1
|
Zhou M, Sun R, Jang J, Martin JG. T cell and airway smooth muscle interaction: a key driver of asthmatic airway inflammation and remodeling. Am J Physiol Lung Cell Mol Physiol 2024; 327:L382-L394. [PMID: 39010821 DOI: 10.1152/ajplung.00121.2024] [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: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
Cross talk between T cells and airway smooth muscle (ASM) may play a role in modulating asthmatic airway inflammation and remodeling. Infiltrating T cells have been observed within the ASM bundles of asthmatics, and a wide range of direct and indirect interactions between T cells and ASM has been demonstrated using various in vitro and in vivo model systems. Contact-dependent mechanisms such as ligation and activation of cellular adhesion and costimulatory molecules, as well as the formation of lymphocyte-derived membrane conduits, facilitate the adhesion, bidirectional communication, and transfer of materials between T and ASM cells. T cell-derived cytokines, particularly of the Th1, Th2, and Th17 subsets, modulate the secretome, proliferation, and contractility of ASM cells. This review summarizes the mechanisms governing T cell-ASM cross talk in the context of asthma. Understanding the underlying mechanistic basis is important for directing future research and developing therapeutic interventions targeted toward this complex interaction.
Collapse
Affiliation(s)
- Muyang Zhou
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rui Sun
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Joyce Jang
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - James G Martin
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
2
|
AbuJabal R, Ramakrishnan RK, Bajbouj K, Hamid Q. Role of IL-5 in asthma and airway remodelling. Clin Exp Allergy 2024; 54:538-549. [PMID: 38938056 DOI: 10.1111/cea.14489] [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: 09/17/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 06/29/2024]
Abstract
Asthma is a common and burdensome chronic inflammatory airway disease that affects both children and adults. One of the main concerns with asthma is the manifestation of irreversible tissue remodelling of the airways due to the chronic inflammatory environment that eventually disrupts the whole structure of the airways. Most people with troublesome asthma are treated with inhaled corticosteroids. However, the development of steroid resistance is a commonly encountered issue, necessitating other treatment options for these patients. Biological therapies are a promising therapeutic approach for people with steroid-resistant asthma. Interleukin 5 is recently gaining a lot of attention as a biological target relevant to the tissue remodelling process. Since IL-5-neutralizing monoclonal antibodies (mepolizumab, reslizumab and benralizumab) are currently available for clinical use, this review aims to revisit the role of IL-5 in asthma pathogenesis at large and airway remodelling in particular, in addition to exploring its role as a target for biological treatments.
Collapse
Affiliation(s)
- Rola AbuJabal
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Rakhee K Ramakrishnan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qutayba Hamid
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada
| |
Collapse
|
3
|
Sun R, Pan X, Ward E, Intrevado R, Morozan A, Lauzon AM, Martin JG. Serum Response Factor Expression in Excess Permits a Dual Contractile-Proliferative Phenotype of Airway Smooth Muscle. Am J Respir Cell Mol Biol 2024; 71:182-194. [PMID: 38775474 DOI: 10.1165/rcmb.2024-0081oc] [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: 02/20/2024] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
The transcription factors (TFs) MyoCD (myocardin) and Elk-1 (ETS Like-1 protein) competitively bind to SRF (serum response factor) and control myogenic- and mitogenic-related gene expression in smooth muscle, respectively. Their functions are therefore mutually inhibitory, which results in a contractile-versus-proliferative phenotype dichotomy. Airway smooth muscle cell (ASMC) phenotype alterations occur in various inflammatory airway diseases, promoting pathological remodeling and contributing to airflow obstruction. We characterized MyoCD and Elk-1 interactions and their roles in phenotype determination in human ASMCs. MyoCD overexpression in ASMCs increased smooth muscle gene expression, force generation, and partially restored the loss of smooth muscle protein associated with prolonged culturing while inhibiting Elk-1 transcriptional activities and proliferation induced by EGF (epidermal growth factor). However, MyoCD overexpression failed to suppress these responses induced by FBS, as FBS also upregulated SRF expression to a degree that allowed unopposed function of both TFs. Inhibition of the RhoA pathway reversed said SRF changes, allowing inhibition of Elk-1 by MyoCD overexpression and suppressing FBS-mediated contractile protein gene upregulation. Our study confirmed that MyoCD in increased abundance can competitively inhibit Elk-1 function. However, SRF upregulation permits a dual contractile-proliferative ASMC phenotype that is anticipated to exacerbate pathological alterations, whereas therapies targeting SRF may inhibit pathological ASMC proliferation and contractile protein gene expression.
Collapse
Affiliation(s)
- Rui Sun
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Xingning Pan
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Erin Ward
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Rafael Intrevado
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Arina Morozan
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| | - James G Martin
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
4
|
Balkrishna A, Sinha S, Karumuri S, Maity M, Dev R, Varshney A. Bronchom assuages airway hyperresponsiveness in house dust mite-induced mouse model of allergic asthma and moderates goblet cell metaplasia, sub-epithelial fibrosis along with changes in Th2 cytokines and chemokines. Front Immunol 2024; 15:1384697. [PMID: 38807596 PMCID: PMC11130375 DOI: 10.3389/fimmu.2024.1384697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/19/2024] [Indexed: 05/30/2024] Open
Abstract
Background Asthma is a common obstructive airway disease with an inflammatory etiology. The main unmet need in the management of asthma is inadequate adherence to pharmacotherapy, leading to a poorly-controlled disease state, necessitating the development of novel therapies. Bronchom is a calcio-herbal formulation, which is purported to treat chronic asthma. The objective of the current study was to examine the in-vivo efficacy of Bronchom in mouse model of allergic asthma. Methods Ultra high performance liquid chromatography was utilized to analyze the phytocompounds in Bronchom. Further, the in-vivo efficacy of Bronchom was evaluated in House dust mite (HDM)-induced allergic asthma in mice. Mice were challenged with aerosolized methacholine to assess airway hyperresponsiveness. Subsequently, inflammatory cell influx was evaluated in bronchoalveolar lavage fluid (BALF) followed by lung histology, wherein airway remodeling features were studied. Simultaneously, the levels of Th2 cytokines and chemokines in the BALF was also evaluated. Additionally, the mRNA expression of pro-inflammatory and Th2 cytokines was also assessed in the lung along with the oxidative stress markers. Results Phytocompounds present in Bronchom included, gallic acid, protocatechuic acid, methyl gallate, rosmarinic acid, glycyrrhizin, eugenol, 6-gingerol and piperine. Bronchom effectively suppressed HDM-induced airway hyperresponsiveness along with the influx of leukocytes in the BALF. Additionally, Bronchom reduced the infiltration of inflammatory cells in the lung and it also ameliorated goblet cell metaplasia, sub-epithelial fibrosis and increase in α-smooth muscle actin. Bronchom decreased Th2 cytokines (IL-4 and IL-5) and chemokines (Eotaxin and IP-10) in the BALF. Likewise, it could also suppress the mRNA expression of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-6 and IL-33), and IL-13. Moreover, Bronchom restored the HDM-induced diminution of endogenous anti-oxidants (GSH and SOD) and the increase in pro-oxidants (GSSG and MDA). Furthermore, Bronchom could also decrease the nitrosative stress by lowering the observed increase in nitrite levels. Conclusion Taken together, the results of the present study data convincingly demonstrate that Bronchom exhibits pharmacological effects in an animal model of allergic asthma. Bronchom mitigated airway hyperresponsiveness, inflammation and airway remodeling evoked by a clinically relevant allergen and accordingly it possesses therapeutic potential for the treatment of asthma.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, India
- Patanjali UK Trust, Glasgow, United Kingdom
- Vedic Acharya Samaj Foundation, Inc., Groveland, FL, United States
| | - Sandeep Sinha
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Shadrakbabu Karumuri
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Madhulina Maity
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Rishabh Dev
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
5
|
Jang JH, Zhou M, Makita K, Sun R, El-Hajjar M, Fonseca G, Lauzon AM, Martin JG. Induction of a memory-like CD4 + T-cell phenotype by airway smooth muscle cells. Eur J Immunol 2024; 54:e2249800. [PMID: 38334162 DOI: 10.1002/eji.202249800] [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: 01/08/2022] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
In asthma, CD4+ T-cell interaction with airway smooth muscle (ASM) may enhance its contractile properties and promote its proliferation. However, less is known about the effects of this interaction on T cells. To explore the consequences of interaction of CD4+ T cells with ASM we placed the cells in co-culture and analyzed the phenotypic and functional changes in the T cells. Effector status as well as cytokine expression was assessed by flow cytometry. An increase in CD45RA-CD45RO+ memory T cells was observed after co-culture; however, these cells were not more responsive to CD3/28 restimulation. A reduction in mitochondrial coupling and an increase in the production of mitochondrial reactive oxygen species by CD4+ T cells post-restimulation suggested altered mitochondrial metabolism after co-culture. RNA sequencing analysis of the T cells revealed characteristic downregulation of effector T-cell-associated genes, but a lack of upregulation of memory T-cell-associated genes. The results of this study demonstrate that ASM cells can induce a phenotypic shift in CD4+ T cells into memory-like T cells but with reduced capacity for activation.
Collapse
Affiliation(s)
- Joyce H Jang
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Michael Zhou
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Kosuke Makita
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Mikal El-Hajjar
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Gregory Fonseca
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Zhou M, Sun R, Chakraborty R, Wang C, Lauzon AM, Martin JG. CD4 + T cell-derived IFN-γ and LIGHT synergistically upregulate chemokine production from airway smooth muscle cells. FASEB J 2024; 38:e23405. [PMID: 38193542 DOI: 10.1096/fj.202301428rr] [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: 07/24/2023] [Revised: 12/02/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Airway smooth muscle (ASM) remodeling in asthmatic airways may contribute to persistent airflow limitation and airway hyperresponsiveness. CD4+ T cells infiltrate the ASM layer where they may induce a proliferative and secretory ASM cell phenotype. We studied the interaction between activated CD4+ T cells and ASM cells in co-culture in vitro and investigated the effects of CD4+ T cells on chemokine production by ASM cells. CD4+ T cells induced marked upregulation of C-X-C motif chemokine ligands (CXCL) 9, 10, and 11 in ASM cells. Blockade of the IFN-γ receptor on ASM cells prevented this upregulation. Furthermore, T cell-derived IFN-γ and LIGHT (lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) synergize in a dose-dependent manner to coordinately enhance CXCL9, 10, and 11 expression. The synergistic property of LIGHT was mediated exclusively through the lymphotoxin-β receptor (LTBR), but not herpes virus entry mediator (HVEM). Disruption of LTBR signaling in ASM cells reduced CXCL9, 10, and 11 production and ASM cell-mediated CD4+ T cell chemotaxis. We conclude that the LIGHT-LTBR signaling axis acts together with IFN-γ to regulate chemokines that mediate lymphocyte infiltration in asthmatics.
Collapse
Affiliation(s)
- Muyang Zhou
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - Rui Sun
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - Rohin Chakraborty
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - Christina Wang
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - Anne-Marie Lauzon
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - James G Martin
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| |
Collapse
|
7
|
Yon C, Thompson DA, Jude JA, Panettieri RA, Rastogi D. Crosstalk between CD4 + T Cells and Airway Smooth Muscle in Pediatric Obesity-related Asthma. Am J Respir Crit Care Med 2023; 207:461-474. [PMID: 36194662 PMCID: PMC12042998 DOI: 10.1164/rccm.202205-0985oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Pediatric obesity-related asthma is a nonatopic asthma phenotype with high disease burden and few effective therapies. RhoGTPase upregulation in peripheral blood T helper (Th) cells is associated with the phenotype, but the mechanisms that underlie this association are not known. Objectives: To investigate the mechanisms by which upregulation of CDC42 (Cell Division Cycle 42), a RhoGTPase, in Th cells is associated with airway smooth muscle (ASM) biology. Methods: Chemotaxis of obese asthma and healthy-weight asthma Th cells, and their adhesion to obese and healthy-weight nonasthmatic ASM, was investigated. Transcriptomics and proteomics were used to determine the differential effect of obese and healthy-weight asthma Th cell adhesion to obese or healthy-weight ASM biology. Measurements and Main Results: Chemotaxis of obese asthma Th cells with CDC42 upregulation was resistant to CDC42 inhibition. Obese asthma Th cells were more adherent to obese ASM compared with healthy-weight asthma Th cells to healthy-weight ASM. Compared with coculture with healthy-weight ASM, obese asthma Th cell coculture with obese ASM was positively enriched for genes and proteins involved in actin cytoskeleton organization, transmembrane receptor protein kinase signaling, and cell mitosis, and negatively enriched for extracellular matrix organization. Targeted gene evaluation revealed upregulation of IFNG, TNF (tumor necrosis factor), and Cluster of Differentiation 247 (CD247) among Th cell genes, and of Ak strain transforming (AKT), Ras homolog family member A (RHOA), and CD38, with downregulation of PRKCA (Protein kinase C-alpha), among smooth muscle genes. Conclusions: Obese asthma Th cells have uninhibited chemotaxis and are more adherent to obese ASM, which is associated with upregulation of genes and proteins associated with smooth muscle proliferation and reciprocal nonatopic Th cell activation.
