1
|
Gong KQ, Brune JE, Guo X, Manicone AM. MAP2K1 dampens cigarette smoke-induced inflammation via suppression of type I interferon pathway activation. Am J Physiol Lung Cell Mol Physiol 2024; 327:L740-L748. [PMID: 39316676 DOI: 10.1152/ajplung.00080.2024] [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: 02/28/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), comprised of chronic bronchitis and emphysema, is a leading cause of morbidity and mortality worldwide. Mitogen-activated protein 2 kinase (MAP2K) pathway activation is present in COPD lung tissue and a genetic polymorphism in Map2k1 associates with FEV1 decline in COPD, suggesting it may contribute to disease pathogenesis. To test the functional contribution of Map2k1 in cigarette smoke (CS)-induced lung inflammation, we used a short-term CS exposure model in mice deficient in myeloid Map2k1 (LysmCre+Mek1fl) and wild-type mice (Mek1fl). Mice deficient in myeloid Map2k1 had enhanced CS-induced lung inflammation characterized by increased neutrophil recruitment, vascular leak, augmented expression of elastolytic matrix metalloproteinases, and increased type I interferon-stimulated gene expression. The augmented neutrophilic inflammatory response could be abrogated by IFNAR1 blockade. These findings indicate that myeloid Map2k1 regulates the immune response to CS via inhibition of the type I interferon pathway. Overall, these results suggest that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation and its expression is protective.NEW & NOTEWORTHY Activation of the mitogen-activated protein kinases (MAPK)-ERK1/2 pathway is present in COPD lung tissue compared with healthy lungs. Our study using mice deficient in myeloid Map2k1 reveals that Map2k1 is a critical determinant in modulating the severity of CS-induced lung inflammation via suppression of type I interferon responses, and its expression is protective.
Collapse
Affiliation(s)
- Ke-Qin Gong
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Jourdan E Brune
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
| | - Xiaoyun Guo
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
2
|
de Fays C, Geudens V, Gyselinck I, Kerckhof P, Vermaut A, Goos T, Vermant M, Beeckmans H, Kaes J, Van Slambrouck J, Mohamady Y, Willems L, Aversa L, Cortesi EE, Hooft C, Aerts G, Aelbrecht C, Everaerts S, McDonough JE, De Sadeleer LJ, Gohy S, Ambroise J, Janssens W, Ceulemans LJ, Van Raemdonck D, Vos R, Hackett TL, Hogg JC, Kaminski N, Gayan-Ramirez G, Pilette C, Vanaudenaerde BM. Mucosal immune alterations at the early onset of tissue destruction in chronic obstructive pulmonary disease. Front Immunol 2023; 14:1275845. [PMID: 37915582 PMCID: PMC10616299 DOI: 10.3389/fimmu.2023.1275845] [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: 08/10/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Rationale COPD is characterized by chronic airway inflammation, small airways changes, with disappearance and obstruction, and also distal/alveolar destruction (emphysema). The chronology by which these three features evolve with altered mucosal immunity remains elusive. This study assessed the mucosal immune defense in human control and end-stage COPD lungs, by detailed microCT and RNA transcriptomic analysis of diversely affected zones. Methods In 11 control (non-used donors) and 11 COPD (end-stage) explant frozen lungs, 4 cylinders/cores were processed per lung for microCT and tissue transcriptomics. MicroCT was used to quantify tissue percentage and alveolar surface density to classify the COPD cores in mild, moderate and severe alveolar destruction groups, as well as to quantify terminal bronchioles in each group. Transcriptomics of each core assessed fold changes in innate and adaptive cells and pathway enrichment score between control and COPD cores. Immunostainings of immune cells were performed for validation. Results In mildly affected zones, decreased defensins and increased mucus production were observed, along CD8+ T cell accumulation and activation of the IgA pathway. In more severely affected zones, CD68+ myeloid antigen-presenting cells, CD4+ T cells and B cells, as well as MHCII and IgA pathway genes were upregulated. In contrast, terminal bronchioles were decreased in all COPD cores. Conclusion Spatial investigation of end-stage COPD lungs show that mucosal defense dysregulation with decreased defensins and increased mucus and IgA responses, start concomitantly with CD8+ T-cell accumulation in mild emphysema zones, where terminal bronchioles are already decreased. In contrast, adaptive Th and B cell activation is observed in areas with more advanced tissue destruction. This study suggests that in COPD innate immune alterations occur early in the tissue destruction process, which affects both the alveoli and the terminal bronchioles, before the onset of an adaptive immune response.
Collapse
Affiliation(s)
- Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Iwein Gyselinck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Astrid Vermaut
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Tinne Goos
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Yousry Mohamady
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Lucia Aversa
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Emanuela E. Cortesi
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Charlotte Hooft
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Gitte Aerts
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Celine Aelbrecht
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Stephanie Everaerts
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - John E. McDonough
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Laurens J. De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jerome Ambroise
- Centre de Technologies Moléculaires Appliquées, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Wim Janssens
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Tillie L. Hackett
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - James C. Hogg
- Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KULeuven, Leuven, Belgium
| |
Collapse
|
3
|
Ranawat P, Kaur N, Koul A. Modulation of cigarette smoke induced alterations by aqueous Ocimum sanctum leaf extract in pulmonary tissue of rodents. Sci Rep 2023; 13:15806. [PMID: 37737272 PMCID: PMC10517011 DOI: 10.1038/s41598-022-26152-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 12/09/2022] [Indexed: 09/23/2023] Open
Abstract
Smoking has been associated with an increased risk of asthma, lung cancer, cardiovascular diseases, chronic bronchitis, and a massive amount of oxidative stress. The present study was undertaken to determine the modulatory effects of Holi Basil/Tulsi, (Ocimum sanctum) leaf extract on cigarette smoke-induced pulmonary damage in mice. Cigarette smoke (CS) inhalation increased the levels of pulmonary lipid peroxidation, and reactive oxygen species and decreased the levels of glutathione. Histoarchitectural alterations and enhanced tissue lactate dehydrogenase (LDH) activity in pulmonary tissue was distinctly indicative of damage. Enhanced mucin production was also observed through mucicarmine and Alcian Blue-Periodic Acid Schiff (PAS) staining. Increased expression of MUC5AC was also observed. Alterations in the lung were also evident through FTIR studies. Administration of Ocimum sanctum leaf extract (80 mg/kg b.w) to CS exposed mice ameliorated these alterations to a greater extent. These findings are suggestive of the fact that Ocimum sanctum leaf extract effectively modulated CS-induced deleterious effects on pulmonary tissue.
Collapse
Affiliation(s)
- Pavitra Ranawat
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Navdeep Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Ashwani Koul
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
4
|
Kim JH, Schaible N, Hall JK, Bartolák-Suki E, Deng Y, Herrmann J, Sonnenberg A, Behrsing HP, Lutchen KR, Krishnan R, Suki B. Multiscale stiffness of human emphysematous precision cut lung slices. SCIENCE ADVANCES 2023; 9:eadf2535. [PMID: 37205750 PMCID: PMC10198632 DOI: 10.1126/sciadv.adf2535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Emphysema is a debilitating disease that remodels the lung leading to reduced tissue stiffness. Thus, understanding emphysema progression requires assessing lung stiffness at both the tissue and alveolar scales. Here, we introduce an approach to determine multiscale tissue stiffness and apply it to precision-cut lung slices (PCLS). First, we established a framework for measuring stiffness of thin, disk-like samples. We then designed a device to verify this concept and validated its measuring capabilities using known samples. Next, we compared healthy and emphysematous human PCLS and found that the latter was 50% softer. Through computational network modeling, we discovered that this reduced macroscopic tissue stiffness was due to both microscopic septal wall remodeling and structural deterioration. Lastly, through protein expression profiling, we identified a wide spectrum of enzymes that can drive septal wall remodeling, which, together with mechanical forces, lead to rupture and structural deterioration of the emphysematous lung parenchyma.
Collapse
Affiliation(s)
- Jae Hun Kim
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Mechanobiologix, LLC, Newton, MA, USA
| | - Niccole Schaible
- Mechanobiologix, LLC, Newton, MA, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Joseph K. Hall
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | | | - Yuqing Deng
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Jacob Herrmann
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- University of Iowa, Iowa City, IA, USA
| | - Adam Sonnenberg
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | | | - Kenneth R. Lutchen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ramaswamy Krishnan
- Mechanobiologix, LLC, Newton, MA, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Mechanobiologix, LLC, Newton, MA, USA
| |
Collapse
|
5
|
Lee KY, Ho SC, Sun WL, Feng PH, Lin CW, Chen KY, Chuang HC, Tseng CH, Chen TT, Wu SM. Lnc-IL7R alleviates PM 2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol Toxicol 2022; 38:1097-1120. [PMID: 35303175 DOI: 10.1007/s10565-022-09709-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Long-term exposure to PM2.5 (particulate matter with an aerodynamic diameter of ≤ 2.5 μm) is associated with pulmonary injury and emphysema in patients with chronic obstructive pulmonary disease (COPD). We investigated mechanisms through which the long noncoding RNA lnc-IL7R contributes to cellular damage by inducing oxidative stress in COPD patients exposed to PM2.5. METHODS Associations of serum lnc-IL7R levels with lung function, emphysema, and previous PM2.5 exposure in COPD patients were analyzed. Reactive oxygen species and lnc-IL7R levels were measured in PM2.5-treated cells. The levels of lnc-IL7R and cellular senescence-associated genes, namely p16INK4a and p21CIP1/WAF1, were determined through lung tissue section staining. The effects of p16INK4a or p21CIP1/WAF1 regulation were examined by performing lnc-IL7R overexpression and knockdown assays. The functions of lnc-IL7R-mediated cell proliferation, cell cycle, senescence, colony formation, and apoptosis were examined in cells treated with PM2.5. Chromatin immunoprecipitation assays were conducted to investigate the epigenetic regulation of p21CIP1/WAF1. RESULTS Lnc-IL7R levels decreased in COPD patients and were negatively correlated with emphysema or PM2.5 exposure. Lnc-IL7R levels were upregulated in normal lung epithelial cells but not in COPD cells exposed to PM2.5. Lower lnc-IL7R expression in PM2.5-treated cells induced p16INK4a and p21CIP1/WAF1 expression by increasing oxidative stress. Higher lnc-IL7R expression protected against cellular senescence and apoptosis, whereas lower lnc-IL7R expression augmented injury in PM2.5-treated cells. Lnc-IL7R and the enhancer of zeste homolog 2 (EZH2) synergistically suppressed p21CIP1/WAF1 expression through epigenetic modulation. CONCLUSION Lnc-IL7R attenuates PM2.5-mediated p21CIP1/WAF1 expression through EZH2 recruitment, and its dysfunction may augment cellular injury in COPD.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
6
|
Suki B, Bates JH, Bartolák-Suki E. Remodeling of the Aged and Emphysematous Lungs: Roles of Microenvironmental Cues. Compr Physiol 2022; 12:3559-3574. [PMID: 35766835 PMCID: PMC11470990 DOI: 10.1002/cphy.c210033] [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] [Indexed: 11/06/2022]
Abstract
Aging is a slow process that affects all organs, and the lung is no exception. At the alveolar level, aging increases the airspace size with thicker and stiffer septal walls and straighter and thickened collagen and elastic fibers. This creates a microenvironment that interferes with the ability of cells in the parenchyma to maintain normal homeostasis and respond to injury. These changes also make the lung more susceptible to disease such as emphysema. Emphysema is characterized by slow but progressive remodeling of the deep alveolar regions that leads to airspace enlargement and increased but disorganized elastin and collagen deposition. This remodeling has been attributed to ongoing inflammation that involves inflammatory cells and the cytokines they produce. Cellular senescence, another consequence of aging, weakens the ability of cells to properly respond to injury, something that also occurs in emphysema. These factors conspire to make alveolar walls more prone to mechanical failure, which can set emphysema in motion by driving inflammation through immune stimulation by protein fragments. Both aging and emphysema are influenced by microenvironmental conditions such as local inflammation, chemical makeup, tissue stiffness, and mechanical stresses. Although aging and emphysema are not equivalent, they have the potential to influence each other in synergistic ways; aging sets up the conditions for emphysema to develop, while emphysema may accelerate cellular senescence and thus aging itself. This article focuses on the similarities and differences between the remodeled microenvironment of the aging and emphysematous lung, with special emphasis on the alveolar septal wall. © 2022 American Physiological Society. Compr Physiol 12:3559-3574, 2022.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jason H.T. Bates
- Depatment of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | |
Collapse
|
7
|
Alsemeh AE, Abdullah DM. Protective effect of alogliptin against cyclophosphamide-induced lung toxicity in rats: Impact on PI3K/Akt/FoxO1 pathway and downstream inflammatory cascades. Cell Tissue Res 2022; 388:417-438. [PMID: 35107620 PMCID: PMC9035424 DOI: 10.1007/s00441-022-03593-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
Cyclophosphamide (CP)-induced lung toxicity is a remaining obstacle against the beneficial use of this chemotherapeutic agent. More considerations were given to the role of Alogliptin (ALO) in ameliorating CP-induced toxicities in many tissues. We designed this study to clarify the protective potential of ALO against CP-induced lung toxicity in rats. ALO was administered for 7 days. Single-dose CP was injected on the 2nd day (200 mg/kg: i.p.) to induce lung toxicity. Rats were divided into four groups: control, ALO-treated, CP-treated and ALO + CP-treated group. Leucocytic count, total proteins, LDH activity, TNF-α, and IL-6 were estimated in the bronchoalveolar lavage fluid (BALF). The oxidative/antioxidants (MDA, Nrf2, TAO and GSH), inflammatory (NFκB), fibrotic (TGF-β1) and apoptotic (PI3K/Akt/FoxO1) markers in pulmonary homogenates were biochemically evaluated. Rat lung sections were examined histologically (light and electron microscopic examination) and immunohistochemically (for iNOS and CD68 positive alveolar macrophages). CP significantly increased oxidative stress, inflammation, fibrosis, and apoptosis markers as well as deteriorated the histopathological pulmonary architecture. These hazardous effects were significantly ameliorated by ALO treatment. ALO protected against CP-induced lung toxicity by mitigating the oxidative, inflammatory and fibrotic impacts making it a promising pharmacological therapy for mitigating CP-induced lung toxicity.
