1
|
Feng L, Wu Z, Jia X, Yang L, Wang M, Huang M, Ma Y. Screening, identification and targeted intervention of necroptotic biomarkers of asthma. Biochem Biophys Res Commun 2024; 735:150674. [PMID: 39270557 DOI: 10.1016/j.bbrc.2024.150674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/22/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND As a pivotal pathway of programmed cell death, necroptosis significantly contributes to the pathogenesis of respiratory disorders. However, its role in asthma is not yet fully elucidated. Therefore, this study aimed to identify markers associated with necroptosis, evaluate their functions in asthma, and explore potential therapeutic agents targeting necroptosis for the management of asthma. METHODS Firstly, machine learning algorithms, including Least Absolute Shrinkage and Selection Operator (LASSO), Random Forest, and Support Vector Machine-Recursive Feature Elimination (SVM-RFE), were utilized to identify necroptosis-related differentially expressed genes (NRDEGs) in asthma patients compared to healthy controls. Concurrently, the expression of NRDEGs was validated using external datasets, Western blot, and quantitative real-time polymerase chain reaction (qPCR). Secondly, the clinical relevance of NRDEGs was assessed through Receiver Operating Characteristic (ROC) curve analysis and correlation with clinical indicators. Thirdly, the relationship between NRDEGs and pulmonary immune cell infiltration, as well as the signaling interactions between different cells types, were analyzed through immune infiltration and single-cell analysis. Fourthly, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA), were conducted to elucidate the functional roles of NRDEGs. Finally, compounds targeting NRDEGs were screened, and their binding affinities were evaluated using molecular docking studies. RESULTS In asthma, necroptosis is activated, leading to the identification of four NRDEGs: NLRP3, PYCARD, ALOX15, and VDAC3. Among these, NLRP3, PYCARD, and ALOX15 are upregulated, whereas VDAC3 is downregulated in asthma. Comprehensive clinical evaluations indicated that NRDEGs hold diagnostic value for asthma. Specifically, NLRP3 was inversely correlated with forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC), while VDAC3 showed an inverse correlation with sputum neutrophils. Conversely, ALOX15 expression was positively correlated with fractional exhaled nitric oxide (FeNO) levels, as well as sputum eosinophils, blood eosinophils, and blood IgE levels. Subsequent immune infiltration analysis revealed associations between NRDEGs and activated dendritic cells, mast cells, and eosinophils. Single-cell RNA sequencing (scRNA-seq) further confirmed the communication signals between myeloid dendritic cells, fibroblasts, neutrophils, and helper T cells, predominantly related to fibrosis and immune-inflammatory responses. Pathway enrichment analysis demonstrated that NRDEGs are involved in ribosomal function, oxidative phosphorylation, and fatty acid metabolism. Finally, resveratrol and triptonide were identified as potential therapeutic agents targeting the proteins encoded by NRDEGs for asthma treatment. CONCLUSIONS The necroptosis pathway is activated in asthma, with NRDEGs-namely PYCARD, NLRP3, ALOX15, and VDAC3-correlated with declines in lung function and airway inflammation. These genes serve as reliable predictors of asthma risk and are involved in the regulation of the immune-inflammatory microenvironment. Resveratrol and triptolide have been identified as promising therapeutic candidates due to their potential to target the proteins encoded by these genes.
