mature in the murine thymus. Murine thymic B10 cells can be induced in vitro and in vivo by IgG and be detected in the spleen and lungs in response to an allergen challenge. Like IgG from atopic individuals, human IgG from nonatopic individuals can induce B10 cells in the infant thymus and adult PBMCs. Our observations suggest that B10 cells may mature in the thymus and that this mechanism may be mediated by IgG in both humans and mice. These observations may support the future development of IgG-based immunoregulatory therapeutic strategies.
Collapse
|
15
|
Bezerra Barros GC, Paiva Ferreira LKD, Ferreira LAMP, Mozzini Monteiro T, Alves AF, Pereira RDA, Piuvezam MR. 4-Carvomenthenol ameliorates the murine combined allergic rhinitis and asthma syndrome by inhibiting IL-13 and mucus production via p38MAPK/NF-κB signaling pathway axis. Int Immunopharmacol 2020; 88:106938. [PMID: 33182052 DOI: 10.1016/j.intimp.2020.106938] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/07/2020] [Accepted: 08/23/2020] [Indexed: 01/11/2023]
Abstract
The aim of this study was to analyze the 4-carvomenthenol (carvo) oral treatment on the experimental model of the combined allergic rhinitis and asthma syndrome (CARAS). BALB/c mice were OVA-sensitized on day zero and 7th (50 μg/mL OVA in 10 mg/mL Al (OH)3) and OVA-challenged (5 mg/mL, 20 μL/animal) for three weeks. In the last week, the animals were dally challenged with aerosol of OVA and the carvo treatment (12.5, 25 or 50 mg/kg) occurred one hour before each OVA-challenge. Data were analyzed and p < 0.05 was considered significant. Carvo (12.5-50 mg/kg) decreased significantly the eosinophil migration into the nasal (NALF) and bronchoalveolar (BALF) cavities as well as on the nasal and lung tissues of sick animals. The treatment also decreased mucus production on both tissue sections stained with PAS (periodic acid-Schiff satin). In addition, the histological analyzes demonstrated that sick mice presented hyperplasia and hypertrophy of the lung smooth muscle layer followed by increasing of extracellular matrix and carvo (50 mg/kg) inhibited these asthmatic parameters. We analyzed the allergic rhinitis signals as nasal frictions and sneezing and observed that carvo decreased these two signals as well as serum OVA-specific IgE titer, type 2 cytokine synthesis, mainly IL-13, with increasing of IL-10 production. Decreasing of IL-13 production corroborated with decreasing of mucus production and these effects were dependent on p38MAPK/NF-κB(p65) signaling pathway inhibition. Therefore, these data demonstrated that a monoterpene of essential oils presents anti-allergic property on an experimental model of CARAS suggesting a new drug prototype to treat this allergic syndrome.
Collapse
Affiliation(s)
- Grasiela Costa Bezerra Barros
- Department of Physiology and Pathology, Federal University of Paraíba, Laboratory of Immunopharmacology, João Pessoa, PB, Brazil
| | - Laércia K D Paiva Ferreira
- Department of Physiology and Pathology, Federal University of Paraíba, Laboratory of Immunopharmacology, João Pessoa, PB, Brazil
| | - Larissa A M P Ferreira
- Department of Physiology and Pathology, Federal University of Paraíba, Laboratory of Immunopharmacology, João Pessoa, PB, Brazil
| | | | - Adriano Francisco Alves
- Department of Physiology and Pathology, Federal University of Paraíba, Laboratory of Pathology, João Pessoa, PB, Brazil
| | - Ramon de Alencar Pereira
- Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, MG, Brazil
| | - Marcia Regina Piuvezam
- Department of Physiology and Pathology, Federal University of Paraíba, Laboratory of Immunopharmacology, João Pessoa, PB, Brazil.
| |
Collapse
|
16
|
Wang B, Hu J, Liu Y, Liu Q, Li D. Food allergy promotes a Th2/Th17 response that drives house dust mite-induced allergic airway inflammation in humanized mice. Clin Exp Immunol 2020; 202:300-307. [PMID: 32757273 DOI: 10.1111/cei.13504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 02/05/2023] Open
Abstract
Food allergy is related to increasing risk of the development of allergic asthma, but the precise interplay between sensitization to different allergens in different compartments of the body is not fully understood. The aim of this study was to develop a novel humanized murine model of mixed food and respiratory allergy that recapitulates the human anaphylactic response and to more clearly understand the impact of food allergies on asthma. Immunodeficient mice transferred with peripheral blood mononuclear cells (PBMCs) from donors with peanut and house dust mite (HDM) allergy were exposed and challenged to peanut. Between peanut exposure and challenge, mice were intranasally treated to HDM. Allergic parameters were analyzed. Allergen-specific immunoglobulin (Ig)E in sera could only be measured in mice treated with peripheral blood mononuclear cells (PBMCs) plus allergen. A preceding peanut exposure increased IgE levels, histamine release, bronchial hyper-responsiveness and lung inflammation. Recruitment of inflammatory cells to the airways was aggravated associated with an enhanced T helper type 2 (Th2)/Th17 cytokine secretion when the two allergies were present. A preceding peanut exposure amplifies allergic asthma in this humanized model, which may contribute to the understanding of underlying immunological mechanism of polysensitization occurring in allergic individuals and evaluation of therapeutic interventions.
Collapse
Affiliation(s)
- B Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - J Hu
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Y Liu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Q Liu
- China Resources Healthcare College, Beijing, China
| | - D Li
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Lee S, Han EH, Lim MK, Lee SH, Yu HJ, Lim YH, Kang S. Fermented Platycodon grandiflorum Extracts Relieve Airway Inflammation and Cough Reflex Sensitivity In Vivo. J Med Food 2020; 23:1060-1069. [PMID: 32758004 DOI: 10.1089/jmf.2019.4595] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Platycodon grandiflorum (PG) has been extensively utilized as an herb to relieve phlegm. In this study, the effects of PG root extracts on airway inflammation and cough reflex were investigated, especially using fermented PG extracts (FPE) to increase an active compound, platycodin D by fermentation. FPE significantly reduced the numbers of eosinophils and total cells in the bronchoalveolar lavage fluid (BALF) obtained from lipopolysaccharide/ovalbumin (LPS/OVA)-induced asthma mice versus those of vehicle control. Moreover, in the BALF and the serum, FPE significantly reduced the concentration of IL-17E, a proinflammatory cytokine that causes TH2 immunity, including eosinophil amplification. It was also demonstrated that FPE might relieve inflammations through histological analysis of the lung separated from each mouse. Furthermore, in cough reflex guinea pigs induced by citric acid treatment, FPE treatment significantly reduced the number of coughs versus that of vehicle control, and consequently decreased cough reflex sensitivity. In addition, the total cell number and eosinophils significantly decreased in the BALF obtained from each guinea pig versus that of vehicle control. In in vitro study, pretreatment with FPE in LPS-stimulated RAW264.7 cells significantly reduced the levels of proinflammatory cytokines such as TNF-α, IL-6, and IL-1β, and inducible nitric oxide synthases (iNOS). Therefore, we demonstrated that FPE relieved airway inflammation and cough reflex sensitivity in vivo, and exhibited anti-inflammatory effects through suppression of iNOS and several proinflammatory cytokines. These findings suggest that FPE might have a beneficial effect on respiratory health, and may be useful as a functional food to prevent respiratory diseases.
Collapse
Affiliation(s)
- Soyeon Lee
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.,Department of Research & Development, Koreaeundan Healthcare Co. Ltd., Ansan, Gyeonggido, Korea
| | - Eun Hye Han
- Department of Research & Development, Koreaeundan Healthcare Co. Ltd., Ansan, Gyeonggido, Korea.,Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul, Korea
| | - Mi-Kyung Lim
- Department of Research & Development, Koreaeundan Co., Seongnam, Gyeonggido, Korea
| | - Sang-Ho Lee
- Department of Research & Development, Koreaeundan Healthcare Co. Ltd., Ansan, Gyeonggido, Korea.,Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul, Korea
| | - Heui Jong Yu
- Research & Development Center, SKbioland Co., Ltd., Ansan, Gyeonggido, Korea
| | - Young Hee Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul, Korea
| | - Seongman Kang
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| |
Collapse
|
18
|
Kolar QK, Waddell LA, Raper A, Rocchi MS, Shaw DJ, Corbishley A, Hope JC. Anatomical distribution of respiratory tract leukocyte cell subsets in neonatal calves. Vet Immunol Immunopathol 2020; 227:110090. [PMID: 32663724 PMCID: PMC7331561 DOI: 10.1016/j.vetimm.2020.110090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/09/2020] [Accepted: 07/01/2020] [Indexed: 11/28/2022]
Abstract
Neonatal calves are highly susceptible to a number of diseases including those that infect via the mucosal surfaces of the respiratory and gastrointestinal tracts. In order to determine appropriate vaccine design and delivery systems, or to identify suitable immunostimulatory methods to combat these infections, a detailed understanding of the immune cell populations present at clinically relevant sites is key. Few studies have assessed the immune cell composition of the neonatal calf lung and comparisons with circulating immune cells in the blood are lacking. We describe immune cell populations present in the peripheral blood, bronchoalveolar lavage (BAL) fluid and lung tissue of young disease-free calves. Flow cytometric analysis revealed significant differences in cell subset distribution between the peripheral blood and respiratory tract, and between compartments within the respiratory tract. Notably, whereas WC1+ γδ TCR + T lymphocytes dominate the peripheral blood, both the BAL fluid and lung tissue contained a high proportion of myeloid cells which expressed CD14 and CD172a (SIRPα). Very low numbers of tissue myeloid cells expressed MHC Class II in comparison to circulating myeloid cells in the blood. Respiratory tract tissues had low frequencies of CD4+ and CD8 + T lymphocytes, which were significantly lower than in the blood. Differences in the proportion of NKp46+ natural killer cells were also observed between tissue compartments. In order to target vaccines or immunostimulatory therapeutics appropriately, these differences in immune cell populations in tissue compartments should be taken into consideration.
