1
|
Yang A, Gu C, Upchurch K, Caffiers A, Millard M, Baert L, Joo H, Oh S. Omalizumab is ineffective in regulating proasthmatic serum cytokine and chemokine levels in nonresponders with high BMI. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100462. [PMID: 40242148 PMCID: PMC12002203 DOI: 10.1016/j.jacig.2025.100462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/13/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025]
Abstract
Background Omalizumab provides clinical benefits to a fraction of patients with asthma. It remains unclear why some patients do not respond to omalizumab therapy. Objective We sought to investigate whether omalizumab could alter serum cytokine and chemokine levels that could be associated with asthma pathogenesis. We also investigated why omalizumab is ineffective in controlling proasthmatic serum cytokine and chemokine levels in nonresponders. Methods Serum cytokine and chemokine levels in patients with moderate to severe asthma (N = 45; 34 responders and 11 nonresponders) were assessed before and after 26 weeks of omalizumab therapy. Correlations between cytokine and chemokine levels and asthma symptoms as well as characteristics of responders and nonresponders were assessed. Nonasthmatic subjects (N = 22) served as controls for patients with asthma (N = 45). Results Omalizumab was more effective in patients with increased serum eotaxin-1 and IL-13 levels than in others at baseline. Omalizumab decreased eotaxin-1 and IL-13, along with levels of most of the cytokines and chemokines tested, including IL-7, CCL17, and CXCL10, in responders, except for CCL5 and CCL22, which can contribute to neutrophilic and type 2 airway inflammation, respectively. In contrast, omalizumab did not decrease such serum cytokine and chemokine levels in nonresponders. Of interest, serum CCL17, CCL22, CXCL10, and IL-7 levels in nonresponders were associated with their body mass index, which could explain why omalizumab was unable to reduce their concentrations in nonresponders. Conclusions Omalizumab can regulate most cytokine and chemokine levels in responders. However, in nonresponders, it is unable to modulate specific proasthmatic cytokines and chemokines due to their association with individual body mass index, which is not influenced by omalizumab.
Collapse
Affiliation(s)
- Agnes Yang
- Department of Immunology, Mayo Clinic, Scottsdale, Ariz
| | - Chao Gu
- Department of Immunology, Mayo Clinic, Scottsdale, Ariz
| | | | | | - Mark Millard
- Martha Foster Lung Care Center, Baylor University Medical Center, Dallas, Tex
| | - Laurie Baert
- Department of Immunology, Mayo Clinic, Scottsdale, Ariz
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, Ariz
- Institute for Biomedical Studies, Baylor University, Waco, Tex
| | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, Ariz
- Institute for Biomedical Studies, Baylor University, Waco, Tex
| |
Collapse
|
2
|
Januskevicius A, Vasyle E, Rimkunas A, Malakauskas K. Integrative Cross-Talk in Asthma: Unraveling the Complex Interactions Between Eosinophils, Immune, and Structural Cells in the Airway Microenvironment. Diagnostics (Basel) 2024; 14:2448. [PMID: 39518415 PMCID: PMC11545034 DOI: 10.3390/diagnostics14212448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Asthma is a chronic inflammatory process that leads to airway narrowing, causing breath loss followed by spasms, wheezing, and shortness of breath. Within the asthmatic lungs, interaction among various immune cells and structural cells plays a significant role in orchestrating an inflammatory response in which eosinophils hold central importance. In these settings, allergens or other environmental exposures commonly drive the immune response to recruit eosinophils to the airways. The appearance of eosinophils in the airways indicates a dynamic interplay of various cell types within lung tissue and does not represent a passive effect of inflammation. The cellular cross-talk causes the persistence of eosinophilic inflammation, and if left untreated, it results in long-term damage to the airway structure and function. Further exacerbation of the condition occurs because of this. We discuss how this complex interplay of eosinophils, immune, and structural cells within the airway microenvironment leads to the distinct pathophysiological features in asthma, the variability in disease severity, and the response to biological treatments.
Collapse
Affiliation(s)
- Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
| | - Egle Vasyle
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
| | - Airidas Rimkunas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
| | - Kestutis Malakauskas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
3
|
Hargitai R, Parráková L, Szatmári T, Monfort-Lanzas P, Galbiati V, Audouze K, Jornod F, Staal YCM, Burla S, Chary A, Gutleb AC, Lumniczky K, Vandebriel RJ, Gostner JM. Chemical respiratory sensitization-Current status of mechanistic understanding, knowledge gaps and possible identification methods of sensitizers. FRONTIERS IN TOXICOLOGY 2024; 6:1331803. [PMID: 39135743 PMCID: PMC11317441 DOI: 10.3389/ftox.2024.1331803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/27/2024] [Indexed: 08/15/2024] Open
Abstract
Respiratory sensitization is a complex immunological process eventually leading to hypersensitivity following re-exposure to the chemical. A frequent consequence is occupational asthma, which may occur after long latency periods. Although chemical-induced respiratory hypersensitivity has been known for decades, there are currently no comprehensive and validated approaches available for the prospective identification of chemicals that induce respiratory sensitization, while the expectations of new approach methodologies (NAMs) are high. A great hope is that due to a better understanding of the molecular key events, new methods can be developed now. However, this is a big challenge due to the different chemical classes to which respiratory sensitizers belong, as well as because of the complexity of the response and the late manifestation of symptoms. In this review article, the current information on respiratory sensitization related processes is summarized by introducing it in the available adverse outcome pathway (AOP) concept. Potentially useful models for prediction are discussed. Knowledge gaps and gaps of regulatory concern are identified.
Collapse
Affiliation(s)
- Rita Hargitai
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Lucia Parráková
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Tünde Szatmári
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Pablo Monfort-Lanzas
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
- Institute of Bioinformatics, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Valentina Galbiati
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università Degli Studi di Milano (UNIMI), Milano, Italy
| | | | | | - Yvonne C. M. Staal
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sabina Burla
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Aline Chary
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Arno C. Gutleb
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Katalin Lumniczky
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Rob J. Vandebriel
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Johanna M. Gostner
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| |
Collapse
|
4
|
Arao Y, Stumpo DJ, Hoenerhoff MJ, Tighe RM, Yu YR, Sutton D, Kashyap A, Beerman I, Blackshear PJ. Lethal eosinophilic crystalline pneumonia in mice expressing a stabilized Csf2 mRNA. FASEB J 2023; 37:e23100. [PMID: 37462673 PMCID: PMC11078221 DOI: 10.1096/fj.202300757r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that stimulates the proliferation and differentiation of granulocyte and macrophage precursors. The mouse gene-encoding GM-CSF, Csf2, is regulated at both transcriptional and post-transcriptional levels. An adenine-uridine-rich element (ARE) within the 3'-untranslated region of Csf2 mRNA was shown in cell transfection studies to confer instability on this transcript. To explore the physiological importance of this element in an intact animal, we generated mice with a knock-in deletion of the 75-nucleotide ARE. Mice heterozygous for this ARE deletion developed severe respiratory distress and death within about 12 weeks of age. There was dense infiltration of lung alveolar spaces by crystal-containing macrophages. Increased stability of Csf2 mRNA was confirmed in bone marrow-derived macrophages, and elevated GM-CSF levels were observed in serum and lung. These mice did not exhibit notable abnormalities in blood or bone marrow, and transplantation of bone marrow from mutant mice into lethally irradiated WT mice did not confer the pulmonary phenotype. Mice with a conditional deletion of the ARE restricted to lung type II alveolar cells exhibited an essentially identical lethal lung phenotype at the same ages as the mice with the whole-body deletion. In contrast, mice with the same conditional ARE deletion in myeloid cells, including macrophages, exhibited lesser degrees of macrophage infiltration into alveolar spaces much later in life, at approximately 9 months of age. Post-transcriptional Csf2 mRNA stability regulation in pulmonary alveolar epithelial cells appears to be essential for normal physiological GM-CSF secretion and pulmonary macrophage homeostasis.
