1
|
Fu M, Lin Y, Yang J, Cheng J, Lin L, Wang G, Long C, Xu S, Lu J, Li G, Yan J, Chen G, Zhuo S, Chen D. Multitask machine learning-based tumor-associated collagen signatures predict peritoneal recurrence and disease-free survival in gastric cancer. Gastric Cancer 2024; 27:1242-1257. [PMID: 39271552 DOI: 10.1007/s10120-024-01551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Accurate prediction of peritoneal recurrence for gastric cancer (GC) is crucial in clinic. The collagen alterations in tumor microenvironment affect the migration and treatment response of cancer cells. Herein, we proposed multitask machine learning-based tumor-associated collagen signatures (TACS), which are composed of quantitative collagen features derived from multiphoton imaging, to simultaneously predict peritoneal recurrence (TACSPR) and disease-free survival (TACSDFS). METHODS Among 713 consecutive patients, with 275 in training cohort, 222 patients in internal validation cohort, and 216 patients in external validation cohort, we developed and validated a multitask machine learning model for simultaneously predicting peritoneal recurrence (TACSPR) and disease-free survival (TACSDFS). The accuracy of the model for prediction of peritoneal recurrence and prognosis as well as its association with adjuvant chemotherapy were evaluated. RESULTS The TACSPR and TACSDFS were independently associated with peritoneal recurrence and disease-free survival in three cohorts, respectively (all P < 0.001). The TACSPR demonstrated a favorable performance for peritoneal recurrence in all three cohorts. In addition, the TACSDFS also showed a satisfactory accuracy for disease-free survival among included patients. For stage II and III diseases, adjuvant chemotherapy improved the survival of patients with low TACSPR and low TACSDFS, or high TACSPR and low TACSDFS, or low TACSPR and high TACSDFS, but had no impact on patients with high TACSPR and high TACSDFS. CONCLUSIONS The multitask machine learning model allows accurate prediction of peritoneal recurrence and survival for GC and could distinguish patients who might benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Meiting Fu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Guangzhou, 510515, People's Republic of China
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
| | - Yuyu Lin
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Junyao Yang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Liyan Lin
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Guangxing Wang
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
| | - Chenyan Long
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuoyu Xu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jianping Lu
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, People's Republic of China
| | - Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
2
|
Usman K, Fouadi M, Nwozor KO, Aminazadeh F, Nair P, Luo H, Sin DD, Osei ET, Hackett TL. Interleukin-1α inhibits transforming growth factor-β1 and β2-induced extracellular matrix production, remodeling and signaling in human lung fibroblasts: Master regulator in lung mucosal repair. Matrix Biol 2024; 132:47-58. [PMID: 39147560 DOI: 10.1016/j.matbio.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-β (TGF-β) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS Stimulation of lung fibroblasts with TGF-β1 and TGF-β2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-β1 and TGF-β2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-β1 or TGF-β2. Mechanistically, IL-1α administration led to the suppression of TGF-β1 and TGF-β2 signaling, through downregulation of mRNA and protein for TGF-β receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION IL-1α acts as a master regulator, modulating TGF-β1 and TGF-β2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-β signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-β1 or TGF-β2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.
Collapse
Affiliation(s)
- Kauna Usman
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - May Fouadi
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kingsley Okechukwu Nwozor
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Fatemeh Aminazadeh
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Parameswaran Nair
- Division of Respirology, St Joseph's Healthcare Hamilton & McMaster University, ON L8N 4A6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Emmanuel Twumasi Osei
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Biology, University of British Columbia, Okanagan, BC V1V 1V7, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
3
|
Hynds RE, Magin CM, Ikonomou L, Aschner Y, Beers MF, Burgess JK, Heise RL, Hume PS, Krasnodembskaya AD, Mei SHJ, Misharin AV, Park JA, Reynolds SD, Tschumperlin DJ, Tanneberger AE, Vaidyanathan S, Waters CM, Zettler PJ, Weiss DJ, Ryan AL. Stem cells, cell therapies, and bioengineering in lung biology and diseases 2023. Am J Physiol Lung Cell Mol Physiol 2024; 327:L327-L340. [PMID: 38772903 PMCID: PMC11442098 DOI: 10.1152/ajplung.00052.2024] [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: 02/14/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/23/2024] Open
Abstract
Repair and regeneration of a diseased lung using stem cells or bioengineered tissues is an exciting therapeutic approach for a variety of lung diseases and critical illnesses. Over the past decade, increasing evidence from preclinical models suggests that mesenchymal stromal cells, which are not normally resident in the lung, can be used to modulate immune responses after injury, but there have been challenges in translating these promising findings to the clinic. In parallel, there has been a surge in bioengineering studies investigating the use of artificial and acellular lung matrices as scaffolds for three-dimensional lung or airway regeneration, with some recent attempts of transplantation in large animal models. The combination of these studies with those involving stem cells, induced pluripotent stem cell derivatives, and/or cell therapies is a promising and rapidly developing research area. These studies have been further paralleled by significant increases in our understanding of the molecular and cellular events by which endogenous lung stem and/or progenitor cells arise during lung development and participate in normal and pathological remodeling after lung injury. For the 2023 Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases Conference, scientific symposia were chosen to reflect the most cutting-edge advances in these fields. Sessions focused on the integration of "omics" technologies with function, the influence of immune cells on regeneration, and the role of the extracellular matrix in regeneration. The necessity for basic science studies to enhance fundamental understanding of lung regeneration and to design innovative translational studies was reinforced throughout the conference.
Collapse
Affiliation(s)
- Robert E Hynds
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Chelsea M Magin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Laertis Ikonomou
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Michael F Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine and PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Patrick S Hume
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States
| | - Susan D Reynolds
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
| | - Alicia E Tanneberger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Sriram Vaidyanathan
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Christopher M Waters
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Patricia J Zettler
- Moritz College of Law, Drug Enforcement and Policy Center, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont, United States
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
4
|
Lee PH, An M, Hwang D, Jang AS. Impact of circulating angiomotin and angiostatin on clinical variables in patients with asthma. Ann Allergy Asthma Immunol 2024; 133:101-103. [PMID: 38556077 DOI: 10.1016/j.anai.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Affiliation(s)
- Pureun-Haneul Lee
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea.
| | - MinHyeok An
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - DaYeon Hwang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - An-Soo Jang
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| |
Collapse
|
5
|
Liu YB, Tan XH, Yang HH, Yang JT, Zhang CY, Jin L, Yang NSY, Guan CX, Zhou Y, Liu SK, Xiong JB. Wnt5a-mediated autophagy contributes to the epithelial-mesenchymal transition of human bronchial epithelial cells during asthma. Mol Med 2024; 30:93. [PMID: 38898476 PMCID: PMC11188189 DOI: 10.1186/s10020-024-00862-3] [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: 02/24/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) of human bronchial epithelial cells (HBECs) is essential for airway remodeling during asthma. Wnt5a has been implicated in various lung diseases, while its role in the EMT of HBECs during asthma is yet to be determined. This study sought to define whether Wnt5a initiated EMT, leading to airway remodeling through the induction of autophagy in HBECs. METHODS Microarray analysis was used to investigate the expression change of WNT5A in asthma patients. In parallel, EMT models were induced using 16HBE cells by exposing them to house dust mites (HDM) or interleukin-4 (IL-4), and then the expression of Wnt5a was observed. Using in vitro gain- and loss-of-function approaches via Wnt5a mimic peptide FOXY5 and Wnt5a inhibitor BOX5, the alterations in the expression of the epithelial marker E-cadherin and the mesenchymal marker protein were observed. Mechanistically, the Ca2+/CaMKII signaling pathway and autophagy were evaluated. An autophagy inhibitor 3-MA was used to examine Wnt5a in the regulation of autophagy during EMT. Furthermore, we used a CaMKII inhibitor KN-93 to determine whether Wnt5a induced autophagy overactivation and EMT via the Ca2+/CaMKII signaling pathway. RESULTS Asthma patients exhibited a significant increase in the gene expression of WNT5A compared to the healthy control. Upon HDM and IL-4 treatments, we observed that Wnt5a gene and protein expression levels were significantly increased in 16HBE cells. Interestingly, Wnt5a mimic peptide FOXY5 significantly inhibited E-cadherin and upregulated α-SMA, Collagen I, and autophagy marker proteins (Beclin1 and LC3-II). Rhodamine-phalloidin staining showed that FOXY5 resulted in a rearrangement of the cytoskeleton and an increase in the quantity of stress fibers in 16HBE cells. Importantly, blocking Wnt5a with BOX5 significantly inhibited autophagy and EMT induced by IL-4 in 16HBE cells. Mechanistically, autophagy inhibitor 3-MA and CaMKII inhibitor KN-93 reduced the EMT of 16HBE cells caused by FOXY5, as well as the increase in stress fibers, cell adhesion, and autophagy. CONCLUSION This study illustrates a new link in the Wnt5a-Ca2+/CaMKII-autophagy axis to triggering airway remodeling. Our findings may provide novel strategies for the treatment of EMT-related diseases.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, 410011, China.
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| | - Jian-Bing Xiong
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
6
|
Roth M, Han B, S’ng CT, Hoang BX, Lambers C. Zinc Iodide Dimethyl Sulfoxide Reduces Collagen Deposition by Increased Matrix Metalloproteinase-2 Expression and Activity in Lung Fibroblasts. Biomedicines 2024; 12:1257. [PMID: 38927463 PMCID: PMC11200730 DOI: 10.3390/biomedicines12061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic inflammatory lung diseases are characterized by disease-specific extracellular matrix accumulation resulting from an imbalance of matrix metalloproteinases (MMPs) and their inhibitors. Zinc is essential for the function of MMPs, and zinc deficiency has been associated with enhanced tissue remodeling. This study assessed if zinc iodide (ZnI) supplementation through dimethyl sulfoxide (DMSO) modifies the action of MMPs in isolated human lung fibroblasts. The expression and activity of two gelatinases, MMP-2 and MMP-9, were determined by gelatin zymography and enzyme-linked immuno-sorbent assay (ELISA). Collagen degradation was determined by cell-based ELISAs. Collagen type I and fibronectin deposition was stimulated by human recombinant tumor growth factor β1 (TGF-β1). Untreated fibroblasts secreted MMP-2 but only minute amounts of MMP-9. TGF-β1 (5 ng/mL) reduced MMP-2 secretion, but stimulated collagen type I and fibronectin deposition. All the effects of TGF-β1 were significantly reduced in cells treated with ZnI-DMSO over 24 h, while ZnI and DMSO alone had a lower reducing effect. ZnI-DMSO alone did not increase MMP secretion but enhanced the ratio of active to inactive of MMP-2. ZnI alone had a lower enhancing effect than ZnI-DMSO on MMP activity. Furthermore, MMP-2 activity was increased by ZnI-DMSO and ZnI in the absence of cells. Soluble collagen type I increased in the medium of ZnI-DMSO- and ZnI-treated cells. Blocking MMP activity counteracted all the effects of ZnI-DMSO. Conclusion: The data suggest that the combination of ZnI with DMSO reduces fibrotic processes by increasing the degradation of collagen type I by up-regulating the activity of gelatinases. Thus, the combination of ZnI with DMSO might be considered for treatment of fibrotic disorders of the lung. DMSO supported the beneficial effects of ZnI.