Collapse
Affiliation(s)
- Quinn K Kolar
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom
| | - Lindsey A Waddell
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom
| | - Anna Raper
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom
| | - Mara S Rocchi
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, EH260PZ, United Kingdom
| | - Darren J Shaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom
| | - Alexander Corbishley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom
| | - Jayne C Hope
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, EH259RG, United Kingdom.
| |
Collapse
|
19
|
de‐Oliveira MG, Lira AAL, Sgnotto FR, Inoue AHS, Santos LS, Nakamatsu BY, Duarte AJS, Leite‐de‐Moraes M, Victor JR. Maternal IgG impairs the maturation of offspring intrathymic IL‐17‐producing γδT cells: Implications for murine and human allergies. Clin Exp Allergy 2019; 49:1000-1012. [DOI: 10.1111/cea.13393] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/20/2019] [Accepted: 03/23/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Marília G. de‐Oliveira
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
| | - Aline A. L. Lira
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
| | - Fábio R. Sgnotto
- Division of Hematology, Medical School University of Sao Paulo Sao Paulo Brazil
| | - Amanda H. S. Inoue
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
| | - Ludimila S. Santos
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
- Division of Environmental Health Faculdades Metropolitanas Unidas (FMU), Laureate International Universities Sao Paulo Brazil
| | - Bernardo Y. Nakamatsu
- Division of Environmental Health Faculdades Metropolitanas Unidas (FMU), Laureate International Universities Sao Paulo Brazil
| | - Alberto J. S. Duarte
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
- Division of Pathology, Medical School University of Sao Paulo Sao Paulo Brazil
| | - Maria Leite‐de‐Moraes
- Laboratory of Immunoregulation and Immunopathology INEM (Institut Necker‐Enfants Malades), CNRS UMR8253, INSERM UMR1151, Université Paris Descartes Paris France
| | - Jefferson R. Victor
- Laboratory of Medical Investigation LIM 56, Division of Clinical Dermatology, Medical School University of Sao Paulo Sao Paulo Brazil
- Division of Environmental Health Faculdades Metropolitanas Unidas (FMU), Laureate International Universities Sao Paulo Brazil
| |
Collapse
|
20
|
Magnusson P, Dziendzikowska K, Oczkowski M, Øvrevik J, Eide DM, Brunborg G, Gutzkow KB, Instanes C, Gajewska M, Wilczak J, Sapierzynski R, Kamola D, Królikowski T, Kruszewski M, Lankoff A, Mruk R, Duale N, Gromadzka-Ostrowska J, Myhre O. Lung effects of 7- and 28-day inhalation exposure of rats to emissions from 1st and 2nd generation biodiesel fuels with and without particle filter - The FuelHealth project. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 67:8-20. [PMID: 30685595 DOI: 10.1016/j.etap.2019.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/22/2018] [Accepted: 01/16/2019] [Indexed: 06/09/2023]
Abstract
Increased use of 1st and 2nd generation biofuels raises concerns about health effects of new emissions. We analyzed cellular and molecular lung effects in Fisher 344 rats exposed to diesel engine exhaust emissions (DEE) from a Euro 5-classified diesel engine running on B7: petrodiesel fuel containing 7% fatty acid methyl esters (FAME), or SHB20 (synthetic hydrocarbon biofuel): petrodiesel fuel containing 7% FAME and 13% hydrogenated vegetable oil. The Fisher 344 rats were exposed for 7 consecutive days (6 h/day) or 28 days (6 h/day, 5 days/week), both with and without diesel particle filter (DPF) treatment of the exhaust in whole body exposure chambers (n = 7/treatment). Histological analysis and analysis of cytokines and immune cell numbers in bronchoalveolar lavage fluid (BALF) did not reveal adverse pulmonary effects after exposure to DEE from B7 or SHB20 fuel. Significantly different gene expression levels for B7 compared to SHB20 indicate disturbed redox signaling (Cat, Hmox1), beta-adrenergic signaling (Adrb2) and xenobiotic metabolism (Cyp1a1). Exhaust filtration induced higher expression of redox genes (Cat, Gpx2) and the chemokine gene Cxcl7 compared to non-filtered exhaust. Exposure time (7 versus 28 days) also resulted in different patterns of lung gene expression. No genotoxic effects in the lungs were observed. Overall, exposure to B7 or SHB20 emissions suggests only minor effects in the lungs.
Collapse
Affiliation(s)
- Pål Magnusson
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | | | - Michał Oczkowski
- Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences, Poland
| | - Johan Øvrevik
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | - Dag M Eide
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | - Gunnar Brunborg
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | - Kristine B Gutzkow
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | - Christine Instanes
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | | | - Jacek Wilczak
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Poland
| | - Rafał Sapierzynski
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Poland
| | - Dariusz Kamola
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Poland
| | - Tomasz Królikowski
- Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland; Department of Molecular Biology and Translational Research, Institute of Rural Health, Lublin, Poland
| | - Anna Lankoff
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland; Jan Kochanowski University, Kielce, Poland
| | - Remigiusz Mruk
- Faculty of Production Engineering, Warsaw University of Life Sciences, Poland
| | - Nur Duale
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway
| | | | - Oddvar Myhre
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Norway.
| |
Collapse
|
21
|
Ng B, Cash-Mason T, Wang Y, Seitzer J, Burchard J, Brown D, Dudkin V, Davide J, Jadhav V, Sepp-Lorenzino L, Cejas PJ. Intratracheal Administration of siRNA Triggers mRNA Silencing in the Lung to Modulate T Cell Immune Response and Lung Inflammation. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:194-205. [PMID: 30901578 PMCID: PMC6426712 DOI: 10.1016/j.omtn.2019.02.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 01/07/2023]
Abstract
Clinical application of siRNA-based therapeutics outside of the liver has been hindered by the inefficient delivery of siRNA effector molecules into extra-hepatic organs and cells of interest. To understand the parameters that enable RNAi activity in vivo, it is necessary to develop a systematic approach to identify which cells within a tissue are permissive to oligonucleotide internalization and activity. In the present study, we evaluate the distribution and activity within the lung of chemically stabilized siRNA to characterize cell-type tropism and structure-activity relationship. We demonstrate intratracheal delivery of fully modified siRNA for RNAi-mediated target knockdown in lung CD11c+ cells (dendritic cells, alveolar macrophages) and alveolar epithelial cells. Finally, we use an allergen-induced model of lung inflammation to demonstrate the capacity of inhaled siRNA to induce target knockdown in dendritic cells and ameliorate lung pathology.
Collapse
Affiliation(s)
- Bruce Ng
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Tanesha Cash-Mason
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Yi Wang
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Jessica Seitzer
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Julja Burchard
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Duncan Brown
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Vadim Dudkin
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Joseph Davide
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | - Vasant Jadhav
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA
| | | | - Pedro J Cejas
- Department of RNA Therapeutics, Merck & Co., Inc., West Point, PA 19486, USA; Department of Infectious Diseases and Vaccines, Merck & Co., Inc., West Point, PA 19486, USA.
| |
Collapse
|
22
|
Paiva Ferreira LKD, Paiva Ferreira LAM, Alves AF, Leite FC, de Araújo Silva LA, Vieira GC, Rodrigues LC, Piuvezam MR. MHTP, 2-Methoxy-4-(7-methoxy-1,2,3,4-tetrahydroisoquinolin-1-yl) phenol, a Synthetic Alkaloid, Induces IFN-γ Production in Murine Model of Ovalbumin-Induced Pulmonary Allergic Inflammation. Inflammation 2019; 41:2116-2128. [PMID: 30032472 DOI: 10.1007/s10753-018-0855-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MHTP [2-methoxy-4-(7-methoxy-1,2,3,4-tetrahydroisoquinolin-1-yl) phenol], a synthetic isoquinolinic alkaloid, presented anti-inflammatory activity in several experimental models of acute inflammation as lipopolysaccharide (LPS)-induced acute lung injury and phlogistic agent-induced edema and presented low preclinical toxicity. The aim of this study was to determine the MHTP effect on ovalbumin (OVA)-induced pulmonary allergic inflammation. In other to realize this study, female BALFB/c mice were sensitized and challenged with OVA (OVA group) and treated with MHTP (MHTP group) by nasal instillation. Inflammatory, allergic, and immunomodulatory parameters such as migration of inflammatory cells to the lung tissue, pulmonary histological analysis, serum level of IgE-allergen specific, cytokine secretion, and lung T cell population characterization were analyzed and the data were considered statistically significant with p < 0.05. OVA-sensitized and OVA-challenged and MHTP (5.0 mg/kg)-treated mice presented reduction on total leukocyte migration into the bronchoalveolar lavage (BALF) dependent of lymphocyte and eosinophil migration (p < 0.001 and p < 0.01) as compared with the OVA group. Flow cytometric analysis showed that MHTP treatment decreased the percentage of granulocytes (p < 0.001) into the BALF and lung tissue histological analyzes demonstrated that the MHTP treatment decreased leukocyte migration and mucus production. In addition, treatment with MHTP decreased the number of CD3+CD4+ T cells independently of CD8+ T cell reduction into the BALF. The treatment also reduced significantly (p < 0.05) the serum level of IgE-OVA specific followed by reduction of IL-4, IL-13, and IL-17 production. Surprisingly, the MHTP treatment increased significantly (p < 0.05) the IFN-γ production in the BALF of these animals. Therefore, the results presented here showed that MHTP treatment, by nasal instillation, in a mouse model of OVA-induced pulmonary allergy has anti-allergic and immunomodulatory effects dependent on a Th1-skewed cytokine production that ameliorate the pulmonary allergic inflammation.
Collapse
Affiliation(s)
- Laércia K D Paiva Ferreira
- Department of Physiology and Pathology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Larissa A M Paiva Ferreira
- Department of Physiology and Pathology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Adriano Francisco Alves
- Department of Physiology and Pathology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Luiz A de Araújo Silva
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Giciane Carvalho Vieira
- Department of Morphology, Health Sciences Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Luís Cezar Rodrigues
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Marcia Regina Piuvezam
- Department of Physiology and Pathology, Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa, PB, Brazil.