Collapse
Affiliation(s)
- Yukitomo Arao
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences/NIH, Durham, North Carolina, USA
| | - Deborah J Stumpo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences/NIH, Durham, North Carolina, USA
| | - Mark J Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert M Tighe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Yen-Rei Yu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Deloris Sutton
- Cellular & Molecular Pathology Branch, National Institute of Environmental Health Sciences/NIH, Durham, North Carolina, USA
| | - Amogh Kashyap
- Epigenetics and Stem Cell Aging Unit, National Institute on Aging/NIH, Baltimore, Maryland, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Aging Unit, National Institute on Aging/NIH, Baltimore, Maryland, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences/NIH, Durham, North Carolina, USA
- Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
5
|
Haggadone MD, Speth J, Hong HS, Penke LR, Zhang E, Lyssiotis CA, Peters-Golden M. ATP citrate lyase links increases in glycolysis to diminished release of vesicular suppressor of cytokine signaling 3 by alveolar macrophages. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166458. [PMID: 35700791 PMCID: PMC9940702 DOI: 10.1016/j.bbadis.2022.166458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are important vectors for intercellular communication. Lung-resident alveolar macrophages (AMs) tonically secrete EVs containing suppressor of cytokine signaling 3 (SOCS3), a cytosolic protein that promotes homeostasis in the distal lung via its actions in recipient neighboring epithelial cells. AMs are metabolically distinct and exhibit low levels of glycolysis at steady state. To our knowledge, whether cellular metabolism influences the packaging and release of an EV cargo molecule has never been explored in any cellular context. Here, we report that increases in glycolysis following in vitro exposure of AMs to the growth and activating factor granulocyte-macrophage colony-stimulating factor inhibit the release of vesicular SOCS3 by primary AMs. Glycolytically diminished SOCS3 secretion requires export of citrate from the mitochondria to the cytosol and its subsequent conversion to acetyl-CoA by ATP citrate lyase. Our data for the first time implicate perturbations in intracellular metabolites in the regulation of vesicular cargo packaging and secretion.
Collapse
Affiliation(s)
- Mikel D Haggadone
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jennifer Speth
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hanna S Hong
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 41809, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 41809, USA
| | - Loka R Penke
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eric Zhang
- Undergraduate Research Opportunity Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 41809, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 41809, USA
| | - Marc Peters-Golden
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Shaw OM, Hurst RD, Cooney J, Sawyer GM, Dinnan H, Martell S. Boysenberry and apple juice concentrate reduced acute lung inflammation and increased M2 macrophage-associated cytokines in an acute mouse model of allergic airways disease. Food Sci Nutr 2021; 9:1491-1503. [PMID: 33747463 PMCID: PMC7958577 DOI: 10.1002/fsn3.2119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Bioactive compounds including anthocyanins and other polyphenols are associated with reduced lung inflammation and improved lung function in asthma and other lung diseases. This study investigated the effects of a Boysenberry and apple juice concentrate, high in cyanidin glycosides, ellagitannins, and chlorogenic acid, on a mouse model of allergic airways inflammation. Male C57BL/6J mice were orally gavaged with 2.5 mg/kg of total anthocyanins (TAC) from BerriQi® Boysenberry and apple juice concentrate (0.2 mg/kg human equivalent dose) or water control 1 hr before an acute intranasal ovalbumin (OVA) challenge and were gavaged again 2 days after the intranasal challenge. Consumption of BerriQi® Boysenberry and apple juice concentrate significantly decreased OVA-induced infiltrating eosinophils, neutrophils, and T cells in the lung, and mucous production. Quantification of gene expression for arginase (Arg1), chitinase 3-like 3 (Ym-1), found in inflammatory zone (Fizz1), which have been associated with an anti-inflammatory macrophage phenotype (M2), found significantly increased Arg1 expression in the lung in the Boysenberry and apple juice concentrate treatment group. There was also increased production of M2-associated cytokines C-X-C motif chemokine ligand (CXCL) 10 and C-C motif chemokine ligand (CCL) 4. These results suggest that consumption of BerriQi® Boysenberry and apple juice concentrate promoted a shift toward an anti-inflammatory environment within the lung leading to reduced immune cell infiltration and tissue damage.
Collapse
Affiliation(s)
- Odette M. Shaw
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Roger D. Hurst
- Food Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Janine Cooney
- Biological Chemistry & Bioactives GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedHamiltonNew Zealand
| | - Gregory M. Sawyer
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Hannah Dinnan
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Sheridan Martell
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| |
Collapse
|
7
|
Jones JT, Liu KW, Wang X, Kowalski CH, Ross BS, Mills KAM, Kerkaert JD, Hohl TM, Lofgren LA, Stajich JE, Obar JJ, Cramer RA. Aspergillus fumigatus Strain-Specific Conidia Lung Persistence Causes an Allergic Broncho-Pulmonary Aspergillosis-Like Disease Phenotype. mSphere 2021; 6:e01250-20. [PMID: 33597172 PMCID: PMC8544898 DOI: 10.1128/msphere.01250-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/27/2021] [Indexed: 11/20/2022] Open
Abstract
Aspergillus fumigatus is a filamentous fungus which can cause multiple diseases in humans. Allergic broncho-pulmonary aspergillosis (ABPA) is a disease diagnosed primarily in cystic fibrosis patients caused by a severe allergic response often to long-term A. fumigatus colonization in the lungs. Mice develop an allergic response to repeated inhalation of A. fumigatus spores; however, no strains have been identified that can survive long-term in the mouse lung and cause ABPA-like disease. We characterized A. fumigatus strain W72310, which was isolated from the expectorated sputum of an ABPA patient, by whole-genome sequencing and in vitro and in vivo viability assays in comparison to a common reference strain, CEA10. W72310 was resistant to leukocyte-mediated killing and persisted in the mouse lung longer than CEA10, a phenotype that correlated with greater resistance to oxidative stressors, hydrogen peroxide, and menadione, in vitro In animals both sensitized and challenged with W72310, conidia, but not hyphae, were viable in the lungs for up to 21 days in association with eosinophilic airway inflammation, airway leakage, serum IgE, and mucus production. W72310-sensitized mice that were recall challenged with conidia had increased inflammation, Th1 and Th2 cytokines, and airway leakage compared to controls. Collectively, our studies demonstrate that a unique strain of A. fumigatus resistant to leukocyte killing can persist in the mouse lung in conidial form and elicit features of ABPA-like disease.IMPORTANCE Allergic broncho-pulmonary aspergillosis (ABPA) patients often present with long-term colonization of Aspergillus fumigatus Current understanding of ABPA pathogenesis has been complicated by a lack of long-term in vivo fungal persistence models. We have identified a clinical isolate of A. fumigatus, W72310, which persists in the murine lung and causes an ABPA-like disease phenotype. Surprisingly, while viable, W72310 showed little to no growth beyond the conidial stage in the lung. This indicates that it is possible that A. fumigatus can cause allergic disease in the lung without any significant hyphal growth. The identification of this strain of A. fumigatus can be used not only to better understand disease pathogenesis of ABPA and potential antifungal treatments but also to identify features of fungal strains that drive long-term fungal persistence in the lung. Consequently, these observations are a step toward helping resolve the long-standing question of when to utilize antifungal therapies in patients with ABPA and fungal allergic-type diseases.