Collapse
Affiliation(s)
- Ling Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenzhen Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuan Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Xu B, Chandrasekar A, olde Heuvel F, Powerski M, Nowak A, Noack L, Omari J, Huber-Lang M, Roselli F, Relja B. Ethanol Intoxication Alleviates the Inflammatory Response of Remote Organs to Experimental Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21218181. [PMID: 33142949 PMCID: PMC7663496 DOI: 10.3390/ijms21218181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) may cause damage to distant organs. Acute ethanol intoxication (EI) induces complex local and systemic anti-inflammatory effects and influences the early outcomes of traumatized patients. Here, we evaluated its effects on the BI-induced expression of local inflammatory mediators in the trauma-remote organs the lungs and liver. Male mice were exposed to ethanol as a single oral dose (5g·kg–1, 32%) before inducing a moderate blunt TBI. Sham groups underwent the same procedures without TBI. Ether 3 or 6h after the TBI, the lung and liver were collected. The gene expression of HMGB1, IL-6, MMP9, IL-1β, and TNF as well as the homogenate protein levels of receptor for advanced glycation end products (RAGE), IL-6, IL-1β, and IL-10 were analyzed. Liver samples were immunohistologically stained for HMGB1. EI decreased the gene expressions of the proinflammatory markers HMGB1, IL-6, and MMP9 in the liver upon TBI. In line with the reduced gene expression, the TBI-induced protein expression of IL-6 in liver tissue homogenates was significantly reduced by EI at 3h after TBI. While the histological HMGB1 expression was enhanced by TBI, the RAGE protein expression in the liver tissue homogenates was diminished after TBI. EI reduced the histological HMGB1 expression and enhanced the hepatic RAGE protein expression at 6h post TBI. With regard to the lungs, EI significantly reduced the gene expressions of HMGB1, IL-6, IL-1β, and TNF upon TBI, without significantly affecting the protein expression levels of inflammatory markers (RAGE, IL-6, IL-1β, and IL-10). At the early stage of TBI-induced inflammation, the gene expression of inflammatory mediators in both the lungs and liver is susceptible to ethanol-induced remote effects. Taken together, EI may alleviate the TBI-induced pro-inflammatory response in the trauma-distant organs, the lungs and liver, via the HMGB1-RAGE axis.
Collapse
Affiliation(s)
- Baolin Xu
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
| | - Akila Chandrasekar
- Department of Neurology, Ulm University, 89081 Ulm, Germany; (A.C.); (F.o.H.)
| | - Florian olde Heuvel
- Department of Neurology, Ulm University, 89081 Ulm, Germany; (A.C.); (F.o.H.)
| | - Maciej Powerski
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
| | - Aleksander Nowak
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
| | - Laurens Noack
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
| | - Jazan Omari
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm University, 89081 Ulm, Germany;
| | - Francesco Roselli
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, 89081 Ulm, Germany;
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany; (B.X.); (M.P.); (A.N.); (L.N.); (J.O.)
- Correspondence:
| |
Collapse
|
3
|
Olde Heuvel F, Holl S, Chandrasekar A, Li Z, Wang Y, Rehman R, Förstner P, Sinske D, Palmer A, Wiesner D, Ludolph A, Huber-Lang M, Relja B, Wirth T, Röszer T, Baumann B, Boeckers T, Knöll B, Roselli F. STAT6 mediates the effect of ethanol on neuroinflammatory response in TBI. Brain Behav Immun 2019; 81:228-246. [PMID: 31207335 DOI: 10.1016/j.bbi.2019.06.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/31/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1β, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1β, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.
Collapse
Affiliation(s)
- Florian Olde Heuvel
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Sarah Holl
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Akila Chandrasekar
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Zhenghui Li
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Yibin Wang
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rida Rehman
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Philip Förstner
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Daniela Sinske
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Diana Wiesner
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Albert Ludolph
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Borna Relja
- Dept. of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tobias Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Francesco Roselli
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
4
|
Hsieh T, Vaickus MH, Stein TD, Lussier BL, Kim J, Stepien DM, Duffy ER, Chiswick EL, Remick DG. The Role of Substance P in Pulmonary Clearance of Bacteria in Comparative Injury Models. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 186:3236-3245. [PMID: 27876152 DOI: 10.1016/j.ajpath.2016.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/11/2016] [Accepted: 08/29/2016] [Indexed: 01/12/2023]
Abstract
Neural input to the immune system can alter its ability to clear pathogens effectively. Patients suffering mild traumatic brain injury (mTBI) have shown reduced rates of pneumonia and a murine model replicated these findings, with better overall survival of TBI mice compared with sham-injured mice. To further investigate the mechanism of improved host response in TBI mice, this study developed and characterized a mild tail trauma model of similar severity to mild TBI. Both mild tail trauma and TBI induced similar systemic changes that normalized within 48 hours, including release of substance P. Examination of tissues showed that injuries are limited to the target tissue (ie, tail in tail trauma, brain in mTBI). Pneumonia challenge showed that mild TBI mice showed improved immune responses, characterized by the following: i) increased survival, ii) increased pulmonary neutrophil recruitment, iii) increased bacterial clearance, and iv) increased phagocytic cell killing of bacteria compared with tail trauma. Administration of a neurokinin-1-receptor antagonist to block substance P signaling eliminated the improved survival of mTBI mice. Neurokinin-1-receptor antagonism did not alter pneumonia mortality in tail trauma mice. These data show that immune benefits of trauma are specific to mTBI and that tail trauma is an appropriate control for future studies aimed at elucidating the mechanisms of improved innate immune responses in mTBI mice.