- Universidade Federal da Paraíba, Campus I, PO 5009, João Pessoa, PB, 58-051-970, Brazil.
| |
Collapse
|
23
|
Ieronymaki E, Theodorakis EM, Lyroni K, Vergadi E, Lagoudaki E, Al-Qahtani A, Aznaourova M, Neofotistou-Themeli E, Eliopoulos AG, Vaporidi K, Tsatsanis C. Insulin Resistance in Macrophages Alters Their Metabolism and Promotes an M2-Like Phenotype. THE JOURNAL OF IMMUNOLOGY 2019; 202:1786-1797. [PMID: 30718296 DOI: 10.4049/jimmunol.1800065] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
Obesity and insulin resistance influences metabolic processes, but whether it affects macrophage metabolism is not known. In this study, we demonstrate that chronic exposure of macrophages to insulin either in culture or in vivo in diet-induced, glucose-intolerant mice rendered them resistant to insulin signals marked by failure to induce Akt2 phosphorylation. Similarly, macrophages lacking Akt2 or IGF1 receptor were also resistant to insulin signals. Insulin-resistant macrophages had increased basal mTORC1 activity, possessed an M2-like phenotype, and reduced LPS responses. Moreover, they exhibited increased glycolysis and increased expression of key glycolytic enzymes. Inhibition of mTORC1 reversed the M2-like phenotype and suppressed glycolysis in insulin-resistant macrophages. In the context of polymicrobial sepsis, mice harboring insulin-resistant macrophages exhibited reduced sepsis-induced lung injury. Thus, macrophages obtain resistance to insulin characterized by increased glycolysis and a unique M2-like phenotype, termed M-insulin resistant, which accounts for obesity-related changes in macrophage responses and a state of trained immunity.
Collapse
Affiliation(s)
- Eleftheria Ieronymaki
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Emmanouel M Theodorakis
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Konstantina Lyroni
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Eleni Vergadi
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| | - Eleni Lagoudaki
- Laboratory of Pathology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Ahmed Al-Qahtani
- Department of Infection and Immunity, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; and
| | - Marina Aznaourova
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Elpida Neofotistou-Themeli
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Aristides G Eliopoulos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece.,Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Katerina Vaporidi
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece; .,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 71110 Crete, Greece
| |
Collapse
|
24
|
van Rijt LS, Logiantara A, Canbaz D, van Ree R. Birch pollen-specific subcutaneous immunotherapy reduces ILC2 frequency but does not suppress IL-33 in mice. Clin Exp Allergy 2018; 48:1402-1411. [PMID: 30126007 DOI: 10.1111/cea.13254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/14/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The underlying mechanism of allergen-specific subcutaneous immunotherapy (SCIT) is not yet fully understood, but suppression of allergen-specific Th2 cells and production of allergen-specific IgG4 antibodies are two hallmarks. The impact on the innate arm of the immune system is far less clear. OBJECTIVE The aim of this study was to investigate the effect of birch pollen (BP) SCIT on the innate immune response in a BP SCIT mouse model. METHODS Mice with birch pollen-induced allergic airway inflammation received weekly subcutaneous immunotherapy injections with birch pollen extract (BPE) adsorbed to alum. The effect of the BP SCIT on innate cytokine levels in lung, the number and the functionality of ILC2s and the airway inflammation was determined. RESULTS Mice with BP allergy had an increased level of the innate cytokines IL-33, IL-25, GM-CSF and IL-5+ ILC2s in the lungs. BP SCIT suppressed the number of IL-5+ ILC2s, mast cell tryptase release, Th2 cytokine production, eosinophil recruitment and peribronchial inflammatory infiltrates. In contrast, innate cytokine production and collagen deposition in the airways were not affected. CONCLUSION AND CLINICAL RELEVANCE BP SCIT is able to suppress the adaptive and part of the innate immune response, but this is not sufficient to inhibit collagen deposition and the IL-33 expression in the airways in mice.
Collapse
Affiliation(s)
- Leonie S van Rijt
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Adrian Logiantara
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Derya Canbaz
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Otorhinolaryngology, Amsterdam UMC, location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Kawa Y, Nagano T, Yoshizaki A, Dokuni R, Katsurada M, Terashita T, Yasuda Y, Umezawa K, Yamamoto M, Kamiryo H, Kobayashi K, Nishimura Y. Role of S1P/S1PR3 axis in release of CCL20 from human bronchial epithelial cells. PLoS One 2018; 13:e0203211. [PMID: 30192865 PMCID: PMC6128515 DOI: 10.1371/journal.pone.0203211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/16/2018] [Indexed: 11/18/2022] Open
Abstract
Background Sphingosine kinase phosphorylates sphingosine to generate sphingosine 1 phosphate (S1P) following stimulation of the five plasma membrane G-protein-coupled receptors. The objective of this study is to clarify the role of S1P and its receptors (S1PRs), especially S1PR3 in airway epithelial cells. Methods The effects of S1P on asthma-related genes expression were examined with the human bronchial epithelial cells BEAS-2B and Calu-3 using a transcriptome analysis and siRNA of S1PRs. To clarify the role of CCL20 in the airway inflammation, BALB/c mice were immunized with ovalbumin (OVA) and subsequently challenged with an OVA-containing aerosol to induce asthma with or without intraperitoneal administration of anti-CCL20. Finally, the anti-inflammatory effect of VPC 23019, S1PR1/3 antagonist, in the OVA-induced asthma was examined. Results S1P induced the expression of some asthma-related genes, such as ADRB2, PTGER4, and CCL20, in the bronchial epithelial cells. The knock-down of SIPR3 suppressed the expression of S1P-inducing CCL20. Anti-CCL20 antibody significantly attenuated the eosinophil numbers in the bronchoalveolar lavage fluid (P<0.01). Upon OVA challenge, VPC23019 exhibited substantially attenuated eosinophilic inflammation. Conclusions S1P/S1PR3 pathways have a role in release of proinflammatory cytokines from bronchial epithelial cells. Our results suggest that S1P/S1PR3 may be a possible candidate for the treatment of bronchial asthma.
Collapse
Affiliation(s)
- Yoshitaka Kawa
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
- * E-mail:
| | - Asuka Yoshizaki
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Ryota Dokuni
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Masahiro Katsurada
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Tomomi Terashita
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Yuichiro Yasuda
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Kanoko Umezawa
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Hiroshi Kamiryo
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Japan
| |
Collapse
|
26
|
Debelleix S, Siao-Him Fa V, Begueret H, Berger P, Kamaev A, Ousova O, Marthan R, Fayon M. Montelukast reverses airway remodeling in actively sensitized young mice. Pediatr Pulmonol 2018; 53:701-709. [PMID: 29493871 DOI: 10.1002/ppul.23980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/10/2018] [Indexed: 12/22/2022]
Abstract
UNLABELLED Asthma is characterized by airway hyperresponsiveness (AHR) and inflammation leading to airway remodeling (AR). In children, AR may occur very early prior to the age of 6 years. Treatments to prevent or reverse AR are unknown. AIM We sought to determine (i) whether short allergenic sensitization at a young age in a mouse model may induce enhanced AR and inflammation compared to adults; (ii) the effect of Montelukast on such AR. METHODS Immature and adult Balb/c mice were sensitized and challenged with ovalbumin. AHR and AR were measured using cultured precision-cut lung slices and inflammation by bronchoalveolar lavage. Experiments were repeated after administration of Montelukast. RESULTS OVA-challenged mice developed AHR to methacholine regardless of age of first exposure to OVA. Young mice developed greater thickened basement membrane, increased smooth muscle mass, and increased area of bronchovascular fibrosis compared with adult mice. Cellular infiltrates in BAL differed depending upon animal age at first exposure with higher eosinophilia measured in younger animals. Montelukast decreased ASM mass, BAL cellularity. CONCLUSION We provide thus evidence for a greater degree of AR after allergenic sensitization and challenge in younger mice versus adults. This study provides proof of concept that airway remodeling can be prevented and reversed in this case by anti-asthmatic drug Montelukast in this model.
Collapse
Affiliation(s)
- Stephane Debelleix
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Valérie Siao-Him Fa
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Hugues Begueret
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Patrick Berger
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Andy Kamaev
- Department of general practice, Pavlov First Saint-Petersburg State Medical University, St. Petersburg, Russia
| | - Olga Ousova
- Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Roger Marthan
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Michael Fayon
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,Inserm, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,CHU de Bordeaux, Service de Pneumo-Pédiatrie, Service d'anatomopathologie, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| |
Collapse
|
27
|
Lin X, Zhuang X, Li C, Wang X. Interactions between dendritic cells and T lymphocytes in pathogenesis of nasal polyps. Exp Ther Med 2018; 15:5167-5172. [PMID: 29904400 PMCID: PMC5996678 DOI: 10.3892/etm.2018.6128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/19/2018] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the functional status of dendritic cells (DCs) in nasal polyps (NP) and their interactions with T lymphocytes. The interactions between DC and T lymphocytes in the pathogenesis of NP was also studied. The expression of cluster of differentiation (CD)1a and CD83 in NP was detected using immunohistochemistry and the ratio of CD83 DC/CD1a+DC was counted. The distribution of DCs in NP and normal inferior turbinate mucosa (nITM) was evaluated using double immunostaining (CD1a/CD40) and low illumination fluorescence microscopy. The number of CD1a+ cells, CD83+ cells and CD1a/CD40-dual positive cells in was significantly higher in NP tissues compared with nITM. Furthermore, the density of DCs observed in NP was significantly greater than that observed in nITM. The ratio of CD83 DC/CD1a+DC in NP was significantly higher compared with in nITM tissues. The results of the present study revealed significant infiltration of DCs in NP, with the majority being mature DCs. DCs are able to interact with T cells via the CD40/CD40L costimulatory factor, thus serving an important role in the development and progression of NP.