Collapse
Affiliation(s)
- Jane T Jones
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Ko-Wei Liu
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Xi Wang
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Caitlin H Kowalski
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Brandon S Ross
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Kathleen A M Mills
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, New York, USA
| | - Joshua D Kerkaert
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, New York, USA
| | - Lotus A Lofgren
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Jason E Stajich
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Joshua J Obar
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Robert A Cramer
- Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
8
|
Nobs SP, Pohlmeier L, Li F, Kayhan M, Becher B, Kopf M. GM-CSF instigates a dendritic cell-T-cell inflammatory circuit that drives chronic asthma development. J Allergy Clin Immunol 2021; 147:2118-2133.e3. [PMID: 33440200 DOI: 10.1016/j.jaci.2020.12.638] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/06/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Steroid-resistant asthma is often characterized by high levels of neutrophils and mixed TH2/TH17 immune profiles. Indeed, neutrophils are key drivers of chronic lung inflammation in multiple respiratory diseases. Their numbers correlate strongly with disease severity, and their presence is often associated with exacerbation of chronic lung inflammation. OBJECTIVE What factors drive development of neutrophil-mediated chronic lung disease remains largely unknown, and we sought to study the role of GM-CSF as a potential regulator in chronic asthma. METHODS Different experimental animal models of chronic asthma were used in combination with alveolar macrophage-reconstitution of global GM-CSF receptor knockout mice as well as cell-type-specific knockout animals to elucidate the role of GM-CSF signaling in chronic airway inflammation. RESULTS We identify GM-CSF signaling as a critical factor regulating pulmonary accumulation of neutrophils. We show that although being not required for intrinsically regulating neutrophil migration, GM-CSF controls lung dendritic cell function, which in turn promotes T-cell-dependent recruitment of neutrophils to the airways. We demonstrate that GM-CSF regulates lung dendritic cell antigen uptake, transport, and TH2/TH17 cell priming in an intrinsic fashion, which in turn drives pulmonary granulocyte recruitment and contributes to development of airway hyperresponsiveness in chronic disease. CONCLUSIONS We identify GM-CSF as a potentially novel therapeutic target in chronic lung inflammation, describing a GM-CSF-dependent lung conventional dendritic cell-T-cell-neutrophil axis that drives chronic lung disease.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lea Pohlmeier
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Fengqi Li
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Painter JD, Galle-Treger L, Akbari O. Role of Autophagy in Lung Inflammation. Front Immunol 2020; 11:1337. [PMID: 32733448 PMCID: PMC7358431 DOI: 10.3389/fimmu.2020.01337] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a cellular recycling system found in almost all types of eukaryotic organisms. The system is made up of a variety of proteins which function to deliver intracellular cargo to lysosomes for formation of autophagosomes in which the contents are degraded. The maintenance of cellular homeostasis is key in the survival and function of a variety of human cell populations. The interconnection between metabolism and autophagy is extensive, therefore it has a role in a variety of different cell functions. The disruption or dysfunction of autophagy in these cell types have been implicated in the development of a variety of inflammatory diseases including asthma. The role of autophagy in non-immune and immune cells both lead to the pathogenesis of lung inflammation. Autophagy in pulmonary non-immune cells leads to tissue remodeling which can develop into chronic asthma cases with long term effects. The role autophagy in the lymphoid and myeloid lineages in the pathology of asthma differ in their functions. Impaired autophagy in lymphoid populations have been shown, in general, to decrease inflammation in both asthma and inflammatory disease models. Many lymphoid cells rely on autophagy for effector function and maintained inflammation. In stark contrast, autophagy deficient antigen presenting cells have been shown to have an activated inflammasome. This is largely characterized by a TH17 response that is accompanied with a much worse prognosis including granulocyte mediated inflammation and steroid resistance. The cell specificity associated with changes in autophagic flux complicates its targeting for amelioration of asthmatic symptoms. Differing asthmatic phenotypes between TH2 and TH17 mediated disease may require different autophagic modulations. Therefore, treatments call for a more cell specific and personalized approach when looking at chronic asthma cases. Viral-induced lung inflammation, such as that caused by SARS-CoV-2, also may involve autophagic modulation leading to inflammation mediated by lung resident cells. In this review, we will be discussing the role of autophagy in non-immune cells, myeloid cells, and lymphoid cells for their implications into lung inflammation and asthma. Finally, we will discuss autophagy's role viral pathogenesis, immunometabolism, and asthma with insights into autophagic modulators for amelioration of lung inflammation.
Collapse
Affiliation(s)
- Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
10
|
Kubo F, Ariestanti DM, Oki S, Fukuzawa T, Demizu R, Sato T, Sabirin RM, Hirose S, Nakamura N. Loss of the adhesion G-protein coupled receptor ADGRF5 in mice induces airway inflammation and the expression of CCL2 in lung endothelial cells. Respir Res 2019; 20:11. [PMID: 30654796 PMCID: PMC6337809 DOI: 10.1186/s12931-019-0973-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/02/2019] [Indexed: 01/09/2023] Open
Abstract
Background Adhesion G-protein coupled receptor F5 (ADGRF5) was recently identified as an essential regulator of pulmonary surfactant homeostasis in alveolar type II cells. We previously showed that in addition to abnormal surfactant accumulation, Adgrf5-deficient (Adgrf5−/−) mice exhibit emphysema-like signs, suggesting a possible role for ADGRF5 in immune regulation. Here, we extended the phenotypic analysis of Adgrf5−/− mice to help understand its biological role in the lung, and especially in immune regulation. Methods Histological features of lungs were evaluated by Alcian blue and Masson’s trichrome staining. Quantitative real-time PCR (qPCR) and western blot analyses were performed to analyze the differential expression of genes/proteins related to airway inflammation in lungs between wildtype and Adgrf5−/− mice. Acid–base status was assessed by performing blood gas tests and urine pH measurements. Inflammatory cell counting was performed using Giemsa-stained bronchoalveolar lavage cells. Serum IgE concentrations were determined by enzyme-linked immunosorbent assay. The expression of Ccl2, S100a8, S100a9, and Saa3 in primary lung endothelial cells (ECs) was determined by qPCR and/or western blotting. Finally, the effect of administrating RS504393 to 2-week-old Adgrf5−/− mice on gene expression in the lungs was analyzed by qPCR. Results Adgrf5−/− mice exhibited several features of chronic airway inflammation (mucous cell metaplasia, mucus hyperproduction, subepithelial fibrosis, respiratory acidosis, high serum IgE, mast cell accumulation, and neutrophilia) in parallel with elevated expression of genes involved in mucous cell metaplasia (Muc5ac, Muc5b, Slc26a4, and Clca1), fibrosis (Tgfb1, Col1a1, Fn1, and Tnc), and type 2 immune response (Il4, Il5, Il13, IL-25, and IL-33) at 12 and/or 30 weeks of age. In contrast, mRNA expression of Ccl2, S100a8, and S100a9 was upregulated in embryonic or neonatal Adgrf5−/− lungs as well as in lung ECs of Adgrf5−/− mice at 1 week of age. RS504393 treatment suppressed the upregulation of S100a8, S100a9, Slc26a4, and Il5 in Adgrf5−/− lungs. Conclusions Targeted disruption of ADGRF5 results in the development of airway inflammation, which is likely mediated by the type 2 immune response and possibly CCL2-mediated inflammation. ADGRF5 also has a potential role in the regulation of genes encoding CCL2 in lung ECs, thereby maintaining immune homeostasis. Electronic supplementary material The online version of this article (10.1186/s12931-019-0973-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fumimasa Kubo
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Donna Maretta Ariestanti
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Souta Oki
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Taku Fukuzawa
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Ryotaro Demizu
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Tomoya Sato
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Rahmaningsih Mara Sabirin
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.,Department of Physiology, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, JI.Farmako Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Shigehisa Hirose
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Nobuhiro Nakamura
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.
| |
Collapse
|
11
|
Nobs SP, Kayhan M, Kopf M. GM-CSF intrinsically controls eosinophil accumulation in the setting of allergic airway inflammation. J Allergy Clin Immunol 2018; 143:1513-1524.e2. [PMID: 30244025 DOI: 10.1016/j.jaci.2018.08.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Eosinophils are a therapeutic target in asthmatic patients, and GM-CSF has been suggested to control various aspects of eosinophil biology, including development, function, and survival. However, to date, the role of GM-CSF signaling in eosinophils in vivo is largely unclear. OBJECTIVE We sought to elucidate the role of GM-CSF signaling in asthmatic inflammation. METHODS Wild-type and GM-CSF receptor α (Csf2ra)-deficient mice reconstituted with Csf2ra-proficient alveolar macrophages were subjected to different models of airway inflammation to evaluate the effect of GM-CSF signaling deficiency on asthmatic inflammation in general and on eosinophils in particular. RESULTS We demonstrate that GM-CSF signaling, although being largely dispensable for eosinophil development at steady state, intrinsically promotes accumulation of eosinophils in the lung during allergic airway inflammation. In contrast, chitin-induced eosinophil accumulation in the peritoneal cavity occurs independent of GM-CSF, indicating organ specificity. We show that GM-CSF induces chemokinesis and promotes eosinophil survival in vitro, which likely contribute to eosinophil accumulation in the airways in vivo. CONCLUSION GM-CSF intrinsically promotes eosinophil accumulation in the setting of pulmonary allergic inflammation.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; the Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Carlier FM, Sibille Y, Pilette C. The epithelial barrier and immunoglobulin A system in allergy. Clin Exp Allergy 2016; 46:1372-1388. [PMID: 27684559 DOI: 10.1111/cea.12830] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway and intestinal epithelial layers represent first-line physical barriers, playing a key role in mucosal immunity. Barrier dysfunction, characterized by alterations such as disruption of cell-cell apical junctions and aberrant epithelial responses, probably constitutes early and key events for chronic immune responses to environmental antigens in the skin and in the gut. For instance, barrier dysfunction drives Th2 responses in atopic disorders or eosinophilic esophagitis. Such epithelial impairment is also a salient feature of allergic asthma and growing evidence indicates that barrier alterations probably play a driving role in this disease. IgA has been identified as the most abundant immunoglobulin in mucosa, where it acts as an active barrier through immune exclusion of inhaled or ingested antigens or pathogens. Historically, it has been thought to represent the serum factor underlying reaginic activity before IgE was discovered. Despite several studies about regulation and major functions of IgA at mucosal surfaces, its role in allergy remains largely unclear. This review aims at summarizing findings about epithelial functions and IgA biology that are relevant to allergy, and to integrate the emerging concepts and the recent developments in mucosal immunology, and how these could translate to clinical observations in allergy.