Collapse
Affiliation(s)
- Michael Roth
- University Hospital of Basel, University of Basel, 4031 Basel, Switzerland
| | - Bo Han
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Ba Xuan Hoang
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher Lambers
- Department of Pneumology, Ordensklinikum Linz Elisabethinen, Fadingerstr. 1, 4020 Linz, Austria;
| |
Collapse
|
7
|
Cheng PP, Yu F, Chen SJ, Feng X, Jia ZH, Hu SH, Cui XL, Zhou YY, Niu Q, Liang LM, Wang M, Song LJ, He XL, Xiong L, Xiang F, Wang X, Ma WL, Ye H. PM2.5 exposure-induced senescence-associated secretory phenotype in airway smooth muscle cells contributes to airway remodeling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123674. [PMID: 38458517 DOI: 10.1016/j.envpol.2024.123674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
Fine particulate matter (PM2.5) has been linked to increased severity and incidence of airway diseases, especially chronic obstructive pulmonary disease (COPD) and asthma. Airway remodeling is an important event in both COPD and asthma, and airway smooth muscle cells (ASMCs) are key cells which directly involved in airway remodeling. However, it was unclear how PM2.5 affected ASMCs. This study investigates the effects of PM2.5 on airway smooth muscle and its mechanism. We first showed that inhaled particulate matter was distributed in the airway smooth muscle bundle, combined with increased airway smooth muscle bundle and collagen deposition in vivo. Then, we demonstrated that PM2.5 induced up-regulation of collagen-I and alpha-smooth muscle actin (α-SMA) expression in rat and human ASMCs in vitro. Next, we found PM2.5 led to rat and human ASMCs senescence and exhibited senescence-associated secretory phenotype (SASP) by autophagy-induced GATA4/TRAF6/NF-κB signaling, which contributed to collagen-I and α-SMA synthesis as well as airway smooth muscle remodeling. Together, our results provided evidence that SASP induced by PM2.5 in airway smooth muscle cells prompted airway remodeling.
Collapse
Affiliation(s)
- Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Shuai-Jun Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zi-Heng Jia
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shi-He Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Lin Cui
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Ya Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Xiaorong Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China.
| |
Collapse
|
8
|
Caputo LDS, Alves CDL, Laranjeira IM, Fonseca-Rodrigues D, da Silva Filho AA, Dias ACP, Pinto-Ribeiro F, Pereira Junior ODS, de Paula ACC, Nagato AC, Corrêa JODA. Copaiba oil minimizes inflammation and promotes parenchyma re-epithelization in acute allergic asthma model induced by ovalbumin in BALB/c mice. Front Pharmacol 2024; 15:1356598. [PMID: 38666018 PMCID: PMC11043548 DOI: 10.3389/fphar.2024.1356598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction: Asthma is a condition of airflow limitation, common throughout the world, with high mortality rates, especially as it still faces some obstacles in its management. As it constitutes a public health challenge, this study aimed to investigate the effect of copaiba oil (e.g., Copaifera langsdorffii), as a treatment resource, at doses of 50 and 100 mg/kg on certain mediators of acute lung inflammation (IL-33, GATA3, FOXP3, STAT3, and TBET) and early mechanisms of lung remodeling (degradation of elastic fiber tissues, collagen deposition, and goblet cell hyperplasia). Methods: Using an ovalbumin-induced acute allergic asthma model in BALB/c mice, we analyzed the inflammatory mediators through immunohistochemistry and the mechanisms of lung remodeling through histopathology, employing orcein, Masson's trichrome, and periodic acid-Schiff staining. Results: Copaiba oil treatment (CO) reduced IL-33 and increased FOXP3 by stimulating the FOXP3/GATA3 and FOXP3/STAT3 pathways. Additionally, it upregulated TBET, suggesting an additional role in controlling GATA3 activity. In the respiratory epithelium, CO decreased the fragmentation of elastic fibers while increasing the deposition of collagen fibers, favoring epithelial restructuring. Simultaneously, CO reduced goblet cell hyperplasia. Discussion: Although additional research is warranted, the demonstrated anti-inflammatory and re-epithelializing action makes CO a viable option in exploring new treatments for acute allergic asthma.
Collapse
Affiliation(s)
- Ludmila de Souza Caputo
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina de Lima Alves
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Inês Martins Laranjeira
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Centre of Molecular and Environmental Biology, CBMA, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Diana Fonseca-Rodrigues
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
| | | | - Alberto Carlos Pires Dias
- Centre of Molecular and Environmental Biology, CBMA, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
| | | | | | - Akinori Cardozo Nagato
- Department of Physiology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, Brazil
| | | |
Collapse
|
9
|
Russell RJ, Boulet LP, Brightling CE, Pavord ID, Porsbjerg C, Dorscheid D, Sverrild A. The airway epithelium: an orchestrator of inflammation, a key structural barrier and a therapeutic target in severe asthma. Eur Respir J 2024; 63:2301397. [PMID: 38453256 PMCID: PMC10991852 DOI: 10.1183/13993003.01397-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Asthma is a disease of heterogeneous pathology, typically characterised by excessive inflammatory and bronchoconstrictor responses to the environment. The clinical expression of the disease is a consequence of the interaction between environmental factors and host factors over time, including genetic susceptibility, immune dysregulation and airway remodelling. As a critical interface between the host and the environment, the airway epithelium plays an important role in maintaining homeostasis in the face of environmental challenges. Disruption of epithelial integrity is a key factor contributing to multiple processes underlying asthma pathology. In this review, we first discuss the unmet need in asthma management and provide an overview of the structure and function of the airway epithelium. We then focus on key pathophysiological changes that occur in the airway epithelium, including epithelial barrier disruption, immune hyperreactivity, remodelling, mucus hypersecretion and mucus plugging, highlighting how these processes manifest clinically and how they might be targeted by current and novel therapeutics.
Collapse
Affiliation(s)
- Richard J Russell
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| | - Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
10
|
Kayalar Ö, Rajabi H, Konyalilar N, Mortazavi D, Aksoy GT, Wang J, Bayram H. Impact of particulate air pollution on airway injury and epithelial plasticity; underlying mechanisms. Front Immunol 2024; 15:1324552. [PMID: 38524119 PMCID: PMC10957538 DOI: 10.3389/fimmu.2024.1324552] [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: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
Air pollution plays an important role in the mortality and morbidity of chronic airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Particulate matter (PM) is a significant fraction of air pollutants, and studies have demonstrated that it can cause airway inflammation and injury. The airway epithelium forms the first barrier of defense against inhaled toxicants, such as PM. Airway epithelial cells clear airways from inhaled irritants and orchestrate the inflammatory response of airways to these irritants by secreting various lipid mediators, growth factors, chemokines, and cytokines. Studies suggest that PM plays an important role in the pathogenesis of chronic airway diseases by impairing mucociliary function, deteriorating epithelial barrier integrity, and inducing the production of inflammatory mediators while modulating the proliferation and death of airway epithelial cells. Furthermore, PM can modulate epithelial plasticity and airway remodeling, which play central roles in asthma and COPD. This review focuses on the effects of PM on airway injury and epithelial plasticity, and the underlying mechanisms involving mucociliary activity, epithelial barrier function, airway inflammation, epithelial-mesenchymal transition, mesenchymal-epithelial transition, and airway remodeling.
Collapse
Affiliation(s)
- Özgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Hadi Rajabi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Deniz Mortazavi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Gizem Tuşe Aksoy
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Jun Wang
- Department of Biomedicine and Biopharmacology, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Türkiye
| |
Collapse
|
11
|
Zhang M, Zhou JX, Huang CQ, Feng KN, Zou XL, Cen JM, Meng P, Li HT, Zhang TT. IL-38 alleviates airway remodeling in chronic asthma via blocking the profibrotic effect of IL-36γ. Clin Exp Immunol 2023; 214:260-274. [PMID: 37586814 PMCID: PMC10719219 DOI: 10.1093/cei/uxad099] [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: 02/18/2023] [Revised: 06/30/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023] Open
Abstract
Airway remodeling is a major feature of asthma. Interleukin (IL)-36γ is significantly upregulated and promotes airway hyper-responsiveness (AHR) in asthma, but its role in airway remodeling is unknown. Here, we aimed to investigate the role of IL-36γ in airway remodeling, and whether IL-38 can alleviate airway remodeling in chronic asthma by blocking the effects of IL-36γ. IL-36γ was quantified in mice inhaled with house dust mite (HDM). Extracellular matrix (ECM) deposition in lung tissues and AHR were assessed following IL-36γ administration to mice. Airway inflammation, AHR, and remodeling were evaluated after IL-38 or blocking IL-36 receptor (IL-36R) treatment in asthmatic mice. The effects of lung fibroblasts stimulated with IL-36γ and IL-38 were quantified in vitro. Increased expression of IL-36γ was detected in lung tissues of HDM-induced asthmatic mice. The intratracheal instillation of IL-36γ to mice significantly enhanced the ECM deposition, AHR, and the number of activated lung fibroblasts around the airways. IL-38 or blocking IL-36R treated asthmatic mice showed a significant alleviation in the airway inflammation, AHR, airway remodeling, and number of activated fibroblasts around airways as compared with the HDM group. In vitro, IL-36γ promoted the activation and migration of human lung fibroblasts (HFL-1). The administration of IL-38 can counteract these biological processes induced by IL-36γ in HFL-1cells. The results indicated that IL-38 can mitigate airway remodeling by blocking the profibrotic effects of IL-36γ in chronic asthma. IL-36γ may be a new therapeutic target, and IL-38 is a potential candidate agent for inhibiting airway remodeling in asthma.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Xia Zhou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chu-Qin Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kang-Ni Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Ling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jie-Mei Cen
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ping Meng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hong-Tao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tian-Tuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Savin IA, Zenkova MA, Sen’kova AV. Bronchial Asthma, Airway Remodeling and Lung Fibrosis as Successive Steps of One Process. Int J Mol Sci 2023; 24:16042. [PMID: 38003234 PMCID: PMC10671561 DOI: 10.3390/ijms242216042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Bronchial asthma is a heterogeneous disease characterized by persistent respiratory system inflammation, airway hyperreactivity, and airflow obstruction. Airway remodeling, defined as changes in airway wall structure such as extensive epithelial damage, airway smooth muscle hypertrophy, collagen deposition, and subepithelial fibrosis, is a key feature of asthma. Lung fibrosis is a common occurrence in the pathogenesis of fatal and long-term asthma, and it is associated with disease severity and resistance to therapy. It can thus be regarded as an irreversible consequence of asthma-induced airway inflammation and remodeling. Asthma heterogeneity presents several diagnostic challenges, particularly in distinguishing between chronic asthma and other pulmonary diseases characterized by disruption of normal lung architecture and functions, such as chronic obstructive pulmonary disease. The search for instruments that can predict the development of irreversible structural changes in the lungs, such as chronic components of airway remodeling and fibrosis, is particularly difficult. To overcome these challenges, significant efforts are being directed toward the discovery and investigation of molecular characteristics and biomarkers capable of distinguishing between different types of asthma as well as between asthma and other pulmonary disorders with similar structural characteristics. The main features of bronchial asthma etiology, pathogenesis, and morphological characteristics as well as asthma-associated airway remodeling and lung fibrosis as successive stages of one process will be discussed in this review. The most common murine models and biomarkers of asthma progression and post-asthmatic fibrosis will also be covered. The molecular mechanisms and key cellular players of the asthmatic process described and systematized in this review are intended to help in the search for new molecular markers and promising therapeutic targets for asthma prediction and therapy.