Collapse
Affiliation(s)
- Amira Ebrahim Alsemeh
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Doaa M Abdullah
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
8
|
Yuan Z, Herrmann J, Murthy S, Peters K, Gerard SE, Nia HT, Lutchen KR, Suki B. A Personalized Spring Network Representation of Emphysematous Lungs From CT Images. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:828157. [PMID: 36926064 PMCID: PMC10013051 DOI: 10.3389/fnetp.2022.828157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022]
Abstract
Emphysema is a progressive disease characterized by irreversible tissue destruction and airspace enlargement, which manifest as low attenuation area (LAA) on CT images. Previous studies have shown that inflammation, protease imbalance, extracellular matrix remodeling and mechanical forces collectively influence the progression of emphysema. Elastic spring network models incorporating force-based mechanical failure have been applied to investigate the pathogenesis and progression of emphysema. However, these models were general without considering the patient-specific information on lung structure available in CT images. The aim of this work was to develop a novel approach that provides an optimal spring network representation of emphysematous lungs based on the apparent density in CT images, allowing the construction of personalized networks. The proposed method takes into account the size and curvature of LAA clusters on the CT images that correspond to a pre-stressed condition of the lung as opposed to a naïve method that excludes the effects of pre-stress. The main findings of this study are that networks constructed by the new method 1) better preserve LAA cluster sizes and their distribution than the naïve method; and 2) predict different course of emphysema progression compared to the naïve method. We conclude that our new method has the potential to predict patient-specific emphysema progression which needs verification using clinical data.
Collapse
Affiliation(s)
- Ziwen Yuan
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Jacob Herrmann
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Samhita Murthy
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Kevin Peters
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Sarah E. Gerard
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Kenneth R. Lutchen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
9
|
Nam JG, Witanto JN, Park SJ, Yoo SJ, Goo JM, Yoon SH. Automatic pulmonary vessel segmentation on noncontrast chest CT: deep learning algorithm developed using spatiotemporally matched virtual noncontrast images and low-keV contrast-enhanced vessel maps. Eur Radiol 2021; 31:9012-9021. [PMID: 34009411 PMCID: PMC8131193 DOI: 10.1007/s00330-021-08036-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/03/2021] [Accepted: 05/03/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To develop a deep learning-based pulmonary vessel segmentation algorithm (DLVS) from noncontrast chest CT and to investigate its clinical implications in assessing vascular remodeling of chronic obstructive lung disease (COPD) patients. METHODS For development, 104 pulmonary CT angiography scans (49,054 slices) using a dual-source CT were collected, and spatiotemporally matched virtual noncontrast and 50-keV images were generated. Vessel maps were extracted from the 50-keV images. The 3-dimensional U-Net-based DLVS was trained to segment pulmonary vessels (with a vessel map as the output) from virtual noncontrast images (as the input). For external validation, vendor-independent noncontrast CT images (n = 14) and the VESSEL 12 challenge open dataset (n = 3) were used. For each case, 200 points were selected including 20 intra-lesional points, and the probability value for each point was extracted. For clinical validation, we included 281 COPD patients with low-dose noncontrast CTs. The DLVS-calculated volume of vessels with a cross-sectional area < 5 mm2 (PVV5) and the PVV5 divided by total vessel volume (%PVV5) were measured. RESULTS DLVS correctly segmented 99.1% of the intravascular points (1,387/1,400) and 93.1% of the extravascular points (1,309/1,400). The areas-under-the receiver-operating characteristic curve (AUROCs) were 0.977 and 0.969 for the two external validation datasets. For the COPD patients, both PPV5 and %PPV5 successfully differentiated severe patients whose FEV1 < 50 (AUROCs; 0.715 and 0.804) and were significantly correlated with the emphysema index (Ps < .05). CONCLUSIONS DLVS successfully segmented pulmonary vessels on noncontrast chest CT by utilizing spatiotemporally matched 50-keV images from a dual-source CT scanner and showed promising clinical applicability in COPD. KEY POINTS • We developed a deep learning pulmonary vessel segmentation algorithm using virtual noncontrast images and 50-keV enhanced images produced by a dual-source CT scanner. • Our algorithm successfully segmented vessels on diseased lungs. • Our algorithm showed promising results in assessing the loss of small vessel density in COPD patients.
Collapse
Affiliation(s)
- Ju Gang Nam
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | | | - Sang Joon Park
- Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- MedicalIp Co., Ltd., Seoul, 03127, Republic of Korea
| | - Seung Jin Yoo
- Department of Radiology, Hanyang University Medical Center and College of Medicine, Seoul, 04763, Republic of Korea
| | - Jin Mo Goo
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soon Ho Yoon
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
10
|
Karsdal MA, Genovese F, Rasmussen DGK, Bay-Jensen AC, Mortensen JH, Holm Nielsen S, Willumsen N, Jensen C, Manon-Jensen T, Jennings L, Reese-Petersen AL, Henriksen K, Sand JM, Bager C, Leeming DJ. Considerations for understanding protein measurements: Identification of formation, degradation and more pathological relevant epitopes. Clin Biochem 2021; 97:11-24. [PMID: 34453894 DOI: 10.1016/j.clinbiochem.2021.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES There is a need for precision medicine and an unspoken promise of an optimal approach for identification of the right patients for value-based medicine based on big data. However, there may be a misconception that measurement of proteins is more valuable than measurement of fewer selected biomarkers. In population-based research, variation may be somewhat eliminated by quantity. However, this fascination of numbers may limit the attention to and understanding of the single. This review highlights that protein measurements (with collagens as examples) may mean different things depending on the targeted epitope - formation or degradation of tissues, and even signaling potential of proteins. DESIGN AND METHODS PubMed was searched for collagen, neo-epitope, biomarkers. RESULTS Ample examples of assays with specific epitopes, either pathological such as HbA1c, or domain specific such as pro-peptides, which total protein arrays would not have identified were evident. CONCLUSIONS We suggest that big data may be considered as the funnel of data points, in which most important parameters will be selected. If the technical precision is low or the biological accuracy is limited, and we include suboptimal quality of biomarkers, disguised as big data, we may not be able to fulfill the promise of helping patients searching for the optimal treatment. Alternatively, if the technical precision of the total protein quantification is high, but we miss the functional domains with the most considerable biological meaning, we miss the most important and valuable information of a given protein. This review highlights that measurements of the same protein in different ways may provide completely different meanings. We need to understand the pathological importance of each epitope quantified to maximize protein measurements.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark.
| | - F Genovese
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D G K Rasmussen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - A C Bay-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J H Mortensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - S Holm Nielsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - N Willumsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - T Manon-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | | | | | - K Henriksen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J M Sand
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Bager
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D J Leeming
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Acute or chronic pulmonary emphysema? Or both?-A contribution to the diagnosis of death due to violent asphyxiation in cases with pre-existing chronic emphysema. Int J Legal Med 2021; 136:133-147. [PMID: 34181078 PMCID: PMC8813827 DOI: 10.1007/s00414-021-02619-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/30/2021] [Indexed: 11/02/2022]
Abstract
The diagnosis of death due to violent asphyxiation may be challenging if external injuries are missing, and a typical acute emphysema (AE) "disappears" in pre-existing chronic emphysema (CE). Eighty-four autopsy cases were systematically investigated to identify a (histo-) morphological or immunohistochemical marker combination that enables the diagnosis of violent asphyxiation in cases with a pre-existing CE ("AE in CE"). The cases comprised four diagnostic groups, namely "AE", "CE", "acute and chronic emphysema (AE + CE)", and "no emphysema (NE)". Samples from all pulmonary lobes were investigated by conventional histological methods as well as with the immunohistochemical markers Aquaporin 5 (AQP-5) and Surfactant protein A1 (SP-A). Particular attention was paid to alveolar septum ends ("dead-ends") suspected as rupture spots, which were additionally analyzed by transmission electron microscopy. The findings in the four diagnostic groups were compared using multivariate analysis and 1-way ANOVA analysis. All morphological findings were found in all four groups. Based on histological and macroscopic findings, a multivariate analysis was able to predict the correct diagnosis "AE + CE" with a probability of 50%, and the diagnoses "AE" and "CE" with a probability of 86% each. Three types of "dead-ends" could be differentiated. One type ("fringed ends") was observed significantly more frequently in AE. The immunohistochemical markers AQP-5 and SP-A did not show significant differences among the examined groups. Though a reliable identification of AE in CE could not be achieved using the examined parameters, our findings suggest that considering many different findings from the macroscopical, histomorphological, and molecular level by multivariate analysis is an approach that should be followed.