Collapse
Affiliation(s)
- F M Carlier
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium.
| | - Y Sibille
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium
| | - C Pilette
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.,Walloon Excellence in Lifesciences and Biotechnology, Wavre, Belgium
| |
Collapse
|
13
|
Sugawara R, Lee EJ, Jang MS, Jeun EJ, Hong CP, Kim JH, Park A, Yun CH, Hong SW, Kim YM, Seoh JY, Jung Y, Surh CD, Miyasaka M, Yang BG, Jang MH. Small intestinal eosinophils regulate Th17 cells by producing IL-1 receptor antagonist. J Exp Med 2016; 213:555-67. [PMID: 26951334 PMCID: PMC4821642 DOI: 10.1084/jem.20141388] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/05/2016] [Indexed: 12/20/2022] Open
Abstract
Jang et al. show that eosinophils in the small intestine can suppress Th17 cell differentiation through the secretion of the IL-1 receptor antagonist. Eosinophils play proinflammatory roles in helminth infections and allergic diseases. Under steady-state conditions, eosinophils are abundantly found in the small intestinal lamina propria, but their physiological function is largely unexplored. In this study, we found that small intestinal eosinophils down-regulate Th17 cells. Th17 cells in the small intestine were markedly increased in the ΔdblGATA-1 mice lacking eosinophils, and an inverse correlation was observed between the number of eosinophils and that of Th17 cells in the small intestine of wild-type mice. In addition, small intestinal eosinophils suppressed the in vitro differentiation of Th17 cells, as well as IL-17 production by small intestinal CD4+ T cells. Unlike other small intestinal immune cells or circulating eosinophils, we found that small intestinal eosinophils have a unique ability to constitutively secrete high levels of IL-1 receptor antagonist (IL-1Ra), a natural inhibitor of IL-1β. Moreover, small intestinal eosinophils isolated from IL-1Ra−deficient mice failed to suppress Th17 cells. Collectively, our results demonstrate that small intestinal eosinophils play a pivotal role in the maintenance of intestinal homeostasis by regulating Th17 cells via production of IL-1Ra.
Collapse
Affiliation(s)
- Reiko Sugawara
- Department of Respiratory Medicine, Allergy, and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 565-0871 Suita, Japan Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, 565-0871 Suita, Japan WPI Immunology Frontier Research Center, Osaka University, 565-0871 Suita, Japan
| | - Eun-Jung Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Min Seong Jang
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141 Daejeon, Republic of Korea
| | - Eun-Ji Jeun
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Chun-Pyo Hong
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Jung-Hwan Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Areum Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Chang Ho Yun
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea
| | - Sung-Wook Hong
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea
| | - You-Me Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea Department of Life Sciences, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Ju-Young Seoh
- Department of Microbiology, Graduate School of Medicine, Ewha Womans University, 158-710 Seoul, Republic of Korea
| | - YunJae Jung
- Department of Microbiology, School of Medicine, Gachon University, 21936 Incheon, Republic of Korea
| | - Charles D Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, 790-784 Pohang, Republic of Korea
| | - Masayuki Miyasaka
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, 565-0871 Suita, Japan WPI Immunology Frontier Research Center, Osaka University, 565-0871 Suita, Japan MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Bo-Gie Yang
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea
| | - Myoung Ho Jang
- Academy of Immunology and Microbiology, Institute for Basic Science, 790-784 Pohang, Republic of Korea WPI Immunology Frontier Research Center, Osaka University, 565-0871 Suita, Japan
| |
Collapse
|
14
|
Naessens T, Schepens B, Smet M, Pollard C, Van Hoecke L, De Beuckelaer A, Willart M, Lambrecht B, De Koker S, Saelens X, Grooten J. GM-CSF treatment prevents respiratory syncytial virus-induced pulmonary exacerbation responses in postallergic mice by stimulating alveolar macrophage maturation. J Allergy Clin Immunol 2015; 137:700-9.e9. [PMID: 26560044 DOI: 10.1016/j.jaci.2015.09.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 08/06/2015] [Accepted: 09/03/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Human respiratory syncytial virus (RSV) is a frequent cause of asthma exacerbations, yet the susceptibility of asthmatic patients to RSV is poorly understood. OBJECTIVE We sought to address the contribution of resident alveolar macrophages (rAMs) to susceptibility to RSV infection in mice that recovered from allergic airway eosinophilia. METHODS Mice were infected with RSV virus after clearance of allergic airway inflammation (AAI). The contribution of post-AAI rAMs was studied in vivo by means of clodronate liposome-mediated depletion, adoptive transfer, and treatment with recombinant cytokines before RSV infection. RESULTS After clearing the allergic bronchial inflammation, post-AAI mice had bronchial hyperreactivity and increased inflammatory cell influx when infected with RSV compared with nonallergic mice, whereas viral clearance was comparable in both mouse groups. Post-AAI rAMs were necessary and sufficient for mediating these proinflammatory effects. In post-AAI mice the residing CD11c(hi) autofluorescent rAM population did not upregulate the terminal differentiation marker sialic acid-binding immunoglobulin-like lectin F and overproduced TNF and IL-6 through increased nuclear factor κB nuclear translocation. In line with these results, post-AAI lungs had reduced levels of the rAM maturation cytokine GM-CSF. Intratracheal administration of GM-CSF induced final rAM maturation in post-AAI mice and prevented the increased susceptibility to RSV-induced hyperreactivity and inflammation. CONCLUSION Defective production of GM-CSF leads to insufficient post-AAI rAM maturation in mice that recovered from an AAI, causing increased susceptibility to RSV-induced immunopathology. Promoting the differentiation of post-AAI rAMs might be a therapeutic option for preventing RSV-induced exacerbations in human asthmatic patients.
Collapse
Affiliation(s)
- Thomas Naessens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bert Schepens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Inflammation Research Center, VIB, Ghent, Belgium
| | - Muriel Smet
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Charlotte Pollard
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ans De Beuckelaer
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Monique Willart
- Inflammation Research Center, VIB, Ghent, Belgium; Department of Pulmonary Medicine, Ghent University, Ghent, Belgium
| | - Bart Lambrecht
- Inflammation Research Center, VIB, Ghent, Belgium; Department of Pulmonary Medicine, Ghent University, Ghent, Belgium
| | - Stefaan De Koker
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Inflammation Research Center, VIB, Ghent, Belgium
| | - Johan Grooten
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
15
|
Liu L, Wang H, Xenakis JJ, Spencer LA. Notch signaling mediates granulocyte-macrophage colony-stimulating factor priming-induced transendothelial migration of human eosinophils. Allergy 2015; 70:805-12. [PMID: 25846339 PMCID: PMC9875669 DOI: 10.1111/all.12624] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Priming with cytokines such as granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances eosinophil migration and exacerbates the excessive accumulation of eosinophils within the bronchial mucosa of asthmatics. However, mechanisms that drive GM-CSF priming are incompletely understood. Notch signaling is an evolutionarily conserved pathway that regulates cellular processes, including migration, by integrating exogenous and cell-intrinsic cues. This study investigates the hypothesis that the priming-induced enhanced migration of human eosinophils requires the Notch signaling pathway. METHODS Using pan Notch inhibitors and newly developed human antibodies that specifically neutralize Notch receptor 1 activation, we investigated a role for Notch signaling in GM-CSF-primed transmigration of human blood eosinophils in vitro and in the airway accumulation of mouse eosinophils in vivo. RESULTS Notch receptor 1 was constitutively active in freshly isolated human blood eosinophils, and inhibition of Notch signaling or specific blockade of Notch receptor 1 activation during GM-CSF priming impaired priming-enhanced eosinophil transendothelial migration in vitro. Inclusion of Notch signaling inhibitors during priming was associated with diminished ERK phosphorylation, and ERK-MAPK activation was required for GM-CSF priming-induced transmigration. In vivo in mice, eosinophil accumulation within allergic airways was impaired following systemic treatment with Notch inhibitor, or adoptive transfer of eosinophils treated ex vivo with Notch inhibitor. CONCLUSIONS These data identify Notch signaling as an intrinsic pathway central to GM-CSF priming-induced eosinophil tissue migration.