Collapse
Affiliation(s)
| | | | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent’ev Ave 8, 630090 Novosibirsk, Russia; (I.A.S.); (M.A.Z.)
| |
Collapse
|
13
|
Jesenak M, Durdik P, Oppova D, Franova S, Diamant Z, Golebski K, Banovcin P, Vojtkova J, Novakova E. Dysfunctional mucociliary clearance in asthma and airway remodeling - New insights into an old topic. Respir Med 2023; 218:107372. [PMID: 37516275 DOI: 10.1016/j.rmed.2023.107372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
Bronchial asthma is a heterogeneous respiratory condition characterized by chronic airway inflammation, airway hyperresponsiveness and airway structural changes (known as remodeling). The clinical symptoms can be evoked by (non)specific triggers, and their intensity varies over time. In the past, treatment was mainly focusing on symptoms' alleviation; in contrast modern treatment strategies target the underlying inflammation, even during asymptomatic periods. Components of airway remodeling include epithelial cell shedding and dysfunction, goblet cell hyperplasia, subepithelial matrix protein deposition, fibrosis, neoangiogenesis, airway smooth muscle cell hypertrophy and hyperplasia. Among the other important, and frequently forgotten aspects of airway remodeling, also loss of epithelial barrier integrity, immune defects in anti-infectious defence and mucociliary clearance (MCC) dysfunction should be pointed out. Mucociliary clearance represents one of the most important defence airway mechanisms. Several studies in asthmatics demonstrated various dysfunctions in MCC - e.g., ciliated cells displaying intracellular disorientation, abnormal cilia and cytoplasmic blebs. Moreover, excessive mucus production and persistent cough are one of the well-recognized features of severe asthma and are also associated with defects in MCC. Damaged airway epithelium and impaired function of the ciliary cells leads to MCC dysfunction resulting in higher susceptibility to infection and inflammation. Therefore, new strategies aimed on restoring the remodeling changes and MCC dysfunction could present a new therapeutic approach for the management of asthma and other chronic respiratory diseases.
Collapse
Affiliation(s)
- Milos Jesenak
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia; Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia; Department of Clinical Immunology and Allergology, University Teaching Hospital in Martin, Martin, Slovakia
| | - Peter Durdik
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia
| | - Dasa Oppova
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia
| | - Sona Franova
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Zuzana Diamant
- Department of Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Belgium; Department of Respiratory Medicine & Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden; Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic; Department of Clinical Pharmacy & Pharmacology, University in Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Kornel Golebski
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter Banovcin
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia
| | - Jarmila Vojtkova
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovakia.
| | - Elena Novakova
- Department of Microbiology and Immunology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia.
| |
Collapse
|
14
|
Brussow J, Feng K, Thiam F, Phogat S, Osei ET. Epithelial-fibroblast interactions in IPF: Lessons from in vitro co-culture studies. Differentiation 2023; 134:11-19. [PMID: 37738701 DOI: 10.1016/j.diff.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/19/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial disease that is characterized by increased cellular proliferation and differentiation together with excessive extracellular matrix (ECM) deposition leading to buildup of scar tissue (fibrosis) and remodeling in the lungs. The activated and differentiated (myo)fibroblasts are one of the main sources of tissue remodeling in IPF and a crucial mechanism known to contribute to this feature is an aberrant crosstalk between pulmonary fibroblasts and the abnormal or injured pulmonary epithelium. This epithelial-fibroblast interaction mimics the temporal, spatial and cell-type specific crosstalk between the endoderm and mesoderm in the so-called epithelial-mesenchymal trophic unit (EMTU) during lung development that is proposed to be activated in healthy lung repair and dysregulated in various lung diseases including IPF. To study the dysregulated lung EMTU in IPF, various complex in vitro models have been established. Hence, in this review, we will provide a summary of studies that have used complex (3-dimensional) in vitro co-culture, and organoid models to assess how abnormal epithelial-fibroblast interactions in lung EMTU contribute to crucial features of the IPF including defective cellular differentiation, proliferation and migration as well as increased ECM deposition.
Collapse
Affiliation(s)
- J Brussow
- Department of Biology, Okanagan Campus, University of British Columbia, Kelowna, BC, Canada
| | - K Feng
- Department of Biology, Okanagan Campus, University of British Columbia, Kelowna, BC, Canada
| | - F Thiam
- Department of Biology, Okanagan Campus, University of British Columbia, Kelowna, BC, Canada
| | - S Phogat
- Department of Biology, Okanagan Campus, University of British Columbia, Kelowna, BC, Canada
| | - E T Osei
- Department of Biology, Okanagan Campus, University of British Columbia, Kelowna, BC, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
15
|
Phogat S, Thiam F, Al Yazeedi S, Abokor FA, Osei ET. 3D in vitro hydrogel models to study the human lung extracellular matrix and fibroblast function. Respir Res 2023; 24:242. [PMID: 37798767 PMCID: PMC10552248 DOI: 10.1186/s12931-023-02548-6] [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: 06/26/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
The pulmonary extracellular matrix (ECM) is a macromolecular structure that provides mechanical support, stability and elastic recoil for different pulmonary cells including the lung fibroblasts. The ECM plays an important role in lung development, remodeling, repair, and the maintenance of tissue homeostasis. Biomechanical and biochemical signals produced by the ECM regulate the phenotype and function of various cells including fibroblasts in the lungs. Fibroblasts are important lung structural cells responsible for the production and repair of different ECM proteins (e.g., collagen and fibronectin). During lung injury and in chronic lung diseases such as asthma, idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD), an abnormal feedback between fibroblasts and the altered ECM disrupts tissue homeostasis and leads to a vicious cycle of fibrotic changes resulting in tissue remodeling. In line with this, using 3D hydrogel culture models with embedded lung fibroblasts have enabled the assessment of the various mechanisms involved in driving defective (fibrotic) fibroblast function in the lung's 3D ECM environment. In this review, we provide a summary of various studies that used these 3D hydrogel models to assess the regulation of the ECM on lung fibroblast phenotype and function in altered lung ECM homeostasis in health and in chronic respiratory disease.
Collapse
Affiliation(s)
- Sakshi Phogat
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Fama Thiam
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Safiya Al Yazeedi
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Filsan Ahmed Abokor
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Emmanuel Twumasi Osei
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada.
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
16
|
Jeon H, Harvey M, Cisek R, Bennett E, Tokarz D. Characterization of pathological stomach tissue using polarization-sensitive second harmonic generation microscopy. BIOMEDICAL OPTICS EXPRESS 2023; 14:5376-5391. [PMID: 37854565 PMCID: PMC10581783 DOI: 10.1364/boe.500335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 09/09/2023] [Indexed: 10/20/2023]
Abstract
Alterations in collagen ultrastructure between human gastric adenocarcinoma and normal gastric tissue were investigated using polarization-resolved second harmonic generation (PSHG) microscopy. Cylindrical and trigonal symmetries were assumed to extract quantitative PSHG parameters, ρ, κ and S, from each image pixel. Statistically significant variations in these values were observed for gastric adenocarcinoma, indicating a higher disorder of collagen. Numerical focal volume simulations of crossing fibrils indicate increased S parameter is due to more intersecting collagen fibrils of varying diameters. These parameters were also able to distinguish between different grades of gastric adenocarcinoma indicating that PSHG may be useful for automated cancer diagnosis.
Collapse
Affiliation(s)
- Hwanhee Jeon
- Department of Chemistry, Saint Mary’s University, 923 Robie Street, Halifax, Nova Scotia, B3H 3C3, Canada
| | - MacAulay Harvey
- Department of Chemistry, Saint Mary’s University, 923 Robie Street, Halifax, Nova Scotia, B3H 3C3, Canada
| | - Richard Cisek
- Department of Chemistry, Saint Mary’s University, 923 Robie Street, Halifax, Nova Scotia, B3H 3C3, Canada
| | - Elisha Bennett
- Department of Chemistry, Saint Mary’s University, 923 Robie Street, Halifax, Nova Scotia, B3H 3C3, Canada
| | - Danielle Tokarz
- Department of Chemistry, Saint Mary’s University, 923 Robie Street, Halifax, Nova Scotia, B3H 3C3, Canada
| |
Collapse
|
17
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
18
|
Hewitt RJ, Puttur F, Gaboriau DCA, Fercoq F, Fresquet M, Traves WJ, Yates LL, Walker SA, Molyneaux PL, Kemp SV, Nicholson AG, Rice A, Roberts E, Lennon R, Carlin LM, Byrne AJ, Maher TM, Lloyd CM. Lung extracellular matrix modulates KRT5 + basal cell activity in pulmonary fibrosis. Nat Commun 2023; 14:6039. [PMID: 37758700 PMCID: PMC10533905 DOI: 10.1038/s41467-023-41621-y] [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: 12/03/2022] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Aberrant expansion of KRT5+ basal cells in the distal lung accompanies progressive alveolar epithelial cell loss and tissue remodelling during fibrogenesis in idiopathic pulmonary fibrosis (IPF). The mechanisms determining activity of KRT5+ cells in IPF have not been delineated. Here, we reveal a potential mechanism by which KRT5+ cells migrate within the fibrotic lung, navigating regional differences in collagen topography. In vitro, KRT5+ cell migratory characteristics and expression of remodelling genes are modulated by extracellular matrix (ECM) composition and organisation. Mass spectrometry- based proteomics revealed compositional differences in ECM components secreted by primary human lung fibroblasts (HLF) from IPF patients compared to controls. Over-expression of ECM glycoprotein, Secreted Protein Acidic and Cysteine Rich (SPARC) in the IPF HLF matrix restricts KRT5+ cell migration in vitro. Together, our findings demonstrate how changes to the ECM in IPF directly influence KRT5+ cell behaviour and function contributing to remodelling events in the fibrotic niche.
Collapse
Affiliation(s)
- Richard J Hewitt
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Franz Puttur
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - David C A Gaboriau
- Facility for Imaging by Light Microscopy, National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | | | - Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - William J Traves
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Laura L Yates
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Samuel V Kemp
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust, City Campus, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Andrew G Nicholson
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Alexandra Rice
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Edward Roberts
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Leo M Carlin
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Keck Medicine of USC, 1510 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
19
|
Xu T, Wu Z, Yuan Q, Zhang X, Liu Y, Wu C, Song M, Wu J, Jiang J, Wang Z, Chen Z, Zhang M, Huang M, Ji N. Proline is increased in allergic asthma and promotes airway remodeling. JCI Insight 2023; 8:e167395. [PMID: 37432745 PMCID: PMC10543727 DOI: 10.1172/jci.insight.167395] [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: 11/28/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Proline and its synthesis enzyme pyrroline-5-carboxylate reductase 1 (PYCR1) are implicated in epithelial-mesenchymal transition (EMT), yet how proline and PYCR1 function in allergic asthmatic airway remodeling via EMT has not yet been addressed to our knowledge. In the present study, increased levels of plasma proline and PYCR1 were observed in patients with asthma. Similarly, proline and PYCR1 in lung tissues were high in a murine allergic asthma model induced by house dust mites (HDMs). Pycr1 knockout decreased proline in lung tissues, with reduced airway remodeling and EMT. Mechanistically, loss of Pycr1 restrained HDM-induced EMT by modulating mitochondrial fission, metabolic reprogramming, and the AKT/mTORC1 and WNT3a/β-catenin signaling pathways in airway epithelial cells. Therapeutic inhibition of PYCR1 in wild-type mice disrupted HDM-induced airway inflammation and remodeling. Deprivation of exogenous proline relieved HDM-induced airway remodeling to some extent. Collectively, this study illuminates that proline and PYCR1 involved with airway remodeling in allergic asthma could be viable targets for asthma treatment.