Collapse
|
13
|
García-Valero J, Olloquequi J, Rodríguez E, Martín-Satué M, Texidó L, Ferrer J. Decreased Expression of EC-SOD and Fibulin-5 in Alveolar Walls of Lungs From COPD Patients. Arch Bronconeumol 2021; 58:S0300-2896(21)00016-8. [PMID: 33640211 DOI: 10.1016/j.arbres.2020.12.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The aim of this study is to analyze the expression of the main oxidant scavenger superoxide dismutase (EC-SOD), its main binding protein Fibulin-5 and several oxidative and nitrosative-derived products in the lung of COPD patients and controls. MATERIALS AND METHODS Lung tissue samples from 19 COPD patients and 20 control subjects were analyzed. The architecture of elastic fibres was assessed by light and electron microscope histochemical techniques, and levels of EC-SOD and fibulin-5 were analyzed by immunohistochemistry and RT-PCR. The impact of oxidative stress on the extracellular matrix was estimated by immunolocalization of 4-hydroxynonenal (4-HNE), malondialdehyde (MDA) and 3-nitrotyrosine (3-NYT) adducts. RESULTS Alveolar walls of COPD patients exhibited abnormal accumulations of collapsing elastic fibres, showing a pierced pattern in the amorphous component. The semiquantitative analysis revealed that COPD patients have a significantly reduced expression of both EC-SOD and fibulin-5 (0.59±0.64 and 0.62±0.61, respectively) in alveolar, bronchiolar and arteriolar walls compared to control subjects (1.39±0.63 and 1.55±0.52, respectively, p<0.05). No significant changes in mRNA levels of these proteins were observed between groups. Among the oxidation markers, malondialdehyde was the best in distinguishing COPD patients. CONCLUSIONS COPD patients show a reduced expression of EC-SOD and fibulin-5 in the lung interstitium. Paralleling the reduction of EC-SOD levels, the decrease of fibulin-5 expression in COPD lungs supports the hypothesis of an impaired pulmonary antioxidant response in COPD patients.
Collapse
Affiliation(s)
- José García-Valero
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Esther Rodríguez
- Department of Pneumology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona and CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Mireia Martín-Satué
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain; Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Laura Texidó
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Jaume Ferrer
- Department of Pneumology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona and CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain.
| |
Collapse
|
14
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
15
|
Wickramasinghe LC, van Wijngaarden P, Johnson C, Tsantikos E, Hibbs ML. An Experimental Model of Bronchopulmonary Dysplasia Features Long-Term Retinal and Pulmonary Defects but Not Sustained Lung Inflammation. Front Pediatr 2021; 9:689699. [PMID: 34527643 PMCID: PMC8435611 DOI: 10.3389/fped.2021.689699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/06/2021] [Indexed: 11/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe lung disease that affects preterm infants receiving oxygen therapy. No standardized, clinically-relevant BPD model exists, hampering efforts to understand and treat this disease. This study aimed to evaluate and confirm a candidate model of acute and chronic BPD, based on exposure of neonatal mice to a high oxygen environment during key lung developmental stages affected in preterm infants with BPD. Neonatal C57BL/6 mouse pups were exposed to 75% oxygen from postnatal day (PN)-1 for 5, 8, or 14 days, and their lungs were examined at PN14 and PN40. While all mice showed some degree of lung damage, mice exposed to hyperoxia for 8 or 14 days exhibited the greatest septal wall thickening and airspace enlargement. Furthermore, when assessed at PN40, mice exposed for 8 or 14 days to supplemental oxygen exhibited augmented septal wall thickness and emphysema, with the severity increased with the longer exposure, which translated into a decline in respiratory function at PN80 in the 14-day model. In addition to this, mice exposed to hyperoxia for 8 days showed significant expansion of alveolar epithelial type II cells as well as the greatest fibrosis when assessed at PN40 suggesting a healing response, which was not seen in mice exposed to high oxygen for a longer period. While evidence of lung inflammation was apparent at PN14, chronic inflammation was absent from all three models. Finally, exposure to high oxygen for 14 days also induced concurrent outer retinal degeneration. This study shows that early postnatal exposure to high oxygen generates hallmark acute and chronic pathologies in mice that highlights its use as a translational model of BPD.
Collapse
Affiliation(s)
- Lakshanie C Wickramasinghe
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Peter van Wijngaarden
- Department of Surgery - Ophthalmology, University of Melbourne, Melbourne, VIC, Australia.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Chad Johnson
- Monash Micro Imaging, Alfred Research Alliance, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Fukuzaki S, Righetti RF, Santos TMD, Camargo LDN, Aristóteles LRCRB, Souza FCR, Garrido AC, Saraiva-Romanholo BM, Leick EA, Prado CM, Martins MDA, Tibério IDFLC. Preventive and therapeutic effect of anti-IL-17 in an experimental model of elastase-induced lung injury in C57Bl6 mice. Am J Physiol Cell Physiol 2020; 320:C341-C354. [PMID: 33326311 DOI: 10.1152/ajpcell.00017.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an important health care issue, and IL-17 can modulate inflammatory responses. We evaluated preventive and therapeutic effect of anti-interleukin (IL)-17 in a model of lung injury induced by elastase, using 32 male C57Bl6 mice, divided into 4 groups: SAL, ELASTASE CONTROL (EC), ELASTASE + PREVENTIVE ANTI-IL-17 (EP), and ELASTASE + THERAPEUTIC ANTI-IL-17 (ET). On the 29th day, animals were anesthetized with thiopental, tracheotomized, and placed on a ventilator to evaluate lung mechanical, exhaled nitric oxide (eNO), and total cells of bronchoalveolar lavage fluid was collected. We performed histological techniques, and linear mean intercept (Lm) was analyzed. Both treatments with anti-IL-17 decreased respiratory resistance and elastance, airway resistance, elastance of pulmonary parenchyma, eNO, and Lm compared with EC. There was reduction in total cells and macrophages in ET compared with EC. Both treatments decreased nuclear factor-кB, inducible nitric oxide synthase, matrix metalloproteinase (MMP)-9, MMP-12, transforming growth factor-β, tumor necrosis factor-α, neutrophils, IL-1β, isoprostane, and IL-17 in airways and alveolar septa; collagen fibers, decorin and lumican in airways; and elastic fibers and fibronectin in alveolar septa compared with EC. There was reduction of collagen fibers in alveolar septa and biglycan in airways in EP and a reduction of eNO synthase in airways in ET. In conclusion, both treatments with anti-IL-17 contributed to improve most of parameters evaluated in inflammation and extracellular matrix remodeling in this model of lung injury.
Collapse
Affiliation(s)
- Silvia Fukuzaki
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Renato Fraga Righetti
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | - Tabata Maruyama Dos Santos
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | - Leandro do Nascimento Camargo
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Flavia C R Souza
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Aurelio C Garrido
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Beatriz Mangueira Saraiva-Romanholo
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Department of Medicine (LIM 20), Hospital Public Employee of São Paulo (Instituto de Assistência Médica ao Servidor Público Estadual de São Paulo), University City of São Paulo, São Paulo, Brazil
| | - Edna Aparecida Leick
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Carla Máximo Prado
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Department of Bioscience, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | | |
Collapse
|
17
|
Karakioulaki M, Papakonstantinou E, Stolz D. Extracellular matrix remodelling in COPD. Eur Respir Rev 2020; 29:29/158/190124. [PMID: 33208482 DOI: 10.1183/16000617.0124-2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/16/2020] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the lung plays several important roles in lung function, as it offers a low resistant pathway that allows the exchange of gases, provides compressive strength and elasticity that supports the fragile alveolar-capillary intersection, controls the binding of cells with growth factors and cell surface receptors and acts as a buffer against retention of water.COPD is a chronic inflammatory respiratory condition, characterised by various conditions that result in progressive airflow limitation. At any stage in the course of the disease, acute exacerbations of COPD may occur and lead to accelerated deterioration of pulmonary function. A key factor of COPD is airway remodelling, which refers to the serious alterations of the ECM affecting airway wall thickness, resistance and elasticity. Various studies have shown that serum biomarkers of ECM turnover are significantly associated with disease severity in patients with COPD and may serve as potential targets to control airway inflammation and remodelling in COPD. Unravelling the complete molecular composition of the ECM in the diseased lungs will help to identify novel biomarkers for disease progression and therapy.
Collapse
Affiliation(s)
- Meropi Karakioulaki
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland.,Dept of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daiana Stolz
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| |
Collapse
|
18
|
Zhang Y, Li Y, Ye Z, Ma H. Expression of Matrix Metalloproteinase-2, Matrix Metalloproteinase-9, Tissue Inhibitor of Metalloproteinase-1, and Changes in Alveolar Septa in Patients with Chronic Obstructive Pulmonary Disease. Med Sci Monit 2020; 26:e925278. [PMID: 33070147 PMCID: PMC7580176 DOI: 10.12659/msm.925278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background This study investigated the relationship between the pathological alteration of alveolar septa and (1) pulmonary function and (2) matrix metalloproteinase (MMP)-2, MMP-9, and tissue inhibitor matrix metalloproteinase 1 (TIMP-1) expression in chronic obstructive pulmonary disease (COPD). Material/Methods Sixty patients with pulmonary disease were divided into control (n=20) and COPD (n=40) groups. Postoperative lung tissue specimens were examined. Hematoxylin and eosin and elastin van Gieson staining detected pathological alterations of pulmonary alveolar septa. Septa thickness was measured. MMP-2, MMP-9, and TIMP-1 expression levels were detected by immunohistochemical staining. Correlations were determined by Pearson analysis. Results Forced expiratory volume in 1 s (FEV1), forced vital capacity, FEV1 percent predicted (FEV1%pre), and diffusion capacity of carbon monoxide percent predicted (DLCO%pre) in COPD patients were significantly lower than in those of the control group (P<0.05). MMP-2, MMP-9, and TIMP-1 expression levels were significantly higher in the COPD group than in control, especially the severe group (P<0.05). Septa thickness was negatively correlated with FEV1%pre (r=−0.335; P<0.05) and positively correlated with MMP-2 and TIMP-1 expression (P<0.05). Proportion of collagenous fiber was negatively correlated with FEV1%pre and DLCO%pre (P<0.01), and positively correlated with MMP-2, MMP-9, and TIMP-1 expression (P<0.01). Proportion of elastic fibers was negatively correlated with collagenous fiber. Conclusions The pathological alteration of alveolar septa was correlated with pulmonary function and expression levels of MMP-2, MMP-9, and TIMP-1, which can play vital roles in COPD progression.
Collapse
Affiliation(s)
- Yongxiang Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China (mainland)
| | - Yuechuan Li
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China (mainland)
| | - Zhen Ye
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China (mainland)
| | - Hui Ma
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China (mainland)
| |
Collapse
|
19
|
Leng L, Cao R, Ma J, Mou D, Zhu Y, Li W, Lv L, Gao D, Zhang S, Gong F, Zhao L, Qiu B, Xiang H, Hu Z, Feng Y, Dai Y, Zhao J, Wu Z, Li H, Zhong W. Pathological features of COVID-19-associated lung injury: a preliminary proteomics report based on clinical samples. Signal Transduct Target Ther 2020; 5:240. [PMID: 33060566 PMCID: PMC7557250 DOI: 10.1038/s41392-020-00355-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has emerged as a global health emergency due to its association with severe pneumonia and relative high mortality. However, the molecular characteristics and pathological features underlying COVID-19 pneumonia remain largely unknown. To characterize molecular mechanisms underlying COVID-19 pathogenesis in the lung tissue using a proteomic approach, fresh lung tissues were obtained from newly deceased patients with COVID-19 pneumonia. After virus inactivation, a quantitative proteomic approach combined with bioinformatics analysis was used to detect proteomic changes in the SARS-CoV-2-infected lung tissues. We identified significant differentially expressed proteins involved in a variety of fundamental biological processes including cellular metabolism, blood coagulation, immune response, angiogenesis, and cell microenvironment regulation. Several inflammatory factors were upregulated, which was possibly caused by the activation of NF-κB signaling. Extensive dysregulation of the lung proteome in response to SARS-CoV-2 infection was discovered. Our results systematically outlined the molecular pathological features in terms of the lung response to SARS-CoV-2 infection, and provided the scientific basis for the therapeutic target that is urgently needed to control the COVID-19 pandemic.