Collapse
Affiliation(s)
- Xinsheng Lin
- Department of Otolaryngology, Head and Neck Surgery, Shantou Central Hospital, Shantou, Guangdong 515031, P.R. China
| | - Xiayan Zhuang
- Department of Otolaryngology, Head and Neck Surgery, Shantou Central Hospital, Shantou, Guangdong 515031, P.R. China
| | - Chuangwei Li
- Department of Otolaryngology, Head and Neck Surgery, Shantou Central Hospital, Shantou, Guangdong 515031, P.R. China
| | - Xin Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
28
|
Wong TH, Lee CL, Su HH, Lee CL, Wu CC, Wang CC, Sheu CC, Lai RS, Leung SY, Lin CC, Wei YF, Wang CJ, Lin YC, Chen HL, Huang MS, Yen JH, Huang SK, Suen JL. A prominent air pollutant, Indeno[1,2,3-cd]pyrene, enhances allergic lung inflammation via aryl hydrocarbon receptor. Sci Rep 2018; 8:5198. [PMID: 29581487 PMCID: PMC5979946 DOI: 10.1038/s41598-018-23542-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/14/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic exposure to ambient polycyclic aromatic hydrocarbons (PAHs) is associated with asthma, but its regulatory mechanisms remain incompletely defined. We report herein that elevated levels of urinary 1-hydroxypyrene, a biomarker of PAH exposure, were found in asthmatic subjects (n = 39) as compared to those in healthy subjects (n = 43) living in an industrial city of Taiwan, where indeno[1,2,3-cd]pyrene (IP) was found to be a prominent PAH associated with ambient PM2.5. In a mouse model, intranasal exposure of mice with varying doses of IP significantly enhanced antigen-induced allergic inflammation, including increased airway eosinophilia, Th2 cytokines, including IL-4 and IL-5, as well as antigen-specific IgE level, which was absent in dendritic cell (DC)-specific aryl hydrocarbon receptor (AhR)-null mice. Mechanistically, IP treatment significantly altered DC's function, including increased level of pro-inflammatory IL-6 and decreased generation of anti-inflammatory IL-10. The IP's effect was lost in DCs from mice carrying an AhR-mutant allele. Taken together, these results suggest that chronic exposure to environmental PAHs may pose a significant risk for asthma, in which IP, a prominent ambient PAH in Taiwan, was shown to enhance the severity of allergic lung inflammation in mice through, at least in part, its ability in modulating DC's function in an AhR-dependent manner.
Collapse
Affiliation(s)
- Tzu-Hsuan Wong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chon-Lin Lee
- Department of Marine Environment and Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiang-Han Su
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Lai Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chao-Chien Wu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chau-Chyun Sheu
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Divison of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ruay-Sheng Lai
- Division of Chest Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Sum-Yee Leung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| | - Chi-Cheng Lin
- Chest Division, Department of Internal Medicine, Antai Medical Care Cooperation Antai Tian-Sheng Memorial Hospital, Ping-Tung, Taiwan
| | - Yu-Feng Wei
- Division of Chest Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chien-Jen Wang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Chun Lin
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Hua-Ling Chen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Ming-Shyan Huang
- Divison of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jeng-Hsien Yen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shau-Ku Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
- Lou-Hu Hospital, Shen-Zhen University, Shen-Zhen, China
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jau-Ling Suen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
29
|
Damle SR, Martin RK, Cockburn CL, Lownik JC, Carlyon JA, Smith AD, Conrad DH. ADAM10 and Notch1 on murine dendritic cells control the development of type 2 immunity and IgE production. Allergy 2018; 73:125-136. [PMID: 28745029 DOI: 10.1111/all.13261] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Allergy and allergic asthma are significant health burdens in developed countries and are increasing in prevalence. Dendritic cells (DCs) initiate immune responses to common aeroallergens, and ADAM10 has been demonstrated to be important for the development of adaptive responses. This study's objective was to understand the role of ADAM10 on DCs in the development of allergic and anaphylactic responses. METHODS In this study, we used mouse models of allergic airway inflammation (house dust mice and Alternaria alternata) and OVA-induced models of active anaphylaxis to determine the DC-specific function of ADAM10 and Notch signaling. To examine TH 1 and TH 17 immunity infection with Anaplasma phagocytophilum and Citrobacter rodentium respectively, were used. RESULTS Mice, which have ADAM10 deleted from DCs, have dramatic reductions in IgE production and do not develop significant TH 2 immune responses. Further, ADAM10DC-/- mice are resistant to IgE-mediated anaphylaxis. This response is selective for TH 2 immunity as TH 1 and TH 17 immunity is largely unaffected. Notch1, a known ADAM10 substrate, when knocked out of DCs (Notch1DC-/- ) demonstrated a similar reduction in anaphylaxis and IgE. Without ADAM10 and Notch1 signaling, DCs were unable to make cytokines that stimulate TH 2 cells and cytokines. Anaphylaxis and allergic lung inflammation were restored in ADAM10DC-/- with the overexpression of the Notch1-intracellular domain, confirming the role of Notch signaling. CONCLUSIONS Targeting ADAM10 and Notch1 on DCs represent a novel strategy for modulating TH 2 immune responses and IgE production.
Collapse
Affiliation(s)
- S. R. Damle
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - R. K. Martin
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - C. L. Cockburn
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - J. C. Lownik
- Center for Clinical and Translational Research Virginia Commonwealth University Richmond VA USA
| | - J. A. Carlyon
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| | - A. D. Smith
- United States Department of Agriculture Diet, Genomics, and Immunology Laboratory, Agricultural Research Service Beltsville Human Nutrition Research Center Beltsville MD USA
| | - D. H. Conrad
- Department of Microbiology and Immunology Virginia Commonwealth University Richmond VA USA
| |
Collapse
|
30
|
Uemura Y, Hagiwara K, Kobayashi K. The intratracheal administration of locked nucleic acid containing antisense oligonucleotides induced gene silencing and an immune-stimulatory effect in the murine lung. PLoS One 2017; 12:e0187286. [PMID: 29107995 PMCID: PMC5673232 DOI: 10.1371/journal.pone.0187286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Locked nucleic acid containing antisense oligonucleotides (LNA-ASOs) have the potential to modulate the disease-related gene expression by the RNaseH-dependent degradation of mRNAs. Pulmonary drug delivery has been widely used for the treatment of lung disease. Thus, the inhalation of LNA-ASOs is expected to be an efficient therapy that can be applied to several types of lung disease. Because the lung has a distinct immune system against pathogens, the immune-stimulatory effect of LNA-ASOs should be considered for the development of novel inhaled LNA-ASOs therapies. However, there have been no reports on the relationship between knock-down (KD) and the immune-stimulatory effects of inhaled LNA-ASOs in the lung. In this report, LNA-ASOs targeting Scarb1 (Scarb1-ASOs) or negative control LNA-ASOs targeting ApoB (ApoB-ASOs) were intratracheally administered to mice to investigate the KD of the gene expression and the immune-stimulatory effects in the lung. We confirmed that the intratracheal administration of Scarb1-ASOs exerted a KD effect in the lung without a drug delivery system. On the other hand, both Scarb1-ASOs and ApoB-ASOs induced neutrophilic infiltration in the alveoli and increased the expression levels of G-CSF and CXCL1 in the lung. The dose required for KD was the same as the dose that induced the neutrophilic immune response. In addition, in our in vitro experiments, Scarb1-ASOs did not increase the G-CSF or CXCL1 expression in primary lung cells, even though Scarb1-ASOs exerted a strong KD effect. Hence, we hypothesize that inhaled LNA-ASOs have the potential to exert a KD effect in the lung, but that they may be associated with a risk of immune stimulation. Further studies about the mechanism underlying the immune-stimulatory effect of LNA-ASOs is necessary for the development of novel inhaled LNA-ASO therapies.
Collapse
Affiliation(s)
- Yasunori Uemura
- Immunology & Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Shizuoka, Japan
- * E-mail:
| | - Kenji Hagiwara
- Innovative Technology Labs, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Machida, Tokyo, Japan
| | - Katsuya Kobayashi
- Immunology & Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Shizuoka, Japan
| |
Collapse
|
31
|
Bandoła J, Richter C, Ryser M, Jamal A, Ashton MP, von Bonin M, Kuhn M, Dorschner B, Alexopoulou D, Navratiel K, Roeder I, Dahl A, Hedrich CM, Bonifacio E, Brenner S, Thieme S. Neurotrophin Receptor p75NTR Regulates Immune Function of Plasmacytoid Dendritic Cells. Front Immunol 2017; 8:981. [PMID: 28861085 PMCID: PMC5562693 DOI: 10.3389/fimmu.2017.00981] [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: 04/28/2017] [Accepted: 07/31/2017] [Indexed: 11/16/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) regulate innate and adaptive immunity. Neurotrophins and their receptors control the function of neuronal tissue. In addition, they have been demonstrated to be part of the immune response but little is known about the effector immune cells involved. We report, for the first time, the expression and immune-regulatory function of the low affinity neurotrophin receptor p75 neurotrophin receptor (p75NTR) by the antigen-presenting pDCs, mediated by toll-like receptor (TLR) 9 activation and differential phosphorylation of interferon regulatory factor 3 and 7. The modulation of p75NTR on pDCs significantly influences disease progression of asthma in an ovalbumin-induced mouse model mediated by the TLR9 signaling pathway. p75NTR activation of pDCs from patients with asthma increased allergen-specific T cell proliferation and cytokine secretion in nerve growth factor concentration-dependent manner. Further, p75NTR activation of pDCs delayed the onset of autoimmune diabetes in RIP-CD80GP mice and aggravated graft-versus-host disease in a xenotransplantation model. Thus, p75NTR signaling on pDCs constitutes a new and critical mechanism connecting neurotrophin signaling and immune response regulation with great therapeutic potential for a variety of immune disorders.