Collapse
Affiliation(s)
- Terry Hsieh
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Max H Vaickus
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Thor D Stein
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts; VA Boston Healthcare System, Boston, Massachusetts; Department of Veterans Affairs Medical Center, Bedford, Massachusetts
| | - Bethany L Lussier
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Jiyoun Kim
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - David M Stepien
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Elizabeth R Duffy
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Evan L Chiswick
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Daniel G Remick
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
5
|
Chandrasekar A, Heuvel FO, Palmer A, Linkus B, Ludolph AC, Boeckers TM, Relja B, Huber-Lang M, Roselli F. Acute ethanol administration results in a protective cytokine and neuroinflammatory profile in traumatic brain injury. Int Immunopharmacol 2017; 51:66-75. [PMID: 28806641 DOI: 10.1016/j.intimp.2017.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/01/2022]
Abstract
Ethanol intoxication is a common comorbidity in traumatic brain injury. To date, the effect of ethanol on TBI pathogenic cascades and resulting outcomes remains debated. A closed blunt weight-drop murine TBI model has been implemented to investigate behavioral (by sensorimotor and neurological tests), and neuro-immunological (by tissue cytokine arrays and immuno-histology) effects of ethanol intoxication on TBI. The effect of the occurrence of traumatic intracerebral hemorrhage was also studied. The results indicate that ethanol pretreatment results in a faster and better recovery after TBI with reduced infiltration of leukocytes and reduced microglia activation. These outcomes correspond to reduced parenchymal levels of GM-CSF, IL-6 and IL-3 and to the transient upregulation of IL-13 and VEGF, indicating an early shift in the cytokine profile towards reduced inflammation. A significant difference in the cytokine profile was still observed 24h post injury in the ethanol pretreated mice, as shown by the delayed peak in IL-6 and by the suppression of GM-CSF, IFN-γ, and IL-3. Seven days post-injury, ethanol-pretreated mice displayed a significant decrease both in CD45+ cells infiltration and in microglial activation. On the other hand, in the case of traumatic intracerebral hemorrhage, the cytokine profile was dominated by KC, CCL5, M-CSF and several interleukins and ethanol pretreatment did not produce any modification. We can thus conclude that ethanol intoxication suppresses the acute neuro-inflammatory response to TBI, an effect which is correlated with a faster and complete neurological recovery, whereas, the presence of traumatic intracerebral hemorrhage overrides the effects of ethanol.
Collapse
Affiliation(s)
| | | | - Annette Palmer
- Institute of Clinical and Experimental Trauma Immunology, University Ulm, Ulm, Germany.
| | - Birgit Linkus
- Dept. of Neurology, University of Ulm, School of Medicine, Germany.
| | - Albert C Ludolph
- Dept. of Neurology, University of Ulm, School of Medicine, Germany.
| | - Tobias M Boeckers
- Dept. of Anatomy and Cell Biology, Ulm University, School of Medicine, Germany.
| | - Borna Relja
- Dept. of General and Visceral Surgery, Goethe University, Frankfurt, Germany.
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Ulm, Ulm, Germany.
| | | |
Collapse
|
6
|
Mehta AJ, Guidot DM. Alcohol and the Lung. Alcohol Res 2017; 38:243-254. [PMID: 28988576 PMCID: PMC5513688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Among the many organ systems affected by harmful alcohol use, the lungs are particularly susceptible to infections and injury. The mechanisms responsible for rendering people with alcohol use disorder (AUD) vulnerable to lung damage include alterations in host defenses of the upper and lower airways, disruption of alveolar epithelial barrier integrity, and alveolar macrophage immune dysfunction. Collectively, these derangements encompass what has been termed the "alcoholic lung" phenotype. Alcohol-related reductions in antioxidant levels also may contribute to lung disease in people with underlying AUD. In addition, researchers have identified several regulatory molecules that may play crucial roles in the alcohol-induced disease processes. Although there currently are no approved therapies to combat the detrimental effects of chronic alcohol consumption on the respiratory system, these molecules may be potential therapeutic targets to guide future investigation.