Collapse
Affiliation(s)
- Ling Leng
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, 102206, Beijing, China
| | - Danlei Mou
- Department of Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, 102206, Beijing, China
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Luye Lv
- Institute of NBC Defense, 102205, Beijing, China
| | - Dunqin Gao
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Shikun Zhang
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, 100850, Beijing, China
| | - Feng Gong
- Department of Stem Cell and Regenerative Medicine Laboratory, Institute of Health Service and Transfusion Medicine, 100850, Beijing, China
| | - Lei Zhao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Bintao Qiu
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Haiping Xiang
- Department of Radiology, Beijing YouAn Hospital, Capital Medical of University, 100069, Beijing, China
| | - Zhongjie Hu
- Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Yingmei Feng
- Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Yan Dai
- Department of Respiratory and Critical Care Medicine, Nanyang Central Hospital, 473000, Henan, China
| | - Jiang Zhao
- Department of Respiratory and Critical Care Medicine, Nanyang Central Hospital, 473000, Henan, China
| | - Zhihong Wu
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China.
| | - Hongjun Li
- Department of Radiology, Beijing YouAn Hospital, Capital Medical of University, 100069, Beijing, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China.
| |
Collapse
|
20
|
Wei A, Wang R, Wei K, Dai C, Huang Y, Xu P, Xu J, Tang H, Zhang Y, Fan Y. LncRNA and mRNA Expression Profiling in the Periurethral Vaginal Wall Tissues of Postmenopausal Women with Stress Urinary Incontinence. Reprod Sci 2020; 27:1490-1501. [PMID: 32046467 DOI: 10.1007/s43032-020-00144-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/20/2019] [Indexed: 10/25/2022]
Abstract
Stress urinary incontinence (SUI) is one of the major pelvic floor disorders affecting postmenopausal women. To investigate the lncRNA and mRNA expression profiling of SUI in postmenopausal women, we used a microarray analysis to examine the differentially expressed long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) in the periurethral vaginal wall of postmenopausal women with SUI. A total of 8840 lncRNAs and 7102 mRNAs were dysregulated in the two groups (absolute fold change ≥ 2 and P < 0.05). The expression levels of five randomly selected lncRNAs and mRNAs were validated by quantitative real-time PCR. A functional analysis revealed that several lncRNAs are involved in the lysosome pathway associated with extracellular matrix (ECM) remodeling. In addition, we also found several mRNAs involved in fibroblast pseudopodia formation, fibroblast growth, and the regulation of smooth muscle cell differentiation in the urinary tract. Our study offers essential data regarding differentially expressed lncRNAs and mRNAs and may provide new potential candidates for the study of SUI.
Collapse
Affiliation(s)
- Aimin Wei
- Department of Gynecology and Obstetrics, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Ruohan Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Kai Wei
- Kangda College of Nanjing Medical University, Lianyungang, 222002, China
| | - Chencheng Dai
- Department of Gynecology and Obstetrics, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Huang
- Kangda College of Nanjing Medical University, Lianyungang, 222002, China
| | - Pengfei Xu
- Department of Gynecology and Obstetrics, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Juan Xu
- Department of Gynecology and Obstetrics, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Hui Tang
- Department of Gynecology and Obstetrics, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Zhang
- Kangda College of Nanjing Medical University, Lianyungang, 222002, China.
| | - Yun Fan
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, China. .,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
21
|
Wu CW, Yau T, Fulgar CC, Mack SM, Revilla AM, Kenyon NJ, Pinkerton KE. Long-Term Sequelae of Smoking and Cessation in Spontaneously Hypertensive Rats. Toxicol Pathol 2019; 48:422-436. [PMID: 31870229 DOI: 10.1177/0192623319893312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Smoking is a major risk factor for heart attack, stroke, and lung cancer. Tobacco smoke (TS) causes bronchitis, emphysema, persistent cough, and dyspnea. Smoking cessation minimizes risks of TS-related disease. To determine whether smoking cessation could reverse TS-induced pulmonary changes, 10-week-old male spontaneously hypertensive rats were exposed to TS or filtered air (FA) for 39 weeks and allowed to live out their normal lifespan. Significantly (P ≤ .05) decreased survival was noted by 21 months in TS versus FA rats. In TS rats, persistent peribronchiolar, perivascular, alveolar, and subpleural inflammation were observed with pervasive infiltration of pigmented foamy macrophages and plausible intra-alveolar fibrosis and osseous metaplasia. Alveolar airspace was significantly (P ≤ .05) increased in TS versus FA rats as was the volume of stored epithelial mucosubstances in the left central axial airway. Increased mucin contributes to airflow obstruction and increased lung infection risks. Findings suggest TS-induced changes do not attenuate with smoking cessation but result in irreversible damage similar to chronic obstructive pulmonary disease. The observed persistent pulmonary changes mirror common TS effects such as chest congestion, sputum production, and shortness of breath long after smoking cessation and represent important targets for treatment of former smokers.
Collapse
Affiliation(s)
- Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, CA, USA.,Forensic Science Graduate Group, University of California, Davis, CA, USA
| | - Tammy Yau
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Ciara C Fulgar
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Savannah M Mack
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Alina M Revilla
- Center for Health and the Environment, University of California, Davis, CA, USA.,Forensic Science Graduate Group, University of California, Davis, CA, USA
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California, Davis, CA, USA.,VA Northern California Healthcare System, Mather, CA, USA
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California, Davis, CA, USA.,Forensic Science Graduate Group, University of California, Davis, CA, USA
| |
Collapse
|
22
|
Mereness JA, Bhattacharya S, Ren Y, Wang Q, Anderson CS, Donlon K, Dylag AM, Haak J, Angelin A, Bonaldo P, Mariani TJ. Collagen VI Deficiency Results in Structural Abnormalities in the Mouse Lung. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:426-441. [PMID: 31837950 DOI: 10.1016/j.ajpath.2019.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/16/2019] [Accepted: 10/11/2019] [Indexed: 01/14/2023]
Abstract
Collagen VI (COL6) is known for its role in a spectrum of congenital muscular dystrophies, which are often accompanied by respiratory dysfunction. However, little is known regarding the function of COL6 in the lung. We confirmed the presence of COL6 throughout the basement membrane region of mouse lung tissue. Lung structure and organization were studied in a previously described Col6a1-/- mouse, which does not produce detectable COL6 in the lung. The Col6a1-/- mouse displayed histopathologic alveolar and airway abnormalities. The airspaces of Col6a1-/- lungs appeared simplified, with larger (29%; P < 0.01) and fewer (31%; P < 0.001) alveoli. These airspace abnormalities included reduced isolectin B4+ alveolar capillaries and surfactant protein C-positive alveolar epithelial type-II cells. Alterations in lung function consistent with these histopathologic changes were evident. Col6a1-/- mice also displayed multiple airway changes, including increased branching (59%; P < 0.001), increased mucosal thickness (34%; P < 0.001), and increased epithelial cell density (13%; P < 0.001). Comprehensive transcriptome analysis revealed that the loss of COL6 is associated with reductions in integrin-paxillin-phosphatidylinositol 3-kinase signaling in vivo. In vitro, COL6 promoted steady-state phosphorylated paxillin levels and reduced cell density (16% to 28%; P < 0.05) at confluence. Inhibition of phosphatidylinositol 3-kinase, or its downstream effectors, resulted in increased cell density to a level similar to that seen on matrices lacking COL6.
Collapse
Affiliation(s)
- Jared A Mereness
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Yue Ren
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Qian Wang
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Christopher S Anderson
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Kathy Donlon
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Andrew M Dylag
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Jeannie Haak
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Alessia Angelin
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York.
| |
Collapse
|
23
|
Rahaghi FN, Argemí G, Nardelli P, Domínguez-Fandos D, Arguis P, Peinado VI, Ross JC, Ash SY, de La Bruere I, Come CE, Diaz AA, Sánchez M, Washko GR, Barberà JA, San José Estépar R. Pulmonary vascular density: comparison of findings on computed tomography imaging with histology. Eur Respir J 2019; 54:1900370. [PMID: 31196942 PMCID: PMC7007984 DOI: 10.1183/13993003.00370-2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 05/11/2018] [Indexed: 11/05/2022]
Abstract
BACKGROUND Exposure to cigarette smoke has been shown to lead to vascular remodelling. Computed tomography (CT) imaging measures of vascular pruning have been associated with pulmonary vascular disease, an important morbidity associated with smoking. In this study we compare CT-based measures of distal vessel loss to histological vascular and parenchymal changes. METHODS A retrospective review of 80 patients who had undergone lung resection identified patients with imaging appropriate for three-dimensional (3D) vascular reconstruction (n=18) and a second group for two-dimensional (2D) analysis (n=19). Measurements of the volume of the small vessels (3D) and the cross-sectional area of the small vessels (<5 mm2 cross-section) were computed. Histological measures of cross-sectional area of the vasculature and loss of alveoli septa were obtained for all subjects. RESULTS The 2D cross-sectional area of the vasculature on CT imaging was associated with the histological vascular cross-sectional area (r=0.69; p=0.001). The arterial small vessel volume assessed by CT correlated with the histological vascular cross-sectional area (r=0.50; p=0.04), a relationship that persisted even when adjusted for CT-derived measures of emphysema in a regression model. CONCLUSIONS Loss of small vessel volume in CT imaging of smokers is associated with histological loss of vascular cross-sectional area. Imaging-based quantification of pulmonary vasculature provides a noninvasive method to study the multiscale effects of smoking on the pulmonary circulation.
Collapse
Affiliation(s)
- Farbod N Rahaghi
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Equal contributors on this article
| | - Gemma Argemí
- Dept of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
- Equal contributors on this article
| | - Pietro Nardelli
- Dept of Radiology, Harvard School of Medicine, Boston, MA, USA
| | - David Domínguez-Fandos
- Dept of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Pedro Arguis
- Dept of Radiology, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Víctor I Peinado
- Dept of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center for Respiratory Diseases (CIBERES), Madrid Spain
| | - James C Ross
- Dept of Radiology, Harvard School of Medicine, Boston, MA, USA
| | - Samuel Y Ash
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Isaac de La Bruere
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Carolyn E Come
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Alejandro A Diaz
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marcelo Sánchez
- Dept of Radiology, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - George R Washko
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Equal contributors on this article
| | - Joan Albert Barberà
- Dept of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center for Respiratory Diseases (CIBERES), Madrid Spain
- Equal contributors on this article
| | - Raúl San José Estépar
- Pulmonary and Critical Care Division, Dept of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Equal contributors on this article
| |
Collapse
|
24
|
Koopsamy Naidoo SV, Bester MJ, Arbi S, Venter C, Dhanraj P, Oberholzer HM. Oral exposure to cadmium and mercury alone and in combination causes damage to the lung tissue of Sprague-Dawley rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 69:86-94. [PMID: 30981014 DOI: 10.1016/j.etap.2019.03.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/20/2019] [Accepted: 03/27/2019] [Indexed: 06/09/2023]
Abstract
Environmental presence and human exposure to heavy metals in air and cigarette smoke has led to a worldwide increase in respiratory disease. The effects of oral exposure to heavy metals in liver and kidney structure and function have been widely investigated and the respiratory system as a target is often overlooked. The aim of the study was to investigate the possible structural changes in the lung tissue of Sprague-Dawley rats after oral exposure for 28 days to cadmium (Cd) and mercury (Hg), alone and in combination at 1000 times the World Health Organization's limit for each metal in drinking water. Following exposure, the general morphology of the bronchiole and lungs as well as collagen and elastin distribution was evaluated using histological techniques and transmission electron microscopy. In the lungs, structural changes to the alveoli included collapsed alveolar spaces, presence of inflammatory cells and thickening of the alveolar walls. In addition, exposure to Cd and Hg caused degeneration of the alveolar structures resulting in confluent alveoli. Changes in bronchiole morphology included an increase in smooth muscle mass with luminal epithelium degeneration, detachment and aggregation. Prominent bronchiole-associated lymphoid tissue was present in the group exposed to Cd and Hg. Ultrastructural examination confirmed the presence of fibrosis where in the Cd exposed group, collagen fibrils arrangement was dense, while in the Hg exposed group, additional prominent elastin was present. This study identified the lungs as target of heavy metals toxicity following oral exposure resulting in cellular damage, inflammation and fibrosis and increased risk of respiratory disease where Hg showed the greatest fibrotic effect, which was further, aggravated in combination with Cd.