Collapse
Affiliation(s)
- Tingting Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenzhen Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Yuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xijie Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chaojie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meijuan Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingxian Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Jiangsu Province Engineering Research Center of Antibody Drug, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Mottais A, Riberi L, Falco A, Soccal S, Gohy S, De Rose V. Epithelial-Mesenchymal Transition Mechanisms in Chronic Airway Diseases: A Common Process to Target? Int J Mol Sci 2023; 24:12412. [PMID: 37569787 PMCID: PMC10418908 DOI: 10.3390/ijms241512412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a reversible process, in which epithelial cells lose their epithelial traits and acquire a mesenchymal phenotype. This transformation has been described in different lung diseases, such as lung cancer, interstitial lung diseases, asthma, chronic obstructive pulmonary disease and other muco-obstructive lung diseases, such as cystic fibrosis and non-cystic fibrosis bronchiectasis. The exaggerated chronic inflammation typical of these pulmonary diseases can induce molecular reprogramming with subsequent self-sustaining aberrant and excessive profibrotic tissue repair. Over time this process leads to structural changes with progressive organ dysfunction and lung function impairment. Although having common signalling pathways, specific triggers and regulation mechanisms might be present in each disease. This review aims to describe the various mechanisms associated with fibrotic changes and airway remodelling involved in chronic airway diseases. Having better knowledge of the mechanisms underlying the EMT process may help us to identify specific targets and thus lead to the development of novel therapeutic strategies to prevent or limit the onset of irreversible structural changes.
Collapse
Affiliation(s)
- Angélique Mottais
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Andrea Falco
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Simone Soccal
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
21
|
Abstract
Collagen provides mechanical and biological support for virtually all human tissues in the extracellular matrix (ECM). Its defining molecular structure, the triple-helix, could be damaged and denatured in disease and injuries. To probe collagen damage, the concept of collagen hybridization has been proposed, revised, and validated through a series of investigations reported as early as 1973: a collagen-mimicking peptide strand may form a hybrid triple-helix with the denatured chains of natural collagen but not the intact triple-helical collagen proteins, enabling assessment of proteolytic degradation or mechanical disruption to collagen within a tissue-of-interest. Here we describe the concept and development of collagen hybridization, summarize the decades of chemical investigations on rules underlying the collagen triple-helix folding, and discuss the growing biomedical evidence on collagen denaturation as a previously overlooked ECM signature for an array of conditions involving pathological tissue remodeling and mechanical injuries. Finally, we propose a series of emerging questions regarding the chemical and biological nature of collagen denaturation and highlight the diagnostic and therapeutic opportunities from its targeting.
Collapse
Affiliation(s)
- Xiaojing Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Qi Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - S. Michael Yu
- Department of Biomedical Engineering, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Yang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| |
Collapse
|
22
|
Erjefält JS. Anatomical and histopathological approaches to asthma phenotyping. Respir Med 2023; 210:107168. [PMID: 36822489 DOI: 10.1016/j.rmed.2023.107168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
Asthma is typically characterized by variable respiratory symptoms and airflow limitation. Along with the pathophysiology and symptoms are immunological and inflammatory processes. The last decades research has revealed that the immunology of asthma is highly heterogeneous. This has clinical consequences and identification of immunological phenotypes is currently used to guide biological treatment. The focus of this review is on another dimension of asthma diversity, namely anatomical heterogeneity. Immunopathological alterations may go beyond the central airways to also involve the distal airways, the alveolar parenchyma, and pulmonary vessels. Also, extrapulmonary tissues are affected. The anatomical distribution of inflammation in asthma has remained relatively poorly discussed despite its potential implication on both clinical presentation and response to treatment. There is today evidence that a significant proportion of the asthma patients has small airway disease with type 2 immunity, eosinophilia and smooth muscle infiltration of mast cells. The small airways in asthma are also subjected to remodelling, constriction, and luminal plugging, events that are likely to contribute to the elevated distal airway resistance seen in some patients. In cases when the inflammation extends into the alveolar parenchyma alveolar FCER1-high mast cells, eosinophilia, type 2 immunity and activated alveolar macrophages, together with modest interstitial remodelling, create a complex immunopathological picture. Importantly, the distal lung inflammation in asthma can be pharmacologically targeted by use of inhalers with more distal drug deposition. Biological treatments, which are readily distributed to the distal lung, may also be beneficial in eligible patients with more severe and anatomically widespread disease.
Collapse
Affiliation(s)
- Jonas S Erjefält
- Unit of Airway Inflammation, Department of Experimental Medical Research, Lund University, Lund, Sweden; Department of Allergology and Respiratory Medicine, Skane University Hospital, Lund, Sweden.
| |
Collapse
|
23
|
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: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
24
|
Feng Y, Hu J, Liu F, Shang Y. Collagen Triple Helix Repeat Containing 1 Deficiency Protects Against Airway Remodeling and Inflammation in Asthma Models In Vivo and In Vitro. Inflammation 2023; 46:925-940. [PMID: 36640227 DOI: 10.1007/s10753-022-01781-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 11/07/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023]
Abstract
Asthma is a chronic inflammatory disease characterized by airway remodeling and lung inflammation. Collagen triple helix repeat containing 1 (CTHRC1), a glycoprotein, is involved in multiple pathological processes, including inflammation and fibrosis. However, the function of CTHRC1 in asthma remains unclear. In the present study, the mouse asthma model was successfully generated by sensitizing and challenging mice with ovalbumin (OVA). CTHRC1 expression at both RNA and protein levels was significantly upregulated in lung tissues of asthmatic mice. Asthmatic mice exhibited significant airway remodeling as evidenced by increased bronchial wall and smooth muscle cell layer thickness, goblet cell hyperplasia and collagen deposition, and epithelial-mesenchymal transition (EMT), but those characteristics were reversed by CTHRC1 silencing. The cell model with transforming growth factor-β1 (TGF-β1) induction in bronchial epithelial cells (BEAS-2B) was conducted to verify the effects of CTHRC1 on EMT, a classic mechanism that mediates airway remodeling. The results showed that TGF-β1 stimulation increased CTHRC1 expression, and CTHRC1 knockdown inhibited TGF-β1-induced EMT. OVA-treated mice also showed increased inflammatory cell infiltration and the production of OVA-specific immunoglobulin E (IgE), interleukin (IL)-4, IL-5, and IL-13, which were decreased by CTHRC1 downregulation. The effects of CTHRC1 on OVA-induced airway inflammation were further determined by treating BEAS-2B cells with IL-13, in which CTHRC1 knockdown reduced the IL-13-induced secretion of pro-inflammatory factors, including IL-4 and IL-5. In conclusion, these results indicate that CTHRC1 silencing attenuates asthmatic airway remodeling and inflammation in vivo and in vitro, suggesting that CTHRC1 may be a potential target for asthma treatment.
Collapse
Affiliation(s)
- Yong Feng
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Liaoning Province, 110004, China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Liaoning Province, 110004, China
| | - Fen Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Liaoning Province, 110004, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Liaoning Province, 110004, China.
| |
Collapse
|
25
|
Khalfaoui L, Pabelick CM. Airway smooth muscle in contractility and remodeling of asthma: potential drug target mechanisms. Expert Opin Ther Targets 2023; 27:19-29. [PMID: 36744401 DOI: 10.1080/14728222.2023.2177533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Asthma is characterized by enhanced airway contractility and remodeling where airway smooth muscle (ASM) plays a key role, modulated by inflammation. Understanding the mechanisms by which ASM contributes to these features of asthma is essential for the development of novel asthma therapies. AREAS COVERED Inflammation in asthma contributes to a multitude of changes within ASM including enhanced airway contractility, proliferation, and fibrosis. Altered intracellular calcium ([Ca2+]i) regulation or Ca2+ sensitization contributes to airway hyperreactivity. Increased airway wall thickness from ASM proliferation and fibrosis contributes to structural changes seen with asthma. EXPERT OPINION ASM plays a significant role in multiple features of asthma. Increased ASM contractility contributes to hyperresponsiveness, while altered ASM proliferation and extracellular matrix production promote airway remodeling both influenced by inflammation of asthma and conversely even influencing the local inflammatory milieu. While standard therapies such as corticosteroids or biologics target inflammation, cytokines, or their receptors to alleviate asthma symptoms, these approaches do not address the underlying contribution of ASM to hyperresponsiveness and particularly remodeling. Therefore, novel therapies for asthma need to target abnormal contractility mechanisms in ASM and/or the contribution of ASM to remodeling, particularly in asthmatics resistant to current therapies.
Collapse
Affiliation(s)
- Latifa Khalfaoui
- Departments of Anesthesiology & Perioperative Medicine Mayo Clinic, Rochester, MN, USA
| | - Christina M Pabelick
- Departments of Anesthesiology & Perioperative Medicine Mayo Clinic, Rochester, MN, USA.,Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Varricchi G, Ferri S, Pepys J, Poto R, Spadaro G, Nappi E, Paoletti G, Virchow JC, Heffler E, Canonica WG. Biologics and airway remodeling in severe asthma. Allergy 2022; 77:3538-3552. [PMID: 35950646 PMCID: PMC10087445 DOI: 10.1111/all.15473] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Asthma is a chronic inflammatory airway disease resulting in airflow obstruction, which in part can become irreversible to conventional therapies, defining the concept of airway remodeling. The introduction of biologics in severe asthma has led in some patients to the complete normalization of previously considered irreversible airflow obstruction. This highlights the need to distinguish a "fixed" airflow obstruction due to structural changes unresponsive to current therapies, from a "reversible" one as demonstrated by lung function normalization during biological therapies not previously obtained even with high-dose systemic glucocorticoids. The mechanisms by which exposure to environmental factors initiates the inflammatory responses that trigger airway remodeling are still incompletely understood. Alarmins represent epithelial-derived cytokines that initiate immunologic events leading to inflammatory airway remodeling. Biological therapies can improve airflow obstruction by addressing these airway inflammatory changes. In addition, biologics might prevent and possibly even revert "fixed" remodeling due to structural changes. Hence, it appears clinically important to separate the therapeutic effects (early and late) of biologics as a new paradigm to evaluate the effects of these drugs and future treatments on airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Sebastian Ferri
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy
| | - Jack Pepys
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Emanuele Nappi
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Giovanni Paoletti
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Enrico Heffler
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Walter G Canonica
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
27
|
Joglekar MM, Nizamoglu M, Fan Y, Nemani SSP, Weckmann M, Pouwels SD, Heijink IH, Melgert BN, Pillay J, Burgess JK. Highway to heal: Influence of altered extracellular matrix on infiltrating immune cells during acute and chronic lung diseases. Front Pharmacol 2022; 13:995051. [PMID: 36408219 PMCID: PMC9669433 DOI: 10.3389/fphar.2022.995051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/19/2022] [Indexed: 10/31/2023] Open
Abstract
Environmental insults including respiratory infections, in combination with genetic predisposition, may lead to lung diseases such as chronic obstructive pulmonary disease, lung fibrosis, asthma, and acute respiratory distress syndrome. Common characteristics of these diseases are infiltration and activation of inflammatory cells and abnormal extracellular matrix (ECM) turnover, leading to tissue damage and impairments in lung function. The ECM provides three-dimensional (3D) architectural support to the lung and crucial biochemical and biophysical cues to the cells, directing cellular processes. As immune cells travel to reach any site of injury, they encounter the composition and various mechanical features of the ECM. Emerging evidence demonstrates the crucial role played by the local environment in recruiting immune cells and their function in lung diseases. Moreover, recent developments in the field have elucidated considerable differences in responses of immune cells in two-dimensional versus 3D modeling systems. Examining the effect of individual parameters of the ECM to study their effect independently and collectively in a 3D microenvironment will help in better understanding disease pathobiology. In this article, we discuss the importance of investigating cellular migration and recent advances in this field. Moreover, we summarize changes in the ECM in lung diseases and the potential impacts on infiltrating immune cell migration in these diseases. There has been compelling progress in this field that encourages further developments, such as advanced in vitro 3D modeling using native ECM-based models, patient-derived materials, and bioprinting. We conclude with an overview of these state-of-the-art methodologies, followed by a discussion on developing novel and innovative models and the practical challenges envisaged in implementing and utilizing these systems.