Collapse
Affiliation(s)
- Changsuek Yon
- Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC; and
| | - David A Thompson
- Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC; and
| | - Joseph A Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Deepa Rastogi
- Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC; and
| |
Collapse
|
8
|
Hsieh A, Assadinia N, Hackett TL. Airway remodeling heterogeneity in asthma and its relationship to disease outcomes. Front Physiol 2023; 14:1113100. [PMID: 36744026 PMCID: PMC9892557 DOI: 10.3389/fphys.2023.1113100] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Asthma affects an estimated 262 million people worldwide and caused over 461,000 deaths in 2019. The disease is characterized by chronic airway inflammation, reversible bronchoconstriction, and airway remodeling. Longitudinal studies have shown that current treatments for asthma (inhaled bronchodilators and corticosteroids) can reduce the frequency of exacerbations, but do not modify disease outcomes over time. Further, longitudinal studies in children to adulthood have shown that these treatments do not improve asthma severity or fixed airflow obstruction over time. In asthma, fixed airflow obstruction is caused by remodeling of the airway wall, but such airway remodeling also significantly contributes to airway closure during bronchoconstriction in acute asthmatic episodes. The goal of the current review is to understand what is known about the heterogeneity of airway remodeling in asthma and how this contributes to the disease process. We provide an overview of the existing knowledge on airway remodeling features observed in asthma, including loss of epithelial integrity, mucous cell metaplasia, extracellular matrix remodeling in both the airways and vessels, angiogenesis, and increased smooth muscle mass. While such studies have provided extensive knowledge on different aspects of airway remodeling, they have relied on biopsy sampling or pathological assessment of lungs from fatal asthma patients, which have limitations for understanding airway heterogeneity and the entire asthma syndrome. To further understand the heterogeneity of airway remodeling in asthma, we highlight the potential of in vivo imaging tools such as computed tomography and magnetic resonance imaging. Such volumetric imaging tools provide the opportunity to assess the heterogeneity of airway remodeling within the whole lung and have led to the novel identification of heterogenous gas trapping and mucus plugging as important predictors of patient outcomes. Lastly, we summarize the current knowledge of modification of airway remodeling with available asthma therapeutics to highlight the need for future studies that use in vivo imaging tools to assess airway remodeling outcomes.
Collapse
Affiliation(s)
- Aileen Hsieh
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Najmeh Assadinia
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada,*Correspondence: Tillie-Louise Hackett,
| |
Collapse
|
9
|
Jackson D, Walum J, Banerjee P, Lewis BW, Prakash YS, Sathish V, Xu Z, Britt RD. Th1 cytokines synergize to change gene expression and promote corticosteroid insensitivity in pediatric airway smooth muscle. Respir Res 2022; 23:126. [PMID: 35578269 PMCID: PMC9109364 DOI: 10.1186/s12931-022-02046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Corticosteroids remain a key therapy for treating children with asthma. Patients with severe asthma are insensitive, resistant, or refractory to corticosteroids and have poorly controlled symptoms that involve airway inflammation, airflow obstruction, and frequent exacerbations. While the pathways that mediate corticosteroid insensitivity in asthma remain poorly defined, recent studies suggest that enhanced Th1 pathways, mediated by TNFα and IFNγ, may play a role. We previously reported that the combined effects of TNFα and IFNγ promote corticosteroid insensitivity in developing human airway smooth muscle (ASM).
Methods
To further understand the effects of TNFα and IFNγ on corticosteroid sensitivity in the context of neonatal and pediatric asthma, we performed RNA sequencing (RNA-seq) on human pediatric ASM treated with fluticasone propionate (FP), TNFα, and/or IFNγ.
Results
We found that TNFα had a greater effect on gene expression (~ 1000 differentially expressed genes) than IFNγ (~ 500 differentially expressed genes). Pathway and transcription factor analyses revealed enrichment of several pro-inflammatory responses and signaling pathways. Interestingly, treatment with TNFα and IFNγ augmented gene expression with more than 4000 differentially expressed genes. Effects of TNFα and IFNγ enhanced several pro-inflammatory genes and pathways related to ASM and its contributions to asthma pathogenesis, which persisted in the presence of corticosteroids. Co-expression analysis revealed several gene networks related to TNFα- and IFNγ-mediated signaling, pro-inflammatory mediator production, and smooth muscle contractility. Many of the co-expression network hubs were associated with genes that are insensitive to corticosteroids.
Conclusions
Together, these novel studies show the combined effects of TNFα and IFNγ on pediatric ASM and implicate Th1-associated cytokines in promoting ASM inflammation and hypercontractility in severe asthma.
Collapse
|
10
|
Lin TY, Chang PJ, Lo CY, Lo YL, Yu CT, Lin SM, Kuo CHS, Lin HC. Interaction Between CD34 + Fibrocytes and Airway Smooth Muscle Promotes IL-8 Production and Akt/PRAS40/mTOR Signaling in Asthma. Front Med (Lausanne) 2022; 9:823994. [PMID: 35547213 PMCID: PMC9081978 DOI: 10.3389/fmed.2022.823994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background The circulating progenitor cells of fibroblasts (fibrocytes) have been shown to infiltrate the airway smooth muscle compartment of asthma patients; however, the pathological significance of this discovery has yet to be elucidated. This study established a co-culture model of airway smooth muscle cells (ASMCs) and fibrocytes from asthmatic or normal subjects to evaluate innate cytokine production, corticosteroid responses, and signaling in ASMCs. Methods CD34+ fibrocytes were purified from peripheral blood of asthmatic (Global Initiative for Asthma treatment step 4–5) and normal subjects and cultured for 5∼7 days. In a transwell plate, ASMCs were co-cultured with fibrocytes at a ratio of 2:1, ASMCs were cultured alone (control condition), and fibrocytes were cultured alone for 48 h. Measurements were obtained of interleukin-8 (IL-8), IL-6, IL-17, thymic stromal lymphopoietin, and IL-33 levels in the supernatant and IL-33 levels in the cell lysate of the co-culture. Screening for intracellular signaling in the ASMCs after stimulation was performed using condition medium from the patients’ co-culture (PtCM) or IL-8. mRNA and western blot analysis were used to analyze AKT/mTOR signaling in ASMCs stimulated via treatment with PtCM or IL-8. Results Compared with ASMCs cultured alone, IL-8 levels in the supernatant and IL-33 levels in the ASMCs lysate were significantly higher in samples co-cultured from asthmatics, but not in those co-cultured from normal subjects. Corticosteroid-induced suppression of IL-8 production was less pronounced in ASMCs co-cultured with fibrocytes from asthma patients than in ASMCs co-cultured from normal subjects. ASMCs stimulated using PtCM and IL-8 presented elevating activated AKT substrate PRAS40. Treatment with IL-8 and PtCM increased mRNA expression of mTOR and P70S6 kinases in ASMCs. Treatment with IL-8 and PtCM also significantly increased phosphorylation of AKT and mTOR subtract S6 ribosomal protein in ASMCs. Conclusion The interaction between ASMCs and fibrocytes from asthmatic patients was shown to increase IL-8 and IL-33 production and promote AKT/mTOR signaling in ASMCs. IL-8 production in the co-culture from asthmatic patients was less affected by corticosteroid than was that in the co-culture from normal subjects. Our results elucidate the novel role of fibrocytes and ASMCs in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Ting-Yu Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jui Chang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Lun Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Teng Yu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-His Scott Kuo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
11
|
Celle A, Esteves P, Cardouat G, Beaufils F, Eyraud E, Dupin I, Maurat E, Lacomme S, Ousova O, Begueret H, Thumerel M, Marthan R, Girodet PO, Berger P, Trian T. Rhinovirus infection of bronchial epithelium induces specific bronchial smooth muscle cell migration of severe asthmatic patients. J Allergy Clin Immunol 2022; 150:104-113. [PMID: 35143808 DOI: 10.1016/j.jaci.2022.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Patients with severe asthma show an increase in both exacerbation frequency and bronchial smooth muscle (BSM) mass. Rhinovirus (RV) infection of the bronchial epithelium (BE) is the main trigger of asthma exacerbations. Histological analysis of biopsies shows that a close connection between BE and hypertrophic BSM is a criterion for severity of asthma. OBJECTIVE We hypothesized that RV infection of BE specifically increases asthmatic BSM cell migration. METHODS Serum samples, biopsies or BSM cells were obtained from 86 patients with severe asthma and 31 non-asthmatic subjects. BE cells from non-asthmatic subjects were cultured in an air-liquid interface and exposed to RV-16. Migration of BSM cells was assessed in response to BE supernatant using chemotaxis assays. Chemokine concentrations were analyzed by transcriptomics and ELISAs. Immunocytochemistry, western blotting and flow cytometry were used to quantify CXCR3 isoform distribution. CXCR3 downstream signaling pathways were assessed by calcium imaging and western blots. RESULTS BSM cells from severe asthmatic patients specifically migrated toward RV-infected BE, whereas those from non-asthmatic subjects did not. This specific migration is driven by BE CXCL10, which was increased in vitro in response to RV infection as well as in vivo in serum from exacerbating patients with severe asthma. The mechanism is related to both decreased expression and activation of the CXCR3-B-specific isoform in severe asthmatic BSM cells. CONCLUSION We have demonstrated a novel mechanism of BSM remodeling in severe asthmatic patients following RV exacerbation. This study highlights the CXCL10/CXCR3-A axis as a potential therapeutic target in severe asthma.
Collapse
Affiliation(s)
- Alexis Celle
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Pauline Esteves
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Guillaume Cardouat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Fabien Beaufils
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Edmée Eyraud
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Isabelle Dupin
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Elise Maurat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Sabrina Lacomme
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France
| | - Olga Ousova
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Hugues Begueret
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Matthieu Thumerel
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Roger Marthan
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Pierre-Olivier Girodet
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Patrick Berger
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Thomas Trian
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France.
| |
Collapse
|
12
|
Luo J, Liu H, Li DKJ, Song B, Zhang Y. Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells. BMC Immunol 2021; 22:74. [PMID: 34876009 PMCID: PMC8650232 DOI: 10.1186/s12865-021-00464-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background Mitochondrial transcription factor A (TFAM) is associated with a number of neurodegenerative diseases and also with asthma. TFAM deficiency-induced mitochondrial DNA stress primes the antiviral innate immune response in mouse embryonic fibroblasts. However, the role of TFAM in asthma related inflammation remains obscure. The purpose of this study was to investigate the regulatory mechanism of TFAM in asthma. Results In this study, we overexpressed TFAM in human lung epithelial cells (A549), then obtained the TFAM-regulated transcriptome by Illumina sequencing technology. Transcriptome analysis revealed that TFAM overexpression down-regulated and up-regulated the expression of 642 and 169 differentially expressed genes (DEGs), respectively. The TFAM-repressed genes were strongly enriched in cytokine-mediated signaling pathway, type I interferon- and INF-γ-mediated signaling pathways, and viral response pathways. We also revealed that 2563 alternative splicing events in 1796 alternative splicing genes (ASGs) were de-regulated upon TFAM overexpression. These TFAM-responding ASGs were enriched in DNA repair, nerve growth factor receptor signaling pathway, and also transcription regulation. Further analysis revealed that the promoters of TFAM-repressed DEGs were enriched by DNA binding motifs of transcription factors whose alternative splicing was regulated by TFAM. Conclusions These findings suggest that TFAM regulates not only immune response gene expression in human lung epithelial cells, but also pre-mRNA alternative splicing which may mediate transcriptional regulation; this TFAM-centered gene regulation network could be targeted in developing therapies against various diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00464-2.