Collapse
Affiliation(s)
| | - Megan Jean Bester
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia, 0007, South Africa
| | - Sandra Arbi
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia, 0007, South Africa
| | - Chantelle Venter
- Laboratory for Microscopy and Microanalysis, University of Pretoria, South Africa
| | - Priyanka Dhanraj
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia, 0007, South Africa
| | - Hester Magdalena Oberholzer
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia, 0007, South Africa.
| |
Collapse
|
25
|
Ito JT, Lourenço JD, Righetti RF, Tibério IFLC, Prado CM, Lopes FDTQS. Extracellular Matrix Component Remodeling in Respiratory Diseases: What Has Been Found in Clinical and Experimental Studies? Cells 2019; 8:E342. [PMID: 30979017 PMCID: PMC6523091 DOI: 10.3390/cells8040342] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 01/09/2023] Open
Abstract
Changes in extracellular matrix (ECM) components in the lungs are associated with the progression of respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome (ARDS). Experimental and clinical studies have revealed that structural changes in ECM components occur under chronic inflammatory conditions, and these changes are associated with impaired lung function. In bronchial asthma, elastic and collagen fiber remodeling, mostly in the airway walls, is associated with an increase in mucus secretion, leading to airway hyperreactivity. In COPD, changes in collagen subtypes I and III and elastin, interfere with the mechanical properties of the lungs, and are believed to play a pivotal role in decreased lung elasticity, during emphysema progression. In ARDS, interstitial edema is often accompanied by excessive deposition of fibronectin and collagen subtypes I and III, which can lead to respiratory failure in the intensive care unit. This review uses experimental models and human studies to describe how inflammatory conditions and ECM remodeling contribute to the loss of lung function in these respiratory diseases.
Collapse
Affiliation(s)
- Juliana T Ito
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Juliana D Lourenço
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Renato F Righetti
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
- Rehabilitation service, Sírio-Libanês Hospital, Sao Paulo 01308-050, Brazil.
| | - Iolanda F L C Tibério
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Carla M Prado
- Department of Bioscience, Laboratory of Studies in Pulmonary Inflammation, Federal University of Sao Paulo, Santos 11015-020, Brazil.
| | - Fernanda D T Q S Lopes
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| |
Collapse
|
26
|
Dougherty SC, Ghaus S, Debesa O. Extracorporeal Membrane Oxygenation in Severe Acute Eosinophilic Pneumonia. Front Med (Lausanne) 2019; 6:65. [PMID: 31024915 PMCID: PMC6467954 DOI: 10.3389/fmed.2019.00065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/15/2019] [Indexed: 01/09/2023] Open
Abstract
Acute Eosinophilic Pneumonia (AEP) is a potentially fatal cause of hypoxemic respiratory failure characterized by fever, diffuse bilateral pulmonary infiltrates, and pulmonary eosinophilia. Shown to be associated with a number of environmental exposures and lifestyle choices, AEP has a good prognosis when diagnosed early and treated with corticosteroids. In this clinical case report, we detail the presentation, evaluation, diagnosis, and management of a 40-year old male who presented to the emergency department with dyspnea, chills, and diaphoresis. He had a history of pulmonary embolism 8 years prior but was otherwise healthy, though he had re-started smoking cigarettes a week prior to presentation. Initial chest CT scan revealed widespread mixed groundglass and solid airspace opacities; over the next 12 hours, he rapidly decompensated and after not responding to other invasive mechanical ventilation, was emergently cannulated for veno-venous extracorporeal membrane oxygenation (V-V ECMO). Bronchoalveolar lavage later revealed pulmonary eosinophilia, and after an infectious workup was negative, a diagnosis of AEP was reached and the patient was started on corticosteroids. To our knowledge, this is one of few published cases of AEP requiring V-V ECMO for clinical stabilization, highlighting the utility of this treatment modality in severe disease.
Collapse
Affiliation(s)
- Sean C Dougherty
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Sophia Ghaus
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Orlando Debesa
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
27
|
Using Hyperpolarized Xenon-129 MRI to Quantify Early-Stage Lung Disease in Smokers. Acad Radiol 2019; 26:355-366. [PMID: 30522808 DOI: 10.1016/j.acra.2018.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/04/2018] [Accepted: 11/16/2018] [Indexed: 12/25/2022]
Abstract
RATIONALE AND OBJECTIVES Hyperpolarized xenon-129 magnetic resonance (MR) provides sensitive tools that may detect early stages of lung disease in smokers before it has progressed to chronic obstructive pulmonary disease (COPD) apparent to conventional spirometric measures. We hypothesized that the functional alveolar wall thickness as assessed by hyperpolarized xenon-129 MR spectroscopy would be elevated in clinically healthy smokers before xenon MR diffusion measurements would indicate emphysematous tissue destruction. MATERIALS AND METHODS Using hyperpolarized xenon-129 MR we measured the functional septal wall thickness and apparent diffusion coefficient of the gas phase in 16 subjects with smoking-related COPD, 9 clinically healthy current or former smokers, and 10 healthy never smokers. All subjects were age-matched and characterized by conventional pulmonary function tests. A total of 11 data sets from younger healthy never smokers were added to determine the age dependence of the septal wall thickness measurements. RESULTS In healthy never smokers the septal wall thickness increased by 0.04 μm per year of age. The healthy smoker cohort exhibited normal pulmonary function test measures that did not significantly differ from the never-smoker cohort. The age-corrected septal wall thickness correlated well with diffusion capacity for carbon monoxide (R2 = 0.56) and showed a highly significant difference between healthy subjects and COPD patients (8.8 μm vs 12.3 μm; p < 0.001), but was the only measure that actually discriminated healthy subjects from healthy smokers (8.8 μm vs 10.6 μm; p < 0.006). CONCLUSION Functional alveolar wall thickness assessed by hyperpolarized xenon-129 MR allows discrimination between healthy subjects and healthy smokers and could become a powerful new measure of early-stage lung disease.
Collapse
|
28
|
Oakley RB, Tingay DG, McCall KE, Perkins EJ, Sourial M, Dargaville PA, Pereira-Fantini PM. Gestational Age Influences the Early Microarchitectural Changes in Response to Mechanical Ventilation in the Preterm Lamb Lung. Front Pediatr 2019; 7:325. [PMID: 31497582 PMCID: PMC6712425 DOI: 10.3389/fped.2019.00325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/19/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Preterm birth is associated with abnormal lung architecture, and a reduction in pulmonary function related to the degree of prematurity. A thorough understanding of the impact of gestational age on lung microarchitecture requires reproducible quantitative analysis of lung structure abnormalities. The objectives of this study were (1) to use quantitative histological software (ImageJ) to map morphological patterns of injury resulting from delivery of an identical ventilation strategy to the lung at varying gestational ages and (2) to identify associations between gestational age-specific morphological alterations and key functional outcomes. Method: Lung morphology was compared after 60 min of a standardized ventilation protocol (40 cm H2O sustained inflation and then volume-targeted positive pressure ventilation with positive end-expiratory pressure 8 cm H2O) in lambs at different gestations (119, 124, 128, 133, 140d) representing the spectrum of premature developmental lung states and the term lung. Age-matched controls were compared at 124 and 128d gestation. Automated and manual functions of Image J were used to measure key histological features. Correlation analysis compared morphological and functional outcomes in lambs aged ≤128 and >128d. Results: In initial studies, unventilated lung was indistinguishable at 124 and 128d. Ventilated lung from lambs aged 124d gestation exhibited increased numbers of detached epithelial cells and lung tissue compared with 128d lambs. Comparing results from saccular to alveolar development (120-140d), lambs aged ≤124d exhibited increased lung tissue, average alveolar area, and increased numbers of detached epithelial cells. Alveolar septal width was increased in lambs aged ≤128d. These findings were mirrored in the measures of gas exchange, lung mechanics, and molecular markers of lung injury. Correlation analysis confirmed the gestation-specific relationships between the histological assessments and functional measures in ventilated lambs at gestation ≤128 vs. >128d. Conclusion: Image J allowed rapid, quantitative assessment of alveolar morphology, and lung injury in the preterm lamb model. Gestational age-specific patterns of injury in response to delivery of an identical ventilation strategy were identified, with 128d being a transition point for associations between morphological alterations and functional outcomes. These results further support the need to develop individualized respiratory support approaches tailored to both the gestational age of the infant and their underlying injury response.
Collapse
Affiliation(s)
- Regina B Oakley
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - David G Tingay
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Neonatology, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Karen E McCall
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.,School of Medicine and Medicinal Sciences, University College Dublin, Dublin, Ireland
| | - Elizabeth J Perkins
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Magdy Sourial
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Peter A Dargaville
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Prue M Pereira-Fantini
- Neonatal Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
29
|
In vivo endomicroscopic features of distal airways in chronic bronchitis and chronic obstructive pulmonary disease. КЛИНИЧЕСКАЯ ПРАКТИКА 2018. [DOI: 10.17816/clinpract9415-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Relevance: Probe-based confocal laser endomicroscopy (CLE) of distal airways is a unique technology that allows real-time visualization of structures containing natural fluorophores, which are emitted by exposure to laser radiation with a wavelength of 488 nm, in vivo. To date, the endomicroscopic features of the distal respiratory tract have not been adequately studied in lung diseases, including chronic obstructive pulmonary disease and chronic bronchitis.
The goal of the present study is to describe the endomicroscopic signs of the distal parts of the respiratory system in chronic inflammatory lung diseases such as chronic obstructive pulmonary disease and chronic bronchitis.
Methods: A total of 21 patients with emphysematous and bronchitic phenotypes of chronic obstructive pulmonary disease and chronic bronchitis were examined. All the patients have undergone CLE of distal airways or alveoloscopy during bronchoscopy. The most pathognomonic changes were evaluated on the obtained endomicroscopic images.
Results: For each studied nosological form of chronic inflammatory lung diseases, by careful morphometric analysis of a significant number of informative images, the most specific endomicroscopic changes were identified with the release of CLE patterns.
Conclusions: CLE of distal airways in patients with chronic inflammatory lung diseases allows visualizing changes in the elastic framework of the acini, as well as identifying pathological intraluminal contents, which can be attributed to valuable additional tools in a row of diagnostic methods of respiratory medicine.
Collapse
|
30
|
Poon J, Al-Halawani M, Dubey G. Spontaneous pneumothorax as a complication of chronic Jet propulsion fuel-8 exposure. Heart Lung 2018; 48:169-172. [PMID: 30185391 DOI: 10.1016/j.hrtlng.2018.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/29/2018] [Accepted: 08/02/2018] [Indexed: 10/28/2022]
Abstract
Jet Propulsion Fuel 8 (JP-8) is a kerosene based fuel commonly used in aviation. Occupational exposure to JP-8 may lead to negative health outcomes, which were described in a small number of studies. We report a case of 33-year-old Caucasian male veteran with a history of JP-8 exposure who presented with chronic dyspnea and recurrent spontaneous pneumothorax. To our knowledge, this is the first case of chronic inhalation injury from JP-8 exposure complicated with recurrent secondary spontaneous pneumothorax.