Collapse
Affiliation(s)
- Mugdha M. Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - YiWen Fan
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Sai Sneha Priya Nemani
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Markus Weckmann
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Simon D. Pouwels
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Barbro N. Melgert
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, Groningen, Netherlands
| | - Janesh Pillay
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Critical Care, Groningen, Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, Netherlands
| |
Collapse
|
28
|
Aghali A, Khalfaoui L, Lagnado AB, Drake LY, Teske JJ, Pabelick CM, Passos JF, Prakash YS. Cellular senescence is increased in airway smooth muscle cells of elderly persons with asthma. Am J Physiol Lung Cell Mol Physiol 2022; 323:L558-L568. [PMID: 36166734 PMCID: PMC9639764 DOI: 10.1152/ajplung.00146.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Senescent cells can drive age-related tissue dysfunction partially via a senescence-associated secretory phenotype (SASP) involving proinflammatory and profibrotic factors. Cellular senescence has been associated with a structural and functional decline during normal lung aging and age-related diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Asthma in the elderly (AIE) represents a major healthcare burden. AIE is associated with bronchial airway hyperresponsiveness and remodeling, which involves increased cell proliferation and higher rates of fibrosis, and resistant to standard therapy. Airway smooth muscle (ASM) cells play a major role in asthma such as remodeling via modulation of inflammation and the extracellular matrix (ECM) environment. Whether senescent ASM cells accumulate in AIE and contribute to airway structural or functional changes is unknown. Lung tissues from elderly persons with asthma showed greater airway fibrosis compared with age-matched elderly persons with nonasthma and young age controls. Lung tissue or isolated ASM cells from elderly persons with asthma showed increased expression of multiple senescent markers including phospho-p53, p21, telomere-associated foci (TAF), as well as multiple SASP components. Senescence and SASP components were also increased with aging per se. These data highlight the presence of cellular senescence in AIE that may contribute to airway remodeling.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony B. Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Li Y. Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jacob J. Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christina M. Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
29
|
Zeng Y, Su X, Takezawa MG, Fichtinger PS, Lee UN, Pippin JW, Shankland SJ, Lim FY, Denlinger LC, Jarjour NN, Mathur SK, Sandbo N, Berthier E, Esnault S, Bernau K, Theberge AB. An open microfluidic coculture model of fibroblasts and eosinophils to investigate mechanisms of airway inflammation. Front Bioeng Biotechnol 2022; 10:993872. [PMID: 36246374 PMCID: PMC9558094 DOI: 10.3389/fbioe.2022.993872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Interactions between fibroblasts and immune cells play an important role in tissue inflammation. Previous studies have found that eosinophils activated with interleukin-3 (IL-3) degranulate on aggregated immunoglobulin G (IgG) and release mediators that activate fibroblasts in the lung. However, these studies were done with eosinophil-conditioned media that have the capacity to investigate only one-way signaling from eosinophils to fibroblasts. Here, we demonstrate a coculture model of primary normal human lung fibroblasts (HLFs) and human blood eosinophils from patients with allergy and asthma using an open microfluidic coculture device. In our device, the two types of cells can communicate via two-way soluble factor signaling in the shared media while being physically separated by a half wall. Initially, we assessed the level of eosinophil degranulation by their release of eosinophil-derived neurotoxin (EDN). Next, we analyzed the inflammation-associated genes and soluble factors using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and multiplex immunoassays, respectively. Our results suggest an induction of a proinflammatory fibroblast phenotype of HLFs following the coculture with degranulating eosinophils, validating our previous findings. Additionally, we present a new result that indicate potential impacts of activated HLFs back on eosinophils. This open microfluidic coculture platform provides unique opportunities to investigate the intercellular signaling between the two cell types and their roles in airway inflammation and remodeling.
Collapse
Affiliation(s)
- Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Xiaojing Su
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Meg G. Takezawa
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Paul S. Fichtinger
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ulri N. Lee
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Jeffery W. Pippin
- Division of Nephrology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Stuart J. Shankland
- Division of Nephrology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Fang Yun Lim
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Loren C. Denlinger
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nizar N. Jarjour
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Sameer K. Mathur
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nathan Sandbo
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Stephane Esnault
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ksenija Bernau
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, WA, United States
- Department of Urology, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
30
|
Novel Lung Growth Strategy with Biological Therapy Targeting Airway Remodeling in Childhood Bronchial Asthma. CHILDREN 2022; 9:children9081253. [PMID: 36010143 PMCID: PMC9406359 DOI: 10.3390/children9081253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022]
Abstract
Anti-inflammatory therapy, centered on inhaled steroids, suppresses airway inflammation in asthma, reduces asthma mortality and hospitalization rates, and achieves clinical remission in many pediatric patients. However, the spontaneous remission rate of childhood asthma in adulthood is not high, and airway inflammation and airway remodeling persist after remission of asthma symptoms. Childhood asthma impairs normal lung maturation, interferes with peak lung function in adolescence, reduces lung function in adulthood, and increases the risk of developing chronic obstructive pulmonary disease (COPD). Early suppression of airway inflammation in childhood and prevention of asthma exacerbations may improve lung maturation, leading to good lung function and prevention of adult COPD. Biological drugs that target T-helper 2 (Th2) cytokines are used in patients with severe pediatric asthma to reduce exacerbations and airway inflammation and improve respiratory function. They may also suppress airway remodeling in childhood and prevent respiratory deterioration in adulthood, reducing the risk of COPD and improving long-term prognosis. No studies have demonstrated a suppressive effect on airway remodeling in childhood severe asthma, and further clinical trials using airway imaging analysis are needed to ascertain the inhibitory effect of biological drugs on airway remodeling in severe childhood asthma. In this review, we describe the natural prognosis of lung function in childhood asthma and the risk of developing adult COPD, the pathophysiology of allergic airway inflammation and airway remodeling via Th2 cytokines, and the inhibitory effect of biological drugs on airway remodeling in childhood asthma.
Collapse
|
31
|
Zhu Y, Esnault S, Ge Y, Jarjour NN, Brasier AR. Airway fibrin formation cascade in allergic asthma exacerbation: implications for inflammation and remodeling. Clin Proteomics 2022; 19:15. [PMID: 35590254 PMCID: PMC9117591 DOI: 10.1186/s12014-022-09351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Airway remodeling in patients with asthma, which leads to a decline in pulmonary function, is likely the result of repeated exacerbations often provoked by aeroallergen exposures. Aeroallegen exposure triggers a stereotypic response orchestrated by growth factor cytokines and other protein mediators. This results in a late-phase allergic reaction characterized by vascular permeability, recruitment of activated leukocytes, and activation of structural cells of the airway. The spectrum of protein mediators and their functions are incompletely understood. METHODS Bronchoalveolar lavage fluid (BALF) samples were obtained from 12 volunteers who exhibited robust eosinophilic recruitment following segmental bronchial provocation with allergen (SBP-Ag). We systematically identified and quantified proteins in BALF using high-performance liquid chromatography-high-resolution mass spectrometry (LC-MS/MS) followed by pathway analysis and correlations with airway physiology. RESULTS Pairwise analysis of protein abundance in BALF pre- vs post-SBP-Ag revealed that 55 proteins were upregulated and 103 proteins were downregulated. We observed enrichment of groups of proteins mapping to hemostasis/fibrin clot, platelet activation, lipoprotein assembly, neutrophil degranulation proteins, and acute-phase inflammation-airway remodeling pathways. The abundances of F2 and Fibrinogen γ (FGG) correlated with eosinophil numbers, whereas SERPINA3 negatively correlated with change in FeNO. The coagulation proteins F2 and KNG negatively correlated with FN1 an index of airway remodeling. Interestingly, patients with lower FEV1 showed distinct allergen-induced patterns of 8 BALF proteins, including MUC1, alarmins (HSPB1), and actin polymerization factors. CONCLUSIONS Protein abundance of the fibrin formation cascade, platelet activation and remodeling are associated with late-phase leukocyte numbers and markers of remodeling. Patients with lower FEV1 have distinct dynamic responses to allergen.
Collapse
Affiliation(s)
- Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Nizar N Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, 53705, USA
| | - Allan R Brasier
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, 715 Highland Ave, Madison, WI, 53705, USA.
| |
Collapse
|
32
|
Savin IA, Markov AV, Zenkova MA, Sen’kova AV. Asthma and Post-Asthmatic Fibrosis: A Search for New Promising Molecular Markers of Transition from Acute Inflammation to Pulmonary Fibrosis. Biomedicines 2022; 10:biomedicines10051017. [PMID: 35625754 PMCID: PMC9138542 DOI: 10.3390/biomedicines10051017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
Asthma is a heterogeneous pulmonary disorder, the progression and chronization of which leads to airway remodeling and fibrogenesis. To understand the molecular mechanisms of pulmonary fibrosis development, key genes forming the asthma-specific regulome and involved in lung fibrosis formation were revealed using a comprehensive bioinformatics analysis. The bioinformatics data were validated using a murine model of ovalbumin (OVA)-induced asthma and post-asthmatic fibrosis. The performed analysis revealed a range of well-known pro-fibrotic markers (Cat, Ccl2, Ccl4, Ccr2, Col1a1, Cxcl12, Igf1, Muc5ac/Muc5b, Spp1, Timp1) and a set of novel genes (C3, C3ar1, Col4a1, Col4a2, Cyp2e1, Fn1, Thbs1, Tyrobp) mediating fibrotic changes in lungs already at the stage of acute/subacute asthma-driven inflammation. The validation of genes related to non-allergic bleomycin-induced pulmonary fibrosis on asthmatic/fibrotic lungs allowed us to identify new universal genes (Col4a1 and Col4a2) associated with the development of lung fibrosis regardless of its etiology. The similarities revealed in the expression profiles of nodal fibrotic genes between asthma-driven fibrosis in mice and nascent idiopathic pulmonary fibrosis in humans suggest a tight association of identified genes with the early stages of airway remodeling and can be considered as promising predictors and early markers of pulmonary fibrosis.