Collapse
Affiliation(s)
- Ashish J Mehta
- Ashish J. Mehta, M.D., is an Assistant Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia. David M. Guidot, M.D., is a Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia
| | - David M Guidot
- Ashish J. Mehta, M.D., is an Assistant Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia. David M. Guidot, M.D., is a Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia
| |
Collapse
|
7
|
De Schryver E, Derycke L, Campo P, Gabriels E, Joos GF, Van Zele T, Bachert C, Hellings PW, Gevaert P. Alcohol hyper-responsiveness in chronic rhinosinusitis with nasal polyps. Clin Exp Allergy 2016; 47:245-253. [DOI: 10.1111/cea.12836] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/02/2016] [Accepted: 10/02/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Els De Schryver
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
| | - Lara Derycke
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
| | - Paloma Campo
- Unidad de Gestión Clínica Allergy-IBIMA; University Hospital; Málaga Spain
| | - Eline Gabriels
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
| | - Guy F. Joos
- Laboratory for Translational Research in Obstructive Pulmonary diseases; Department of Respiratory Medicine; Ghent University Hospital; Ghent Belgium
| | - Thibaut Van Zele
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
- Division of ENT Diseases; Clintec, Karolinska Institutet; Stockholm Sweden
| | - Peter W. Hellings
- Clinical Division of ENT, Head & Neck Surgery; Laboratory of Clinical Immunology; University Hospitals Leuven; Leuven Belgium
- Department of Otorhinolaryngology; Academic Medical Center; Amsterdam the Netherlands
| | - Philippe Gevaert
- Upper Airways Research Laboratory; Department Otorhinolaryngology; Ghent University Hospital; Ghent Belgium
| |
Collapse
|
8
|
Stevenson DD, White AA. Clinical Characteristics of Aspirin-Exacerbated Respiratory Disease. Immunol Allergy Clin North Am 2016; 36:643-655. [PMID: 27712760 DOI: 10.1016/j.iac.2016.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aspirin-exacerbated respiratory disease is a significant endotype of both asthma and chronic rhinosinusitis with nasal polyps. The disease demonstrates what seems to be a unified inflammatory mechanism culminating in highly eosinophilic nasal polyp disease and asthma. The rate of polyp recurrence and morbidity from asthma exacerbations are significant and warrant separating this group diagnostically from aspirin-tolerant peers. Given the unique anti-inflammatory effects of aspirin and the evolving landscape of new, targeted biologic treatments, it is even more incumbent to consider this diagnosis and offer patients treatment specific for the disease.
Collapse
Affiliation(s)
- Donald D Stevenson
- Scripps Clinic, Division of Allergy, Asthma and Immunology, 3811 Valley Centre Drive S99, San Diego, CA 92130, USA
| | - Andrew A White
- Scripps Clinic, Division of Allergy, Asthma and Immunology, 3811 Valley Centre Drive S99, San Diego, CA 92130, USA.
| |
Collapse
|
9
|
Cannon AR, Morris NL, Hammer AM, Curtis B, Remick DG, Yeligar SM, Poole L, Burnham EL, Wyatt TA, Molina PE, So-Armah K, Cisneros T, Wang G, Lang CH, Mandrekar P, Kovacs EJ, Choudhry MA. Alcohol and inflammatory responses: Highlights of the 2015 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2016; 54:73-7. [PMID: 27522326 DOI: 10.1016/j.alcohol.2016.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/24/2016] [Indexed: 01/04/2023]
Abstract
On September 27, 2015 the 20th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite symposium at the annual meeting of the Society for Leukocyte Biology in Raleigh, NC. The 2015 meeting focused broadly on adverse effects of alcohol and alcohol-use disorders in multiple organ systems. Divided into two plenary sessions, AIRIG opened with the topic of pulmonary inflammation as a result of alcohol consumption, which was followed by alcohol's effect on multiple organs, including the brain and liver. With presentations showing the diverse range of underlying pathology and mechanisms associated with multiple organs as a result of alcohol consumption, AIRIG emphasized the importance of continued alcohol research, as its detrimental consequences are not limited to one or even two organs, but rather extend to the entire host as a whole.