Collapse
Affiliation(s)
- Jinsong Luo
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Hong Liu
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Daniel K Jun Li
- ABLife BioBigData Institute, Wuhan, Hubei, China.,Department of Biology and Biotechnology, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, Hubei, China
| | - Bin Song
- ABLife BioBigData Institute, Wuhan, Hubei, China
| | - Yi Zhang
- ABLife BioBigData Institute, Wuhan, Hubei, China
| |
Collapse
|
13
|
Oliver BG. Food for thought: why is there more airway smooth muscle in asthma? Eur Respir J 2021; 58:58/5/2101565. [PMID: 34824128 DOI: 10.1183/13993003.01565-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/24/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia .,Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Sydney, Australia
| |
Collapse
|
14
|
Xue Y, Bao W, Zhou Y, Fu Q, Hao H, Han L, Yin D, Zhang Y, Zhang X, Zhang M. Small-Airway Dysfunction is Involved in the Pathogenesis of Asthma: Evidence from Two Mouse Models. J Asthma Allergy 2021; 14:883-896. [PMID: 34285515 PMCID: PMC8286250 DOI: 10.2147/jaa.s312361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
Background There has been growing evidence of small-airway dysfunction in patients with asthma. Few studies have evaluated the mechanism of small-airway dysfunction in mouse models of asthma. Purpose We explored the correlation between small-airway spirometric variables and large-airway function or inflammation in different endotypes of asthma. Methods Ovalbumin (OVA) sensitization/challenge was used to produce a type 2 (T2)-high asthma model, and OVA combined with ozone exposure (OVA + ozone) was used for the T2-low asthma model with increased neutrophils. Spirometry, airway responsiveness, cytokine levels in bronchoalveolar lavage fluid (BALF), and pathological analyses of lung slices stained with hematoxylin-eosin, periodic acid–Schiff, and Masson’s trichrome stain were all determined. Muc5ac expression in lung tissue was evaluated by the reverse transcription-polymerase chain reaction (RT-PCR), and alpha-smooth muscle actin was measured by immunohistochemistry. Results Inflammatory cells infiltrated the lung tissue and inflammatory cytokines were increased in the BALF of both the OVA and OVA + ozone groups, compared with the control group. Peribronchial hypersecretion and collagen deposition were evident in the models. The OVA + ozone group showed greater neutrophilic infiltration and peribronchial smooth muscle proliferation than the OVA group. Large-airway obstruction, small-airway dysfunction, and airway hyperresponsiveness were confirmed in both models. Small-airway functional variables, such as MMEF (mean midexpiratory flow, average flow from 25 to 75% forced vital capacity [FVC]) and FEF50 (forced expiratory flow at 50% of FVC), were positively correlated with large-airway function and had a stronger negative correlation with airway inflammation, mucus secretion, and responsiveness than large-airway function. Conclusion Small-airway dysfunction was evident in the two endotypes of asthma and was correlated with severe airway inflammation, mucus hypersecretion, and airway hyperresponsiveness. The small airways may be an important target in asthma treatment, and further research in the role of small-airway variables in the pathogenesis of asthma is warranted.
Collapse
Affiliation(s)
- Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Qiang Fu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Huijuan Hao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Dongning Yin
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yingying Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Xue Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| |
Collapse
|
15
|
Sun R, Jang JH, Lauzon AM, Martin JG. Interferon-γ amplifies airway smooth muscle-mediated CD4+ T cell recruitment by promoting the secretion of C-X-C-motif chemokine receptor 3 ligands. FASEB J 2021; 35:e21228. [PMID: 33337555 DOI: 10.1096/fj.202001480r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 11/11/2022]
Abstract
Asthmatic airways feature increased ASM mass that is largely attributable to hyperplasia, and which potentially contributes to excessive airway narrowing. T cells induce ASMC proliferation via contact-dependent mechanisms in vitro that may have importance for asthmatic ASM growth, as CD4+ T cells infiltrate ASM bundles in asthmatic human airways. In this study, we used an in vitro migration assay to investigate the pathways responsible for the trafficking of human CD4+ T cells to ASM. ASMCs induced chemotaxis of activated CD4+ T cells, which was inhibited by the CXCR3 antagonist AMG487 and neutralizing antibodies against its ligands CXCL10 and 11, but not CCR3 or CCR5 antagonists. CXCR3 expression was upregulated among all T cells following anti-CD3/CD28-activation. CD4+ T cells upregulated CXCL9, 10, and 11 expression in ASMCs in an IFN-γ/STAT1-dependent manner. Disruption of IFN-γ-signaling resulted in reduced T cell migration, along with the inhibition of CD4+ T cell-mediated STAT1 activation and CXCR3 ligand secretion by ASMCs. ASMCs derived from healthy and asthmatic donors demonstrated similar T cell-recruiting capacities. In vivo CXCL10 and 11 expression by asthmatic ASM was confirmed by immunostaining. We conclude that the CXCL10/11-CXCR3 axis causes CD4+ T cell recruitment to ASM that is amplified by T cell-derived IFN-γ.
Collapse
Affiliation(s)
- Rui Sun
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Joyce H Jang
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
16
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
17
|
Esteves P, Blanc L, Celle A, Dupin I, Maurat E, Amoedo N, Cardouat G, Ousova O, Gales L, Bellvert F, Begueret H, Thumerel M, Dupuy JW, Desbenoit N, Marthan R, Girodet PO, Rossignol R, Berger P, Trian T. Crucial role of fatty acid oxidation in asthmatic bronchial smooth muscle remodelling. Eur Respir J 2021; 58:13993003.04252-2020. [PMID: 33833033 DOI: 10.1183/13993003.04252-2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/26/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Bronchial smooth muscle (BSM) remodelling in asthma is related to an increased mitochondrial biogenesis and enhanced BSM cell proliferation in asthma. Since (i) mitochondria produce the highest levels of cellular energy and (ii) fatty acid beta-oxidation is the most powerful way to produce ATP, we hypothesized that, in asthmatic BSM cells, energetic metabolism is shifted towards the beta-oxidation of fatty acids. OBJECTIVES We aimed to characterize BSM cell metabolism in asthma both in vitro and ex vivo to identify a novel target for reducing BSM cell proliferation. METHODS Twenty-one asthmatic and 31 non-asthmatic patients were enrolled. We used metabolomic and proteomic approaches to study BSM cells. Oxidative stress, ATP synthesis, fatty acid endocytosis, metabolite production, metabolic capabilities, mitochondrial networks, cell proliferation and apoptosis were assessed on BSM cells. Fatty acid content was assessed in vivo using MALDI-spectrometry imaging. RESULTS Asthmatic BSM cells were characterized by an increased rate of mitochondrial respiration with a stimulated ATP production and mitochondrial β-oxidation. Fatty acid consumption was increased in asthmatic BSM both in vitro and ex vivo. Proteome remodelling of asthmatic BSM occurred via 2 canonical mitochondrial pathways. The levels of CPT2 and LDL-receptor, which internalize fatty acids through mitochondrial and cell membranes, respectively, were both increased in asthmatic BSM cells. Blocking CPT2 or LDL-receptor drastically and specifically reduced asthmatic BSM cell proliferation. CONCLUSION This study demonstrates a metabolic switch towards mitochondrial beta-oxidation in asthmatic BSM and identifies fatty acid metabolism as a new key target to reduce BSM remodelling in asthma.
Collapse
Affiliation(s)
- Pauline Esteves
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Landry Blanc
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Alexis Celle
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Elise Maurat
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Nivea Amoedo
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Guillaume Cardouat
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Olga Ousova
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Lara Gales
- CNRS 5504, INRA 792, INSA Toulouse, Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, MetaToul, Toulouse, France
| | - Florian Bellvert
- CNRS 5504, INRA 792, INSA Toulouse, Toulouse Biotechnology Institute, Bio & Chemical Engineering, Université de Toulouse, MetaToul, Toulouse, France
| | - Hugues Begueret
- Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Matthieu Thumerel
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Jean-William Dupuy
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Nicolas Desbenoit
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,CNRS, UMR5248, Institute of Chemistry & Biology of Membranes & Nano objects, Functional Genomics Center (CGFB), Proteomics Facility, Université de Bordeaux, Bordeaux, France
| | - Roger Marthan
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Pierre-Olivier Girodet
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France
| | - Rodrigue Rossignol
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France
| | - Patrick Berger
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France.,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Service d'exploration fonctionnelle respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, CIC 1401, CHU de Bordeaux, Bordeaux, France.,Co-last author
| | - Thomas Trian
- Centre de Recherche Cardio-thoracique de Bordeaux, U1045, MRGM, Functional Genomics Center (CGFB), CIC 1401, CELLOMET, Univ-Bordeaux, Bordeaux, France .,Centre de Recherche Cardio-thoracique de Bordeaux, U1045, U1211, CIC 1401, INSERM, Bordeaux, France.,Co-last author
| |
Collapse
|
18
|
Beaufils F, Esteves P, Enaud R, Germande O, Celle A, Marthan R, Trian T, Fayon M, Berger P. Mitochondria are involved in bronchial smooth muscle remodeling in severe preschool wheezers. J Allergy Clin Immunol 2021; 148:645-651.e11. [PMID: 33819511 DOI: 10.1016/j.jaci.2021.03.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Bronchial remodeling is a key feature of asthma that is already present in preschoolers with wheezing. Moreover, bronchial smooth muscle (BSM) remodeling at preschool age is predictive of asthma at school age. However, the mechanism responsible for BSM remodeling in preschoolers with wheezing remains totally unknown. In contrast, in adult asthma, BSM remodeling has been associated with an increase in BSM cell proliferation related to increased mitochondrial mass and biogenesis triggered by an altered calcium homeostasis. Indeed, BSM cell proliferation was decreased in vitro by the calcium channel blocker gallopamil. OBJECTIVE Our aim was to investigate the mechanisms involved in BSM cell proliferation in preschoolers with severe wheezing, with special attention to the role of mitochondria and calcium signaling. METHODS Bronchial tissue samples obtained from 12 preschool controls without wheezing and 10 preschoolers with severe wheezing were used to measure BSM mass and establish primary BSM cell cultures. BSM cell proliferation was assessed by manual counting and flow cytometry, ATP content was assessed by bioluminescence, mitochondrial respiration was assessed by using either the Seahorse or Oroboros technique, mitochondrial mass and biogenesis were assessed by immunoblotting, and calcium response to carbachol was assessed by confocal microscopy. The effect of gallopamil was also evaluated. RESULTS BSM mass, cell proliferation, ATP content, mitochondrial respiration, mass and biogenesis, and calcium response were all increased in preschoolers with severe wheezing compared with in the controls. Gallopamil significantly decreased BSM mitochondrial biogenesis and mass, as well as cell proliferation. CONCLUSION Mitochondria are key players in BSM cell proliferation in preschoolers with severe wheezing and could represent a potential target to treat BSM remodeling at an early stage of the disease.
Collapse
Affiliation(s)
- Fabien Beaufils
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de Pédiatrie médicale, Bordeaux, France
| | - Pauline Esteves
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Raphaël Enaud
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de Pédiatrie médicale, Bordeaux, France
| | - Ophélie Germande
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Alexis Celle
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Roger Marthan
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de Pédiatrie médicale, Bordeaux, France
| | - Thomas Trian
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Michael Fayon
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de Pédiatrie médicale, Bordeaux, France
| | - Patrick Berger
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de Pédiatrie médicale, Bordeaux, France.
| |
Collapse
|
19
|
Zhu C, Shi Y, You J. Immune Cell Connection by Tunneling Nanotubes: The Impact of Intercellular Cross-Talk on the Immune Response and Its Therapeutic Applications. Mol Pharm 2021; 18:772-786. [PMID: 33529022 DOI: 10.1021/acs.molpharmaceut.0c01248] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Direct intercellular communication is an important prerequisite for the development of multicellular organisms, the regeneration of tissue, and the maintenance of various physiological activities. Tunnel nanotubes (TNTs), which have diameters of approximately 50-1500 nm and lengths of up to several cell diameters, can connect cells over long distances and have emerged as one of the most important recently discovered types of efficient communication between cells. Moreover, TNTs can also directly transfer organelles, vehicles, proteins, genetic material, ions, and small molecules from one cell to adjacent and even distant cells. However, the mechanism of intercellular communication between various immune cells within the complex immune system has not been fully elucidated. Studies in the past decades have confirmed the existence of TNTs in many types of cells, especially in various kinds of immune cells. TNTs display different structural and functional characteristics between and within different immunocytes, playing a major role in the transmission of signals across various kinds of immune cells. In this review, we introduce the discovery and structure of TNTs, as well as their different functional properties within different immune cells. We also discuss the roles of TNTs in potentiating the immune response and their potential therapeutic applications.