Collapse
|
33
|
Janulaityte I, Januskevicius A, Rimkunas A, Palacionyte J, Vitkauskiene A, Malakauskas K. Asthmatic Eosinophils Alter the Gene Expression of Extracellular Matrix Proteins in Airway Smooth Muscle Cells and Pulmonary Fibroblasts. Int J Mol Sci 2022; 23:4086. [PMID: 35456903 PMCID: PMC9031271 DOI: 10.3390/ijms23084086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 11/26/2022] Open
Abstract
The impaired production of extracellular matrix (ECM) proteins by airway smooth muscle cells (ASMC) and pulmonary fibroblasts (PF) is a part of airway remodeling in asthma. This process might be influenced by eosinophils that migrate to the airway and abundantly secrete various cytokines, including TGF-β. We aimed to investigate the effect of asthmatic eosinophils on the gene expression of ECM proteins in ASMC and PF. A total of 34 study subjects were recruited: 14 with allergic asthma (AA), 9 with severe non-allergic eosinophilic asthma (SNEA), and 11 healthy subjects (HS). All AA patients underwent bronchial allergen challenge with D. pteronyssinus. The peripheral blood eosinophils were isolated using high-density centrifugation and magnetic separation. The individual cell cultures were made using hTERT ASMC and MRC-5 cell lines and the subjects' eosinophils. The gene expression of ECM and the TGF-β signaling pathway was analyzed using qRT-PCR. We found that asthmatic eosinophils significantly promoted collagen I, fibronectin, versican, tenascin C, decorin, vitronectin, periostin, vimentin, MMP-9, ADAM33, TIMP-1, and TIMP-2 gene expression in ASMC and collagen I, collagen III, fibronectin, elastin, decorin, MMP-2, and TIMP-2 gene expression in PF compared with the HS eosinophil effect. The asthmatic eosinophils significantly increased the gene expression of several canonical and non-canonical TGF-β signaling pathway components in ASMC and PF compared with the HS eosinophil effect. The allergen-activated AA and SNEA eosinophils had a greater effect on these changes. In conclusion, asthmatic eosinophils, especially SNEA and allergen-activated eosinophils, imbalanced the gene expression of ECM proteins and their degradation-regulating proteins. These changes were associated with increased gene expression of TGF-β signaling pathway molecules in ASMC and PF.
Collapse
Affiliation(s)
- Ieva Janulaityte
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (A.R.); (K.M.)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (A.R.); (K.M.)
| | - Airidas Rimkunas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (A.R.); (K.M.)
| | - Jolita Palacionyte
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | - Astra Vitkauskiene
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | - Kestutis Malakauskas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (A.R.); (K.M.)
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| |
Collapse
|
34
|
Burgess JK, Harmsen MC. Chronic lung diseases: entangled in extracellular matrix. Eur Respir Rev 2022; 31:31/163/210202. [PMID: 35264410 DOI: 10.1183/16000617.0202-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/17/2021] [Indexed: 01/10/2023] Open
Abstract
The extracellular matrix (ECM) is the scaffold that provides structure and support to all organs, including the lung; however, it is also much more than this. The ECM provides biochemical and biomechanical cues to cells that reside or transit through this micro-environment, instructing their responses. The ECM structure and composition changes in chronic lung diseases; how such changes impact disease pathogenesis is not as well understood. Cells bind to the ECM through surface receptors, of which the integrin family is one of the most widely recognised. The signals that cells receive from the ECM regulate their attachment, proliferation, differentiation, inflammatory secretory profile and survival. There is extensive evidence documenting changes in the composition and amount of ECM in diseased lung tissues. However, changes in the topographical arrangement, organisation of the structural fibres and stiffness (or viscoelasticity) of the matrix in which cells are embedded have an undervalued but strong impact on cell phenotype. The ECM in diseased lungs also changes in physical and biomechanical ways that drive cellular responses. The characteristics of these environments alter cell behaviour and potentially orchestrate perpetuation of lung diseases. Future therapies should target ECM remodelling as much as the underlying culprit cells.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands .,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, KOLFF Institute - REGENERATE, Groningen, The Netherlands
| | - Martin C Harmsen
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, KOLFF Institute - REGENERATE, Groningen, The Netherlands
| |
Collapse
|
35
|
Hackett TL, Vriesde NRTF, AL-Fouadi M, Mostaco-Guidolin L, Maftoun D, Hsieh A, Coxson N, Usman K, Sin DD, Booth S, Osei ET. The Role of the Dynamic Lung Extracellular Matrix Environment on Fibroblast Morphology and Inflammation. Cells 2022; 11:cells11020185. [PMID: 35053300 PMCID: PMC8773771 DOI: 10.3390/cells11020185] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) supports lung tissue architecture and physiology by providing mechanical stability and elastic recoil. Over the last several decades, it has become increasingly clear that the stiffness of the ECM governs many cellular processes, including cell-phenotype and functions during development, healing, and disease. Of all the lung ECM proteins, collagen-I is the most abundant and provides tensile strength. In many fibrotic lung diseases, the expression of collagen is increased which affects the stiffness of the surrounding environment. The goal of this study was to assess the effect on fibroblast morphology, cell death, and inflammation when exposed to 2D and 3D low (0.4 mg/mL) versus high (2.0 mg/mL) collagen-I-matrix environments that model the mechanics of the breathing lung. This study demonstrates that human fetal lung fibroblasts (HFL1), grown in a 3D collagen type-I environment compared to a 2D one, do not form cells with a myofibroblast morphology, express less F-actin stress fibers, exhibit less cell death, and significantly produce less pro-inflammatory IL-6 and IL-8 cytokines. Exposure to mechanical strain to mimic breathing (0.2 Hz) led to the loss of HFL1 fibroblast dendritic extensions as well as F-actin stress fibers within the cell cytoskeleton, but did not influence cytokine production or cell death. This dynamic assay gives researchers the ability to consider the assessment of the mechanodynamic nature of the lung ECM environment in disease-relevant models and the potential of mechano-pharmacology to identify therapeutic targets for treatment.
Collapse
Affiliation(s)
- Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Noamie R. T. F. Vriesde
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
| | - May AL-Fouadi
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Leila Mostaco-Guidolin
- Department of Systems and Computer Engineering, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Delaram Maftoun
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Aileen Hsieh
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Nicole Coxson
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kauna Usman
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Don D. Sin
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Steve Booth
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
| | - Emmanuel T. Osei
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V5Z 1M9, Canada; (T.-L.H.); (N.R.T.F.V.); (M.A.-F.); (D.M.); (A.H.); (N.C.); (K.U.); (D.D.S.); (S.B.)
- Department of Biology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Correspondence:
| |
Collapse
|
36
|
Kim KH, Hur J, Lee HY, Lee EG, Lee SY. Cyclo-VEGI inhibits bronchial artery remodeling in a murine model of chronic asthma. Exp Lung Res 2021; 47:494-506. [PMID: 34890282 DOI: 10.1080/01902148.2021.2015011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Purpose/Aim: In the context of asthma, airway bronchial remodeling and angiogenesis in the bronchial mucosa are well established. Cyclopeptidic-vascular endothelial growth inhibitor (cyclo-VEGI) is an inhibitor of the vascular endothelial growth factor (VEGF) receptor that increases the proliferation of endothelial cells and the formation of new vessels. However, changes in the bronchial arteries of patients with asthma have not been clearly elucidated. We investigated whether structural changes occurred in bronchial arteries, as well as the effects of cyclo-VEGI in a mouse model of chronic asthma (in vivo) and human fibroblasts (in vitro). Materials and Methods: A validated mouse model of allergic airway inflammation with ovalbumin (OVA) as the causative allergen was used for the study. Mice were treated with cyclo-VEGI or fluticasone during OVA challenge. In vitro experiments were conducted to determine whether fibroblasts proliferated following elastin exposure and the effects of cyclo-VEGI on them. Results: OVA sensitization and challenge led to greater perivascular smooth muscle area, more elastic fibers, and elevated expression of vascular cell adhesion molecule (VCAM)-1 antigen. These phenomena indicated changes to bronchial arteries. Cyclo-VEGI and fluticasone treatment both inhibited airway hyper-responsiveness and inflammation. Cyclo-VEGI-treated mice exhibited decreased perivascular smooth muscle area, elastin fibers, and VCAM-1 expression. Fluticasone-treated mice exhibited reductions in perivascular smooth muscle but not in perivascular elastin or VCAM-1 expression. In vitro, fibroblast proliferation was enhanced by elastin treatment, which was inhibited by cyclo-VEGI treatment. Eotaxin expression was elevated in elastin-treated fibroblasts and decreased with cyclo-VEGI treatment. Conclusions: Vascular remodeling occurred in our mouse model of chronic asthma. Cyclo-VEGI could reduce airway inflammation and hyper-responsiveness by inhibiting VCAM-1 expression and elastin deposition around the bronchial arteries.
Collapse
Affiliation(s)
- Kyung Hoon Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of South Korea
| | - Jung Hur
- Division of Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of South Korea
| | - Hwa Young Lee
- Division of Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of South Korea
| | - Eung Gu Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of South Korea
| | - Sook Young Lee
- Division of Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of South Korea
| |
Collapse
|
37
|
Xi G, Qiu L, Xu S, Guo W, Fu F, Kang D, Zheng L, He J, Zhang Q, Li L, Wang C, Chen J. Computer-assisted quantification of tumor-associated collagen signatures to improve the prognosis prediction of breast cancer. BMC Med 2021; 19:273. [PMID: 34789257 PMCID: PMC8600902 DOI: 10.1186/s12916-021-02146-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Collagen fibers play an important role in tumor initiation, progression, and invasion. Our previous research has already shown that large-scale tumor-associated collagen signatures (TACS) are powerful prognostic biomarkers independent of clinicopathological factors in invasive breast cancer. However, they are observed on a macroscale and are more suitable for identifying high-risk patients. It is necessary to investigate the effect of the corresponding microscopic features of TACS so as to more accurately and comprehensively predict the prognosis of breast cancer patients. METHODS In this retrospective and multicenter study, we included 942 invasive breast cancer patients in both a training cohort (n = 355) and an internal validation cohort (n = 334) from one clinical center and in an external validation cohort (n = 253) from a different clinical center. TACS corresponding microscopic features (TCMFs) were firstly extracted from multiphoton images for each patient, and then least absolute shrinkage and selection operator (LASSO) regression was applied to select the most robust features to build a TCMF-score. Finally, the Cox proportional hazard regression analysis was used to evaluate the association of TCMF-score with disease-free survival (DFS). RESULTS TCMF-score is significantly associated with DFS in univariate Cox proportional hazard regression analysis. After adjusting for clinical variables by multivariate Cox regression analysis, the TCMF-score remains an independent prognostic indicator. Remarkably, the TCMF model performs better than the clinical (CLI) model in the three cohorts and is particularly outstanding in the ER-positive and lower-risk subgroups. By contrast, the TACS model is more suitable for the ER-negative and higher-risk subgroups. When the TACS and TCMF are combined, they could complement each other and perform well in all patients. As expected, the full model (CLI+TCMF+TACS) achieves the best performance (AUC 0.905, [0.873-0.938]; 0.896, [0.860-0.931]; 0.882, [0.840-0.925] in the three cohorts). CONCLUSION These results demonstrate that the TCMF-score is an independent prognostic factor for breast cancer, and the increased prognostic performance (TCMF+TACS-score) may help us develop more appropriate treatment protocols.