Collapse
|
10
|
Beceiro C, Campos J, Valcarcel MA, Fenger RV, Lojo S, Linneberg A, Vidal C, Gonzalez-Quintela A. Serum concentrations of mast cell tryptase are reduced in heavy drinkers. Alcohol Clin Exp Res 2016; 39:672-8. [PMID: 25833028 DOI: 10.1111/acer.12682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/19/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Baseline serum tryptase concentrations are commonly used in clinical practice as a marker of the body's mast cell burden. This study aimed to investigate serum tryptase concentrations in heavy drinkers. METHODS Serum tryptase concentrations were determined in 126 heavy drinkers (75% males, median age 47 years) who were admitted to the hospital because of alcohol withdrawal syndrome (n = 60), general symptoms with abnormalities on biochemical tests that indicated acute liver disease (n = 19), complications of advanced liver disease (n = 33), and miscellaneous reasons (n = 14). Results were compared with those of 70 healthy controls (66% males, median age 40 years). RESULTS Serum tryptase concentrations were lower in heavy drinkers than in healthy controls (median 2.23 μg/l vs. median 3.25 μg/l, p < 0.001). Ten heavy drinkers (7.9%) had undetectable (<1 μg/l) serum tryptase levels versus none of the healthy controls (p = 0.01). The association of low tryptase levels with heavy drinking was independent of age, gender, and smoking status. Among heavy drinkers, the lowest tryptase concentrations were observed in patients with alcohol withdrawal syndrome and patients with general symptoms with abnormalities on biochemical tests that indicated acute liver disease. Furthermore, serum tryptase concentrations were negatively correlated with markers of acute liver damage or alcohol consumption (serum aspartate aminotransferase and gamma-glutamyl transferase). Atopy (skin prick test positivity) was not associated with serum tryptase concentrations in heavy drinkers. CONCLUSIONS Serum concentrations of mast cell tryptase are lower in heavy drinkers than in healthy controls.
Collapse
Affiliation(s)
- Carmen Beceiro
- Department of Internal Medicine, Hospital Clinico Universitario, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Alcoholic Liver Disease: A Mouse Model Reveals Protection by Lactobacillus fermentum. Clin Transl Gastroenterol 2016; 7:e138. [PMID: 26795070 PMCID: PMC4737872 DOI: 10.1038/ctg.2015.66] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/04/2015] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Alcoholism is one of the most devastating diseases with high incidence, but knowledge of its pathology and treatment is still plagued with gaps mostly because of the inherent limitations of research with patients. We developed an animal model for studying liver histopathology, Hsp (heat-shock protein)-chaperones involvement, and response to treatment. METHODS The system was standardized using mice to which ethanol was orally administered alone or in combination with Lactobacillus fermentum following a precise schedule over time and applying, at predetermined intervals, a battery of techniques (histology, immunohistochemistry, western blotting, real-time PCR, immunoprecipitation, 3-nitrotyrosine labeling) to assess liver pathology (e.g., steatosis, fibrosis), and Hsp60 and iNOS (inducible form of nitric oxide synthase) gene expression and protein levels, and post-translational modifications. RESULTS Typical ethanol-induced liver pathology occurred and the effect of the probiotic could be reliably monitored. Steatosis score, iNOS levels, and nitrated proteins (e.g., Hsp60) decreased after probiotic intake. CONCLUSIONS We describe a mouse model useful for studying liver disease induced by chronic ethanol intake and for testing pertinent therapeutic agents, e.g., probiotics. We tested L. fermentum, which reduced considerably ethanol-induced tissue damage and deleterious post-translational modifications of the chaperone Hsp60. The model is available to test other agents and probiotics with therapeutic potential in alcoholic liver disease.
Collapse
|
12
|
Summary of the 2014 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2015; 49:767-72. [PMID: 26520175 DOI: 10.1016/j.alcohol.2015.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/21/2015] [Accepted: 09/21/2015] [Indexed: 02/08/2023]
Abstract
On November 21, 2014 the 19th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held at Loyola University Chicago Health Sciences Campus in Maywood, Illinois. The meeting focused broadly on inflammatory cell signaling responses in the context of alcohol and alcohol-use disorders, and was divided into four plenary sessions focusing on the gut and liver, lung infections, general systemic effects of alcohol, and neuro-inflammation. One common theme among many talks was the differential roles of macrophages following both chronic and acute alcohol intoxication. Macrophages were shown to play significant roles in regulating inflammation, oxidative stress, and viral infection following alcohol exposure in the liver, lungs, adipose tissue, and brain. Other work examined the role of alcohol on disease progression in a variety of pathologies including psoriasis, advanced stage lung disease, and cancer.