Collapse
Affiliation(s)
- Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| |
Collapse
|
20
|
Amrani Y, Panettieri RA, Ramos-Ramirez P, Schaafsma D, Kaczmarek K, Tliba O. Important lessons learned from studies on the pharmacology of glucocorticoids in human airway smooth muscle cells: Too much of a good thing may be a problem. Pharmacol Ther 2020; 213:107589. [PMID: 32473159 PMCID: PMC7434707 DOI: 10.1016/j.pharmthera.2020.107589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are the treatment of choice for chronic inflammatory diseases such as asthma. Despite proven effective anti-inflammatory and immunosuppressive effects, long-term and/or systemic use of GCs can potentially induce adverse effects. Strikingly, some recent experimental evidence suggests that GCs may even exacerbate some disease outcomes. In asthma, airway smooth muscle (ASM) cells are among the targets of GC therapy and have emerged as key contributors not only to bronchoconstriction, but also to airway inflammation and remodeling, as implied by experimental and clinical evidence. We here will review the beneficial effects of GCs on ASM cells, emphasizing the differential nature of GC effects on pro-inflammatory genes and on other features associated with asthma pathogenesis. We will also summarize evidence describing how GCs can potentially promote pro-inflammatory and remodeling features in asthma with a specific focus on ASM cells. Finally, some of the possible solutions to overcome these unanticipated effects of GCs will be discussed.
Collapse
Affiliation(s)
- Yassine Amrani
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, Leicester Biomedical Research Center Respiratory, Leicester, UK
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Patricia Ramos-Ramirez
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | | | - Klaudia Kaczmarek
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - Omar Tliba
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA.
| |
Collapse
|
21
|
Liu Y, Li X, He C, Chen R, Wei L, Meng L, Zhang C. Emodin ameliorates ovalbumin-induced airway remodeling in mice by suppressing airway smooth muscle cells proliferation. Int Immunopharmacol 2020; 88:106855. [PMID: 32777676 DOI: 10.1016/j.intimp.2020.106855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 01/15/2023]
Abstract
Increased number of airway smooth muscle cells (ASMCs) is a characteristic of airway remodeling in asthma. In this study we investigated whether emodin alleviated airway remodeling in a murine asthma model and reduced the proliferation of ASMCs in vitro. We provided in vivo evidence suggesting that intraperitoneal injection of emodin (20 mg/kg) 1 h prior to OVA challenge apparently alleviated the thickness of airway smooth muscle, the mass of alpha-smooth muscle actin (α-SMA), collagen deposition, epithelial damage, goblet cell hyperplasia, airway inflammation and airway hyperresponsiveness (AHR) in lung tissue. Meanwhile, we found that emodin suppressed the activation of the Akt pathway in lungtissue of allergic mouse models. Additionally, we found that emodin inhibited cellular proliferation and Akt activation in a dose-dependent manner in vitro. Furthermore, LY294002, an inhibitor for PI3K, abrogated serum-induced phosphorylation of Akt, and decreased the proliferation of ASMCs. These findings indicated that emodin alleviated ASMCs proliferation by inhibiting PI3K/Akt pathway in vivo and in vitro, which may provide a potential therapeutic option for airway smooth muscle remodeling in asthma.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Xin Li
- Department of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Chao He
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Ran Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Li Wei
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Ling Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China.
| | - Caiqing Zhang
- Department of Respiratory Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
22
|
Wang KCW, Donovan GM, James AL, Noble PB. Asthma: Pharmacological degradation of the airway smooth muscle layer. Int J Biochem Cell Biol 2020; 126:105818. [PMID: 32707120 DOI: 10.1016/j.biocel.2020.105818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/23/2022]
Abstract
Asthma: A disease characterised by excessive and variable airway narrowing, and pathologies of inflammation and remodelling, particularly thickening of the airway smooth muscle (ASM). Treatment approaches dilate narrowed airways and reduce inflammation; however, remodelling seems largely neglected. This review considers the evolution of remodelling in asthma and whether conventional hypotheses that inflammation causes ASM thickening has mislead the medical community into thinking that anti-inflammatories will remedy this ASM defect. There is instead reasonable evidence that ASM thickening occurs independently of inflammation, such that therapies should employ strategies to directly modify ASM growth. Lessons have been learned from the use of untargeted bronchial thermoplasty and there should also be consideration of pharmacological therapies to ablate ASM. We discuss several new approaches to target ASM remodelling in asthma. A major current obstacle is our inability to image the ASM layer and assess treatment response. In this regard, polarisation-sensitive optical coherence tomography offers future promise.
Collapse
Affiliation(s)
- Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, 6009, Western Australia, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.
| | - Graham M Donovan
- Department of Mathematics, University of Auckland, Auckland, 1142, New Zealand
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, 6009, Western Australia, Australia; Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, 6009, Western Australia, Australia
| |
Collapse
|
23
|
Molecular mechanisms of action of naringenin in chronic airway diseases. Eur J Pharmacol 2020; 879:173139. [PMID: 32343971 DOI: 10.1016/j.ejphar.2020.173139] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
Chronic airway inflammatory diseases are characterized by persistent proinflammatory responses in the respiratory tract. Although, several treatment strategies are currently available, lifelong therapy is necessary for most of these diseases. In recent years, phytophenols, namely, flavonoids, derived from fruits and vegetables have been gaining tremendous interest and have been extensively studied due to their low toxicological profile. Naringenin is a bioflavonoid abundantly found in citrus fruits. This substance has shown notable therapeutic potential in various diseases due to its promising diverse biological activities. In this review, we have attempted to review the published studies from the available literature, discussing the molecular level mechanisms of naringenin in different experimental models of airway inflammatory diseases including asthma, chronic obstructive pulmonary disease (COPD), lung cancer, pulmonary fibrosis and cystic fibrosis. Current evidences have proposed that the anti-inflammatory properties of naringenin play a major role in ameliorating inflammatory disease states. In addition, naringenin also possesses several other biological properties. Despite the proposed mechanisms suggesting remarkable therapeutic benefits, the clinical use of naringenin is, however, hampered by its low solubility and bioavailability. Furthermore, this review also discusses on the studies that utilise nanocarriers as a drug delivery system to address the issue of poor solubility.
Collapse
|
24
|
Esteves P, Celle A, Berger P, Trian T. [Bronchial smooth muscle mitochondria: A new target for asthma therapy?]. Rev Mal Respir 2020; 37:201-204. [PMID: 32139106 DOI: 10.1016/j.rmr.2020.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 01/12/2020] [Indexed: 12/30/2022]
Abstract
The main purpose of this review is to highlight mitochondria as a new therapeutic target to prevent bronchial smooth muscle (BSM) remodeling in asthma. Severe asthmatic patients, representing 5-10% of all asthmatics, are characterized by an increased BSM mass which is highly correlated with the severity of the disease and the rate of exacerbations. None of the current asthma therapies are effective in reducing BSM remodelling. This review, based on the current literature, reports the role of mitochondria in BSM, particularly in calcium signaling.
Collapse
Affiliation(s)
- P Esteves
- Université Bordeaux, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France; Inserm, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France.
| | - A Celle
- Université Bordeaux, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France; Inserm, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France
| | - P Berger
- Université Bordeaux, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France; Inserm, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France; CHU de Bordeaux, service d'exploration fonctionnelle respiratoire, CIC 1401, 33604 Pessac, France
| | - T Trian
- Université Bordeaux, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France; Inserm, centre de recherche cardio-thoracique de Bordeaux, U1045, CIC 1401, 33600 Pessac, France
| |
Collapse
|
25
|
Allard B, Levardon H, Esteves P, Celle A, Maurat E, Thumerel M, Girodet PO, Trian T, Berger P. Asthmatic Bronchial Smooth Muscle Increases CCL5-Dependent Monocyte Migration in Response to Rhinovirus-Infected Epithelium. Front Immunol 2020; 10:2998. [PMID: 31969885 PMCID: PMC6956660 DOI: 10.3389/fimmu.2019.02998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022] Open
Abstract
Asthma exacerbations, a major concern in therapeutic strategies, are most commonly triggered by viral respiratory infections, particularly with human rhinovirus (HRV). Infection of bronchial epithelial (BE) cells by HRV triggers inflammation, notably monocyte recruitment. The increase of bronchial smooth muscle (BSM) mass in asthma, a hallmark of bronchial remodeling, is associated with the annual rate of exacerbations. The aim of the present study was to assess whether or not BSM could increase monocyte migration induced by HRV-infected BE. We used an advanced in vitro model of co-culture of human BE cells in air-liquid interface with human BSM cells from control and asthmatic patients. Inflammation triggered by HRV infection (HRV-16, MOI 0.1, 1 h) was assessed at 24 h with transcriptomic analysis and multiplex ELISA. In vitro CD14+ monocyte migration was evaluated with modified Boyden chamber. Results showed that HRV-induced monocyte migration was substantially increased in the co-culture model with asthmatic BSM, compared with control BSM. Furthermore, the well-known monocyte migration chemokine, CCL2, was not involved in this increased migration. However, we demonstrated that CCL5 was further increased in the asthmatic BSM co-culture and that anti-CCL5 blocking antibody significantly decreased monocyte migration induced by HRV-infected BE. Taken together, our findings highlight a new role of BSM cells in HRV-induced inflammation and provide new insights in mucosal immunology which may open new opportunities for prevention and/or treatment of asthma exacerbation.
Collapse
Affiliation(s)
- Benoit Allard
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Hannah Levardon
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Pauline Esteves
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Alexis Celle
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Elise Maurat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Matthieu Thumerel
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France.,CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de Chirurgie Thoracique, CIC 1401, Pessac, France
| | - Pierre Olivier Girodet
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France.,CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de Chirurgie Thoracique, CIC 1401, Pessac, France
| | - Thomas Trian
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France
| | - Patrick Berger
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC 1401, Bordeaux, France.,INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, Bordeaux, France.,CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de Chirurgie Thoracique, CIC 1401, Pessac, France
| |
Collapse
|
26
|
Yap HM, Israf DA, Harith HH, Tham CL, Sulaiman MR. Crosstalk Between Signaling Pathways Involved in the Regulation of Airway Smooth Muscle Cell Hyperplasia. Front Pharmacol 2019; 10:1148. [PMID: 31649532 PMCID: PMC6794426 DOI: 10.3389/fphar.2019.01148] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Increased ASM mass, primarily due to ASM hyperplasia, has been recognized as a hallmark of airway remodeling in asthma. Increased ASM mass is the major contributor to the airway narrowing, thus worsening the bronchoconstriction in response to stimuli. Inflammatory mediators and growth factors released during inflammation induce increased ASM mass surrounding airway wall via increased ASM proliferation, diminished ASM apoptosis and increased ASM migration. Several major pathways, such as MAPKs, PI3K/AKT, JAK2/STAT3 and Rho kinase, have been reported to regulate these cellular activities in ASM and were reported to be interrelated at certain points. This article aims to provide an overview of the signaling pathways/molecules involved in ASM hyperplasia as well as the mapping of the interplay/crosstalk between these major pathways in mediating ASM hyperplasia. A more comprehensive understanding of the complexity of cellular signaling in ASM cells will enable more specific and safer drug development in the control of asthma.
Collapse
Affiliation(s)
- Hui Min Yap
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Mohd Roslan Sulaiman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
27
|
Jendzjowsky NG, Kelly MM. The Role of Airway Myofibroblasts in Asthma. Chest 2019; 156:1254-1267. [PMID: 31472157 DOI: 10.1016/j.chest.2019.08.1917] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/14/2019] [Accepted: 08/11/2019] [Indexed: 12/17/2022] Open
Abstract
Airway remodeling is a characteristic feature of asthma and is thought to play an important role in the pathogenesis of airway hyperresponsiveness. Myofibroblasts are key structural cells involved in injury and repair, and there is evidence that dysregulation of their normal function contributes to airway remodeling. Despite the importance of myofibroblasts, a lack of specific cellular markers and inconsistent nomenclature have limited recognition of their key role in airway remodeling. Myofibroblasts are increased several-fold in the airways in asthma, in proportion to the severity of the disease. Myofibroblasts are postulated to be derived from both tissue-resident and bone marrow-derived cells, depending on the stage of injury and the tissue. A small number of studies have demonstrated attenuation of myofibroblast numbers and also reversal of established myofibroblast populations in asthma and other inflammatory processes. In this article, we review what is currently known about the biology of myofibroblasts in the airways in asthma and identify potential targets to reduce or reverse the remodeling process. However, further translational research is required to better understand the mechanistic role of the myofibroblast in asthma.