Collapse
Affiliation(s)
- Gangqin Xi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Lida Qiu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.,College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, 350108, China
| | - Shuoyu Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenhui Guo
- Breast Surgery Ward, Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Fangmeng Fu
- Breast Surgery Ward, Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Jiajia He
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.
| | - Chuan Wang
- Breast Surgery Ward, Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.
| |
Collapse
|
38
|
Novak C, Ballinger MN, Ghadiali S. Mechanobiology of Pulmonary Diseases: A Review of Engineering Tools to Understand Lung Mechanotransduction. J Biomech Eng 2021; 143:110801. [PMID: 33973005 PMCID: PMC8299813 DOI: 10.1115/1.4051118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/01/2021] [Indexed: 12/17/2022]
Abstract
Cells within the lung micro-environment are continuously subjected to dynamic mechanical stimuli which are converted into biochemical signaling events in a process known as mechanotransduction. In pulmonary diseases, the abrogated mechanical conditions modify the homeostatic signaling which influences cellular phenotype and disease progression. The use of in vitro models has significantly expanded our understanding of lung mechanotransduction mechanisms. However, our ability to match complex facets of the lung including three-dimensionality, multicellular interactions, and multiple simultaneous forces is limited and it has proven difficult to replicate and control these factors in vitro. The goal of this review is to (a) outline the anatomy of the pulmonary system and the mechanical stimuli that reside therein, (b) describe how disease impacts the mechanical micro-environment of the lung, and (c) summarize how existing in vitro models have contributed to our current understanding of pulmonary mechanotransduction. We also highlight critical needs in the pulmonary mechanotransduction field with an emphasis on next-generation devices that can simulate the complex mechanical and cellular environment of the lung. This review provides a comprehensive basis for understanding the current state of knowledge in pulmonary mechanotransduction and identifying the areas for future research.
Collapse
Affiliation(s)
- Caymen Novak
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Megan N. Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210
| | - Samir Ghadiali
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210; Department of Biomedical Engineering, The Ohio State University, 2124N Fontana Labs, 140 West 19th Avenue, Columbus, OH 43210
| |
Collapse
|
39
|
Chen D, Chen H, Chi L, Fu M, Wang G, Wu Z, Xu S, Sun C, Xu X, Lin L, Cheng J, Jiang W, Dong X, Lu J, Zheng J, Chen G, Li G, Zhuo S, Yan J. Association of Tumor-Associated Collagen Signature With Prognosis and Adjuvant Chemotherapy Benefits in Patients With Gastric Cancer. JAMA Netw Open 2021; 4:e2136388. [PMID: 34846524 PMCID: PMC8634059 DOI: 10.1001/jamanetworkopen.2021.36388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE The current TNM staging system provides limited information for prognosis prediction and adjuvant chemotherapy benefits for patients with gastric cancer (GC). OBJECTIVE To develop a tumor-associated collagen signature of GC (TACSGC) in the tumor microenvironment to predict prognosis and adjuvant chemotherapy benefits in patients with GC. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study included a training cohort of 294 consecutive patients treated between January 1, 2012, and December 31, 2013, from Nanfang Hospital, Southern Medical University, People's Republic of China, and a validation cohort of 225 consecutive patients treated between October 1, 2010, and December 31, 2012, from Fujian Provincial Cancer Hospital, Fujian Medical University, People's Republic of China. In total, 146 collagen features in the tumor microenvironment were extracted with multiphoton imaging. A TACSGC was then constructed using the least absolute shrinkage and selection operator Cox proportional hazards regression model in the training cohort. Data analysis was conducted from October 1, 2020, to April 30, 2021. MAIN OUTCOMES AND MEASURES The association of TACSGC with disease-free survival (DFS) and overall survival (OS) was assessed. An independent external cohort was included to validate the results. Interactions between TACSGC and adjuvant chemotherapy were calculated. RESULTS This study included 519 patients (median age, 57 years [IQR, 49-65 years]; 360 [69.4%] male). A 9 feature-based TACSGC was built. A higher TACSGC level was significantly associated with worse DFS and OS in both the training (DFS: hazard ratio [HR], 3.57 [95% CI, 2.45-5.20]; OS: HR, 3.54 [95% CI, 2.41-5.20]) and validation (DFS: HR, 3.10 [95% CI, 2.26-4.27]; OS: HR, 3.24 [95% CI, 2.33-4.50]) cohorts (continuous variable, P < .001 for all comparisons). Multivariable analyses found that carbohydrate antigen 19-9, depth of invasion, lymph node metastasis, distant metastasis, and TACSGC were independent prognostic predictors of GC, and 2 integrated nomograms that included the 5 predictors were established for predicting DFS and OS. Compared with clinicopathological models that included only the 4 clinicopathological predictors, the integrated nomograms yielded an improved discrimination for prognosis prediction in a C index comparison (training cohort: DFS, 0.80 [95% CI, 0.73-0.88] vs 0.78 [95% CI, 0.71-0.85], P = .03; OS, 0.81 [95% CI, 0.75-0.88] vs 0.80 [95% CI, 0.73-0.86], P = .03; validation cohort: DFS, 0.78 [95% CI, 0.70-0.87] vs 0.76 [95% CI, 0.67-0.84], P = .006; OS, 0.78 [95% CI, 0.69-0.86] vs 0.75 [95% CI, 0.67-0.84], P = .002). Patients with stage II and III GC and low TACSGC levels rather than high TACSGC levels had a favorable response to adjuvant chemotherapy (DFS: HR, 0.65 [95% CI, 0.43-0.96]; P = .03; OS: HR, 0.55 [95% CI, 0.36-0.82]; P = .004; dichotomized variable, P < .001 for interaction for both comparisons). CONCLUSIONS AND RELEVANCE The findings suggest that TACSGC provides additional prognostic information for patients with GC and may distinguish patients with stage II and III disease who are more likely to derive benefits from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- School of Science, Jimei University, Xiamen, People’s Republic of China
| | - Hao Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Liangjie Chi
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
| | - Meiting Fu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Guangxing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, People’s Republic of China
| | - Zhida Wu
- Department of Pathology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Shuoyu Xu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Caihong Sun
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, People’s Republic of China
| | - Xueqin Xu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, People’s Republic of China
| | - Liyan Lin
- Department of Pathology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Wei Jiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaoyu Dong
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jianping Lu
- Department of Pathology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Jixiang Zheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Medical University Cancer Hospital and Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, People’s Republic of China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
40
|
Modeling Extracellular Matrix-Cell Interactions in Lung Repair and Chronic Disease. Cells 2021; 10:cells10082145. [PMID: 34440917 PMCID: PMC8394761 DOI: 10.3390/cells10082145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/18/2021] [Indexed: 01/11/2023] Open
|
41
|
The Role of miRNAs in Extracellular Matrix Repair and Chronic Fibrotic Lung Diseases. Cells 2021; 10:cells10071706. [PMID: 34359876 PMCID: PMC8304879 DOI: 10.3390/cells10071706] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
The lung extracellular matrix (ECM) plays a key role in the normal architecture of the lung, from embryonic lung development to mechanical stability and elastic recoil of the breathing adult lung. The lung ECM can modulate the biophysical environment of cells through ECM stiffness, porosity, topography and insolubility. In a reciprocal interaction, lung ECM dynamics result from the synthesis, degradation and organization of ECM components by the surrounding structural and immune cells. Repeated lung injury and repair can trigger a vicious cycle of aberrant ECM protein deposition, accompanied by elevated ECM stiffness, which has a lasting effect on cell and tissue function. The processes governing the resolution of injury repair are regulated by several pathways; however, in chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary disease (IPF) these processes are compromised, resulting in impaired cell function and ECM remodeling. Current estimates show that more than 60% of the human coding transcripts are regulated by miRNAs. miRNAs are small non-coding RNAs that regulate gene expressions and modulate cellular functions. This review is focused on the current knowledge of miRNAs in regulating ECM synthesis, degradation and topography by cells and their dysregulation in asthma, COPD and IPF.
Collapse
|
42
|
Faiz A, Harkness LM, Tjin G, Bernal V, Horvatovich P, James A, Elliot JG, Burgess JK, Ashton AW. Angiogenic regulatory influence of extracellular matrix deposited by resting state asthmatic and non-asthmatic airway smooth muscle cells is similar. J Cell Mol Med 2021; 25:6438-6447. [PMID: 34146379 PMCID: PMC8256353 DOI: 10.1111/jcmm.16648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/26/2022] Open
Abstract
The extracellular matrix (ECM) is the tissue microenvironment that regulates the characteristics of stromal and systemic cells to control processes such as inflammation and angiogenesis. Despite ongoing anti-inflammatory treatment, low levels of inflammation exist in the airways in asthma, which alters ECM deposition by airway smooth muscle (ASM) cells. The altered ECM causes aberrant behaviour of cells, such as endothelial cells, in the airway tissue. We therefore sought to characterize the composition and angiogenic potential of the ECM deposited by asthmatic and non-asthmatic ASM. After 72 hours under non-stimulated conditions, the ECM deposited by primary human asthmatic ASM cells was equal in total protein, collagen I, III and fibronectin content to that from non-asthmatic ASM cells. Further, the matrices of non-asthmatic and asthmatic ASM cells were equivalent in regulating the growth, activity, attachment and migration of primary human umbilical vein endothelial cells (HUVECs). Under basal conditions, asthmatic and non-asthmatic ASM cells intrinsically deposit an ECM of equivalent composition and angiogenic potential. Previous findings indicate that dysregulation of the airway ECM is driven even by low levels of inflammatory provocation. This study suggests the need for more effective anti-inflammatory therapies in asthma to maintain the airway ECM and regulate ECM-mediated aberrant angiogenesis.
Collapse
Affiliation(s)
- Alen Faiz
- Respiratory Cellular and Molecular BiologyWoolcock Institute of Medical Research. SydneyNSWAustralia
- Emphysema CenterWoolcock Institute of Medical ResearchThe University of SydneyGlebeNSWAustralia
- Respiratory Bioinformatics and Molecular BiologyFaculty of ScienceUniversity of Technology SydneyUltimoNSWAustralia
- Department of Pathology and Medical BiologyGroningen Research Institute for Asthma and COPDUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of PulmonologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Discipline of PharmacologySchool of Medical SciencesThe University of SydneySydneyNSWAustralia
- Central Clinical SchoolThe University of SydneySydneyNSWAustralia
| | - Louise M. Harkness
- Respiratory Cellular and Molecular BiologyWoolcock Institute of Medical Research. SydneyNSWAustralia
- Department of PulmonologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Discipline of PharmacologySchool of Medical SciencesThe University of SydneySydneyNSWAustralia
| | - Gavin Tjin
- Respiratory Cellular and Molecular BiologyWoolcock Institute of Medical Research. SydneyNSWAustralia
- Discipline of PharmacologySchool of Medical SciencesThe University of SydneySydneyNSWAustralia
- Central Clinical SchoolThe University of SydneySydneyNSWAustralia
- Present address:
St Vincent’s Institute Medical ResearchFitzroyVic.Australia
| | - Victor Bernal
- Bernoulli Institute (BI)University of GroningenGroningenThe Netherlands
- Department of PharmacyAnalytical BiochemistryUniversity of GroningenGroningenThe Netherlands
| | - Peter Horvatovich
- Department of PharmacyAnalytical BiochemistryUniversity of GroningenGroningenThe Netherlands
| | - Alan James
- Department of Pulmonary Physiology and Sleep MedicineWest Australian Sleep Disorders Research InstituteSir Charles Gairdner HospitalPerthWAAustralia
- School of Medicine and PharmacologyUniversity of Western AustraliaPerthWAAustralia
| | - John G. Elliot
- Department of Pulmonary Physiology and Sleep MedicineWest Australian Sleep Disorders Research InstituteSir Charles Gairdner HospitalPerthWAAustralia
| | - Janette K. Burgess
- Respiratory Cellular and Molecular BiologyWoolcock Institute of Medical Research. SydneyNSWAustralia
- Department of Pathology and Medical BiologyGroningen Research Institute for Asthma and COPDUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of PulmonologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Discipline of PharmacologySchool of Medical SciencesThe University of SydneySydneyNSWAustralia
- Central Clinical SchoolThe University of SydneySydneyNSWAustralia
- Department of Pathology and Medical BiologyKOLFF InstituteUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Anthony W. Ashton
- Division of Perinatal ResearchKolling Institute of Medical ResearchSydneyNSWAustralia
| |
Collapse
|
43
|
Mostaço-Guidolin LB, Yang CX, Hackett TL. Pulmonary Vascular Remodeling Is an Early Feature of Fatal and Nonfatal Asthma. Am J Respir Cell Mol Biol 2021; 65:114-118. [PMID: 34241585 DOI: 10.1165/rcmb.2020-0339le] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Leila B Mostaço-Guidolin
- St. Paul's Hospital Vancouver, British Columbia, Canada.,University of British Columbia Vancouver, British Columbia, Canada, and.,Carleton University Ottawa, Ontario, Canada
| | - Chen Xi Yang
- St. Paul's Hospital Vancouver, British Columbia, Canada.,University of British Columbia Vancouver, British Columbia, Canada, and
| | - Tillie-Louise Hackett
- St. Paul's Hospital Vancouver, British Columbia, Canada.,University of British Columbia Vancouver, British Columbia, Canada, and
| |
Collapse
|
44
|
Bennet TJ, Randhawa A, Hua J, Cheung KC. Airway-On-A-Chip: Designs and Applications for Lung Repair and Disease. Cells 2021; 10:1602. [PMID: 34206722 PMCID: PMC8304815 DOI: 10.3390/cells10071602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022] Open
Abstract
The lungs are affected by illnesses including asthma, chronic obstructive pulmonary disease, and infections such as influenza and SARS-CoV-2. Physiologically relevant models for respiratory conditions will be essential for new drug development. The composition and structure of the lung extracellular matrix (ECM) plays a major role in the function of the lung tissue and cells. Lung-on-chip models have been developed to address some of the limitations of current two-dimensional in vitro models. In this review, we describe various ECM substitutes utilized for modeling the respiratory system. We explore the application of lung-on-chip models to the study of cigarette smoke and electronic cigarette vapor. We discuss the challenges and opportunities related to model characterization with an emphasis on in situ characterization methods, both established and emerging. We discuss how further advancements in the field, through the incorporation of interstitial cells and ECM, have the potential to provide an effective tool for interrogating lung biology and disease, especially the mechanisms that involve the interstitial elements.