Collapse
Affiliation(s)
- Joanna Bandoła
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany
| | - Cornelia Richter
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany
| | - Martin Ryser
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany
| | - Arshad Jamal
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany.,Department of Medical Laboratory Sciences, Imperial College of Business Studies, Lahore, Pakistan
| | - Michelle P Ashton
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universitaet Dresden, Dresden, Germany
| | - Malte von Bonin
- Medical Clinic I, University Clinic Dresden, Dresden, Germany.,DKTK-German Cancer Consortium, Partner Site Dresden, University Clinic Dresden, Dresden, Germany.,DKFZ-German Cancer Research Center, Heidelberg, Germany
| | - Matthias Kuhn
- Faculty of Medicine, Institute for Medical Informatics and Biometry, Technische Universitaet Dresden, Dresden, Germany
| | | | - Dimitra Alexopoulou
- BIOTEChnology Center/DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universitaet Dresden, Dresden, Germany
| | - Katrin Navratiel
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany
| | - Ingo Roeder
- Faculty of Medicine, Institute for Medical Informatics and Biometry, Technische Universitaet Dresden, Dresden, Germany
| | - Andreas Dahl
- BIOTEChnology Center/DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universitaet Dresden, Dresden, Germany
| | | | - Ezio Bonifacio
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universitaet Dresden, Dresden, Germany
| | - Sebastian Brenner
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany.,DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universitaet Dresden, Dresden, Germany
| | - Sebastian Thieme
- Department of Pediatrics, University Clinic Dresden, Dresden, Germany
| |
Collapse
|
32
|
Magnusson P, Oczkowski M, Øvrevik J, Gajewska M, Wilczak J, Biedrzycki J, Dziendzikowska K, Kamola D, Królikowski T, Kruszewski M, Lankoff A, Mruk R, Brunborg G, Instanes C, Gromadzka-Ostrowska J, Myhre O. No adverse lung effects of 7- and 28-day inhalation exposure of rats to emissions from petrodiesel fuel containing 20% rapeseed methyl esters (B20) with and without particulate filter – the FuelHealth project. Inhal Toxicol 2017; 29:206-218. [DOI: 10.1080/08958378.2017.1339149] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Pål Magnusson
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Michał Oczkowski
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Johan Øvrevik
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Malgorzata Gajewska
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Jacek Wilczak
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | | | - Katarzyna Dziendzikowska
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Dariusz Kamola
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Tomasz Królikowski
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Lublin, Poland
| | - Anna Lankoff
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- Department of Radiobiology and Immunology, Jan Kochanowski University, Kielce, Warsaw, Poland
| | - Remigiusz Mruk
- Faculty of Production Engineering, Warsaw University of Life Sciences, Warsaw, Poland
| | - Gunnar Brunborg
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Christine Instanes
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Oddvar Myhre
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
33
|
Corthésy B, Bioley G. Therapeutic intranasal instillation of allergen-loaded microbubbles suppresses experimental allergic asthma in mice. Biomaterials 2017; 142:41-51. [PMID: 28727997 DOI: 10.1016/j.biomaterials.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022]
Abstract
Despite proven efficiency, subcutaneous immunotherapy for aeroallergens is impaired by the duration of the protocol, the repeated injections and potential side-effects associated with the doses of allergen administered. Intranasal delivery of immunotherapeutic agents may overcome several of these drawbacks, provided that an efficient allergen delivery vehicle can be identified. This study evaluates whether intranasally delivered gas-filled microbubble (MB)-associated ovalbumin (OVA), used as a model allergen, can serve as a therapeutic treatment in a mouse model of established allergic asthma. Lung and systemic production of pro-tolerogenic markers, including Foxp3+ CD4 T cells, IL-10, and TGF-β, as well as the Th1-type cytokine IFN-γ, was observed after intranasal immunization with OVA-MB. Post-treatment, aerosol-sensitized mice exhibited the same pattern of markers. Moreover, decrease of eosinophils and neutrophils in BALs, lower frequencies of Th2 cytokine- and IL-17-producing CD4 T cells in lungs and reduced specific IgE in BALs and sera after allergen challenge were observed. Concomitantly, lung resistance and mucus production diminished in OVA-MB-treated animals. Thus, therapeutic intranasal administration of OVA-MBs in established experimental allergic asthma allows modulating pathology-associated immune and physiological parameters usually triggered after exposure to the allergen.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland.
| |
Collapse
|
34
|
Dua K, Shukla SD, Hansbro PM. Aspiration techniques for bronchoalveolar lavage in translational respiratory research: Paving the way to develop novel therapeutic moieties. J Biol Methods 2017; 4:e73. [PMID: 31453230 PMCID: PMC6706109 DOI: 10.14440/jbm.2017.174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 05/04/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Bronchoalveolar lavage (BAL) is a simple, yet informative tool in understanding the immunopathology of various lung diseases via quantifying various inflammatory cells, cytokines and growth factors. At present, this traditional method is often blended with several robust and sophisticated molecular and biological techniques sustaining the significance and longevity of this technique. Crucially, the existence of slightly distinct approaches and variables employed at different laboratories around the globe in performing BAL aspiration indeed demands an utmost need to optimize and develop an effective, cost-effective and a reproducible technique. This mini review will be of importance to the biological translational scientist, particularly respiratory researchers in understanding the fundamentals and approaches to apply and consider with BAL aspiration techniques. This will ensure generating a meaningful and clinically relevant data which in turn accelerate the development of new and effective therapeutic moieties for major respiratory conditions.
Collapse
Affiliation(s)
- Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton Heights, Newcastle, NSW 2305, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
35
|
Daubeuf F, Becker J, Aguilar-Pimentel JA, Ebel C, Hrabě de Angelis M, Hérault Y, Frossard N. A Fast, Easy, and Customizable Eight-Color Flow Cytometric Method for Analysis of the Cellular Content of Bronchoalveolar Lavage Fluid in the Mouse. ACTA ACUST UNITED AC 2017. [PMID: 28628216 DOI: 10.1002/cpmo.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cell composition of bronchoalveolar lavage fluid (BAL) is an important indicator of airway inflammation. It is commonly determined by cytocentrifuging leukocytes on slides, then staining, identifying, and counting them as eosinophils, neutrophils, macrophages, or lymphocytes according to morphological criteria under light microscopy, where it is not always easy to distinguish macrophages from lymphocytes. We describe here a one-step, easy-to-use, and easy-to-customize 8-color flow cytometric method for performing differential cell count and comparing it to morphological counts on stained cytospins. This method identifies BAL cells by a simultaneous one-step immunolabeling procedure using antibodies to identify T cells, B cells, neutrophils, eosinophils, and macrophages. Morphological analysis of flow-sorted cell subsets is used to validate this protocol. An important advantage of this basic flow cytometry protocol is the ability to customize it by the addition of antibodies to study receptor expression at leukocyte cell surfaces and identify subclasses of inflammatory cells as needed. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- François Daubeuf
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7200, Illkirch Cedex, France
| | - Julien Becker
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch-Graffenstaden, France
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Claudine Ebel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Yann Hérault
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch-Graffenstaden, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7200, Illkirch Cedex, France
| |
Collapse
|
36
|
Forkuo GS, Nieman AN, Yuan NY, Kodali R, Yu OB, Zahn NM, Jahan R, Li G, Stephen MR, Guthrie ML, Poe MM, Hartzler BD, Harris TW, Yocum GT, Emala CW, Steeber DA, Stafford DC, Cook JM, Arnold LA. Alleviation of Multiple Asthmatic Pathologic Features with Orally Available and Subtype Selective GABA A Receptor Modulators. Mol Pharm 2017; 14:2088-2098. [PMID: 28440659 PMCID: PMC5497587 DOI: 10.1021/acs.molpharmaceut.7b00183] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We describe pharmacokinetic and pharmacodynamic properties of two novel oral drug candidates for asthma. Phenolic α4β3γ2 GABAAR selective compound 1 and acidic α5β3γ2 selective GABAAR positive allosteric modulator compound 2 relaxed airway smooth muscle ex vivo and attenuated airway hyperresponsiveness (AHR) in a murine model of asthma. Importantly, compound 2 relaxed acetylcholine contracted human tracheal airway smooth muscle strips. Oral treatment of compounds 1 and 2 decreased eosinophils in bronchoalveolar lavage fluid in ovalbumin sensitized and challenged mice, thus exhibiting anti-inflammatory properties. Additionally, compound 1 reduced the number of lung CD4+ T lymphocytes and directly modulated their transmembrane currents by acting on GABAARs. Excellent pharmacokinetic properties were observed, including long plasma half-life (up to 15 h), oral availability, and extremely low brain distribution. In conclusion, we report the selective targeting of GABAARs expressed outside the brain and demonstrate reduction of AHR and airway inflammation with two novel orally available GABAAR ligands.