Collapse
|
13
|
Gomez-Pinilla PJ, Farro G, Di Giovangiulio M, Stakenborg N, Némethova A, de Vries A, Liston A, Feyerabend TB, Rodewald HR, Boeckxstaens GE, Matteoli G. Mast cells play no role in the pathogenesis of postoperative ileus induced by intestinal manipulation. PLoS One 2014; 9:e85304. [PMID: 24416383 PMCID: PMC3887017 DOI: 10.1371/journal.pone.0085304] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/25/2013] [Indexed: 01/15/2023] Open
Abstract
Introduction Intestinal manipulation (IM) during abdominal surgery results in intestinal inflammation leading to hypomotility or ileus. Mast cell activation is thought to play a crucial role in the pathophysiology of postoperative ileus (POI). However, this conclusion was mainly drawn using mast cell-deficient mouse models with abnormal Kit signaling. These mice also lack interstitial cells of Cajal (ICC) resulting in aberrant gastrointestinal motility even prior to surgery, compromising their use as model to study POI. To avoid these experimental weaknesses we took advantage of a newly developed knock-in mouse model, Cpa3Cre/+, devoid of mast cells but with intact Kit signaling. Design The role of mast cells in the development of POI and intestinal inflammation was evaluated assessing gastrointestinal transit and muscularis externa inflammation after IM in two strains of mice lacking mast cells, i.e. KitW-sh/W-sh and Cpa3Cre/+ mice, and by use of the mast cell stabilizer cromolyn. Results KitW-sh/W-sh mice lack ICC networks and already revealed significantly delayed gastrointestinal transit even before surgery. IM did not further delay intestinal transit, but induced infiltration of myeloperoxidase positive cells, expression of inflammatory cytokines and recruitment of monocytes and neutrophils into the muscularis externa. On the contrary, Cpa3Cre/+ mice have a normal network of ICC and normal gastrointestinal. Surprisingly, IM in Cpa3Cre/+ mice caused delay in gut motility and intestinal inflammation as in wild type littermates mice (Cpa3+/+). Furthermore, treatment with the mast cell inhibitor cromolyn resulted in an inhibition of mast cells without preventing POI. Conclusions Here, we confirm that IM induced mast cell degranulation. However, our data demonstrate that mast cells are not required for the pathogenesis of POI in mice. Although there might be species differences between mouse and human, our results argue against mast cell inhibitors as a therapeutic approach to shorten POI.
Collapse
Affiliation(s)
- Pedro J. Gomez-Pinilla
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Giovanna Farro
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Martina Di Giovangiulio
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Andrea Némethova
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Annick de Vries
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Adrian Liston
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Autoimmune Genetics Laboratory, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Thorsten B. Feyerabend
- Division for Cellular Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Reimwer Rodewald
- Division for Cellular Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guy E. Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
14
|
Kim MJ, Nepal S, Lee ES, Jeong TC, Kim SH, Park PH. Ethanol increases matrix metalloproteinase-12 expression via NADPH oxidase-dependent ROS production in macrophages. Toxicol Appl Pharmacol 2013; 273:77-89. [DOI: 10.1016/j.taap.2013.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 07/30/2013] [Accepted: 08/06/2013] [Indexed: 12/12/2022]
|
15
|
Cockroach allergen Bla g 7 promotes TIM4 expression in dendritic cells leading to Th2 polarization. Mediators Inflamm 2013; 2013:983149. [PMID: 24204099 PMCID: PMC3800592 DOI: 10.1155/2013/983149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/21/2013] [Indexed: 11/29/2022] Open
Abstract
As one of the most common sources of indoor aeroallergens worldwide, cockroach is important in causing rhinitis and asthma while the mechanisms underlying remain obscure. Since T helper (Th) type 2 polarization plays an important role in the pathogenesis of allergic diseases, we investigated the effect of Bla g 7, a pan-allergen from Blattella germanica (B. germanica), on Th polarization which is controlled by monocyte-derived dendritic cells (DCs). Challenged by recombinant Bla g 7 (rBla g 7), immature DCs obtained from human exhibited upregulated levels of TIM4, CD80, and CD86 and increased IL-13 secretion. Cocultured with CD4+ T cells, challenged DCs increased the ratio of IL-4+ versus IFN-γ+ of CD4+ T cells, suggesting a balance shift from Th1 to Th2. Moreover, antibodies against TIM4, CD80, and CD86 reversed the enhancement of IL-4+/IFN-γ+ ratio and alleviated the IL-13 release induced by rBla g 7, indicating that the Th2 polarization provoked by rBla g 7 challenged DCs is via TIM4-, CD80-, and CD86-dependent mechanisms. In conclusion, the present findings implied a crucial role of Bla g 7 in the development of cockroach allergy and highlighted an involvement of DCs-induced Th2 polarization in cockroach allergy.