Collapse
Affiliation(s)
- L.Y. Liu
- Division of Allergy and Inflammation; Department of Medicine; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston MA USA
| | - H. Wang
- Division of Allergy and Inflammation; Department of Medicine; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston MA USA
| | - J. J. Xenakis
- Division of Allergy and Inflammation; Department of Medicine; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston MA USA
| | - L. A. Spencer
- Division of Allergy and Inflammation; Department of Medicine; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston MA USA
| |
Collapse
|
16
|
Wang HY, Dai Y, Wang JL, Yang XY, Jiang XG. Anti-CD69 monoclonal antibody treatment inhibits airway inflammation in a mouse model of asthma. J Zhejiang Univ Sci B 2015; 16:622-631. [PMID: 26160720 PMCID: PMC4506953 DOI: 10.1631/jzus.b1400285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/01/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Airway inflammation and airway hyper-responsiveness (AHR) are principle pathological manifestations of asthma. Cluster of differentiation 69 (CD69) is a well-known co-stimulatory factor associated with the activation, proliferation as well as apoptosis of immune cells. This study aims to examine the effect of anti-CD69 monoclonal antibody (mAb) on the pathophysiology of a mouse model of asthma. METHODS A murine model of ovalbumin (OVA)-induced allergic airway inflammation was used in this study. Briefly, mice were injected with 20 μg chicken OVA intraperitoneally on Days 0 and 14, followed by aerosol provocation with 1% (0.01 g/ml) OVA on Days 24, 25, and 26. Anti-CD69 mAb or isotype IgG was injected intraperitoneally after OVA challenge; dexamethasone (DXM) was administrated either before or after OVA challenge. AHR, mucus production, and eosinophil infiltration in the peribronchial area were examined. The levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-5 (IL-5) in bronchoalveolar lavage fluid (BALF) were also assayed as indices of airway inflammation on Day 28 following OVA injection. RESULTS Pretreatment with DXM together with anti-CD69 mAb treatment after OVA provocation completely inhibited AHR, eosinophil infiltration and mucus overproduction, and significantly reduced BALF IL-5. However, treatment with DXM alone after OVA challenge only partially inhibited AHR, eosinophil infiltration and mucus overproduction, and did not diminish BALF IL-5. Treatment with either DXM or anti-CD69 mAb did not alter the concentration of BALF GM-CSF. CONCLUSIONS Anti-CD69 mAb treatment inhibits established airway inflammation as effectively as DXM pretreatment. This study provides a potential alternative therapeutic opportunity for the clinical management of asthma and its exacerbation.
Collapse
Affiliation(s)
- Hui-ying Wang
- Department of Allergy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yu Dai
- Department of Clinical Laboratory, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jiao-li Wang
- Department of Respiratory Medicine, Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Xu-yan Yang
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xin-guo Jiang
- Department of Medicine, VA Palo Alto Health Care System/Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Subramaniam R, Hillberry Z, Chen H, Feng Y, Fletcher K, Neuenschwander P, Shams H. Delivery of GM-CSF to Protect against Influenza Pneumonia. PLoS One 2015; 10:e0124593. [PMID: 25923215 PMCID: PMC4414562 DOI: 10.1371/journal.pone.0124593] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/16/2015] [Indexed: 01/10/2023] Open
Abstract
Background Since adaptive immunity is thought to be central to immunity against influenza A virus (IAV) pneumonias, preventive strategies have focused primarily on vaccines. However, vaccine efficacy has been variable, in part because of antigenic shift and drift in circulating influenza viruses. Recent studies have highlighted the importance of innate immunity in protecting against influenza. Methods Granulocyte-macrophage colony stimulating factor (GM-CSF) contributes to maturation of mononuclear phagocytes, enhancing their capacity for phagocytosis and cytokine production. Results Overexpression of granulocyte macrophage-colony stimulating factor (GM-CSF) in the lung of transgenic mice provides remarkable protection against IAV, which depends on alveolar macrophages (AM). In this study, we report that pulmonary delivery of GM-CSF to wild type young and aged mice abrogated mortality from IAV. Conclusion We also demonstrate that protection is species specific and human GM-CSF do not protect the mice nor stimulates mouse immunity. We also show that IAV-induced lung injury is the culprit for side-effects of GM-CSF in treating mice after IAV infection, and introduce a novel strategy to deliver the GM-CSF to and retain it in the alveolar space even after IAV infection.
Collapse
Affiliation(s)
- Renuka Subramaniam
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
| | - Zachary Hillberry
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
| | - Han Chen
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
| | - Yan Feng
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
| | - Kalyn Fletcher
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
| | - Pierre Neuenschwander
- Biomedical Research, The University of Texas Health Science Center at Tyler, U.S. Highway 271, Tyler, TX, USA
| | - Homayoun Shams
- Center for Pulmonary and Infectious Diseases Control (CPIDC), The University of Texas Health Science Center at Tyler, 11937 U.S. Highway 271, Tyler, TX, United States of America
- * E-mail:
| |
Collapse
|
18
|
Dual proinflammatory and antiviral properties of pulmonary eosinophils in respiratory syncytial virus vaccine-enhanced disease. J Virol 2014; 89:1564-78. [PMID: 25410867 DOI: 10.1128/jvi.01536-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human respiratory syncytial virus (RSV) is a major cause of morbidity and severe lower respiratory tract disease in the elderly and very young, with some infants developing bronchiolitis, recurrent wheezing, and asthma following infection. Previous studies in humans and animal models have shown that vaccination with formalin-inactivated RSV (FI-RSV) leads to prominent airway eosinophilic inflammation following RSV challenge; however, the roles of pulmonary eosinophilia in the antiviral response and in disease pathogenesis are inadequately understood. In vivo studies in mice with eotaxin and/or interleukin 5 (IL-5) deficiency showed that FI-RSV vaccination did not lead to enhanced pulmonary disease, where following challenge there were reduced pulmonary eosinophilia, inflammation, Th2-type cytokine responses, and altered chemokine (TARC and CCL17) responses. In contrast to wild-type mice, RSV was recovered at high titers from the lungs of eotaxin- and/or IL-5-deficient mice. Adoptive transfer of eosinophils to FI-RSV-immunized eotaxin- and IL-5-deficient (double-deficient) mice challenged with RSV was associated with potent viral clearance that was mediated at least partly through nitric oxide. These studies show that pulmonary eosinophilia has dual outcomes: one linked to RSV-induced airway inflammation and pulmonary pathology and one with innate features that contribute to a reduction in the viral load. IMPORTANCE This study is critical to understanding the mechanisms attributable to RSV vaccine-enhanced disease. This study addresses the hypothesis that IL-5 and eotaxin are critical in pulmonary eosinophil response related to FI-RSV vaccine-enhanced disease. The findings suggest that in addition to mediating tissue pathology, eosinophils within a Th2 environment also have antiviral activity.