Collapse
Affiliation(s)
- Gloria S. Forkuo
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Amanda N. Nieman
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Nina Y. Yuan
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Revathi Kodali
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Olivia B. Yu
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Nicolas M. Zahn
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Rajwana Jahan
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Guanguan Li
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Michael Rajesh Stephen
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Margaret L. Guthrie
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Michael M. Poe
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Benjamin D. Hartzler
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Ted W. Harris
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Gene T. Yocum
- Department of Anesthesiology, Columbia University, New York, New York, 10032
| | - Charles W. Emala
- Department of Anesthesiology, Columbia University, New York, New York, 10032
| | - Douglas A. Steeber
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Douglas C. Stafford
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - James M. Cook
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| |
Collapse
|
37
|
Preconception allergen sensitization can induce B10 cells in offspring: a potential main role for maternal IgG. Allergy Asthma Clin Immunol 2017; 13:22. [PMID: 28428801 PMCID: PMC5392917 DOI: 10.1186/s13223-017-0195-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 04/08/2017] [Indexed: 11/18/2022] Open
Abstract
Background The mechanisms through which allergies can be inhibited after preconception immunization with allergens are not fully understood. We aimed to evaluate whether maternal immunization can induce a regulatory B (B10) cell population in offspring in concert with allergy inhibition. Methods C57BL/6 females were or were not immunized with OVA and were mated with normal WT males. Their offspring were evaluated at 3 days of age or 20 days after neonatal immunization. Human peripheral B cells from atopic and non-atopic individuals were also evaluated. Results Preconception OVA immunization induced B10 cells in offspring, and IL-10 production appeared to be critical for FcγRIIB upregulation in offspring B cells. Murine and human IL-10-producing B cells responded in vitro to IgG according to the atopic repertoire of the cells. Conclusions Our results reveal that maternal immunization induces allergen-specific B10 cells in offspring and a pivotal role for the IgG repertoire in IL-10 production by murine and human B cells. Electronic supplementary material The online version of this article (doi:10.1186/s13223-017-0195-8) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Dong J, Liao W, Peh HY, Chan TK, Tan WSD, Li L, Yong A, Wong WSF. Ribosomal protein S3 gene silencing protects against experimental allergic asthma. Br J Pharmacol 2017; 174:540-552. [PMID: 28093718 DOI: 10.1111/bph.13717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 01/11/2017] [Accepted: 01/11/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND AND PURPOSE Ribosomal protein S3 (RPS3) is a 40S ribosomal protein of the S3P family essential for implementing protein translation. RPS3 has recently been found to interact with the p65 subunit of the NF-κB complex and promote p65 DNA-binding activity. Persistent activation of the NF-κB pathway is evident in allergic asthma. We hypothesized that gene silencing of lung RPS3 can ameliorate allergic airway inflammation. EXPERIMENTAL APPROACH The gene silencing efficacy of RPS3 siRNA was screened in three different mouse cell lines by real-time PCR and immunoblotting. Protective effects of intratracheal RPS3 siRNA in a house dust mite (HDM) mouse asthma model were determined by measuring cell counts in lung lavage fluid and lung sections, lung cytokine profiles and airway hyperresponsiveness (AHR). KEY RESULTS RPS3 siRNA markedly knocked down RPS3 levels in all mouse cell lines tested, and in mouse lung tissues, blocked TNF-α- or HDM-induced release of mediators by the cultured cells and reduced eosinophil counts in lung lavage fluid from the HDM mouse asthma model. RPS3 siRNA lessened HDM-induced airway mucus hypersecretion, cytokine production and serum IgE elevation. Moreover, RPS3 knockdown significantly suppressed methacholine-induced AHR in experimental asthma. RPS3 siRNA disrupted TNF-α-induced NF-κB activation in a NF-κB reporter gene assay in vitro and prevented the nuclear accumulation of p65 subunit and p65 transcriptional activation in HDM-challenged lungs and cells. CONCLUSIONS AND IMPLICATIONS RPS3 gene silencing ameliorates experimental asthma, probably by disrupting NF-κB activity. RPS3 could be a novel therapeutic target for allergic airway inflammation.
Collapse
Affiliation(s)
- Jinrui Dong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Hong Yong Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Tze Khee Chan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology (SMART), Singapore
| | - W S Daniel Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Li Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Amy Yong
- Department of Pharmacology and Therapeutics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Immunology Program, Life Science Institute, National University of Singapore, Singapore
| |
Collapse
|
39
|
Differential Macrophage Polarization from Pneumocystis in Immunocompetent and Immunosuppressed Hosts: Potential Adjunctive Therapy during Pneumonia. Infect Immun 2017; 85:IAI.00939-16. [PMID: 27993972 DOI: 10.1128/iai.00939-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/09/2016] [Indexed: 11/20/2022] Open
Abstract
We explored differential polarization of macrophages during infection using a rat model of Pneumocystis pneumonia. We observed enhanced pulmonary M1 macrophage polarization in immunosuppressed (IS) hosts, but an M2 predominant response in immunocompetent (IC) hosts following Pneumocystis carinii challenge. Increased inflammation and inducible nitric oxide synthase (iNOS) levels characterized the M1 response. However, macrophage ability to produce nitric oxide was defective. In contrast, the lungs of IC animals revealed a prominent M2 gene signature, and these macrophages effectively elicited an oxidative burst associated with clearance of Pneumocystis In addition, during P. carinii infection the expression of Dectin-1, a critical receptor for recognition and clearance of P. carinii, was upregulated in macrophages of IC animals but suppressed in IS animals. In the absence of an appropriate cytokine milieu for M2 differentiation, Pneumocystis induced an M1 response both in vitro and in vivo The M1 response induced by P. carinii was plastic in nature and reversible with appropriate cytokine stimuli. Finally, we tested whether macrophage polarization can be modulated in vivo and used to help manage the pathogenesis of Pneumocystis pneumonia by adoptive transfer. Treatment with both M1 and M2 cells significantly improved survival of P. carinii-infected IS hosts. However, M2 treatment provided the best outcomes with efficient clearance of P. carinii and reduced inflammation.
Collapse
|
40
|
Curcumin inhibits paraquat induced lung inflammation and fibrosis by extracellular matrix modifications in mouse model. Inflammopharmacology 2016; 24:335-345. [PMID: 27766504 DOI: 10.1007/s10787-016-0286-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/30/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Paraquat (PQ), a potent herbicide can cause severe toxicity. We report here that fibroproliferation phase of acute lung injury (ALI) is initiated much earlier (within 48 h) after PQ intoxication than previously reported (after 2 weeks) and we aimed to study the protective effects of intranasal curcumin as new therapeutic strategy in mouse model. METHODS Mice (Park's strain) were divided into five experimental groups (I) control, received only saline (0.9 % NaCl) (II) PQ, mice intoxicated with PQ (50 mg/kg, i.p., single dose); (III) curcumin, treated with curcumin (5 mg/kg, i.n) an hour before PQ administration; (IV)Veh, DMSO (equal volume to curcumin) given an hour before PQ exposure; (V) DEXA, mice treated with dexamethasone (1 mg/kg, i.p) before an hour of PQ intoxication. After 48 h of the PQ exposure, all mice were sacrificed and samples were analyzed. RESULTS Pretreatment with intranasal curcumin (5 mg/kg) could modify the PQ-intoxication (50 mg/kg, i.p) induced structural remodeling of lung parenchyma at an early phase of acute lung injury. Significant increase in inflammatory cell count, reactive oxygen species and hydroxyproline levels were decreased after curcumin pretreatment (all p < 0.05). Histological examination and zymography results were also found consistent. CONCLUSION Our results show that curcumin pretreatment decreased the expression of alpha smooth muscle actin (α-SMA), matrix metalloproteinases-9 (MMP-9) and changed the expression of tissue inhibitors of metalloproteinase (TIMP-1) after PQ intoxication. Single toxic dose of PQ has initiated fibroproliferation within 48 h and intranasal curcumin may prove as new therapeutic strategy for PQ induced ALI and fibroproliferation.
Collapse
|
41
|
Protein kinase Cθ controls type 2 innate lymphoid cell and T H2 responses to house dust mite allergen. J Allergy Clin Immunol 2016; 139:1650-1666. [PMID: 27746240 DOI: 10.1016/j.jaci.2016.08.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 07/14/2016] [Accepted: 08/08/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Protein kinase C (PKC) θ, a serine/threonine kinase, is involved in TH2 cell activation and proliferation. Type 2 innate lymphoid cells (ILC2s) resemble TH2 cells and produce the TH2 cytokines IL-5 and IL-13 but lack antigen-specific receptors. The mechanism by which PKC-θ drives innate immune cells to instruct TH2 responses in patients with allergic lung inflammation remains unknown. OBJECTIVES We hypothesized that PKC-θ contributes to ILC2 activation and might be necessary for ILC2s to instruct the TH2 response. METHODS PRKCQ gene expression was assessed in innate lymphoid cell subsets purified from human PBMCs and mouse lung ILC2s. ILC2 activation and eosinophil recruitment, TH2-related cytokine and chemokine production, lung histopathology, interferon regulatory factor 4 (IRF4) mRNA expression, and nuclear factor of activated T cells (NFAT1) protein expression were determined. Adoptive transfer of ILC2s from wild-type mice was performed in wild-type and PKC-θ-deficient (PKC-θ-/-) mice. RESULTS Here we report that PKC-θ is expressed in both human and mouse ILC2s. Mice lacking PKC-θ had reduced ILC2 numbers, TH2 cell numbers and activation, airway hyperresponsiveness, and expression of the transcription factors IRF4 and NFAT1. Importantly, adoptive transfer of ILC2s restored eosinophil influx and IL-4, IL-5 and IL-13 production in lung tissue, as well as TH2 cell activation. The pharmacologic PKC-θ inhibitor (Compound 20) administered during allergen challenge reduced ILC2 numbers and activation, as well as airway inflammation and IRF4 and NFAT1 expression. CONCLUSIONS Therefore our findings identify PKC-θ as a critical factor for ILC2 activation that contributes to TH2 cell differentiation, which is associated with IRF4 and NFAT1 expression in allergic lung inflammation.
Collapse
|
42
|
Utsch L, Logiantara A, van Ree R, van Rijt LS. Experimental food allergy to peanut enhances the immune response to house dust mite in the airways of mice. Clin Exp Allergy 2016; 47:121-128. [PMID: 27533916 DOI: 10.1111/cea.12799] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/26/2016] [Accepted: 08/09/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Food allergy has been associated with an increased risk for the development of allergic asthma. Asthma is a risk factor for the development of an anaphylactic response to food allergens. An immunological interplay between sensitization to different allergens in different compartments of the body might be involved. OBJECTIVE To evaluate the immunological interplay between intragastrical peanut (PE) sensitization and respiratory sensitization to house dust mite (HDM) allergens. METHODS BALB/c mice were intragastrically sensitized to peanut or sham-sensitized and challenged systemically to PE. Between sensitization and challenge, mice were intranasally exposed to HDM extract or PBS, as a control. The response to HDM (eosinophil recruitment, cytokine response, HDM-specific immunoglobulins and airway hyper-reactivity) and to PE (cytokine response, mast cells in gut, mMCP-1 in serum and body temperature) was assessed. RESULTS A preceding PE sensitization increased HDM-induced production of IL-4, IL-5, IL-13 and IFNγ in lung-draining lymph nodes and total IgE levels in HDM-sensitized mice. However, recruitment of inflammatory cells to the airways or airway hyper-reactivity was not aggravated in PE/HDM double-sensitized mice. Alternatively, HDM-induced airway inflammation did not significantly affect the immune response or the anaphylactic response to a systemic challenge with peanut. CONCLUSION AND CLINICAL RELEVANCE Our data show that a preceding peanut sensitization boosted IgE- and HDM-specific Th2 response in the airways in mice. It contributes to the understanding of the underlying immunological mechanism of polysensitization which often occurs in allergic individuals over time.