Collapse
Affiliation(s)
- Nicholas G Jendzjowsky
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Margaret M Kelly
- Airway Inflammation Research Group, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
28
|
Abstract
Current management of severe asthma relying either on guidelines (bulk approach) or on disease phenotypes (stratified approach) did not improve the burden of the disease. Several severe phenotypes are described: clinical, functional, morphological, inflammatory, molecular and microbiome-related. However, phenotypes do not necessarily relate to or give insights into the underlying pathogenetic mechanisms which are described by the disease endotypes. Based on the major immune-inflammatory pathway involved type-2 high, type-2 low and mixed endotypes are described for severe asthma, with several shared pathogenetic pathways such as genetic and epigenetic, metabolic, neurogenic and remodelling subtypes. The concept of multidimensional endotyping as un unbiased approach to severe asthma is discussed, together with new tools and targets facilitating the shift from the stratified to the precision medicine approach.
Collapse
|
29
|
Kalinauskaite-Zukauske V, Januskevicius A, Janulaityte I, Miliauskas S, Malakauskas K. Expression of eosinophil β chain-signaling cytokines receptors, outer-membrane integrins, and type 2 inflammation biomarkers in severe non-allergic eosinophilic asthma. BMC Pulm Med 2019; 19:158. [PMID: 31438916 PMCID: PMC6706886 DOI: 10.1186/s12890-019-0904-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 07/23/2019] [Indexed: 01/21/2023] Open
Abstract
Background Severe non-allergic eosinophilic asthma (SNEA) is a rare asthma phenotype associated with severe clinical course, frequent exacerbations, and resistance to therapy, including high steroid doses. The key feature is type 2 inflammation with predominant airway eosinophilia. Eosinophil maturation, activation, survivability, and recruitment are mainly induced by interleukin (IL)-3, IL-5 and granulocyte–macrophage colony-stimulating factor (GM-CSF) through their receptors on eosinophil surface and related with integrins activation states. The aim of the study was to estimate the expression of eosinophil β chain-signaling cytokines receptors, outer-membrane integrins, and serum-derived type 2 inflammation biomarkers in SNEA. Methods We examined 8 stable SNEA patients with high inhaled steroid doses, 12 steroid-free patients with non-severe allergic asthma (AA), 12 healthy subjects (HS). Blood eosinophils were isolated using Ficol gradient centrifugation and magnetic separation. Eosinophils were lysed, and mRNA was isolated. Gene expressions of IL-5Rα, IL-3Rα, GM-CSFRα, and α4β1, αMβ2 integrins were analyzed using quantitative real-time reverse transcription polymerase chain reaction. Type 2 inflammation activity was evaluated measuring exhaled nitric oxide concentration (FeNO) collected with the electrochemical sensing device. Serum IL-5, IL-3, GM-CSF, periostin, chemokine ligand (CCL) 17 and eotaxin concentrations were assessed by enzyme-linked immunosorbent assay. Results Eosinophils from SNEA patients demonstrated significantly increased gene expression of IL-3Rα, IL-5Rα and GM-CSFRα as well as α4, β1 and αM integrin subunits compared with the AA group. The highest IL-5 serum concentration was in the SNEA group; it significantly differed compared with AA and HS. GM-CSF serum levels were similar in the SNEA and AA groups and were significantly lower in the HS group. No differences in serum IL-3 concentration were found among all groups. Furthermore, serum levels of eotaxin, CCL17 and FeNO, but not periostin, differed in all groups, with the highest levels in SNEA patients. Conclusions Eosinophil demonstrated higher expression of IL-3, IL-5, GM-CSF α-chain receptors and α4, β1, αM integrins subunits in SNEA compared with the AA group. Additionally, SNEA patients had increased serum levels of IL-5, eotaxin and CCL-17. Trial registration ClinicalTrials.gov Identifier NCT03388359.
Collapse
Affiliation(s)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ieva Janulaityte
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Skaidrius Miliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kestutis Malakauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
30
|
PARP-1 Is Critical for Recruitment of Dendritic Cells to the Lung in a Mouse Model of Asthma but Dispensable for Their Differentiation and Function. Mediators Inflamm 2019; 2019:1656484. [PMID: 31178661 PMCID: PMC6507252 DOI: 10.1155/2019/1656484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/15/2018] [Accepted: 01/02/2019] [Indexed: 02/01/2023] Open
Abstract
Dendritic cells (DCs) are critical in asthma and many other immune diseases. We previously demonstrated a role for PARP-1 in asthma. Evidence on PARP-1 playing a role in Th2-associated DC function is not clear. In this study, we examined whether PARP-1 is critical for DC differentiation and function using bone marrow progenitors and their migration to the lung in an ovalbumin-based mouse model of asthma. Results show that changes in PARP-1 levels during GM-CSF-induced DC differentiation from bone marrow progenitors were cyclic and appear to be part of an array of changes that included STAT3/STAT5/STAT6/GRAIL/RAD51. Interestingly, PARP-1 gene deletion affected primarily STAT6 and γH2AX. PARP-1 inhibition significantly reduced the migration of DCs to the lungs of ovalbumin-challenged mice, which was associated with a concomitant reduction in lung levels of the adhesion molecule VCAM-1. The requirement of PARP-1 for VCAM-1 expression was confirmed using endothelial and lung smooth muscle cells. PARP-1 expression and activity were also required for VCAM-1 in differentiated DCs. An assessment of CD11b+/CD11c+/MHCIIhigh DCs in spleens and lymph nodes of OVA-sensitized mice revealed that PARP-1 inhibition genetically or by olaparib exerted little to no effect on DC differentiation, percentage of CD80+/CD86+/CD40+-expressing cells, or their capacity to promote proliferation of ovalbumin-primed (OTII) CD4+ T cells. These findings were corroborated using GM-CSF-induced differentiation of DCs from the bone marrow. Surprisingly, the PARP-1−/− DCs exhibited a higher intrinsic capacity to induce OTII CD4+ T cell proliferation in the absence of ovalbumin. Overall, our results show that PARP-1 plays little to no role in DC differentiation and function and that the protective effect of PARP-1 inhibition against asthma is associated with a prevention of DC migration to the lung through a reduction in VCAM-1 expression. Given the current use of PARP inhibitors (e.g., olaparib) in the clinic, the present results may be of interest for the relevant therapies.
Collapse
|
31
|
Shi F, Zhang Y, Qiu C, Xiong Y, Li M, Shan A, Yang Y, Li B. Effects of inhaled corticosteroids on the expression of TNF family molecules in murine model of allergic asthma. Exp Lung Res 2018; 43:301-310. [PMID: 29140131 DOI: 10.1080/01902148.2017.1376129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The tumor necrosis factor superfamily member LIGHT (the official gene symbol approved by NCBI Gene Database), an inflammatory factor secreted by T cells after allergen exposure, recently discovered to play crucial roles in asthmatic airway remodeling. However, it is unclear whether LIGHT could be controlled by inhaled corticosteroids, a key component of asthma management. This study was to investigate the effects and potential mechanisms of inhaled budesonide on the expressions of LIGHT and its receptors (LTβR and HVEM) of lung tissues in ovalbumin-sensitized mice. MATERIALS AND METHODS Thirty-three BALB/c mice were randomly divided into the control, asthma model, and budesonide treatment groups (11 in each group). Mice were sensitized and challenged by OVA to develop mouse model of chronic asthma, and treated with aerosolized budesonide before OVA challenge. Bronchoalveolar lavage fluid (BALF) and lungs were obtained after the final OVA challenge. Protein and mRNA Levels of LIGHT, LTβR, and HVEM in the lungs were investigated by immunohistochemistry, image analysis, and real-time PCR. Expressions of IL-6 and IFN-γ in BALF were measured by ELISA. RESULTS Inhaled budesonide significantly reduced protein and mRNA levels of lung LIGHT, LTβR, and HVEM in asthmatic mice. Correspondingly, the number of eosinophils and neutrophils and IL-6 levels in BALF after budesonide treatment were found to be decreased, whereas the IFN-γ levels in BALF were increased. Moreover, the expressions of LIGHT and HVEM mRNA showed positive correlation with IL-6 levels in the treatment group. CONCLUSIONS Inhaled budesonide can down-regulate the expressions of LIGHT, LTβR, and HVEM in the lungs of asthmatic mice, and LIGHT/LTβR/HVEM interactions may be a potentially key target for asthma treatment.
Collapse
Affiliation(s)
- Fei Shi
- a Emergency Department , Jinan University, The Second Clinical College , NO. 1017 Dongmen north Road, Shenzhen , China
| | - Yarui Zhang
- b Biomedical Research Institute , Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , NO. 1120 Lianhua Road, Shenzhen , China
| | - Chen Qiu
- c Pulmonary Department , Jinan University, The Second Clinical College , NO. 1017 Dongmen north Road, Shenzhen , China
| | - Yi Xiong
- b Biomedical Research Institute , Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , NO. 1120 Lianhua Road, Shenzhen , China
| | - Manhui Li
- b Biomedical Research Institute , Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , NO. 1120 Lianhua Road, Shenzhen , China
| | - Aijun Shan
- a Emergency Department , Jinan University, The Second Clinical College , NO. 1017 Dongmen north Road, Shenzhen , China
| | - Ying Yang
- a Emergency Department , Jinan University, The Second Clinical College , NO. 1017 Dongmen north Road, Shenzhen , China
| | - Binbin Li
- a Emergency Department , Jinan University, The Second Clinical College , NO. 1017 Dongmen north Road, Shenzhen , China
| |
Collapse
|
32
|
James AL, Noble PB, Drew SA, Mauad T, Bai TR, Abramson MJ, McKay KO, Green FHY, Elliot JG. Airway smooth muscle proliferation and inflammation in asthma. J Appl Physiol (1985) 2018; 125:1090-1096. [PMID: 30024335 DOI: 10.1152/japplphysiol.00342.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In asthma, it is unclear if the airway smooth muscle cells proliferate more or are increased at the onset of asthma and remain stable. This study aimed to compare smooth muscle cell proliferation in individuals with and without asthma and correlate proliferation rates with cell size and number and with granulocytic airway inflammation. Postmortem airway sections were labeled with proliferating cell nuclear antigen (PCNA) and percent positive muscle cells calculated. On the same sections, smooth muscle cell size and number and the number of eosinophils and neutrophils were estimated and compared in cases of nonfatal ( n = 15) and fatal ( n = 15) asthma and control subjects ( n = 15). The %PCNA+ muscle cells was not significantly different in fatal (29.4 ± 7.7%, mean ± SD), nonfatal asthma (28.6 ± 8.3%), or control subjects (24.6 ± 6.7%) and not related to mean muscle cell size ( r = 0.09), number ( r = 0.36), thickness of the muscle layer ( r = 0.05), or eosinophil numbers ( r = 0.04) in the asthma cases. These data support the hypothesis that in asthma the increased thickness of the smooth muscle layer may be present before or at the onset of asthma and independent of concurrent granulocytic inflammation or exacerbation. NEW & NOTEWORTHY There is debate regarding the origins of the increased airway smooth muscle in asthma. It may be independent of inflammation or arise as a proliferative response to inflammation. The present study found no increase in the proportion of proliferating smooth muscle cells in asthma and no relation of proliferation to numbers of airway smooth muscle cells or inflammation. These results support a stable increase in smooth muscle in asthma that is independent of airway inflammation.