Collapse
Affiliation(s)
- Justin Poon
- Internal Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Moh'd Al-Halawani
- Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA.
| | - Gangacharan Dubey
- Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA; Pulmonary and Critical Care Medicine, VA New York Harbor Health Care System - Brooklyn Campus, Brooklyn, NY 11203, USA
| |
Collapse
|
31
|
Sakinah S, Kumari S, Munisvaradass R, Mok PL, Chee HY, Seenichamy A, P.V K, Higuchi A, Rajan M, Seenivasan KN, Marlina , Arulselvan P, Benelli G, Suresh Kumar S. Repeated infections of dengue (serotype DENV-2) in lung cells of BALB/c mice lead to severe histopathological consequences. Pathog Glob Health 2018. [DOI: 10.1080/20477724.2018.1492765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Affiliation(s)
- S. Sakinah
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sharmilah Kumari
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Selangor, Malaysia
| | | | - Pooi-Ling Mok
- Department of Biomedical Science, Universiti Putra Malaysia, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
| | - Hui-Yee Chee
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Selangor, Malaysia
| | - Arivudainambi Seenichamy
- Microbiology & Immunology Unit, Faculty Of Medicine, Lincoln University College, Petaling Jaya, Malaysia
| | - Kiruthiga P.V
- Department of Medical Biotechnology, Division of Applied Biomedical Sciences and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhong-li, Taiwan
- Department of Reproduction, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Mariappan Rajan
- Biomaterials in Medicinal Chemistry Laboratory, Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, India
| | - K. Nataraja Seenivasan
- Medical Microbiology Laboratory, Department of Microbiology, Bharathidasan University, Tiruchirappalli, India
| | - Marlina
- Faculty of Pharmacy, Universitas Andalas, Padang, Indonesia
| | - Palanisamy Arulselvan
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, India
| | - Giovanni Benelli
- Department of Agriculture, Food and Environment, University of Pisa, Pisa, Italy
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Pisa, Italy
| | - S. Suresh Kumar
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, Selangor, Malaysia
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, India
| |
Collapse
|
32
|
Wang Z, Li R, Zhong R. Extracellular matrix promotes proliferation, migration and adhesion of airway smooth muscle cells in a rat model of chronic obstructive pulmonary disease via upregulation of the PI3K/AKT signaling pathway. Mol Med Rep 2018; 18:3143-3152. [PMID: 30066869 PMCID: PMC6102654 DOI: 10.3892/mmr.2018.9320] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrix (ECM) creates the tissue microenvironment and serves a role in airway wall remodeling in chronic obstructive pulmonary disease (COPD). However, the biological function of ECM in COPD remains to be elucidated. In the present study, 24 healthy Sprague Dawley rats were randomized to normal and COPD groups. COPD was established by intratracheal injection with lipopolysaccharide over 30 days. Subsequently, airway smooth muscle cells (ASMCs) were isolated from rats and served as a model to assess the effects of three ECM components, including collagen type I, laminin and collagen type III (COL‑3). Functional analysis in vitro, using cell counting kit‑8, flow cytometry, wound healing and cell adhesion assays indicated that the ECM components could promote cell proliferation, cell cycle progression, migration and adhesion ability, respectively. Furthermore, as demonstrated by ELISA, treatment with ECM components increased levels of C‑X‑C motif chemokine ligand 1 (CXCL1), CXCL8 and interleukin‑6 in ASMCs. Expression of transforming growth factor β1 (TGFβ1), fibroblast growth factor‑1 (FGF‑1) and tissue inhibitor of metalloproteinase 1 (TIMP1) was increased, and expression of matrix metalloproteinase‑9 (MMP‑9) was decreased following treatment with ECM components, as demonstrated by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Additionally, specific activation of phosphoinositide 3‑kinase (PI3K) signaling, using insulin‑like growth factor‑1 (IGF‑1), promoted cell proliferation and cell cycle progression, increased expression of TGFβ1, FGF‑1, PI3K, AKT, phospho‑AKT, serine/threonine‑protein kinase mTOR (mTOR), phospho‑mTOR and TIMP1, promoted cell migration capacity and reduced the expression level of MMP‑9 in cells from COPD rats. Consistently, PI3K inhibitor LY294002 exerted the opposite effect to IGF‑1. In conclusion, ECM proteins promoted proliferation, migration and adhesion of ASMCs form rat models of COPD through activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zhengyan Wang
- Department of Respiratory Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Rui Li
- Department of Orthopedics, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Rui Zhong
- Second Affiliated Hospital of Hubei University of Medicine, Suizhou, Hubei 442000, P.R. China
| |
Collapse
|
33
|
Balansky R, Ganchev G, Iltcheva M, Dimitrova E, Micale RT, La Maestra S, De Flora S. Carcinogenic response and other histopathological alterations in mice exposed to cigarette smoke for varying time periods after birth. Carcinogenesis 2018; 39:580-587. [PMID: 29370344 DOI: 10.1093/carcin/bgy013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/15/2018] [Indexed: 01/01/2023] Open
Abstract
In spite of the outstanding role of tobacco smoking in human carcinogenesis, it is difficult to reproduce its effects in experimental animals. Based on the knowledge that a variety of mechanisms account for a higher susceptibility to carcinogens early in life, we have developed a murine model in which mainstream cigarette smoke becomes convincingly carcinogenic. The standard model involves exposure to smoke for 4 months, starting after birth, followed by an additional 3-4 months in filtered air. We evaluated herein the time- and dose-dependent response, at 7.5 months of life, of Swiss H mice that had been exposed to smoke for either 1, 2 or 4 months after birth. A one-month exposure, corresponding to a period of intense alveolarization, was sufficient to induce most inflammatory, degenerative and preneoplastic pulmonary lesions, including emphysema and alveolar epithelial hyperplasia, blood vessel proliferation and hemangiomas, reflecting an early proangiogenic role of smoking, and microadenomas bearing ki-67-positive proliferating cells as well as urinary bladder epithelial hyperplasia. Two months of exposure were needed to induce pulmonary adenomas and urinary bladder papillomas in males only, which highlights a protective role of estrogens in urinary bladder carcinogenesis. Four months, which in humans would correspond to the postnatal period, puberty, adolescence and early adulthood, were needed to induce other lesions, including tubular epithelial hyperplasia of kidney, bronchial epithelial hyperplasia and especially pulmonary malignant tumors. These findings highlight the concept that preneoplastic and neoplastic lesions occurring in adulthood can be induced by exposure to smoke early in life.
Collapse
Affiliation(s)
| | | | | | | | - Rosanna T Micale
- Department of Health Sciences, University of Genoa, Genoa, Italy
- National Center of Oncology, Sofia, Bulgaria
| | - Sebastiano La Maestra
- Department of Health Sciences, University of Genoa, Genoa, Italy
- National Center of Oncology, Sofia, Bulgaria
| | - Silvio De Flora
- Department of Health Sciences, University of Genoa, Genoa, Italy
- National Center of Oncology, Sofia, Bulgaria
| |
Collapse
|
34
|
Gharib SA, Manicone AM, Parks WC. Matrix metalloproteinases in emphysema. Matrix Biol 2018; 73:34-51. [PMID: 29406250 DOI: 10.1016/j.matbio.2018.01.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/14/2017] [Accepted: 01/24/2018] [Indexed: 02/07/2023]
Abstract
Several studies have implicated a causative role for specific matrix metalloproteinases (MMPs) in the development and progression of cigarette smoke-induced chronic obstructive pulmonary disease (COPD) and its severe sequela, emphysema. However, the precise function of any given MMP in emphysema remains an unanswered question. Emphysema results from the degradation of alveolar elastin - among other possible mechanisms - a process that is often thought to be caused by elastolytic proteinases made by macrophages. In this article, we discuss the data suggesting, supporting, or refuting causative roles of macrophage-derived MMPs, with a focus on MMPs-7, -9, -10, -12, and 28, in both the human disease and mouse models of emphysema. Findings from experimental models suggest that some MMPs, such as MMP-12, may directly breakdown elastin, whereas others, particularly MMP-10 and MMP-28, promote the development of emphysema by influencing the proteolytic and inflammatory activities of macrophages.
Collapse
Affiliation(s)
- Sina A Gharib
- Center for Lung Biology, University of Washington, Seattle, WA, USA
| | - Anne M Manicone
- Center for Lung Biology, University of Washington, Seattle, WA, USA
| | - William C Parks
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
36
|
Burgstaller G, Oehrle B, Gerckens M, White ES, Schiller HB, Eickelberg O. The instructive extracellular matrix of the lung: basic composition and alterations in chronic lung disease. Eur Respir J 2017; 50:50/1/1601805. [PMID: 28679607 DOI: 10.1183/13993003.01805-2016] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
The pulmonary extracellular matrix (ECM) determines the tissue architecture of the lung, and provides mechanical stability and elastic recoil, which are essential for physiological lung function. Biochemical and biomechanical signals initiated by the ECM direct cellular function and differentiation, and thus play a decisive role in lung development, tissue remodelling processes and maintenance of adult homeostasis. Recent proteomic studies have demonstrated that at least 150 different ECM proteins, glycosaminoglycans and modifying enzymes are expressed in the lung, and these assemble into intricate composite biomaterials. These highly insoluble assemblies of interacting ECM proteins and their glycan modifications can act as a solid phase-binding interface for hundreds of secreted proteins, which creates an information-rich signalling template for cell function and differentiation. Dynamic changes within the ECM that occur upon injury or with ageing are associated with several chronic lung diseases. In this review, we summarise the available data about the structure and function of the pulmonary ECM, and highlight changes that occur in idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension (PAH), chronic obstructive pulmonary disease (COPD), asthma and lung cancer. We discuss potential mechanisms of ECM remodelling and modification, which we believe are relevant for future diagnosis and treatment of chronic lung disease.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Bettina Oehrle
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Michael Gerckens
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Herbert B Schiller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Oliver Eickelberg
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| |
Collapse
|
37
|
Li FJ, Surolia R, Li H, Wang Z, Liu G, Liu RM, Mirov SB, Athar M, Thannickal VJ, Antony VB. Low-dose cadmium exposure induces peribronchiolar fibrosis through site-specific phosphorylation of vimentin. Am J Physiol Lung Cell Mol Physiol 2017; 313:L80-L91. [PMID: 28450285 PMCID: PMC5538875 DOI: 10.1152/ajplung.00087.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/18/2017] [Indexed: 02/08/2023] Open
Abstract
Exposure to cadmium (Cd) has been associated with development of chronic obstructive lung disease (COPD). The mechanisms and signaling pathways whereby Cd causes pathological peribronchiolar fibrosis, airway remodeling, and subsequent airflow obstruction remain unclear. We aimed to evaluate whether low-dose Cd exposure induces vimentin phosphorylation and Yes-associated protein 1 (YAP1) activation leading to peribronchiolar fibrosis and subsequent airway remodeling. Our data demonstrate that Cd induces myofibroblast differentiation and extracellular matrix (ECM) deposition around small (<2 mm in diameter) airways. Upon Cd exposure, α-smooth muscle actin (α-SMA) expression and the production of ECM proteins, including fibronectin and collagen-1, are markedly induced in primary human lung fibroblasts. Cd induces Smad2/3 activation and the translocation of both Smad2/3 and Yes-associated protein 1 (YAP1) into the nucleus. In parallel, Cd induces AKT and cdc2 phosphorylation and downstream vimentin phosphorylation at Ser39 and Ser55, respectively. AKT and cdc2 inhibitors block Cd-induced vimentin fragmentation and secretion in association with inhibition of α-SMA expression, ECM deposition, and collagen secretion. Furthermore, vimentin silencing abrogates Cd-induced α-SMA expression and decreases ECM production. Vimentin-deficient mice are protected from Cd-induced peribronchiolar fibrosis and remodeling. These findings identify two specific sites on vimentin that are phosphorylated by Cd and highlight the functional significance of vimentin phosphorylation in YAP1/Smad3 signaling that mediates Cd-induced peribronchiolar fibrosis and airway remodeling.