Collapse
Affiliation(s)
- L Utsch
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - A Logiantara
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - R van Ree
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - L S van Rijt
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Park EJ, Hong YS, Lee BS, Yoon C, Jeong U, Kim Y. Single-walled carbon nanotubes disturbed the immune and metabolic regulation function 13-weeks after a single intratracheal instillation. ENVIRONMENTAL RESEARCH 2016; 148:184-195. [PMID: 27078092 DOI: 10.1016/j.envres.2016.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/20/2016] [Accepted: 03/21/2016] [Indexed: 06/05/2023]
Abstract
Due to their unique physicochemical properties, the potential health effects of single-walled carbon nanotubes (SWCNTs) have attracted continuous attention together with their extensive application. In this study, we aimed to identify local and systemic health effects following pulmonary persistence of SWCNTs. As expected, SWCNTs remained in the lung for 13 weeks after a single intratracheal instillation (50, 100, and 200μg/kg). In the lung, the total number of cells and the percentages of lymphocytes and neutrophils significantly increased at 200μg/kg compared to the control, and the Th1-polarized immune response was induced accompanying enhanced expression of tissue damage-related genes and increased release of chemokines. Additionally, SWCNTs enhanced the expression of antigen presentation-related proteins on the surface of antigen-presenting cells, however, maturation of dendritic cells was inhibited by their persistence. As compared to the control, a significant increase in the percentage of neutrophils and a remarkable decrease of BUN and potassium level were observed in the blood of mice treated with the highest dose. This was accompanied by the down-regulation of the expression of antigen presentation-related proteins on splenocytes. Moreover, protein and glucose metabolism were disturbed with an up-regulation of fatty acid β-oxidation. Taken together, we conclude that SWCNTs may induce adverse health effects by disturbing immune and metabolic regulation functions in the body. Therefore, careful application of SWCNTs is necessary for the enforcement of safety in nano-industries.
Collapse
Affiliation(s)
- Eun-Jung Park
- Myunggok Eye Research Institute, Konyang University, Daejeon 302-718, Republic of Korea.
| | - Young-Shick Hong
- Division of Food and Nutrition, Chonnam National University, Yongbong-Ro, Buk-Gu, Gwangju 500-757, Republic of Korea
| | - Byoung-Seok Lee
- Toxicologic Pathology Center, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Cheolho Yoon
- Seoul Center, Korea Basic Science Institute, Seoul 126-16, Republic of Korea
| | - Uiseok Jeong
- Department of Chemical Engineering, Kwangwoon University, Seoul 139-701, Republic of Korea
| | - Younghun Kim
- Department of Chemical Engineering, Kwangwoon University, Seoul 139-701, Republic of Korea
| |
Collapse
|
44
|
Suzuki S, Ogawa M, Ohta S, Arima K, Nunomura S, Nanri Y, Mitamura Y, Yoshihara T, Nakamura Y, Yamauchi K, Chibana K, Ishii Y, Lee JJ, Aratani Y, Kakuta S, Kubo S, Iwakura Y, Yoshida H, Izuhara K. The potential for repositioning antithyroid agents as antiasthma drugs. J Allergy Clin Immunol 2016; 138:1458-1461.e8. [PMID: 27444178 DOI: 10.1016/j.jaci.2016.04.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 04/12/2016] [Accepted: 04/20/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Shoichi Suzuki
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | - Masahiro Ogawa
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | - Shoichiro Ohta
- Department of Laboratory Medicine, Saga Medical School, Saga, Japan
| | - Kazuhiko Arima
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | - Satoshi Nunomura
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | - Yasuhiro Nanri
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | | | | | - Yutaka Nakamura
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Kohei Yamauchi
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Kazuyuki Chibana
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Yoshiki Ishii
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - James J Lee
- Division of Pulmonary Medicine, Mayo Clinic Arizona, Scottsdale, Ariz
| | - Yasuaki Aratani
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
| | - Shigeru Kakuta
- Department of Biomedical Science, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan; Center for Experimental Medicine and Systems Biology, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Sachiko Kubo
- Center for Experimental Medicine and Systems Biology, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan; Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Center for Experimental Medicine and Systems Biology, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan; Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Hiroki Yoshida
- Division of Immunology, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Kenji Izuhara
- Division of Medical Biochemistry, Saga Medical School, Saga, Japan.
| |
Collapse
|
45
|
Debeuf N, Haspeslagh E, van Helden M, Hammad H, Lambrecht BN. Mouse Models of Asthma. ACTA ACUST UNITED AC 2016; 6:169-184. [PMID: 27248433 DOI: 10.1002/cpmo.4] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allergic asthma is a chronic inflammatory disease of the conducting airways characterized by the presence of allergen-specific IgE, Th2 cytokine production, eosinophilic airway inflammation, bronchial hyperreactivity, mucus overproduction, and structural changes in the airways. Investigators have tried to mimic these features of human allergic asthma in murine models. Whereas the surrogate allergen ovalbumin has been extremely valuable for unravelling underlying mechanisms of the disease, murine asthma models depend nowadays on naturally occurring allergens, such as house dust mite (HDM), cockroach, and Alternaria alternata. Here we describe a physiologically relevant model of acute allergic asthma based on sensitization and challenge with HDM extracts, and compare it with the ovalbumin/alum-induced asthma model. Moreover, we propose a detailed readout of the asthma phenotype, determining the degree of eosinophilia in bronchoalveolar lavage fluids by flow cytometry, visualizing goblet cell metaplasia, and measuring Th cytokine production by lung-draining mediastinal lymph node cells restimulated with HDM. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Nincy Debeuf
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Eline Haspeslagh
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Mary van Helden
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
46
|
Mozzini Monteiro T, Ferrera Costa H, Carvalho Vieira G, Rodrigues Salgado PR, da Silva Stiebbe Salvadori MG, de Almeida RN, de Fatima Vanderlei de Souza M, Neves Matias W, Andrade Braga V, Nalivaiko E, Piuvezam MR. Anti-asthmatic and anxiolytic effects of Herissantia tiubae, a Brazilian medicinal plant. Immun Inflamm Dis 2016; 4:201-212. [PMID: 27957328 PMCID: PMC4879466 DOI: 10.1002/iid3.107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/03/2016] [Accepted: 02/29/2016] [Indexed: 12/30/2022] Open
Abstract
Herissantia tiubae (HtE) is a Brazilian plant used in folk medicine to treat inflammatory diseases. Our aim was to determine whether the HtE has anti-inflammatory and anxiolytic effects in a murine model of asthma. Ovalbumin (OVA)-sensitized BALB/c mice were treated with HtE (50, 100, or 200 mg/kg) or dexamethasone before each OVA challenge. After the last challenge, animals were subjected to anxiety tests and respiratory measurements. Following euthanasia, we quantified immune cells in the bronchoalveolar lavage (BAL), serum IgE titer and cytokine levels, cellular infiltration and mucus content in the lung tissues, and cellular composition of the mediastinal lymph nodes. OVA challenge in sensitized animals caused: (1) reduction of mean respiratory and dominant respiratory rate (from 398 ± 12 to 286 ± 20 cicles per minute (cpm) and from 320 ± 14 to 162 ± 15 cpm, respectively); (2) increase in behavioral markers of anxiety tests; (3) substantial pro-inflammatory effects, including rise in OVA-specific IgE titer (from 0 to 1:2048) and these inflammatory effect diminished the titer to 1:512 after HtE treatment; rise in plasma IL-13 (from 13 ng/mL in saline to 227 ng/mL in OVA and HtE treatment restored to 1.29 ng/mL; rise in total BAL cell count (from 0.742 cells/mL in saline to 11.77 cells/mL in OVA), with prominent eosinophilia. H. tiubae extract affected respiratory parameters similarly to aminophylline, behavioral changes comparable to diazepam, and inflammation being as efficient as dexamethasone. H. tiubae extract (HtE) possesses both anti-inflammatory and anxiolytic properties in the murine model of asthma.
Collapse
Affiliation(s)
- Talissa Mozzini Monteiro
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | - Hermann Ferrera Costa
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | - Giciane Carvalho Vieira
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | - Paula Regina Rodrigues Salgado
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | | | - Reinaldo Nobrega de Almeida
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | | | - Wemerson Neves Matias
- Department of PharmacyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| | - Valdir Andrade Braga
- Department of Biotechnology, Center of BiotechnologyFederal University of ParaibaJoão PessoaParaibaBrazil
| | - Eugene Nalivaiko
- School of Biomedical Sciences and PharmacyUniversity of NewcastleNewcastleNew South WalesAustralia
| | - Marcia Regina Piuvezam
- Laboratory of Immunopharmacology, Department of Physiology and PathologyFederal University of ParaíbaJoão PessoaParaíba58051‐970Brazil
| |
Collapse
|
47
|
Canbaz D, Logiantara A, Hamers T, van Ree R, van Rijt LS. Indoor Pollutant Hexabromocyclododecane Has a Modest Immunomodulatory Effect on House Dust Mite Induced Allergic Asthma in Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:405-411. [PMID: 26633745 DOI: 10.1021/acs.est.5b05348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Hexabromocyclododecane (HBCD) has been recognized as an indoor pollutant. HBCD is added as a flame retardant to many consumer products and leaches from the products into house dust. HBCD might be potentially hazardous to the airways because of inhalation of house dust. Sensitization to house dust mite (HDM) is a risk factor for the development of allergic asthma. In this study, we examined whether HBCD can affect the immune response to HDM allergens. Bone-marrow-derived dendritic cells (BMDCs) were exposed simultaneously to HBCD and HDM in vitro. HBCD enhanced oxidative stress in HDM-pulsed BMDCs, which was accompanied by a higher production of Interleukin (IL)-6 and -10. Adoptive transfer of HDM/HBCD-exposed BMDCs into naı̈ve mice resulted in enhanced levels of IL-17A after inhalational challenge with HDM. Direct mucosal exposure to HBCD during HDM inhalation enhanced IL-4 or IL-17A production, depending on the HDM extract used, but did not aggravate the eosinophilic airway inflammation or airway hyper-reactivity. Our results indicate that exposure to HBCD can have a mild immune-modulating effect by enhancing the inflammatory cytokine production in response to inhaled HDM in mice.