Collapse
|
16
|
Bouchard JC, Beal DR, Kim J, Vaickus LJ, Remick DG. Chemokines mediate ethanol-induced exacerbations of murine cockroach allergen asthma. Clin Exp Immunol 2013; 172:203-16. [PMID: 23574317 DOI: 10.1111/cei.12048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2012] [Indexed: 10/27/2022] Open
Abstract
Asthma imposes considerable patient and economic burdens, with the most severe cases causing the greatest affliction. Identifying stimuli that worsen asthma severity is an essential step to controlling both disease morbidity and the lessening economic impact. This study provides the first mechanistic investigation into how acute ethanol exposure will increase asthma severity in a murine model of mild cockroach allergen (CRA)-induced asthma. Outbred mice were sensitized to induce mild allergic asthma, with intratracheal CRA exposures on days 0 and 14. On day 21 mice were gavaged with water or 32% ethanol, and the third allergen exposure was given 30 min post-gavage. Asthmatic responses were measured at several time-points up to 42 h after the third allergen challenge. Ethanol-gavaged mice showed increased asthma severity within 90 min post-allergen challenge, with exacerbations lasting for 24 h. Ethanol caused greater airways obstruction, including an eightfold increase in epithelial cell mucin and increased mucus plugs, resulting in a 50% reduction in bronchiole patency. Ethanol gavage also induced significant increases in airways hyperreactivity. While T helper type 1 (Th1) and Th2 cytokines were not altered by ethanol gavage, pulmonary neutrophil and eosinophil recruitment were augmented. This increase was associated with increased chemokine production. Administration 2 h prior to ethanol gavage of a neutralizing antibody cocktail to keratinocyte-derived chemokine, macrophage inflammatory protein-2, eotaxin-1 and eotaxin-2 prevented ethanol-induced eosinophil recruitment and airways hyperreactivity. These data provide evidence that acute alcohol exposure immediately prior to a mild allergen-triggered asthmatic episode will exacerbate asthma severity mediated by increased production of chemokines.
Collapse
Affiliation(s)
- J C Bouchard
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | |
Collapse
|
17
|
Loftis JM, Huckans M. Substance use disorders: psychoneuroimmunological mechanisms and new targets for therapy. Pharmacol Ther 2013; 139:289-300. [PMID: 23631821 DOI: 10.1016/j.pharmthera.2013.04.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/15/2013] [Indexed: 12/28/2022]
Abstract
An estimated 76.4 million people worldwide meet criteria for alcohol use disorders, and 15.3 million meet criteria for drug use disorders. Given the high rates of addiction and the associated health, economic, and social costs, it is essential to develop a thorough understanding of the impact of substance abuse on mental and physical health outcomes and to identify new treatment approaches for substance use disorders (SUDs). Psychoneuroimmunology is a rapidly expanding, multidisciplinary area of research that may be of particular importance to addiction medicine, as its focus is on the dynamic and complex interactions among behavioral factors, the central nervous system, and the endocrine and immune systems (Ader, 2001). This review, therefore, focuses on: 1) the psychoneuroimmunologic effects of SUDs by substance type and use pattern, and 2) the current and future treatment strategies, including barriers that can impede successful recovery outcomes. Evidence-based psychosocial and pharmacotherapeutic treatments are reviewed. Psychological factors and central nervous system correlates that impact treatment adherence and response are discussed. Several novel therapeutic approaches that are currently under investigation are introduced; translational data from animal and human studies is presented, highlighting immunotherapy as a promising new direction for addiction medicine.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research and Development Service, Portland VA Medical Center, Portland, OR, USA.
| | | |
Collapse
|