Collapse
Affiliation(s)
- Fu Jun Li
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ranu Surolia
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Huashi Li
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zheng Wang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gang Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sergey B Mirov
- Department of Physics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Veena B Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
38
|
Matrix Metalloproteinase-28 Is a Key Contributor to Emphysema Pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1288-1300. [PMID: 28399390 DOI: 10.1016/j.ajpath.2017.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/16/2017] [Indexed: 10/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) comprises chronic bronchitis and emphysema, and is a leading cause of morbidity and mortality. Because tissue destruction is the prominent characteristic of emphysema, extracellular proteinases, particularly those with elastolytic ability, are often considered to be key drivers in this disease. Several human and mouse studies have implicated roles for matrix metalloproteinases (MMPs), particularly macrophage-derived proteinases, in COPD pathogenesis. MMP-28 is expressed by the pulmonary epithelium and macrophage, and we have found that it regulates macrophage recruitment and polarization. We hypothesized that MMP-28 has contributory roles in emphysema via alteration of macrophage numbers and activation. Because of the established association of emphysema pathogenesis to macrophage influx, we evaluated the inflammatory changes and lung histology of Mmp28-/- mice exposed to 3 and 6 months of cigarette smoke. At earlier time points, we found altered macrophage polarization in the smoke-exposed Mmp28-/- lung consistent with other published findings that MMP-28 regulates macrophage activation. At both 3 and 6 months, Mmp28-/- mice had blunted inflammatory responses more closely resembling nonsmoked mice, with a reduction in neutrophil recruitment and CXCL1 chemokine expression. By 6 months, Mmp28-/- mice were protected from emphysema. These results highlight a previously unrecognized role for MMP-28 in promoting chronic lung inflammation and tissue remodeling induced by cigarette smoke and highlight another potential target to modulate COPD.
Collapse
|
39
|
Washko GR, Kinney GL, Ross JC, San José Estépar R, Han MK, Dransfield MT, Kim V, Hatabu H, Come CE, Bowler RP, Silverman EK, Crapo J, Lynch DA, Hokanson J, Diaz AA. Lung Mass in Smokers. Acad Radiol 2017; 24:386-392. [PMID: 27940230 DOI: 10.1016/j.acra.2016.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 01/21/2023]
Abstract
RATIONALE AND OBJECTIVE Emphysema is characterized by airspace dilation, inflammation, and irregular deposition of elastin and collagen in the interstitium. Computed tomographic studies have reported that lung mass (LM) may be increased in smokers, a finding attributed to inflammatory and parenchymal remodeling processes observed on histopathology. We sought to examine the epidemiologic and clinical associations of LM in smokers. MATERIALS AND METHODS Baseline epidemiologic, clinical, and computed tomography (CT) data (n = 8156) from smokers enrolled into the COPDGene Study were analyzed. LM was calculated from the CT scan. Changes in lung function at 5 years' follow-up were available from 1623 subjects. Regression analysis was performed to assess for associations of LM with forced expiratory volume in 1 second (FEV1) and FEV1 decline. RESULTS Subjects with Global Initiative for Chronic Obstructive Lung Disease (GOLD) 1 chronic obstructive pulmonary disease had greater LM than either smokers with normal lung function or those with GOLD 2-4 chronic obstructive pulmonary disease (P < 0.001 for both comparisons). LM was predictive of the rate of the decline in FEV1 (decline per 100 g, -4.7 ± 1.7 mL/y, P = 0.006). CONCLUSIONS Our cross-sectional data suggest the presence of a biphasic radiological remodeling process in smokers: the presence of such nonlinearity must be accounted for in longitudinal computed tomographic studies. Baseline LM predicts the decline in lung function.
Collapse
Affiliation(s)
- George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado
| | - James C Ross
- Surgical Planning Laboratory, Laboratory of Mathematics in Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Raúl San José Estépar
- Surgical Planning Laboratory, Laboratory of Mathematics in Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - MeiLan K Han
- Department of Medicine, Division of Pulmonary and Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Mark T Dransfield
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Hiroto Hatabu
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Carolyn E Come
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115
| | - Russell P Bowler
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - James Crapo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, Colorado
| | - John Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115.
| |
Collapse
|
40
|
Hogg JC, Paré PD, Hackett TL. The Contribution of Small Airway Obstruction to the Pathogenesis of Chronic Obstructive Pulmonary Disease. Physiol Rev 2017; 97:529-552. [PMID: 28151425 PMCID: PMC6151481 DOI: 10.1152/physrev.00025.2015] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hypothesis that the small conducting airways were the major site of obstruction to airflow in normal lungs was introduced by Rohrer in 1915 and prevailed until Weibel introduced a quantitative method of studying lung anatomy in 1963. Green repeated Rohrer's calculations using Weibels new data in 1965 and found that the smaller conducting airways offered very little resistance to airflow. This conflict was resolved by seminal experiments conducted by Macklem and Mead in 1967, which confirmed that a small proportion of the total lower airways resistance is attributable to small airways <2 mm in diameter. Shortly thereafter, Hogg, Macklem, and Thurlbeck used this technique to show that small airways become the major site of obstruction in lungs affected by emphysema. These and other observations led Mead to write a seminal editorial in 1970 that postulated the small airways are a silent zone within normal lungs where disease can accumulate over many years without being noticed. This review provides a progress report since the 1970s on methods for detecting chronic obstructive pulmonary disease, the structural nature of small airways' disease, and the cellular and molecular mechanisms that are thought to underlie its pathogenesis.
Collapse
Affiliation(s)
- James C Hogg
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| | - Peter D Paré
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia (UBC), British Columbia, Canada; Department of Pathology and Laboratory Medicine, UBC, British Columbia, Canada; Respiratory Division, Department of Medicine, UBC, British Columbia, Canada; and Department of Anesthesiology, Pharmacology and Therapeutics, UBC, British Columbia, Canada
| |
Collapse
|
41
|
Mondoñedo JR, Suki B. Predicting Structure-Function Relations and Survival following Surgical and Bronchoscopic Lung Volume Reduction Treatment of Emphysema. PLoS Comput Biol 2017; 13:e1005282. [PMID: 28182686 PMCID: PMC5300131 DOI: 10.1371/journal.pcbi.1005282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/06/2016] [Indexed: 11/26/2022] Open
Abstract
Lung volume reduction surgery (LVRS) and bronchoscopic lung volume reduction (bLVR) are palliative treatments aimed at reducing hyperinflation in advanced emphysema. Previous work has evaluated functional improvements and survival advantage for these techniques, although their effects on the micromechanical environment in the lung have yet to be determined. Here, we introduce a computational model to simulate a force-based destruction of elastic networks representing emphysema progression, which we use to track the response to lung volume reduction via LVRS and bLVR. We find that (1) LVRS efficacy can be predicted based on pre-surgical network structure; (2) macroscopic functional improvements following bLVR are related to microscopic changes in mechanical force heterogeneity; and (3) both techniques improve aspects of survival and quality of life influenced by lung compliance, albeit while accelerating disease progression. Our model predictions yield unique insights into the microscopic origins underlying emphysema progression before and after lung volume reduction. Surgical and, more recently, bronchoscopic lung volume reduction is the only available treatments for patients with advanced stage emphysema. Several large-scale, clinical studies have outlined appropriate selection criteria based on patient outcomes; however, the underlying mechanisms determining disease progression and response to these treatments are not well-understood. To answer this question, we have developed a network model of the lung to compare immediate and long-term response to each treatment. This approach allows us to directly study macroscopic changes in function related to microscopic changes in the local structural and mechanical environment. In addition, it facilitates direct comparisons between surgical and bronchoscopic lung volume reduction given identical initial conditions, which is not feasible in a clinical study. We propose here a mechanism suggesting that lung volume reduction efficacy is intimately linked to changes in microscopic force heterogeneity within the lung. Such an understanding of the mechanisms driving emphysema has the potential to greatly improve current therapies for this condition through more rationalized, patient-specific treatment strategies.
Collapse
Affiliation(s)
- Jarred R. Mondoñedo
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- School of Medicine, Boston University, Boston, MA, United States of America
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
42
|
Burgess JK, Mauad T, Tjin G, Karlsson JC, Westergren-Thorsson G. The extracellular matrix - the under-recognized element in lung disease? J Pathol 2016; 240:397-409. [PMID: 27623753 PMCID: PMC5129494 DOI: 10.1002/path.4808] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/17/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022]
Abstract
The lung is composed of airways and lung parenchyma, and the extracellular matrix (ECM) contains the main building blocks of both components. The ECM provides physical support and stability to the lung, and as such it has in the past been regarded as an inert structure. More recent research has provided novel insights revealing that the ECM is also a bioactive environment that orchestrates the cellular responses in its environs. Changes in the ECM in the airway or parenchymal tissues are now recognized in the pathological profiles of many respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Only recently have we begun to investigate whether these ECM changes result from the disease process, or whether they constitute a driving factor that orchestrates the pathological outcomes. This review summarizes our current knowledge of the alterations in the ECM in asthma, COPD, and IPF, and the contributions of these alterations to the pathologies. Emerging data suggest that alterations in the composition, folding or rigidity of ECM proteins may alter the functional responses of cells within their environs, and in so doing change the pathological outcomes. These characteristics highlight potential avenues for targeting lung pathologies in the future. This may ultimately contribute to a better understanding of chronic lung diseases, and novel approaches for finding therapeutic solutions. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Centre Groningen, GRIAC Research Institute, Department of Pathology and Medical Biology, Groningen, The Netherlands.,Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Discipline of Pharmacology, The University of Sydney, NSW, Australia.,Central Clinical School, The University of Sydney, NSW, Australia
| | - Thais Mauad
- Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Gavin Tjin
- Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Central Clinical School, The University of Sydney, NSW, Australia
| | - Jenny C Karlsson
- Lung Biology, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| | | |
Collapse
|
43
|
Lai PS, Hang JQ, Zhang FY, Sun J, Zheng BY, Su L, Washko GR, Christiani DC. Imaging Phenotype of Occupational Endotoxin-Related Lung Function Decline. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1436-1442. [PMID: 27138294 PMCID: PMC5010398 DOI: 10.1289/ehp195] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/04/2015] [Accepted: 04/19/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Although occupational exposures contribute to a significant proportion of obstructive lung disease, the phenotype of obstructive lung disease associated with work-related organic dust exposure independent of smoking remains poorly defined. OBJECTIVE We identified the relative contributions of smoking and occupational endotoxin exposure to parenchymal and airway remodeling as defined by quantitative computed tomography (CT). METHODS The Shanghai Textile Worker Study is a longitudinal study of endotoxin-exposed cotton workers and endotoxin-unexposed silk workers that was initiated in 1981. Spirometry, occupational endotoxin exposure, and smoking habits were assessed at 5-year intervals. High-resolution computed tomography (CT) was performed in 464 retired workers in 2011, along with quantitative lung densitometric and airway analysis. RESULTS Significant differences in all CT measures were noted across exposure groups. Occupational endotoxin exposure was associated with a decrease (-1.3%) in percent emphysema (LAAI-950), a 3.3-Hounsfield unit increase in 15th percentile density, an 18.1-g increase in lung mass, and a 2.3% increase in wall area percent. Current but not former smoking was associated with a similar CT phenotype. Changes in LAAI-950 were highly correlated with 15th percentile density (correlation -1.0). Lung mass was the only measure associated with forced expiratory volume in 1 sec (FEV1) decline, with each 10-g increase in lung mass associated with an additional loss (-6.1 mL) of FEV1 (p = 0.001) between 1981 and 2011. CONCLUSIONS There are many similarities between the effects of occupational endotoxin exposure and those of tobacco smoke exposure on lung parenchyma and airway remodeling. The effects of occupational endotoxin exposure appear to persist even after the cessation of exposure. LAAI-950 may not be a reliable indicator of emphysema in subjects without spirometric impairment. Lung mass is a CT-based biomarker of accelerated lung function decline. CITATION Lai PS, Hang J, Zhang F, Sun J, Zheng BY, Su L, Washko GR, Christiani DC. 2016. Imaging phenotype of occupational endotoxin-related lung function decline. Environ Health Perspect 124:1436-1442; http://dx.doi.org/10.1289/EHP195.