Collapse
|
19
|
Makihara S, Okano M, Fujiwara T, Noda Y, Higaki T, Miyateke T, Kanai K, Haruna T, Kariya S, Nishizaki K. Local expression of interleukin-17a is correlated with nasal eosinophilia and clinical severity in allergic rhinitis. ALLERGY & RHINOLOGY 2014; 5:22-7. [PMID: 24758732 PMCID: PMC4019741 DOI: 10.2500/ar.2014.5.0078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interleukin (IL)-17A is a major cytokine produced by Th17 cells, which are associated with chronic inflammations. The local expression of IL-17A in allergic rhinitis (AR) remains to be characterized. We sought to determine the role of IL-17A expression in human inferior turbinate mucosa in the pathophysiology of AR. Inferior turbinate mucosa was sampled from medical treatment–resistant, surgery-required patients with perennial AR (PAR, n = 21), nonallergic rhinitis with eosinophilia syndrome (NARES, n = 7), and nonallergic hypertrophic rhinitis (HR, n = 13). IL-17A expression was determined with immunohistochemical staining. The mean number of IL-17A+ cells and eosinophils per field were counted. Total serum immunoglobulin E (IgE) levels, blood eosinophil count, and forced expiratory volume in 1 second (FEV1)/forced vital capacity (FVC) ratio were also examined in each patient. IL-17A was primarily expressed in infiltrating inflammatory cells. The number of IL-17A+ cells in nasal mucosa was significantly higher in the PAR group compared with HR (p = 0.002) and NARES (p = 0.021) groups. There was a significant and positive correlation between the number of IL-17A+ cells and total nasal symptom score (rho = 0.403; p = 0.011), especially sneezing score (rho = 0.471; p = 0.003). The number of IL-17A+ cells was significantly and positively correlated with the degree of eosinophil infiltration (rho = 0.623; p < 0.001), but not with total serum IgE levels (rho = 0.284; p = 0.098), blood eosinophil counts (rho = 0.302; p = 0.056), or FEV1/FVC ratio (rho = 0.092; p = 0.569). The present study provides evidence that IL-17A expression in the nasal mucosa is associated with the pathophysiology of AR, including disease severity and nasal eosinophilia.
Collapse
Affiliation(s)
- Seiichiro Makihara
- Department of Otolaryngology-Head and Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Komatsuzaki T, Suzaki I, Hirano K, Kanai KI, Asano K, Suzaki H. Suppression of osteopontin functions by levocetirizine, a histamine H1 receptor antagonist, in vitro. BIOMED RESEARCH INTERNATIONAL 2013; 2013:735835. [PMID: 24490170 PMCID: PMC3893813 DOI: 10.1155/2013/735835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Osteopontin (OPN), a multifunctional glycoprotein secreted from a wide variety of cells after inflammatory stimulation, is well accepted to contribute to the development of allergic diseases. However, the influence of histamine H1 receptor antagonists (antihistamines) on OPN functions is not well understood. The present study was undertaken to examine the influence of antihistamines on OPN functions in vitro. METHODS Human nasal epithelial cells (5 × 10(5) cells) were stimulated with 250 ng/mL OPN in the presence of either desloratadine (DL), fexofenadine (FEX), or levocetirizine (LCT). The levels of OPN, GM-CSF, Eotaxin, and RANTES in 24 h culture supernatants were examined by ELISA. The influence of LCT on mRNA expression and transcription factor activation in cells were also examined by real-time RT-PCR and ELISA, respectively. KEY FINDINGS The antihistamines examined significantly suppressed the production of GM-CSF, Eotaxin, and RANTES from cells after OPN stimulation. LCT also exhibited the suppression of mRNA expression for chemokines and transcription factor, NF- κ B and AP-1, activation, which were increased by the stimulation of cells with OPN. CONCLUSIONS The suppressive activity of LCT on OPN functions on nasal epithelial cells may be responsible for the attenuating effect of the agent on allergic diseases.
Collapse
Affiliation(s)
- Toshimitsu Komatsuzaki
- Department of Otorhinolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Isao Suzaki
- Department of Otorhinolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Kojiro Hirano
- Department of Otorhinolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Ken-Ichi Kanai
- Department of Otorhinolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Kazuhito Asano
- Division of Physiology, School of Nursing and Rehabilitation Sciences, Showa University, Yokohama 226-8555, Japan
| | - Harumi Suzaki
- Department of Otorhinolaryngology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
21
|
The role of macrophages in obstructive airways disease: chronic obstructive pulmonary disease and asthma. Cytokine 2013; 64:613-25. [PMID: 24084332 DOI: 10.1016/j.cyto.2013.09.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/04/2013] [Accepted: 09/06/2013] [Indexed: 12/15/2022]
Abstract
Macrophages are a major cellular component of the innate immune system, and play an important role in the recognition of microbes, particulates, and immunogens and to the regulation of inflammatory responses. In the lung, macrophages react with soluble proteins that bind microbial products in order to remove pathogens and particles and to maintain the sterility of the airway tract. Chronic obstructive pulmonary disease and asthma are both obstructive airway diseases that involve chronic inflammation of the respiratory tract which contributes to disease progression. In the case of COPD, there is increasing evidence that lung macrophages orchestrate inflammation through the release of chemokines that attract neutrophils, monocytes and T cells and the release of several proteases. On the other hand, in asthma, it seems that alveolar macrophages are inappropriately activated and are implicated in the development and progression of the disease. In this review we summarize the current basic and clinical research studies which highlight the role of macrophages in asthma and COPD.
Collapse
|
22
|
Dalrymple H, Barna BP, Malur A, Malur AG, Kavuru MS, Thomassen MJ. Alveolar macrophages of GM-CSF knockout mice exhibit mixed M1 and M2 phenotypes. BMC Immunol 2013; 14:41. [PMID: 24044676 PMCID: PMC3848434 DOI: 10.1186/1471-2172-14-41] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 08/29/2013] [Indexed: 11/24/2022] Open
Abstract
Background Activin A is a pleiotrophic regulatory cytokine, the ablation of which is neonatal lethal. Healthy human alveolar macrophages (AMs) constitutively express activin A, but AMs of patients with pulmonary alveolar proteinosis (PAP) are deficient in activin A. PAP is an autoimmune lung disease characterized by neutralizing autoantibodies to Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF). Activin A can be stimulated, however, by GM-CSF treatment of AMs in vitro. To further explore pulmonary activin A regulation, we examined AMs in bronchoalveolar lavage (BAL) from wild-type C57BL/6 compared to GM-CSF knockout mice which exhibit a PAP-like histopathology. Both human PAP and mouse GM-CSF knockout AMs are deficient in the transcription factor, peroxisome proliferator activated receptor gamma (PPARγ). Results In sharp contrast to human PAP, activin A mRNA was elevated in mouse GM-CSF knockout AMs, and activin A protein was increased in BAL fluid. Investigation of potential causative factors for activin A upregulation revealed intrinsic overexpression of IFNγ, a potent inducer of the M1 macrophage phenotype, in GM-CSF knockout BAL cells. IFNγ mRNA was not elevated in PAP BAL cells. In vitro studies confirmed that IFNγ stimulated activin A in wild-type AMs while antibody to IFNγ reduced activin A in GM-CSF knockout AMs. Both IFNγ and Activin A were also reduced in GM-CSF knockout mice in vivo after intratracheal instillation of lentivirus-PPARγ compared to control lentivirus vector. Examination of other M1 markers in GM-CSF knockout mice indicated intrinsic elevation of the IFNγ-regulated gene, inducible Nitrogen Oxide Synthetase (iNOS), CCL5, and interleukin (IL)-6 compared to wild-type. The M2 markers, IL-10 and CCL2 were also intrinsically elevated. Conclusions Data point to IFNγ as the primary upregulator of activin A in GM-CSF knockout mice which in addition, exhibit a unique mix of M1-M2 macrophage phenotypes.
Collapse
Affiliation(s)
- Heidi Dalrymple
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care Medicine and Sleep Medicine, East Carolina University, Brody School of Medicine, 3E-149 Brody Medical Sciences Building, Greenville, NC 27834, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Asthma is increasing in prevalence worldwide. It is characterized by typical symptoms and variable airway obstruction punctuated with episodes of worsening symptoms known as exacerbations. Underlying this clinical expression of disease is airway inflammation and remodeling. Cytokines and their networks are implicated in the innate and adaptive immune responses driving airway inflammation in asthma and are modulated by host-environment interactions. Asthma is a complex heterogeneous disease, and the paradigm of Th2 cytokine-mediated eosinophilic inflammation as a consequence of allergic sensitization has been challenged and probably represents a subgroup of asthma. Indeed, as attention has switched to the importance of severe asthma, which represents the highest burden both to the patient and health care provider, there is an increasing recognition of inflammatory subphenotypes that are likely to be driven by different cytokine networks. Interestingly, these networks may be specific to aspects of clinical expression as well as inflammatory cell profiles and therefore present novel phenotype-specific therapeutic strategies. Here, we review the breadth of cytokines implicated in the pathogenesis of asthma and focus upon the outcomes of early clinical trials conducted using cytokines or cytokine-blocking therapies.
