1
|
Samarelli AV, Tonelli R, Raineri G, Mastrolia I, Costantini M, Fabbiani L, Catani V, Petrachi T, Bruzzi G, Andrisani D, Gozzi F, Marchioni A, Masciale V, Aramini B, Ruggieri V, Grisendi G, Dominici M, Cerri S, Clini E. Expression of HOXB7 in the Lung of Patients with Idiopathic Pulmonary Fibrosis: A Proof-of-Concept Study. Biomedicines 2024; 12:1321. [PMID: 38927528 PMCID: PMC11201217 DOI: 10.3390/biomedicines12061321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The molecular pathways involved in the onset and progression of idiopathic pulmonary fibrosis (IPF) still need to be fully clarified as some are shared with lung cancer development. HOXB7, a member of the homeobox (Hox) gene family, has been found involved in various cancers. METHODS Immunohistochemical (IHC) analysis was run on lung tissue samples from surgical lung biopsy (SLB) of 19 patients with IPF, retrospectively selected from the IPF database of the University Hospital of Modena. HOXB7 expression was analyzed and compared with that of five patients with no evidence of pulmonary fibrosis as controls. RESULTS The semi-quantitative analysis of IHC showed that HOXB7 protein expression was higher in IPF patients compared to controls (difference between means = 6.2 ± 2.37, p = 0.0157). Further, HOXB7 expression was higher in IPF patients with a higher extent of fibrosis (50-75%)-measured with high-resolution computer tomography-compared to those with a lower extent (0-25%) (difference between means = 25.74 ± 6.72, p = 0.004). CONCLUSIONS The expression of HOXB7 is higher in the lung of IPF patients compared to controls, and was represented in different cellular compartments within the lung niche. Further investigations are needed to clarify its role in the pathogenesis and progression of IPF.
Collapse
Affiliation(s)
- Anna Valeria Samarelli
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Roberto Tonelli
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Giulia Raineri
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Ilenia Mastrolia
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (I.M.); (V.C.); (V.M.); (G.G.); (M.D.)
| | - Matteo Costantini
- Pathology Unit, University Hospital of Modena, 41124 Modena, Italy; (M.C.); (L.F.)
| | - Luca Fabbiani
- Pathology Unit, University Hospital of Modena, 41124 Modena, Italy; (M.C.); (L.F.)
| | - Virginia Catani
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (I.M.); (V.C.); (V.M.); (G.G.); (M.D.)
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
| | - Tiziana Petrachi
- Technopole “Mario Veronesi”, Via 29 Maggio 6, 41037 Mirandola, Italy;
| | - Giulia Bruzzi
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
| | - Dario Andrisani
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Filippo Gozzi
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
| | - Alessandro Marchioni
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Valentina Masciale
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (I.M.); (V.C.); (V.M.); (G.G.); (M.D.)
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, DIMEC of the Alma Mater Studiorum, University of Bologna, GB Morgagni-L Pierantoni Hospital, 47121 Forlì, Italy;
| | - Valentina Ruggieri
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (I.M.); (V.C.); (V.M.); (G.G.); (M.D.)
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (I.M.); (V.C.); (V.M.); (G.G.); (M.D.)
- Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Stefania Cerri
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Enrico Clini
- Respiratory Diseases Unit, University Hospital of Modena, 41124 Modena, Italy; (A.V.S.); (G.R.); (G.B.); (D.A.); (F.G.); (A.M.); (V.R.); (S.C.); (E.C.)
- Laboratory of Cellular Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41124 Modena, Italy
| |
Collapse
|
2
|
Chong L, Zou L, Xiang L, Song X, Miao W, Yan X, Xu M, Ling G, El Agha E, Bellusci S, Lou Z, Zhang H, Zhang JS. WSB1, a Hypoxia-Inducible E3 Ligase, Promotes Myofibroblast Accumulation and Attenuates Alveolar Epithelial Regeneration in Mouse Lung Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:656-672. [PMID: 38325552 DOI: 10.1016/j.ajpath.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/19/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Idiopathic pulmonary fibrosis is a progressive interstitial lung disease for which there is no curative therapy available. Repetitive alveolar epithelial injury repair, myofibroblast accumulation, and excessive collagen deposition are key pathologic features of idiopathic pulmonary fibrosis, eventually leading to cellular hypoxia and respiratory failure. The precise mechanism driving this complex maladaptive process remains inadequately understood. WD repeat and suppressor of cytokine signaling box containing 1 (WSB1) is an E3 ubiquitin ligase, the expression of which is associated strongly with hypoxia, and forms a positive feedback loop with hypoxia-inducible factor 1α (HIF-1α) under anoxic condition. This study explored the expression, cellular distribution, and function of WSB1 in bleomycin (BLM)-induced mouse lung injury and fibrosis. WSB1 expression was highly induced by BLM injury and correlated with the progression of lung fibrosis. Significantly, conditional deletion of Wsb1 in adult mice ameliorated BLM-induced pulmonary fibrosis. Phenotypically, Wsb1-deficient mice showed reduced lipofibroblast to myofibroblast transition, but enhanced alveolar type 2 proliferation and differentiation into alveolar type 1 after BLM injury. Proteomic analysis of mouse lung tissues identified caveolin 2 as a potential downstream target of WSB1, contributing to BLM-induced epithelial injury repair and fibrosis. These findings unravel a vital role for WSB1 induction in lung injury repair, thus highlighting it as a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Lei Chong
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lihui Zou
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liyan Xiang
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyue Song
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanqi Miao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, and Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Wenzhou, China
| | - Xihua Yan
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Xu
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gongxia Ling
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Elie El Agha
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Institute for Lung Health, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Hailin Zhang
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jin-San Zhang
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, and Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Wenzhou, China.