Collapse
Affiliation(s)
- Tanya J. Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Avineet Randhawa
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Hua
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
45
|
Xing Q, You Y, Zhao X, Ji J, Yan H, Dong Y, Ren L, Ding Y, Hou S. iTRAQ-Based Proteomics Reveals Gu-Ben-Fang-Xiao Decoction Alleviates Airway Remodeling via Reducing Extracellular Matrix Deposition in a Murine Model of Chronic Remission Asthma. Front Pharmacol 2021; 12:588588. [PMID: 34194321 PMCID: PMC8237094 DOI: 10.3389/fphar.2021.588588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Airway remodeling is a primary pathological feature of asthma. The current therapy for asthma mainly targets reducing inflammation but not particularly airway remodeling. Therefore, it is worthwhile to develop alternative and more effective therapies to attenuate remodeling. Gu-Ben-Fang-Xiao Decoction (GBFXD) has been used to effectively and safely treat asthma for decades. In this study, GBFXD regulated airway inflammation, collagen deposition, and the molecules relevant to airway remodeling such as Vimentin, α-SMA, hydroxyproline, and E-cadherin in chronic remission asthma (CRA) murine model. Proteomic analysis indicated that the overlapping differentially expressed proteins (DEPs) (Model/Control and GBFXD/Model) were mainly collagens and laminins, which were extracellular matrix (ECM) proteins. In addition, the KEGG analysis showed that GBFXD could regulate pathways related to airway remodeling including ECM-receptor interactions, focal adhesion, and the PI3K/AKT signaling pathway, which were the top three significantly enriched pathways containing the most DEPs for both Model/Control and GBFXD/Model. Further validation research showed that GBFXD regulated reticulon-4 (RTN4) and suppressed the activation of the PI3K/AKT pathway to alleviate ECM proteins deposition. In conclusion, our findings indicate that GBFXD possibly regulate the PI3K/AKT pathway via RTN4 to improve airway remodeling, which provides a new insight into the molecular mechanism of GBFXD for the treatment of CRA.
Collapse
Affiliation(s)
- Qiongqiong Xing
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yannan You
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Xia Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Jianjian Ji
- Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Hua Yan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yingmei Dong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Lishun Ren
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yuanyuan Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Shuting Hou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| |
Collapse
|
46
|
Bazan-Socha S, Jakiela B, Zuk J, Zarychta J, Soja J, Okon K, Dziedzina S, Zareba L, Dropinski J, Wojcik K, Padjas A, Marcinkiewicz C, Bazan JG. Interactions via α 2β 1 Cell Integrin May Protect against the Progression of Airway Structural Changes in Asthma. Int J Mol Sci 2021; 22:ijms22126315. [PMID: 34204767 PMCID: PMC8231566 DOI: 10.3390/ijms22126315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/25/2022] Open
Abstract
Increased airway wall thickness and remodeling of bronchial mucosa are characteristic of asthma and may arise from altered integrin signaling on airway cells. Here, we analyzed the expression of β1-subfamily integrins on blood and airway cells (flow cytometry), inflammatory biomarkers in serum and bronchoalveolar lavage, reticular basement membrane (RBM) thickness and collagen deposits in the mucosa (histology), and airway geometry (CT-imaging) in 92 asthma patients (persistent airflow limitation subtype: n = 47) and 36 controls. Persistent airflow limitation was associated with type-2 inflammation, elevated soluble α2 integrin chain, and changes in the bronchial wall geometry. Both subtypes of asthma showed thicker RBM than control, but collagen deposition and epithelial α1 and α2 integrins staining were similar. Type-I collagen accumulation and RBM thickness were inversely related to the epithelial expression of the α2 integrin chain. Expression of α2β1 integrin on T-cells and eosinophils was not altered in asthma. Collagen I deposits were, however, more abundant in patients with lower α2β1 integrin on blood and airway CD8+ T-cells. Thicker airway walls in CT were associated with lower α2 integrin chain on blood CD4+ T-cells and airway eosinophils. Our data suggest that α2β1 integrin on inflammatory and epithelial cells may protect against airway remodeling advancement in asthma.
Collapse
Affiliation(s)
- Stanislawa Bazan-Socha
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
- Correspondence: ; Tel.: +48-12-4248023; Fax: +48-12-4248041
| | - Bogdan Jakiela
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Joanna Zuk
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Jacek Zarychta
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
- Pulmonary Hospital, 34-500 Zakopane, Poland
| | - Jerzy Soja
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Krzysztof Okon
- Faculty of Medicine, Department of Pathology, Jagiellonian University Medical College, 31-531 Krakow, Poland;
| | - Sylwia Dziedzina
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Lech Zareba
- College of Natural Sciences, Institute of Computer Science, University of Rzeszów, 35-310 Rzeszów, Poland; (L.Z.); (J.G.B.)
| | - Jerzy Dropinski
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Krzysztof Wojcik
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Agnieszka Padjas
- Faculty of Medicine, Department of Internal Medicine, Jagiellonian University Medical College, 31-066 Krakow, Poland; (B.J.); (J.Z.); (J.Z.); (J.S.); (S.D.); (J.D.); (K.W.); (A.P.)
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA;
| | - Jan G. Bazan
- College of Natural Sciences, Institute of Computer Science, University of Rzeszów, 35-310 Rzeszów, Poland; (L.Z.); (J.G.B.)
| |
Collapse
|
47
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
48
|
Jamieson RR, Stasiak SE, Polio SR, Augspurg RD, McCormick CA, Ruberti JW, Parameswaran H. Stiffening of the extracellular matrix is a sufficient condition for airway hyperreactivity. J Appl Physiol (1985) 2021; 130:1635-1645. [PMID: 33792403 DOI: 10.1152/japplphysiol.00554.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The current therapeutic approach to asthma focuses exclusively on targeting inflammation and reducing airway smooth muscle force to prevent the recurrence of symptoms. However, even when inflammation is brought under control, airways in an asthmatic can still hyperconstrict when exposed to a low dose of agonist. This suggests that there are mechanisms at play that are likely triggered by inflammation and eventually become self-sustaining so that even when airway inflammation is brought back under control, these alternative mechanisms continue to drive airway hyperreactivity in asthmatics. In this study, we hypothesized that stiffening of the airway extracellular matrix is a core pathological change sufficient to support excessive bronchoconstriction even in the absence of inflammation. To test this hypothesis, we increased the stiffness of the airway extracellular matrix by photo-crosslinking collagen fibers within the airway wall of freshly dissected bovine rings using riboflavin (vitamin B2) and Ultraviolet-A radiation. In our experiments, collagen crosslinking led to a twofold increase in the stiffness of the airway extracellular matrix. This change was sufficient to cause airways to constrict to a greater degree, and at a faster rate when they were exposed to 10-5 M acetylcholine for 5 min. Our results show that stiffening of the extracellular matrix is sufficient to drive excessive airway constriction even in the absence of inflammatory signals.NEW & NOTEWORTHY Targeting inflammation is the central dogma on which current asthma therapy is based. Here, we show that a healthy airway can be made to constrict excessively and at a faster rate in response to the same stimulus by increasing the stiffness of the extracellular matrix, without the use of inflammatory agents. Our results provide an independent mechanism by which airway remodeling in asthma can sustain airway hyperreactivity even in the absence of inflammatory signals.
Collapse
Affiliation(s)
- Ryan R Jamieson
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Suzanne E Stasiak
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Samuel R Polio
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Ralston D Augspurg
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | | | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | | |
Collapse
|
49
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
50
|
Interleukin-1α Is a Critical Mediator of the Response of Human Bronchial Fibroblasts to Eosinophilic Inflammation. Cells 2021; 10:cells10030528. [PMID: 33801398 PMCID: PMC7998867 DOI: 10.3390/cells10030528] [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: 02/11/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/08/2023] Open
Abstract
Eosinophils contribute to allergic inflammation in asthma in part via elaboration of a complex milieu of soluble mediators. Human bronchial fibroblasts (HBF) respond to stimulation by these mediators by acquiring a pro-inflammatory profile including induction of interleukin 6 (IL6) and IL8. This study sought to determine key component(s) of eosinophil soluble factors that mediate IL6 and IL8 induction in HBF. HBF treated with eosinophil-derived soluble mediators were analyzed for gene expression, intracellular signaling, and IL6 and IL8 secretion following inhibition of inflammatory signaling. Segmental allergen bronchoprovocation (SBP-Ag) was performed in mild asthmatics and bronchoalveolar lavage fluid was analyzed for eosinophils and cytokines. We found that signaling via the IL1α/IL1 receptor is an essential component of the response of HBF to eosinophil-derived soluble factors. IL1α-dependent activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling is required to induce IL6 secretion. However, NFκB signaling is dispensable for the induction of IL8, whereas Src is required. IL1α is associated with eosinophilic inflammation in human airways after SBP-Ag. Conclusions: IL1α appears to be a critical component of the soluble eosinophil-derived milieu that drives pro-inflammatory bronchial fibroblast responses and associates with eosinophilic inflammation following SBP-Ag. Disruption of IL1α-signaling could modify the downstream effects of eosinophilic inflammation on airway remodeling.
Collapse
|