Collapse
Affiliation(s)
- Dhananjay Desai
- Department of Infection, Inflammation and Immunity, Institute for Lung Health, University of Leicester, Glenfield Hospital, Leicester, UK
| | | |
Collapse
|
24
|
Sheikh Bahaie N, Rao SP, Massoud A, Sriramarao P. GM-CSF differentially regulates eosinophil and neutrophil adhesive interactions with vascular endothelium in vivo. IRANIAN JOURNAL OF ALLERGY, ASTHMA AND IMMUNOLOGY 2011; 9:207-17. [PMID: 21131700 DOI: 09.04/ijaai.207217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Allergic airway inflammation is characterized by elaboration of cytokines and chemokines leading to recruitment of inflammatory leukocytes, predominantly eosinophils, to the airways. Granulocyte macrophage colony stimulating factor (GM-CSF) is generated in the lungs of human subjects with asthma in response to allergen challenge and is necessary for the development of allergen-induced bronchial eosinophilia in mice. The effect of GM-CSF on human eosinophil and neutrophil interactions with the vascular endothelium under conditions of blood flow was investigated in post-capillary venules of the rabbit mesentery by intravital microscopy.While GM-CSF significantly reduced the rolling fraction of neutrophils in vivo and induced consistent shedding of neutrophil L-selectin in vitro, its effect on eosinophil rolling was variable. Eosinophils from 57% of the donors demonstrated inhibition of rolling, while eosinophils from the remaining 43% of donors demonstrated no inhibition or increased rolling. The variable effect of GM-CSF on inhibition of eosinophil rolling was associated with variable shedding of L-selectin in vitro. In contrast to the differential effect of GM-CSF on neutrophils versus eosinophils, stimulation with phorbol myristate acetate demonstrated a similar degree of inhibition of rolling and L-selectin shedding by neutrophils and eosinophils suggesting that there was no defect in L-selectin shedding in the eosinophil donors who did not respond to GM-CSF. Overall, these studies demonstrate that GM-CSF consistently inhibits interaction of neutrophils with endothelium in vivo, whereas its effect on eosinophil-endothelial interactions is variable. GM-CSF may thus be one factor accounting for the varying percentage of eosinophils and neutrophils recruited to sites of allergic inflammation in different individuals.
Collapse
Affiliation(s)
- Nooshin Sheikh Bahaie
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota, St. Paul, MN, USA
| | | | | | | |
Collapse
|
25
|
Moreira AP, Hogaboam CM. Macrophages in allergic asthma: fine-tuning their pro- and anti-inflammatory actions for disease resolution. J Interferon Cytokine Res 2011; 31:485-91. [PMID: 21631355 DOI: 10.1089/jir.2011.0027] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Macrophages exert prominent effects in the defense of the respiratory tract from airborne pathogens. These cells are specialized to recognize, phagocytose, and destroy these infectious agents and then promote appropriate tissue repair after successful pathogen clearance. For reasons that are not presently clear, macrophages appear to be inappropriately activated during asthma responses. Evidence stems from the appearance of either classically (or M1) and alternatively activated (or M2) cells in the alveolar compartment of asthmatic lung. Macrophages localized in the interstitial area of the lung appear to be less prone to polarization toward either the M1 or M2 phenotype as these cells predominately express interleukin-10 and exhibit immunoregulatory properties. Effective treatment of clinical asthma, regardless of severity, might depend on restoring an appropriate balance between M1, M2, and immunoregulatory macrophages in the lung.
Collapse
Affiliation(s)
- Ana Paula Moreira
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | |
Collapse
|
26
|
Protection against Nippostrongylus brasiliensis infection in mice is independent of GM-CSF. Immunol Cell Biol 2011; 90:553-8. [PMID: 21844882 DOI: 10.1038/icb.2011.69] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Granulocyte macrophage-colony stimulating factor (GM-CSF) is a cytokine with the capacity to promote inflammation in a wide variety of infectious and inflammatory diseases. These conditions include allergic airway inflammation, which is driven by T-helper 2 (Th2) cells. Because of the importance of Th2 cells in parasite infections, we have investigated the role of GM-CSF in mice infected with the nematode Nippostrongylus brasiliensis. The effect of primary and secondary infection was investigated in mice lacking functional genes for GM-CSF (CSF2 genes) (ΔGM-CSF mice), and in mice lacking the cytokine receptor common β chain (Δβ mice), the latter being unable to signal in response to GM-CSF and interleukin (IL)-5. ΔGM-CSF mice showed no significant defect in parasite immunity, measured by larval numbers in the lungs, worm numbers in the intestine or egg numbers in the faeces, in either primary or secondary infection. By contrast, the Δβ mice showed increased parasite burden, with higher numbers of lung larvae after secondary infection and higher numbers of intestinal worms and faecal eggs after both primary and secondary infection. Unexpectedly, there were increased numbers of circulating eosinophils in the ΔGM-CSF mice, associated with significantly reduced larval numbers in the lungs. These results indicate that GM-CSF is redundant in protection against N. brasiliensis infection, and that the increased susceptibility of Δβ mice to infection is likely to be attributed to the lack of IL-5 signalling in these mice. The results suggest that clinical use of agents that neutralise GM-CSF may not be associated with increased risk of parasite infection.
Collapse
|
27
|
Makihara S, Okano M, Fujiwara T, Kariya S, Noda Y, Higaki T, Nishizaki K. Regulation and characterization of IL-17A expression in patients with chronic rhinosinusitis and its relationship with eosinophilic inflammation. J Allergy Clin Immunol 2010; 126:397-400, 400.e1-11. [PMID: 20621345 DOI: 10.1016/j.jaci.2010.05.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Accepted: 05/03/2010] [Indexed: 10/19/2022]
|
28
|
Huvenne W, Callebaut I, Reekmans K, Hens G, Bobic S, Jorissen M, Bullens DMA, Ceuppens JL, Bachert C, Hellings PW. Staphylococcus aureus enterotoxin B augments granulocyte migration and survival via airway epithelial cell activation. Allergy 2010; 65:1013-20. [PMID: 20132156 DOI: 10.1111/j.1398-9995.2009.02313.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Staphylococcus aureus enterotoxin B (SEB) has recently been postulated to be involved in the pathology of granulocyte-dominated disease. Studying the immunologic interaction between SEB and airway epithelial cells in immortalized cell lines or long-term epithelial cell cultures has obvious disadvantages. METHODS We used a novel technique of freshly isolated and purified human nasal epithelial cells (HNEC) from healthy, nonallergic individuals, which were incubated for 24 h without/with SEB at different concentrations. Chemokine production was evaluated in the supernatant using Cytometric Bead Array. The chemotactic activity of the supernatant was studied in vitro using a Boyden chamber. Survival was evaluated with flow cytometry, using propidium iodide to identify dead cells. RESULTS Staphylococcus aureus enterotoxin B showed a dose-dependent induction of interferon-inducible protein-10, monokine induced by interferon-gamma, regulated upon activation normal T cell expressed and secreted, monocyte chemoattractant protein 1 (MCP-1) and granulocyte colony stimulating factor production by epithelial cells in vitro. The supernatant of epithelial cells had chemotactic activity for granulocytes in vitro, which was enhanced in the supernatant of SEB-stimulated epithelial cells. Reduced number of propidium iodide positive granulocytes was found in the conditions where supernatant of SEB-stimulated epithelial cells was applied. CONCLUSION Staphylococcus aureus enterotoxin B exerts a direct pro-inflammatory effect on HNEC, with induction of chemokine and growth factor release, resulting in the migration and prolonged survival of granulocytes in vitro.