Collapse
Affiliation(s)
- Peggy S. Lai
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jing-qing Hang
- Shanghai Putuo District People’s Hospital, Shanghai, China
| | | | - J. Sun
- Shanghai Putuo District People’s Hospital, Shanghai, China
| | - Bu-Yong Zheng
- Shanghai Putuo District People’s Hospital, Shanghai, China
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - George R. Washko
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - David C. Christiani
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Jones RL, Noble PB, Elliot JG, James AL. Airway remodelling in COPD: It's not asthma! Respirology 2016; 21:1347-1356. [PMID: 27381663 DOI: 10.1111/resp.12841] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/03/2016] [Accepted: 04/30/2016] [Indexed: 11/29/2022]
Abstract
COPD is defined as airflow limitation that is not reversed by treatment. In asthma, airflow limitation is not only reversible, but also inducible. This is called 'airway hyperresponsiveness' (AHR) and is associated with thickening of the airway wall, predominantly the layer of airway smooth muscle, due to more cells, bigger cells and more extracellular matrix (ECM) in proportion to the increase in smooth muscle. AHR is also observed in COPD if the changes in airflow are expressed as a percent of the baseline lung function. However, the absolute change in baseline lung function that can be induced in COPD is actually less than that seen in normal subjects, suggesting that the airways in COPD are resistant not only to opening, but also to closing. This observation agrees with physiological measures showing increased airway wall stiffness in COPD. Like asthma, airway wall thickness is increased in COPD, including the layer of smooth muscle. Unlike asthma, however, fixed airflow obstruction appears to be characterized by a disproportionate increase in the ECM within the smooth muscle layer. In this review, we summarize the studies of airway matrix deposition in COPD and put forward the proposal that the airway remodelling in COPD is different from that in asthma and call for a systematic analysis of airway matrix deposition in COPD.
Collapse
Affiliation(s)
- Robyn L Jones
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia. .,School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Western Australia, Australia.
| | - Peter B Noble
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Western Australia, Australia.,Centre for Neonatal Research and Education, School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - John G Elliot
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
45
|
Chronic obstructive pulmonary disease: A guide for the primary care physician. Dis Mon 2016; 62:164-87. [DOI: 10.1016/j.disamonth.2016.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
46
|
Bidan CM, Veldsink AC, Meurs H, Gosens R. Airway and Extracellular Matrix Mechanics in COPD. Front Physiol 2015; 6:346. [PMID: 26696894 PMCID: PMC4667091 DOI: 10.3389/fphys.2015.00346] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common lung diseases worldwide, and is characterized by airflow obstruction that is not fully reversible with treatment. Even though airflow obstruction is caused by airway smooth muscle contraction, the extent of airway narrowing depends on a range of other structural and functional determinants that impact on active and passive tissue mechanics. Cells and extracellular matrix in the airway and parenchymal compartments respond both passively and actively to the mechanical stimulation induced by smooth muscle contraction. In this review, we summarize the factors that regulate airway narrowing and provide insight into the relative contributions of different constituents of the extracellular matrix and their biomechanical impact on airway obstruction. We then review the changes in extracellular matrix composition in the airway and parenchymal compartments at different stages of COPD, and finally discuss how these changes impact airway narrowing and the development of airway hyperresponsiveness. Finally, we position these data in the context of therapeutic research focused on defective tissue repair. As a conclusion, we propose that future works should primarily target mild or early COPD, prior to the widespread structural changes in the alveolar compartment that are more characteristic of severe COPD.
Collapse
Affiliation(s)
- Cécile M Bidan
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands ; Laboratoire Interdisciplinaire de Physique (LIPhy), Université Grenoble Alpes Grenoble, France ; Centre National de la Recherche Scientifique, LIPhy Grenoble, France
| | - Annemiek C Veldsink
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| |
Collapse
|
47
|
Hidvegi T, Stolz DB, Alcorn JF, Yousem SA, Wang J, Leme AS, Houghton AM, Hale P, Ewing M, Cai H, Garchar EA, Pastore N, Annunziata P, Kaminski N, Pilewski J, Shapiro SD, Pak SC, Silverman GA, Brunetti-Pierri N, Perlmutter DH. Enhancing Autophagy with Drugs or Lung-directed Gene Therapy Reverses the Pathological Effects of Respiratory Epithelial Cell Proteinopathy. J Biol Chem 2015; 290:29742-57. [PMID: 26494620 DOI: 10.1074/jbc.m115.691253] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 11/06/2022] Open
Abstract
Recent studies have shown that autophagy mitigates the pathological effects of proteinopathies in the liver, heart, and skeletal muscle but this has not been investigated for proteinopathies that affect the lung. This may be due at least in part to the lack of an animal model robust enough for spontaneous pathological effects from proteinopathies even though several rare proteinopathies, surfactant protein A and C deficiencies, cause severe pulmonary fibrosis. In this report we show that the PiZ mouse, transgenic for the common misfolded variant α1-antitrypsin Z, is a model of respiratory epithelial cell proteinopathy with spontaneous pulmonary fibrosis. Intracellular accumulation of misfolded α1-antitrypsin Z in respiratory epithelial cells of the PiZ model resulted in activation of autophagy, leukocyte infiltration, and spontaneous pulmonary fibrosis severe enough to elicit functional restrictive deficits. Treatment with autophagy enhancer drugs or lung-directed gene transfer of TFEB, a master transcriptional activator of the autophagolysosomal system, reversed these proteotoxic consequences. We conclude that this mouse is an excellent model of respiratory epithelial proteinopathy with spontaneous pulmonary fibrosis and that autophagy is an important endogenous proteostasis mechanism and an attractive target for therapy.
Collapse
Affiliation(s)
- Tunda Hidvegi
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | - John F Alcorn
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | | | | | | | - Pamela Hale
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Michael Ewing
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Houming Cai
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Evelyn Akpadock Garchar
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Nunzia Pastore
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | - Patrizia Annunziata
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | | | | | | | - Stephen C Pak
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Gary A Silverman
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138 Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy, 80131, and
| | - David H Perlmutter
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| |
Collapse
|
48
|
Lung structure and function in elastase-treated rats: A follow-up study. Respir Physiol Neurobiol 2015; 215:13-9. [DOI: 10.1016/j.resp.2015.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 11/19/2022]
|
49
|
Gould NS, Min E, Huang J, Chu HW, Good J, Martin RJ, Day BJ. Glutathione Depletion Accelerates Cigarette Smoke-Induced Inflammation and Airspace Enlargement. Toxicol Sci 2015; 147:466-74. [PMID: 26149495 DOI: 10.1093/toxsci/kfv143] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The study objective was to assess age-related changes in glutathione (GSH) adaptive response to cigarette smoke (CS) exposure. Older cigarette smokers show a decline (67%) in lung epithelial lining fluid (ELF) GSH and a 1.8-fold decreased GSH adaptive response to cigarette smoking with a concomitant elevation (47%) of exhaled nitric oxide compared with younger smokers. In order to isolate the changes in tissue GSH from other age-related effects, pharmacological inhibition of the rate limiting step in GSH synthesis was employed to examine the lung's response to CS exposure in young mice. The γ-glutamylcysteine ligase inhibitor L-buthionine-sulfoximine (BSO) was administered in the drinking water (20 mM) to decrease by half the in vivo GSH levels to those found in aged mice and humans. Mice were then exposed to CS (3 h/day) for 5 or 15 days. Biochemical analysis of the ELF and lung tissue revealed an inhibition of the CS-induced GSH adaptive response by BSO with a concurrent increase in mixed protein-GSH disulfides indicating increased cysteine oxidation. The prevention of the GSH adaptive response led to an increase in pro-inflammatory cytokines present in the lung. Airspace enlargement is a hallmark of lung emphysema and was observed in mice treated with BSO and exposed to CS for as little as 15 days, whereas these types of changes normally take up to 6 months in this model. BSO treatment potentiated both lung elastase and matrix metalloproteinase activity in the CS group. These data suggest that age-related decline in the GSH adaptive response can markedly accelerate many of the factors thought to drive CS-induced emphysema.
Collapse
Affiliation(s)
- Neal S Gould
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206; Departments of Pharmaceutical Sciences
| | - Elysia Min
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Jie Huang
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Hong Wei Chu
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206; Medicine and Immunology, University of Colorado at Denver, Aurora, Colorado 80045
| | - Jim Good
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Richard J Martin
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206; Medicine and
| | - Brian J Day
- *Department of Medicine, National Jewish Health, Denver, Colorado 80206; Departments of Pharmaceutical Sciences, Medicine and Immunology, University of Colorado at Denver, Aurora, Colorado 80045
| |
Collapse
|
50
|
Taguchi L, Pinheiro NM, Olivo CR, Choqueta-Toledo A, Grecco SS, Lopes FDTQS, Caperuto LC, Martins MA, Tiberio IFLC, Câmara NO, Lago JHG, Prado CM. A flavanone from Baccharis retusa (Asteraceae) prevents elastase-induced emphysema in mice by regulating NF-κB, oxidative stress and metalloproteinases. Respir Res 2015; 16:79. [PMID: 26122092 PMCID: PMC4489216 DOI: 10.1186/s12931-015-0233-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/08/2015] [Indexed: 01/02/2023] Open
Abstract
Background Pulmonary emphysema is characterized by irreversible airflow obstruction, inflammation, oxidative stress imbalance and lung remodeling, resulting in reduced lung function and a lower quality of life. Flavonoids are plant compounds with potential anti-inflammatory and antioxidant effects that have been used in folk medicine. Our aim was to determine whether treatment with sakuranetin, a flavonoid extracted from the aerial parts of Baccharis retusa, interferes with the development of lung emphysema. Methods Intranasal saline or elastase was administered to mice; the animals were then treated with sakuranetin or vehicle 2 h later and again on days 7, 14 and 28. We evaluated lung function and the inflammatory profile in bronchoalveolar lavage fluid (BALF). The lungs were removed to evaluate alveolar enlargement, extracellular matrix fibers and the expression of MMP-9, MMP-12, TIMP-1, 8-iso-PGF-2α and p65-NF-κB in the fixed tissues as well as to evaluate cytokine levels and p65-NF-κB protein expression. Results In the elastase-treated animals, sakuranetin treatment reduced the alveolar enlargement, collagen and elastic fiber deposition and the number of MMP-9- and MMP-12-positive cells but increased TIMP-1 expression. In addition, sakuranetin treatment decreased the inflammation and the levels of TNF-α, IL-1β and M-CSF in the BALF as well as the levels of NF-κB and 8-iso-PGF-2α in the lungs of the elastase-treated animals. However, this treatment did not affect the changes in lung function. Conclusion These data emphasize the importance of oxidative stress and metalloproteinase imbalance in the development of emphysema and suggest that sakuranetin is a potent candidate that should be further investigated as an emphysema treatment. This compound may be useful for counteracting lung remodeling and oxidative stress and thus attenuating the development of emphysema. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0233-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Taguchi
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil
| | - Nathalia M Pinheiro
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Clarice R Olivo
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Simone S Grecco
- Department of Exact and Earth Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Fernanda D T Q S Lopes
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luciana C Caperuto
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil
| | - Mílton A Martins
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tiberio
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Niels O Câmara
- Department of Immunology, Biological Institute, Universidade de São Paulo, São Paulo, Brazil
| | - João Henrique G Lago
- Department of Exact and Earth Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Carla M Prado
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil. .,Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|