Collapse
Affiliation(s)
- Alan L James
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital , Nedlands, WA , Australia.,School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA , Australia
| | - Peter B Noble
- School of Human Sciences, University of Western Australia , Nedlands, WA , Australia.,Centre for Neonatal Research and Education, School of Paediatrics and Child Health, University of Western Australia , Perth, WA , Australia
| | - Su-Ann Drew
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital , Nedlands, WA , Australia.,School of Human Sciences, University of Western Australia , Nedlands, WA , Australia
| | - Thais Mauad
- University Medical School , Sao Paulo , Brazil
| | - Tony R Bai
- University of British Columbia , Vancouver, BC , Canada
| | - Michael J Abramson
- Department of Epidemiology & Preventive Medicine, Monash University , Melbourne, VIC , Australia
| | - Karen O McKay
- Children's Hospital at Westmead , Sydney, NSW , Australia
| | - Francis H Y Green
- Department of Pathology and Laboratory Medicine, University of Calgary , Calgary, AB , Canada
| | - John G Elliot
- West Australian Sleep Disorders Research Institute, Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital , Nedlands, WA , Australia
| |
Collapse
|
33
|
Rastogi D, Nico J, Johnston AD, Tobias TAM, Jorge Y, Macian F, Greally JM. CDC42-related genes are upregulated in helper T cells from obese asthmatic children. J Allergy Clin Immunol 2018; 141:539-548.e7. [PMID: 28479334 PMCID: PMC5671374 DOI: 10.1016/j.jaci.2017.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pediatric obesity-related asthma is more severe and less responsive to medications than asthma in normal-weight children. Obese asthmatic children have nonatopic TH1-polarized systemic inflammation that correlates with pulmonary function deficits, but the pathways underlying TH1-polarized inflammation are not well understood. OBJECTIVE We compared the CD4+ T-cell transcriptome in obese children with asthma with that in normal-weight children with asthma to identify key differentially expressed genes associated with TH1-polarized inflammation. METHODS CD4+ T-cell transcriptome-wide differential gene expression was compared between 21 obese and 21 normal-weight children by using directional RNA sequencing. High-confidence differentially expressed genes were verified in the first cohort and validated in a second cohort of 20 children (10 obese and 10 normal-weight children) by using quantitative RT-PCR. RESULTS Transcriptome-wide differential gene expression among obese asthmatic children was enriched for genes, including VAV2, DOCK5, PAK3, PLD1, CDC42EP4, and CDC42PBB, which are associated with CDC42, a small guanosine triphosphate protein linked to T-cell activation. Upregulation of MLK3 and PLD1, genes downstream of CDC42 in the mitogen-activated protein kinase and mammalian target of rapamycin pathways and the inverse correlation of CDC42EP4 and DOCK5 transcript counts with FEV1/FVC ratio together support a role of CDC42 in the TH1 polarization and pulmonary function deficits found in patients with obesity-related asthma. CONCLUSIONS Our study identifies the CDC42 pathway as a novel target that is upregulated in TH cells of obese asthmatic children, suggesting its role in nonatopic TH1-polarized systemic inflammation and pulmonary function deficits found in patients with pediatric obesity-related asthma.
Collapse
Affiliation(s)
- Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY.
| | - John Nico
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Andrew D Johnston
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| | | | - Yurydia Jorge
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
| | - John M Greally
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
34
|
Bronchial Thermoplasty in Severe Asthma. CURRENT PULMONOLOGY REPORTS 2017. [DOI: 10.1007/s13665-017-0191-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
35
|
Salter B, Pray C, Radford K, Martin JG, Nair P. Regulation of human airway smooth muscle cell migration and relevance to asthma. Respir Res 2017; 18:156. [PMID: 28814293 PMCID: PMC5559796 DOI: 10.1186/s12931-017-0640-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/10/2017] [Indexed: 01/15/2023] Open
Abstract
Airway remodelling is an important feature of asthma pathogenesis. A key structural change inherent in airway remodelling is increased airway smooth muscle mass. There is emerging evidence to suggest that the migration of airway smooth muscle cells may contribute to cellular hyperplasia, and thus increased airway smooth muscle mass. The precise source of these cells remains unknown. Increased airway smooth muscle mass may be collectively due to airway infiltration of myofibroblasts, neighbouring airway smooth muscle cells in the bundle, or circulating hemopoietic progenitor cells. However, the relative contribution of each cell type is not well understood. In addition, although many studies have identified pro and anti-migratory agents of airway smooth muscle cells, whether these agents can impact airway remodelling in the context of human asthma, remains to be elucidated. As such, further research is required to determine the exact mechanism behind airway smooth muscle cell migration within the airways, how much this contributes to airway smooth muscle mass in asthma, and whether attenuating this migration may provide a therapeutic avenue for asthma. In this review article, we will discuss the current evidence with respect to the regulation of airway smooth muscle cell migration in asthma.
Collapse
Affiliation(s)
- Brittany Salter
- Firestone Institute for Respiratory Health, St Joseph’s Healthcare and Department of Medicine, 50 Charlton Avenue, East, Hamilton, ON L8N 4A6 Canada
| | - Cara Pray
- Firestone Institute for Respiratory Health, St Joseph’s Healthcare and Department of Medicine, 50 Charlton Avenue, East, Hamilton, ON L8N 4A6 Canada
| | - Katherine Radford
- Firestone Institute for Respiratory Health, St Joseph’s Healthcare and Department of Medicine, 50 Charlton Avenue, East, Hamilton, ON L8N 4A6 Canada
| | - James G. Martin
- Meakins Christie Laboratories, McGill University, Montreal, QC Canada
| | - Parameswaran Nair
- Firestone Institute for Respiratory Health, St Joseph’s Healthcare and Department of Medicine, 50 Charlton Avenue, East, Hamilton, ON L8N 4A6 Canada
| |
Collapse
|
36
|
Johansson MW. Eosinophil Activation Status in Separate Compartments and Association with Asthma. Front Med (Lausanne) 2017; 4:75. [PMID: 28660189 PMCID: PMC5466952 DOI: 10.3389/fmed.2017.00075] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023] Open
Abstract
Asthma is frequently characterized by eosinophil-rich airway inflammation. Airway eosinophilia is associated with asthma exacerbations and likely plays a part in airway remodeling. Eosinophil recruitment from the bloodstream depends on circulating eosinophils becoming activated, which leads to eosinophil arrest on activated endothelium, extravasation, and continued movement through the bronchial tissue by interaction with the extracellular matrix (ECM). Circulating eosinophils can exist at different activation levels, which include non-activated or pre-activated (sensitized or “primed”). Further, the bloodstream may lack pre-activated cells, due to such eosinophils having arrested on endothelium or extravasated into tissue. Increased expression, and in some instances, decreased expression of cell-surface proteins, including CD44, CD45, CD45R0, CD48, CD137, neuropeptide S receptor, cytokine receptors, Fc receptors, and integrins (receptors mediating cell adhesion and migration by interacting with ligands on other cells or in the ECM), and activated states of integrins or Fc receptors on blood eosinophils have been reported to correlate with aspects of asthma. A subset of these proteins has been reported to respond to intervention, e.g., with anti-interleukin (IL)-5. How these surface proteins and the activation state of the eosinophil respond to other interventions, e.g., with anti-IL-4 receptor alpha or anti-IL-13, is unknown. Eosinophil surface proteins suggested to be biomarkers of activation, particularly integrins, and reports on correlations between eosinophil activation and aspects of asthma are described in this review. Intermediate activation of beta1 and beta2 integrins on circulating eosinophils correlates with decreased pulmonary function, airway inflammation, or airway lumen eosinophils in non-severe asthma. The correlation does not appear in severe asthma, likely due to a higher degree of extravasation of pre-activated eosinophils in more severe disease. Bronchoalveolar lavage (BAL) eosinophils have highly activated integrins and other changes in surface proteins compared to blood eosinophils. The activation state of eosinophils in lung tissue, although likely very important in asthma, is largely unknown. However, some recent articles, mainly on mice but partly on human cells, indicate that tissue eosinophils may have a surface phenotype(s) different from that of sputum or BAL eosinophils.
Collapse
Affiliation(s)
- Mats W Johansson
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
37
|
Ijpma G, Panariti A, Lauzon AM, Martin JG. Directional preference of airway smooth muscle mass increase in human asthmatic airways. Am J Physiol Lung Cell Mol Physiol 2017; 312:L845-L854. [PMID: 28360113 DOI: 10.1152/ajplung.00353.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 11/22/2022] Open
Abstract
Airway smooth muscle (ASM) orientation and morphology determine the ability of the muscle to constrict the airway. In asthma, ASM mass is increased, but it is unknown whether ASM orientation and morphology are altered as well or whether the remodeling at the source of the mass increase is ongoing. We dissected human airway trees from asthmatic and control lungs. Stained, intact airway sections were imaged in axial projection to show ASM bundle orientation, whereas cross-sectional histological slides were used to assess ASM area, bundle thickness, and ASM bundle-to-basement membrane distance. We also used these slides to assess cell size, proliferation, and apoptosis. We showed that ASM mass increase in cartilaginous airways is primarily the result of an increase of ASM bundle thickness (as measured radially in an airway cross section) and coincides with an increased distance of the ASM bundles to the airway perimeter. ASM orientation was unchanged in all airways. Apoptosis markers and cell size did not show differences between asthmatics and controls. Our findings show that ASM mass increase likely contributes to the airway-constricting capacity of the muscle. Both the increased bundle thickness and increased thickness of the airway wall inwards of the ASM bundles could further enhance this capacity. Turnover of ASM appears to be the same in airways and biopsies, but the lack of correlation between different markers of proliferation casts doubt on the specificity of markers generally used to assess proliferation.
Collapse
Affiliation(s)
- Gijs Ijpma
- Department of Medicine, McGill University, Montreal, Quebec, Canada; and.,Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Montreal, Quebec, Canada
| | - Alice Panariti
- Department of Medicine, McGill University, Montreal, Quebec, Canada; and.,Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Montreal, Quebec, Canada
| | - Anne-Marie Lauzon
- Department of Medicine, McGill University, Montreal, Quebec, Canada; and.,Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Montreal, Quebec, Canada
| | - James G Martin
- Department of Medicine, McGill University, Montreal, Quebec, Canada; and .,Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Montreal, Quebec, Canada
| |
Collapse
|
38
|
Yang M, Wang HY, Chen JC, Zhao J. Regulation of airway inflammation and remodeling in asthmatic mice by TLR3/TRIF signal pathway. Mol Immunol 2017; 85:265-272. [PMID: 28342933 DOI: 10.1016/j.molimm.2017.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/02/2017] [Accepted: 03/04/2017] [Indexed: 12/17/2022]
Abstract
This paper aims to investigate the effect of Toll-like receptors 3 (TLR3)/TIR-domain-containing adapter-inducing interferon-β (TRIF) signal pathway on the airway inflammation and remodeling in asthmatic mice. C57BL/6 and TLR3-/- mice were randomly divided into three groups (10 mice per group), including Control group (mice inhaled phosphate buffer saline (PBS)), Asthma group (mice inhaled ovalbumin (OVA)) and polyriboinosinic-ribocytidylic acid (poly (I: C)) group (asthmatic mice were injected intraperitoneally with TLR3 agonist poly (I: C)). Hematoxylin-eosin (HE) staining, Wright-Giemsa staining, Enzyme-linked immunosorbent assay (ELISA), Immunohistochemistry, Hydroxyproline assay, quantitative real time polymerase chain reaction (qRT-PCR) and Western blot were used to assess for the indices of airway inflammation and remodeling. In terms of WT mice, all asthma groups with or without the addition of poly (I: C) showed exaggerated inflammation and remodeling in the airways as compared to Control group, which were more seriously in poly (I: C) group than Asthma group. Furthermore, we observed the significant inhibition of airway inflammation and remodeling in the TLR3-/- mice in both Asthma no matter with or without addition of poly (I: C) than the WT mice. TLR3 knockout could obviously relieve the airway inflammation and remodeling in asthma through inhibiting TLR3/TRIF signaling pathway.