Collapse
Affiliation(s)
- W Huvenne
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Finkelman FD, Hogan SP, Hershey GKK, Rothenberg ME, Wills-Karp M. Importance of cytokines in murine allergic airway disease and human asthma. THE JOURNAL OF IMMUNOLOGY 2010; 184:1663-74. [PMID: 20130218 DOI: 10.4049/jimmunol.0902185] [Citation(s) in RCA: 229] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Asthma is a common, disabling inflammatory respiratory disease that has increased in frequency and severity in developed nations. We review studies of murine allergic airway disease (MAAD) and human asthma that evaluate the importance of Th2 cytokines, Th2 response-promoting cytokines, IL-17, and proinflammatory and anti-inflammatory cytokines in MAAD and human asthma. We discuss murine studies that directly stimulate airways with specific cytokines or delete, inactivate, neutralize, or block specific cytokines or their receptors, as well as controversial issues including the roles of IL-5, IL-17, and IL-13Ralpha2 in MAAD and IL-4Ralpha expression by specific cell types. Studies of human asthmatic cytokine gene and protein expression, linkage of cytokine polymorphisms to asthma, cytokine responses to allergen stimulation, and clinical responses to cytokine antagonists are discussed as well. Results of these analyses establish the importance of specific cytokines in MAAD and human asthma and have therapeutic implications.
Collapse
Affiliation(s)
- Fred D Finkelman
- Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220, USA.
| | | | | | | | | |
Collapse
|
30
|
Desai D, Brightling C. Cytokine and anti-cytokine therapy in asthma: ready for the clinic? Clin Exp Immunol 2009; 158:10-9. [PMID: 19737225 PMCID: PMC2759053 DOI: 10.1111/j.1365-2249.2009.03998.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2009] [Indexed: 01/13/2023] Open
Abstract
Asthma is a common disease with an increasing prevalence worldwide. Up to 10% of these patients have asthma that is refractory to current therapy. This group have a disproportionate use of health care resources attributed to asthma, have significant morbidity and mortality and therefore represent an unmet clinical need. Asthma is a complex heterogeneous condition that is characterized by typical symptoms and disordered airway physiology set against a background of airway inflammation and remodelling. The inflammatory process underlying asthma is co-ordinated by a cytokine network. Modulating this network with biological therapy presents a new paradigm for asthma treatment. Clinical trials undertaken to date have underscored the complexity of the inflammatory profile and its relationship to the clinical features of the disease and have raised the importance of safety considerations related to these novel therapies. T helper type 2 cytokine blockade remains the most promising strategy, with anti-interleukin-5 reducing asthma exacerbations. Although anti-cytokine therapy is not yet ready for the clinic, the long-awaited possibility of new treatments for severe asthma is moving ever closer.
Collapse
Affiliation(s)
- D Desai
- Department of Infection, Inflammation and Immunity, University of Leicester, Leicester, UK
| | | |
Collapse
|
31
|
Malur A, Mccoy AJ, Arce S, Barna BP, Kavuru MS, Malur AG, Thomassen MJ. Deletion of PPARγ in Alveolar Macrophages Is Associated with a Th-1 Pulmonary Inflammatory Response. THE JOURNAL OF IMMUNOLOGY 2009; 182:5816-22. [DOI: 10.4049/jimmunol.0803504] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Saha S, Doe C, Mistry V, Siddiqui S, Parker D, Sleeman M, Cohen ES, Brightling CE. Granulocyte-macrophage colony-stimulating factor expression in induced sputum and bronchial mucosa in asthma and COPD. Thorax 2009; 64:671-6. [PMID: 19213775 PMCID: PMC2712140 DOI: 10.1136/thx.2008.108290] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Granulocyte–macrophage colony-stimulating factor (GM-CSF) has been implicated as an important mediator in the pathogenesis of asthma and chronic obstructive pulmonary disease (COPD). However, the expression of GM-CSF and its receptor in airway samples in asthma and COPD across disease severity needs to be further defined. Methods: Sputum GM-CSF was measured in 18 control subjects, 45 subjects with asthma and 47 subjects with COPD. Enumeration of GM-CSF+ cells in the bronchial submucosa and airway smooth muscle bundle was performed in 29 control subjects, 36 subjects with asthma and 10 subjects with COPD. Results: The proportion of subjects with measurable GM-CSF in the sputum was raised in those with moderate (7/14) and severe (11/18) asthma, and in those with COPD GOLD (Global Initiative for Chronic Obstructive Lung Disease) stage II (7/16), III (8/17) and IV (7/14) compared with controls (1/18) and those with mild asthma (0/13); p = 0.001. The sputum GM-CSF concentration was correlated with the sputum eosinophilia in subjects with moderate to severe asthma (rs = 0.41; p = 0.018). The median (interquartile range) GM-CSF+ and GM-CSFR+ cells/mm2 of submucosa was increased in severe asthma (1.4 (3.0) and 2.1 (8.4)) compared with those with mild to moderate asthma (0 (2.5) and 1.1 (5)) and healthy controls (0 (0.5) and 0 (1.6)), (p = 0.004 and p = 0.02, respectively). Conclusions: The findings support a potential role for GM-CSF in asthma and COPD and suggest that overexpression of GM-CSF in sputum and the bronchial mucosa is a particular feature of severe asthma.
Collapse
Affiliation(s)
- S Saha
- Institute for Lung Health, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Groby Road, Leicester, UK
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Bogaert P, Tournoy KG, Naessens T, Grooten J. Where asthma and hypersensitivity pneumonitis meet and differ: noneosinophilic severe asthma. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:3-13. [PMID: 19074616 PMCID: PMC2631313 DOI: 10.2353/ajpath.2009.071151] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/17/2008] [Indexed: 11/20/2022]
Abstract
Asthma is a type-I allergic airway disease characterized by Th(2) cells and IgE. Episodes of bronchial inflammation, eosinophilic in nature and promoting bronchoconstriction, may become chronic and lead to persistent respiratory symptoms and irreversible structural airway changes. Representative mostly of mild to moderate asthma, this clinical definition fails to account for the atypical and often more severe phenotype found in a considerable proportion of asthmatics who have increased neutrophil cell counts in the airways as a distinguishing trait. Neutrophilic inflammation is a hallmark of another type of allergic airway pathology, hypersensitivity pneumonitis. Considered as an immune counterpart of asthma, hypersensitivity pneumonitis is a prototypical type-III allergic inflammatory reaction involving the alveoli and lung interstitium, steered by Th(1) cells and IgG and, in its chronic form, accompanied by fibrosis. Although pathologically very different and commonly approached as separate disorders, as discussed in this review, clinical studies as well as data from animal models reveal undeniable parallels between both airway diseases. Danger signaling elicited by the allergenic agent or by accompanying microbial patterns emerges as critical in enabling immune sensitization and in determining the type of sensitization and ensuing allergic disease. On this basis, we propose that asthma allergens cause severe noneosinophilic asthma because of sensitization in the presence of hypersensitivity pneumonitis-promoting danger signaling.
Collapse
Affiliation(s)
- Pieter Bogaert
- Department of Molecular Biomedical Research, Ghent University, Ghent, Belgium
| | | | | | | |
Collapse
|
34
|
Abstract
Although they were originally defined as haematopoietic-cell growth factors, colony-stimulating factors (CSFs) have been shown to have additional functions by acting directly on mature myeloid cells. Recent data from animal models indicate that the depletion of CSFs has therapeutic benefit in many inflammatory and/or autoimmune conditions and as a result, early-phase clinical trials targeting granulocyte/macrophage colony-stimulating factor and macrophage colony-stimulating factor have now commenced. The distinct biological features of CSFs offer opportunities for specific targeting, but with some associated risks. Here, I describe these biological features, discuss the probable specific outcomes of targeting CSFs in vivo and highlight outstanding questions that need to be addressed.
Collapse
|
35
|
Mechanisms in allergic airway inflammation - lessons from studies in the mouse. Expert Rev Mol Med 2008; 10:e15. [PMID: 18503727 DOI: 10.1017/s1462399408000707] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Asthma is a chronic inflammatory disease of the airways, involving recurrent episodes of airway obstruction and wheezing. A common pathological feature in asthma is the presence of a characteristic allergic airway inflammatory response involving extensive leukocyte infiltration, mucus overproduction and airway hyper-reactivity. The pathogenesis of allergic airway inflammation is complex, involving multiple cell types such as T helper 2 cells, regulatory T cells, eosinophils, dendritic cells, mast cells, and parenchymal cells of the lung. The cellular response in allergic airway inflammation is controlled by a broad range of bioactive mediators, including IgE, cytokines and chemokines. The asthmatic allergic inflammatory response has been a particular focus of efforts to develop novel therapeutic agents. Animal models are widely used to investigate inflammatory mechanisms. Although these models are not perfect replicas of clinical asthma, such studies have led to the development of numerous novel therapeutic agents, of which some have already been successful in clinical trials.
Collapse
|