| |
Collapse
|
3
|
Silva AR, de Souza e Souza KFC, Souza TBD, Younes-Ibrahim M, Burth P, de Castro Faria Neto HC, Gonçalves-de-Albuquerque CF. The Na/K-ATPase role as a signal transducer in lung inflammation. Front Immunol 2024; 14:1287512. [PMID: 38299144 PMCID: PMC10827986 DOI: 10.3389/fimmu.2023.1287512] [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: 09/01/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is marked by damage to the capillary endothelium and alveolar epithelium following edema formation and cell infiltration. Currently, there are no effective treatments for severe ARDS. Pathologies such as sepsis, pneumonia, fat embolism, and severe trauma may cause ARDS with respiratory failure. The primary mechanism of edema clearance is the epithelial cells' Na/K-ATPase (NKA) activity. NKA is an enzyme that maintains the electrochemical gradient and cell homeostasis by transporting Na+ and K+ ions across the cell membrane. Direct injury on alveolar cells or changes in ion transport caused by infections decreases the NKA activity, loosening tight junctions in epithelial cells and causing edema formation. In addition, NKA acts as a receptor triggering signal transduction in response to the binding of cardiac glycosides. The ouabain (a cardiac glycoside) and oleic acid induce lung injury by targeting NKA. Besides enzymatic inhibition, the NKA triggers intracellular signal transduction, fostering proinflammatory cytokines production and contributing to lung injury. Herein, we reviewed and discussed the crucial role of NKA in edema clearance, lung injury, and intracellular signaling pathway activation leading to lung inflammation, thus putting the NKA as a protagonist in lung injury pathology.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Thamires Bandeira De Souza
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Mauricio Younes-Ibrahim
- Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Zhang X, Su L, Pan P. Advances and Applications of Lung Organoids in the Research on Acute Respiratory Distress Syndrome (ARDS). J Clin Med 2024; 13:346. [PMID: 38256480 PMCID: PMC10816077 DOI: 10.3390/jcm13020346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a sudden onset of lung injury characterized by bilateral pulmonary edema, diffuse inflammation, hypoxemia, and a low P/F ratio. Epithelial injury and endothelial injury are notable in the development of ARDS, which is more severe under mechanical stress. This review explains the role of alveolar epithelial cells and endothelial cells under physiological and pathological conditions during the progression of ARDS. Mechanical injury not only causes ARDS but is also a side effect of ventilator-supporting treatment, which is difficult to model both in vitro and in vivo. The development of lung organoids has seen rapid progress in recent years, with numerous promising achievements made. Multiple types of cells and construction strategies are emerging in the lung organoid culture system. Additionally, the lung-on-a-chip system presents a new idea for simulating lung diseases. This review summarizes the basic features and critical problems in the research on ARDS, as well as the progress in lung organoids, particularly in the rapidly developing microfluidic system-based organoids. Overall, this review provides valuable insights into the three major factors that promote the progression of ARDS and how advances in lung organoid technology can be used to further understand ARDS.