Collapse
Affiliation(s)
| | | | - Timo Hamers
- Institute for Environmental Studies, VU University Amsterdam , De Boelelaan 1087, Amsterdam 1087HV, The Netherlands
| | | | | |
Collapse
|
48
|
Kurakula K, Vos M, Logiantara A, Roelofs JJTH, Nieuwenhuis MA, Koppelman GH, Postma DS, Brandsma CA, Sin DD, Bossé Y, Nickle DC, van Rijt LS, de Vries CJM. Deficiency of FHL2 attenuates airway inflammation in mice and genetic variation associates with human bronchial hyper-responsiveness. Allergy 2015. [PMID: 26222912 DOI: 10.1111/all.12709] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Asthma is an inflammatory disease that involves airway hyper-responsiveness and mucus hypersecretion. The LIM-only protein FHL2 is a crucial modulator of multiple signal transduction pathways and functions as a scaffold in specific protein-protein interactions. OBJECTIVE We sought to investigate the role of FHL2 in airway inflammation. METHODS Allergic airway inflammation was induced in WT and FHL2-knock out (FHL2-KO) mice with ovalbumin (OVA). Lung tissue, bronchoalveolar lavage fluid (BALF) and draining lymph node cells were analysed for inflammation. FHL2 loss and gain of function studies were performed in lung epithelial cells. RESULTS FHL2-deficient mice challenged with OVA show significantly reduced airway inflammation as evidenced by reduced infiltration of inflammatory cells including eosinophils, dendritic cells, B cells and T cells. Furthermore, mucus production was decreased in FHL2-KO mice. In BALF, the levels of IL-5, IL-13, eotaxin-1 and eotaxin-2 were significantly lower in FHL2-KO mice. In addition, draining lymph node cells from FHL2-KO mice show reduced levels of IL-5 and IL-13. Consistent with this, OVA-specific serum IgG and IgE levels were reduced in FHL2-KO mice. We also found that phosphorylation of ERK1/2 is markedly attenuated in FHL2-KO lung. Knock-down of FHL2 in human lung epithelial cells resulted in a striking decrease in ERK1/2 phosphorylation and mRNA levels of inflammatory cytokines and MUC5AC, whereas FHL2 overexpression exhibited opposite effects. Finally, the SNP rs4851765 shows an association with the severity of bronchial hyper-responsiveness. CONCLUSION These results highlight functional involvement of FHL2 in airway inflammation and identify FHL2 as a novel gene associated with asthma severity in human.
Collapse
Affiliation(s)
- K. Kurakula
- Deartment of Medical Biochemistry; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - M. Vos
- Deartment of Medical Biochemistry; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - A. Logiantara
- Department of Experimental Immunology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - J. J. T. H. Roelofs
- Department of Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - M. A. Nieuwenhuis
- University of Groningen; University Medical Center Groningen; Department of Pulmonology; GRIAC Research Institute; Groningen The Netherlands
| | - G. H. Koppelman
- University of Groningen; University Medical Center Groningen; Department of Pediatric Pulmonology; Beatrix Children's Hospital; GRIAC Research Institute; Groningen The Netherlands
| | - D. S. Postma
- University of Groningen; University Medical Center Groningen; Department of Pulmonology; GRIAC Research Institute; Groningen The Netherlands
| | - C. A. Brandsma
- University of Groningen; University Medical Center Groningen; Department of Pathology and Medical Biology; GRIAC Research Institute; Groningen The Netherlands
| | - D. D. Sin
- The University of British Columbia James Hogg Research Laboratory; St Paul's Hospital; Vancouver Canada
- Respiratory Division; Department of Medicine; University of British Columbia; Vancouver Canada
| | - Y. Bossé
- Department of Molecular Medicine; Laval University; Quebec City Canada
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec; Laval University; Quebec City Canada
| | - D. C. Nickle
- Genetics; Rosetta Inpharmatics; Merck Seattle WA USA
- Merck Research Laboratories; Boston MA USA
| | - L. S. van Rijt
- Department of Experimental Immunology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - C. J. M. de Vries
- Deartment of Medical Biochemistry; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
49
|
Oyeniran C, Sturgill JL, Hait NC, Huang WC, Avni D, Maceyka M, Newton J, Allegood JC, Montpetit A, Conrad DH, Milstien S, Spiegel S. Aberrant ORM (yeast)-like protein isoform 3 (ORMDL3) expression dysregulates ceramide homeostasis in cells and ceramide exacerbates allergic asthma in mice. J Allergy Clin Immunol 2015; 136:1035-46.e6. [PMID: 25842287 PMCID: PMC4591101 DOI: 10.1016/j.jaci.2015.02.031] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/28/2015] [Accepted: 02/27/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Asthma, a chronic inflammatory condition defined by episodic shortness of breath with expiratory wheezing and cough, is a serious health concern affecting more than 250 million persons. Genome-wide association studies have identified ORM (yeast)-like protein isoform 3 (ORMDL3) as a gene associated with susceptibility to asthma. Although its yeast ortholog is a negative regulator of de novo ceramide biosynthesis, how ORMDL3 contributes to asthma pathogenesis is not known. OBJECTIVES We sought to decipher the molecular mechanism for the pathologic functions of ORMDL3 in asthma and the relationship to its evolutionarily conserved role in regulation of ceramide homeostasis. METHODS We determined the relationship between expression of ORMDL3 and ceramide in epithelial and inflammatory cells and in asthma pathogenesis in mice. RESULTS Although small increases in ORMDL3 expression decrease ceramide levels, remarkably, higher expression in lung epithelial cells and macrophages in vitro and in vivo increased ceramide production, which promoted chronic inflammation, airway hyperresponsiveness, and mucus production during house dust mite-induced allergic asthma. Moreover, nasal administration of the immunosuppressant drug FTY720/fingolimod reduced ORMDL3 expression and ceramide levels and mitigated airway inflammation and hyperreactivity and mucus hypersecretion in house dust mite-challenged mice. CONCLUSIONS Our findings demonstrate that overexpression of ORMDL3 regulates ceramide homeostasis in cells in a complex manner and suggest that local FTY720 administration might be a useful therapeutic intervention for the control of allergic asthma.
Collapse
Affiliation(s)
- Clement Oyeniran
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Jamie L Sturgill
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Va; School of Nursing, Virginia Commonwealth University, Richmond, Va
| | - Nitai C Hait
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Wei-Ching Huang
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Dorit Avni
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Michael Maceyka
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Jason Newton
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Jeremy C Allegood
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Alison Montpetit
- School of Nursing, Virginia Commonwealth University, Richmond, Va
| | - Daniel H Conrad
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va.
| |
Collapse
|
50
|
Utsch L, Folisi C, Akkerdaas JH, Logiantara A, van de Pol MA, van der Zee JS, Krop EJM, Lutter R, van Ree R, van Rijt LS. Allergic sensitization is associated with inadequate antioxidant responses in mice and men. Allergy 2015; 70:1246-58. [PMID: 26081441 DOI: 10.1111/all.12674] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND Allergies arise from aberrant Th2 responses to allergens. The processes involved in the genesis of allergic sensitization remain elusive. Some allergens such as derived from house dust mites have proteolytic activity which can induce oxidative stress in vivo. A reduced capacity of the host to control oxidative stress might prime for allergic sensitization. METHODS Two different strains of mice were compared for their antioxidant and immune response to HDM. Protease activity of the HDM extract was reduced to investigate its role in oxidative stress induction in the airways and whether this induction could determine allergic sensitization and inflammation. The role of oxidative stress in allergic sensitization was also investigated in humans. An occupational cohort of animal workers was followed for the development of sensitization to rodent urinary proteins. Levels of oxidative stress in serum and antioxidant responses by PBMCs were determined. RESULTS Susceptibility to allergic sensitization to mite allergens in mice was highly dependent on host genetic background and was associated with oxidative stress in the lungs before allergen exposure and poor antioxidant response after allergen exposure. Reduction in mite protease activity limited its capacity to induce oxidative stress and allergic inflammation in mice. We showed that also in human subjects, oxidative stress before allergen exposure and poor antioxidant responses were associated with predisposition to occupational allergy. CONCLUSION Our study indicates that oxidative stress condition before allergen exposure due to an inadequate antioxidant response may prime for allergic Th2 responses.
Collapse
Affiliation(s)
- L. Utsch
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - C. Folisi
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
- Department of Respiratory Medicine; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - J. H. Akkerdaas
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - A. Logiantara
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - M. A. van de Pol
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
- Department of Respiratory Medicine; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | | | - E. J. M. Krop
- Institute for Risk Assessment Sciences; Utrecht University; Utrecht the Netherlands
| | - R. Lutter
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
- Department of Respiratory Medicine; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - R. van Ree
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
- Department of Otorhinolaryngology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| | - L. S. van Rijt
- Department of Experimental Immunology; Academic Medical Center/University of Amsterdam; Amsterdam the Netherlands
| |
Collapse
|