Collapse
Affiliation(s)
- Mei Yang
- Department of critical care medicine, The Third People's Hospital of Jinan, Jinan 250132, Shandong, PR China.
| | - Hao-Ying Wang
- Department of critical care medicine, The Third People's Hospital of Jinan, Jinan 250132, Shandong, PR China
| | - Jian-Chang Chen
- Department of emergency, Shandong Provincial Western Hospital, Jinan 250021, Shandong, PR China
| | - Jing Zhao
- Department of cardiology, Qilu Hospital Affiliated to Shandong University, Jinan 250012, PR China
| |
Collapse
|
39
|
Girodet PO, Allard B, Thumerel M, Begueret H, Dupin I, Ousova O, Lassalle R, Maurat E, Ozier A, Trian T, Marthan R, Berger P. Bronchial Smooth Muscle Remodeling in Nonsevere Asthma. Am J Respir Crit Care Med 2016; 193:627-33. [PMID: 26540234 DOI: 10.1164/rccm.201507-1404oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
RATIONALE Increased bronchial smooth muscle (BSM) mass is a key feature of airway remodeling that classically distinguishes severe from nonsevere asthma. Proliferation of BSM cells involves a specific mitochondria-dependent pathway in individuals with severe asthma. However, BSM remodeling and mitochondrial biogenesis have not been examined in nonsevere asthma. OBJECTIVES We aimed to assess whether an increase in BSM mass was also implicated in nonsevere asthma and its relationship with mitochondria and clinical outcomes. METHODS We enrolled 34 never-smoker subjects with nonsevere asthma. In addition, we recruited 56 subjects with nonsevere asthma and 19 subjects with severe asthma as comparative groups (COBRA cohort [Cohorte Obstruction Bronchique et Asthme; Bronchial Obstruction and Asthma Cohort; sponsored by the French National Institute of Health and Medical Research, INSERM]). A phenotypic characterization was performed using questionnaires, atopy and pulmonary function testing, exhaled nitric oxide measurement, and blood collection. Bronchial biopsy specimens were processed for immunohistochemistry and electron microscopy analysis. After BSM remodeling assessment, subjects were monitored over a 12-month period. MEASUREMENTS AND MAIN RESULTS We identified characteristic features of remodeling (BSM area >26.6%) and increased mitochondrial number within BSM in a subgroup of subjects with nonsevere asthma. The number of BSM mitochondria was positively correlated with BSM area (r = 0.78; P < 0.001). Follow-up analysis showed that subjects with asthma with high BSM had worse asthma control and a higher rate of exacerbations per year compared with subjects with low BSM. CONCLUSIONS This study reveals that BSM remodeling and mitochondrial biogenesis may play a critical role in the natural history of nonsevere asthma (Mitasthme study). Clinical trial registered with www.clinicaltrials.gov (NCT00808730).
Collapse
Affiliation(s)
- Pierre-Olivier Girodet
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and.,3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| | - Benoit Allard
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Matthieu Thumerel
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and.,3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| | - Hugues Begueret
- 3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| | - Isabelle Dupin
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Olga Ousova
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Régis Lassalle
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Elise Maurat
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Annaig Ozier
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and.,3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| | - Thomas Trian
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and
| | - Roger Marthan
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and.,3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| | - Patrick Berger
- 1 University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France.,2 INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC1401, Bordeaux, France; and.,3 CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC1401, Service d'Anatomopathologie, Pessac, France
| |
Collapse
|
40
|
Seidel P, Sun Q, Costa L, Lardinois D, Tamm M, Roth M. The MNK-1/eIF4E pathway as a new therapeutic pathway to target inflammation and remodelling in asthma. Cell Signal 2016; 28:1555-62. [PMID: 27418099 DOI: 10.1016/j.cellsig.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/30/2016] [Accepted: 07/08/2016] [Indexed: 01/31/2023]
Abstract
Therapeutic targets in asthma are reduction of airway inflammation and remodelling, the latter is not affected by available drugs. Here we present data that inhibition of MAPK-activated protein kinase (MNK)-1 reduces inflammation and remodelling. MNK-1 regulates protein expression by controlling mRNA stability, nuclear export and translation through the eukaryotic initiation factor 4E (eIF4E). Airway smooth muscle cells were derived from asthmatic and non-asthmatic donors. Cells were pre-treated with CGP57380 (MNK-1 inhibitor) or MNK-1 siRNA, before TNF-α stimulation. Cytokine and protein expression was analysed by ELISA, real time PCR and immunoblotting. Proliferation was monitored by cell counts. TNF-α activated MNK-1 phosphorylation between 15 and 30min. and subsequently eIF4E between 15 and 60min. EIF4E activity was inhibited by CGP57380 dose-dependently. Inhibition of MNK-1 by CGP57380 or MNK-1 siRNA significantly reduced TNF-α induced CXCL10 and eotaxin mRNA expression and secretion, but had no effect on IL-8. However, CXCL10 mRNA stability or NF-κB activity were not affected by MNK-1 inhibition. Furthermore, eIF4E was detected in the cytosol and the nucleus, but TNF-α did not affected its export from the nucleus. Cytokine array assessment showed that in addition to eotaxin and CXCL10, asthma relevant GRO α and RANTES were down-regulated by MNK-1 inhibition. In addition, MNK-1 inhibition significantly reduced FCS and PDGF-BB induced cell proliferation. We are the first to report that MNK-1 controls chemokine secretion and proliferation in human airway smooth muscle cells. Therefore we suggest that MNK-1 inhibition may present a new target to limit inflammation and remodelling in asthmatic airways.
Collapse
Affiliation(s)
- Petra Seidel
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Qingzhu Sun
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Department of Biochemistry and Molecular Biology, School of Basic Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China
| | - Luigi Costa
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Didier Lardinois
- Thoracic Surgery, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Michael Tamm
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Pneumology Clinic, Internal Medicine, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Pneumology Clinic, Internal Medicine, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland.
| |
Collapse
|
41
|
Mattoli S. Involvement of fibrocytes in asthma and clinical implications. Clin Exp Allergy 2016; 45:1497-509. [PMID: 25752439 DOI: 10.1111/cea.12525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bloodborne fibrocytes are bone marrow-derived cells that participate in immune responses and exhibit pro-inflammatory and matrix remodelling properties. In patients with asthma receiving an adequate treatment, the blood fibrocyte count is very low and comparable to that obtained in healthy individuals. In these patients, a transient increase in fibrocyte numbers in the peripheral blood and in the airways occurs in concomitance with increased bronchial inflammation and reflects disease worsening and the need for more intensive treatment. Persistently elevated numbers of fibrocytes in the peripheral blood and in the bronchial mucosa are observed in chronically undertreated or corticosteroid-resistant asthma and are associated with persistent airway inflammation and ongoing remodelling of the bronchial wall. The asthmatic bronchial epithelium is the main source of fibrocyte chemoattractants in asthma and contributes with T helper type 2 lymphocytes and eosinophils to promote the proliferation and pro-remodelling function of recruited fibrocytes. The presence of elevated numbers of fibrocytes in the bronchial mucosa of allergic patients with undertreated or treatment-resistant asthma may also increase the risk of acute exacerbations because these cells can amplify T helper type 2 lymphocyte-driven inflammation on every exposure to the clinically relevant allergen and can promote further inflammation on rhinovirus infections by allowing viral replication and releasing additional pro-inflammatory factors. Improved methods for the isolation and functional analysis of pure populations of viable circulating fibrocytes have allowed a better understanding of the effector role of these cells. A reliable and clinically applicable assay has been developed to measure blood fibrocyte counts as outcome measure in future clinical trials. New therapeutic agents are needed to block both persistent inflammation and fibrocytosis in corticosteroid-resistant asthma.
Collapse
Affiliation(s)
- S Mattoli
- Avail Biomedical Research Institute, Scientific Direction and Project Management Centre, Basel, Switzerland
| |
Collapse
|
42
|
Abstract
Airway hyperresponsiveness (AHR) is a defining characteristic of asthma that refers to the capacity of the airways to undergo exaggerated narrowing in response to stimuli that do not result in comparable degrees of airway narrowing in healthy subjects. Airway smooth muscle (ASM) contraction mediates airway narrowing, but it remains uncertain as to whether the smooth muscle is intrinsically altered in asthmatic subjects or is responding abnormally as a result of the milieu in which it sits. ASM in the trachea or major bronchi does not differ in its contractile characteristics in asthmatics, but the more pertinent peripheral airways await complete exploration. The mass of ASM is increased in many but not all asthmatics and therefore cannot be a unifying hypothesis for AHR, although when increased in mass it may contribute to AHR. The inability of a deep breath to reverse or prevent bronchial narrowing in asthma may reflect an intrinsic difference in the mechanisms that lead to softening of contracted ASM when subjected to stretch. Cytokines such as interleukin-13 and tumor necrosis factor-α promote a more contractile ASM phenotype. The composition and increased stiffness of the matrix in which ASM is embedded promotes a more proliferative and pro-inflammatory ASM phenotype, but the expected dedifferentiation and loss of contractility have not been shown. Airway epithelium may drive ASM proliferation and/or molecular remodeling in ways that may lead to AHR. In conclusion, AHR is likely multifactorial in origin, reflecting the plasticity of ASM properties in the inflammatory environment of the asthmatic airway.
Collapse
Affiliation(s)
- Anne-Marie Lauzon
- Meakins-Christie Laboratories, McGill University Health Center Research Institute, Montreal, QC, Canada; Department of Medicine, McGill University, Montreal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University Health Center Research Institute, Montreal, QC, Canada; Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Hall SC, Fischer KD, Agrawal DK. The impact of vitamin D on asthmatic human airway smooth muscle. Expert Rev Respir Med 2015; 10:127-35. [PMID: 26634624 DOI: 10.1586/17476348.2016.1128326] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Asthma is a chronic heterogeneous disorder, which involves airway inflammation, airway hyperresponsiveness (AHR) and airway remodeling. The airway smooth muscle (ASM) bundle regulates the broncho-motor tone and plays a critical role in AHR as well as orchestrating inflammation. Vitamin D deficiency has been linked to increased severity and exacerbations of symptoms in asthmatic patients. It has been shown to modulate both immune and structural cells, including ASM cells, in inflammatory diseases. Given that current asthma therapies have not been successful in reversing airway remodeling, vitamin D supplementation as a potential therapeutic option has gained a great deal of attention. Here, we highlight the potential immunomodulatory properties of vitamin D in regulating ASM function and airway inflammation in bronchial asthma.
Collapse
Affiliation(s)
- Sannette C Hall
- a Department of Biomedical Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Kimberly D Fischer
- b Department of Medical Microbiology and Immunology , Creighton University School of Medicine , Omaha , NE , USA
| | - Devendra K Agrawal
- a Department of Biomedical Science , Creighton University School of Medicine , Omaha , NE , USA.,b Department of Medical Microbiology and Immunology , Creighton University School of Medicine , Omaha , NE , USA.,c Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| |
Collapse
|
44
|
Trian T, Allard B, Ozier A, Maurat E, Dupin I, Thumerel M, Ousova O, Gillibert-Duplantier J, Le Morvan V, Begueret H, Girodet PO, Marthan R, Berger P. Selective dysfunction of p53 for mitochondrial biogenesis induces cellular proliferation in bronchial smooth muscle from asthmatic patients. J Allergy Clin Immunol 2015; 137:1717-1726.e13. [PMID: 26688517 DOI: 10.1016/j.jaci.2015.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Increase of bronchial smooth muscle (BSM) mass is a crucial feature of asthma remodeling. The mechanisms of such an increased BSM mass are complex but involve enhanced mitochondrial biogenesis, leading to increased proliferation of BSM cells in asthmatic patients. The major tumor suppressor protein p53 is a key cell regulator involved in cell proliferation and has also been implicated in mitochondrial biogenesis. However, the role of p53 in BSM cell proliferation and mitochondrial biogenesis has not been investigated thus far. OBJECTIVE We sought to evaluate the role of p53 in proliferation of BSM cells in asthmatic patients and mitochondrial biogenesis. METHODS The expression of p53 was assessed both in vitro by using flow cytometry and Western blotting and ex vivo by using RT-PCR after laser microdissection. The role of p53 was assessed with small hairpin RNA lentivirus in both asthmatic patients and control subjects with BSM cell proliferation by using 5-bromo-2'-deoxyuridine and cell counting and in the expression of p21, BCL2-associated X protein, mitochondrial transcription factor A (TFAM), and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). RESULTS Twenty-nine patients with moderate-to-severe asthma and 26 control subjects were enrolled in the study. p53 expression was increased in BSM from asthmatic patients both ex vivo and in vitro, with a decreased interaction with mouse double minute 2 homolog (Mdm2) and an increased phosphorylation of serine 20. p53 did not inhibit the transcription of both TFAM and PGC-1α in BSM cells from asthmatic patients. As a consequence, p53 is unable to slow the increased mitochondrial biogenesis and hence the subsequent increased proliferation of BSM cells in asthmatic patients. CONCLUSION This study suggests that p53 might act as a new potential therapeutic target against BSM remodeling in asthmatic patients.
Collapse
Affiliation(s)
- Thomas Trian
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France.
| | - Benoit Allard
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
| | - Annaig Ozier
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France; CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| | - Elise Maurat
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
| | - Isabelle Dupin
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
| | - Matthieu Thumerel
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France; CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| | - Olga Ousova
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
| | - Jennifer Gillibert-Duplantier
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
| | | | - Hugues Begueret
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| | - Pierre-Olivier Girodet
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France; CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| | - Roger Marthan
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France; CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| | - Patrick Berger
- Université Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France; CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, Service de chirurgie thoracique, Service d'anatomopathologie, Pessac, France
| |
Collapse
|
45
|
Lin TY, Venkatesan N, Nishioka M, Kyoh S, Al-Alwan L, Baglole CJ, Eidelman DH, Ludwig MS, Hamid Q. Monocyte-derived fibrocytes induce an inflammatory phenotype in airway smooth muscle cells. Clin Exp Allergy 2015; 44:1347-60. [PMID: 25255717 DOI: 10.1111/cea.12421] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 08/30/2014] [Accepted: 09/04/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Infiltration of fibrocytes (FC) in the airway smooth muscle is a feature of asthma, but the pathological significance is unknown. OBJECTIVE We sought to explore whether FC modulate the phenotype of airway smooth muscle cells (ASMC) in asthmatic vs. control subjects. METHODS Fibrocytes were isolated from CD14+ monocytes from asthmatic and normal subjects. Proliferation of ASMC of asthmatic or normal subjects was analysed by (3) H-thymidine incorporation, cell number counting and Ki-67 expression after treatment of ASMC with FC-conditioned medium (FCCM) or co-culture with FC. ASMC-associated cytokines/chemokines implicated in asthma (TGF-β1, eotaxin, IL-6 and IL-8) were measured in co-culture or transwell culture of ASMC + FC by ELISA. Immunofluorescence staining was performed to localize these cytokines in ASMC. Cytokine secretion was measured in the transwell culture of ASMC + FC, where NF-κB-p65 or ERK1/2 in ASMC was silenced by siRNA. Contractile phenotype of ASMC in transwell culture was assessed by immunoblotting of α-smooth muscle actin (α-SMA) and myosin light chain kinase (MLCK). RESULTS Fibrocytes did not affect ASMC proliferation and expression of TGF-β1, eotaxin, α-SMA and MLCK; however, ASMC production of IL-8 and IL-6 was increased in the co-culture and transwell culture by FC. ASMC treated with FCCM were immunopositive for IL-8/IL-6 and produced more IL-8/IL-6. Furthermore, siRNA silencing of NF-κB-p65 or ERK1/2 in transwell cultures of asthmatic ASMC with normal subject FC decreased IL-8 and IL-6 production. CONCLUSIONS AND CLINICAL RELEVANCE Fibrocytes promoted IL-8 and IL-6 production by ASMC, demonstrating a proinflammatory role for FC and a possible mechanism of the inflammatory phenotype in asthma.
Collapse
Affiliation(s)
- T-Y Lin
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada; Department of Thoracic Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Smooth muscle CaMKIIδ promotes allergen-induced airway hyperresponsiveness and inflammation. Pflugers Arch 2015; 467:2541-54. [PMID: 26089028 DOI: 10.1007/s00424-015-1713-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/02/2015] [Accepted: 06/04/2015] [Indexed: 12/28/2022]
Abstract
Airway smooth muscle (ASM) is a key target cell in allergen-induced asthma known to contribute to airway hyperresponsiveness (AHR) and chronic airway remodeling. Changes in ASM calcium homeostasis have been shown to contribute to AHR although the mechanisms and Ca(2+) signal effectors are incompletely understood. In the present study, we tested the function of ASM multifunctional protein kinase Ca(2+)/calmodulin-dependent kinase II (CaMKII) isoforms CaMKIIδ and CaMKIIγ in allergen-induced AHR and airway remodeling in vivo. Using a murine model of atopic asthma, we demonstrate that CaMKIIδ protein is upregulated in ASM derived from ovalbumin (OVA)-treated animals compared to controls. A genetic approach to conditionally knock out smooth muscle CaMKIIδ and CaMKIIγ in separate Cre-loxp systems was validated, and using this loss-of-function approach, the function of these CaMKII isoforms was tested in ovalbumin (OVA)-induced airway remodeling and AHR. OVA treatment in control mice had no effect on ASM remodeling in this model of AHR, and CaMKIIδ knockouts had no independent effects on ASM content. However, at 1 day post-final OVA challenge, OVA-induced AHR was eliminated in the CaMKIIδ knockouts. OVA-induced peribronchial inflammation and bronchoalveolar lavage fluid (BALF) levels of the Th2 cytokine IL-13 were significantly decreased in the CaMKIIδ knockouts. Unexpectedly, we found increased peribronchial eosinophils in the smooth muscle CaMKIIδ knockouts compared to control animals at 1 day post-final challenge, suggesting that lack of ASM CaMKIIδ delays the progression of AHR rather than inhibiting it. Indeed, when AHR was determined at 7 days post-final OVA challenge, CaMKIIδ knockouts showed robust AHR while AHR was fully resolved in OVA-challenged control mice. These in vivo studies demonstrate a role for smooth muscle CaMKIIδ in promoting airway inflammation and AHR and suggest a complex signaling role for CaMKIIδ in regulating ASM function. These studies confirm the diverse roles of ASM cells as immune effectors that control AHR and call for further studies into CaMKIIδ-mediated signaling in ASM cells during disease.
Collapse
|
47
|
Girodet PO, Dournes G, Thumerel M, Begueret H, Dos Santos P, Ozier A, Dupin I, Trian T, Montaudon M, Laurent F, Marthan R, Berger P. Calcium channel blocker reduces airway remodeling in severe asthma. A proof-of-concept study. Am J Respir Crit Care Med 2015; 191:876-83. [PMID: 25633090 DOI: 10.1164/rccm.201410-1874oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Severe asthma is a major public health issue throughout the world. Increased bronchial smooth muscle (BSM) mass, a characteristic feature of airway remodeling in severe asthma, is associated with resistance to high-intensity treatment and poor prognosis. In vitro, the Ca(2+)-channel blocker gallopamil decreased the proliferation of BSM cells from patients with severe asthma. OBJECTIVES We conducted a double-blind, randomized, placebo-controlled study to evaluate the effect of gallopamil on airway remodeling in patients with severe asthma. METHODS Subjects received either gallopamil (n = 16) or placebo (n = 15) for 1 year and were monitored for an additional 3-month period. Airway remodeling was analyzed at baseline and after treatment phase using both fiberoptic bronchoscopy and computed tomography scan. The primary end point was the BSM area. Secondary end points included normalized BSM thickness and frequency of asthma exacerbations. MEASUREMENTS AND MAIN RESULTS BSM area was reduced in the gallopamil group (baseline vs. end of treatment) but was unchanged in the placebo group. Between-group differences in BSM area were not significantly different in gallopamil versus placebo groups. By contrast, between-group differences in normalized BSM thickness were significantly different between the two groups. The mean number of exacerbations per month was not different during the treatment phase in gallopamil versus placebo group but was significantly lower in patients previously treated with gallopamil during the follow-up period. There were no differences between the groups with respect to overall side effects. CONCLUSIONS Gallopamil treatment for 12 months reduces BSM remodeling and prevents the occurrence of asthma exacerbations. Clinical trial registered with www.clinicaltrials.gov (NCT 00896428).
Collapse
Affiliation(s)
- Pierre-Olivier Girodet
- 1 Université de Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Bordeaux, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Asthma remains a major health problem with significant morbidity, mortality and economic costs. In asthma, airway remodelling, which refers to all the microscopic structural changes seen in the airway tissue, has been recognised for many decades and remains one of the defining characteristics of the disease; however, it is still poorly understood. The detrimental pathophysiological consequences of some features of remodelling, like increased airway smooth muscle mass and subepithelial fibrosis, are well documented. However, whether targeting these by therapy would be beneficial is unknown. Although the prevailing thinking is that remodelling is an abnormal response to persistent airway inflammation, recent evidence, especially from studies of remodelling in asthmatic children, suggests that the two processes occur in parallel. The effects of asthma therapy on airway remodelling have not been studied extensively due to the challenges of obtaining airway tissue in the context of clinical trials. Corticosteroids remain the cornerstone of asthma therapy, and their effects on remodelling have been better studied than other drugs. Bronchial thermoplasty is the only asthma therapy to primarily target remodelling, although how it results in the apparent clinical benefits seen is not exactly clear. In this article we discuss the mechanisms of airway remodelling in asthma and review the effects of conventional and novel asthma therapies on the process.
Collapse
Affiliation(s)
- Rachid Berair
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, Glenfield Hospital, University of Leicester, Leicester, LE3 9QP, UK
| | | |
Collapse
|
49
|
Trian T, Allard B, Dupin I, Carvalho G, Ousova O, Maurat E, Bataille J, Thumerel M, Begueret H, Girodet PO, Marthan R, Berger P. House dust mites induce proliferation of severe asthmatic smooth muscle cells via an epithelium-dependent pathway. Am J Respir Crit Care Med 2015; 191:538-46. [PMID: 25569771 DOI: 10.1164/rccm.201409-1582oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Asthma is a frequent airway disease, and asthma control determinants have been associated with indoor allergen sensitization. The most frequent allergens are house dust mites (HDM), which act in vivo on the bronchial epithelial layer. Severe asthma has also been associated with bronchial remodeling and more specifically with increased mass of bronchial smooth muscle (BSM). However, the relationship between HDM stimulation of the bronchial epithelial layer and BSM remodeling is unknown. OBJECTIVES To evaluate whether epithelial stimulation with HDM induces BSM cell proliferation in subjects with severe asthma. METHODS A total of 22 subjects with severe asthma and 27 subjects with no asthma were recruited. We have developed an in vitro culture model combining an epithelium layer in air-liquid interface (ALI) interacting with BSM. We assessed BSM proliferation using BrdU incorporation. We explored the role of epithelium-derived mediators using reverse-transcriptase polymerase chain reaction (RT-PCR) and ELISA in vitro and in vivo. Finally, leukotrienes receptor expression was assessed in vitro by flow cytometry and RT-PCR and ex vivo by laser microdissection and RT-PCR. MEASUREMENTS AND MAIN RESULTS We found that epithelial stimulation by HDM selectively increased the proliferation of asthmatic BSM cells and not that of nonasthmatic cells. The mechanism involved epithelial protease-activated receptor-2-dependent production of leukotrienes C4 associated with an overexpression of leukotrienes receptor CysLTR1 by asthmatic BSM cells in vitro and ex vivo. CONCLUSIONS This work demonstrates the selective role of HDM on BSM remodeling in patients with severe asthma and points out different therapeutic targets at epithelial and smooth muscle levels.
Collapse
|
50
|
Al Heialy S, Zeroual M, Farahnak S, McGovern T, Risse PA, Novali M, Lauzon AM, Roman HN, Martin JG. Nanotubes connect CD4+ T cells to airway smooth muscle cells: novel mechanism of T cell survival. THE JOURNAL OF IMMUNOLOGY 2015; 194:5626-34. [PMID: 25934863 DOI: 10.4049/jimmunol.1401718] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 04/11/2015] [Indexed: 11/19/2022]
Abstract
Contact between airway smooth muscle (ASM) cells and activated CD4(+) T cells, a key interaction in diseases such as asthma, triggers ASM cell proliferation and enhances T cell survival. We hypothesized that direct contact between ASM and CD4(+) T cells facilitated the transfer of anti-apoptotic proteins via nanotubes, resulting in increased survival of activated CD4(+) T cells. CD4(+) T cells, isolated from PBMCs of healthy subjects, when activated and cocultured with ASM cells for 24 h, formed nanotubes that were visualized by immunofluorescence and atomic force microscopy. Cell-to-cell transfer of the fluorescent dye calcein-AM confirmed cytoplasmic communication via nanotubes. Immunoreactive B cell lymphoma 2 (Bcl-2) and induced myeloid leukemia cell differentiation protein (Mcl-1), two major anti-apoptotic proteins, were present within the nanotubes. Downregulation of Mcl-1 by small interfering RNA in ASM cells significantly increased T cell apoptosis, whereas downregulation of Bcl-2 had no effect. Transfer of GFP-tagged Mcl-1 from ASM cells to CD4(+) T cells via the nanotubes confirmed directionality of transfer. In conclusion, activated T cells communicate with ASM cells via nanotube formation. Direct transfer of Mcl-1 from ASM to CD(+) T cells via nanotubes is involved in T cell survival. This study provides a novel mechanism of survival of CD4(+) T cells that is dependent on interaction with a structural cell.
Collapse
Affiliation(s)
- Saba Al Heialy
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Melissa Zeroual
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Soroor Farahnak
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Toby McGovern
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Paul-André Risse
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Mauro Novali
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - Horia N Roman
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada; andResearch Institute, McGill University Health Centre, Montreal, Quebec H2X 2P2, Canada
| |
Collapse
|