Collapse
Affiliation(s)
- Xingwu Zhang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing 100091, China;
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Longxiang Su
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| | - Pan Pan
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing 100091, China;
| |
Collapse
|
5
|
Mishra S, Kalra N, Botlagunta M, Rajasekaran S. MicroRNA-195-5p mediates arsenic-induced cytotoxicity in human lung epithelial cells: Beneficial role of plant-derived tannic acid. Toxicol Appl Pharmacol 2024; 482:116775. [PMID: 38042305 DOI: 10.1016/j.taap.2023.116775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Arsenic (As), a highly toxic metalloid, which causes environmental lung diseases and affects millions of people worldwide. Respiratory epithelial cells are essential for maintaining lung homeostasis, aberrant epithelial damage and death due to exposure to a wide range of environmental pollutants, which are considered to be the initial trigger for many pulmonary diseases. Accumulating evidence has shown that microRNAs (miRNAs) appear to be important players in various normal physiological and pathological processes. Therefore, the present study was carried out to examine the cytotoxic effects of a trivalent form of As (As3+) in normal human bronchial (BEAS-2B) and adenocarcinoma alveolar basal (A549) epithelial cells and the role of miR-195-5p. Further, we also explored the protective effects of a natural dietary polyphenol tannic acid (TA). As3+ (1 μM) treatment in BEAS-2B cells for 24 h induced cytotoxicity by decreasing the cell viability, mitochondrial membrane potential (ΔΨm) and inducing reactive oxygen species (ROS) generation, lipid peroxidation (LPO), cell cycle arrest, and apoptosis, which was associated with a significantly higher level of miR-195-5p expression compared with vehicle control. Forced expression of miR-195-5p alone suppressed cell survival, ΔΨm, regulated cell cycle distribution and induced ROS generation in BEAS-2B cells. As expected, miR-195-5p inhibition effectively rescued BEAS-2B cells from As3+-mediated toxicity, confirming the involvement of miR-195-5p in the cytotoxic effects of As3+. Further, TA pre-treatment expressively alleviated As3+-induced toxicity by suppressing ROS production, miR-195-5p expression, and increasing ΔΨm. These in vitro results indicate that miR-195-5p may be useful as a therapeutic target for treating As3+ toxicity.
Collapse
Affiliation(s)
- Sehal Mishra
- Division of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India; School of Bioengineering, VIT Bhopal University, Kothrikalan, Sehore, Madhya Pradesh, India
| | - Neetu Kalra
- School of Bioengineering, VIT Bhopal University, Kothrikalan, Sehore, Madhya Pradesh, India
| | - Mahendran Botlagunta
- School of Bioengineering, VIT Bhopal University, Kothrikalan, Sehore, Madhya Pradesh, India
| | - Subbiah Rajasekaran
- Division of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
6
|
Chen XY, Kao C, Peng SW, Chang JH, Lee YL, Laiman V, Chung KF, Bhavsar PK, Heriyanto DS, Chuang KJ, Chuang HC. Role of DCLK1/Hippo pathway in type II alveolar epithelial cells differentiation in acute respiratory distress syndrome. Mol Med 2023; 29:159. [PMID: 37996782 PMCID: PMC10668445 DOI: 10.1186/s10020-023-00760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Delay in type II alveolar epithelial cell (AECII) regeneration has been linked to higher mortality in patients with acute respiratory distress syndrome (ARDS). However, the interaction between Doublecortin-like kinase 1 (DCLK1) and the Hippo signaling pathway in ARDS-associated AECII differentiation remains unclear. Therefore, the objective of this study was to understand the role of the DCLK1/Hippo pathway in mediating AECII differentiation in ARDS. MATERIALS AND METHODS AECII MLE-12 cells were exposed to 0, 0.1, or 1 μg/mL of lipopolysaccharide (LPS) for 6 and 12 h. In the mouse model, C57BL/6JNarl mice were intratracheally (i.t.) injected with 0 (control) or 5 mg/kg LPS and were euthanized for lung collection on days 3 and 7. RESULTS We found that LPS induced AECII markers of differentiation by reducing surfactant protein C (SPC) and p53 while increasing T1α (podoplanin) and E-cadherin at 12 h. Concurrently, nuclear YAP dynamic regulation and increased TAZ levels were observed in LPS-exposed AECII within 12 h. Inhibition of YAP consistently decreased cell levels of SPC, claudin 4 (CLDN-4), galectin 3 (LGALS-3), and p53 while increasing transepithelial electrical resistance (TEER) at 6 h. Furthermore, DCLK1 expression was reduced in isolated human AECII of ARDS, consistent with the results in LPS-exposed AECII at 6 h and mouse SPC-positive (SPC+) cells after 3-day LPS exposure. We observed that downregulated DCLK1 increased p-YAP/YAP, while DCLK1 overexpression slightly reduced p-YAP/YAP, indicating an association between DCLK1 and Hippo-YAP pathway. CONCLUSIONS We conclude that DCLK1-mediated Hippo signaling components of YAP/TAZ regulated markers of AECII-to-AECI differentiation in an LPS-induced ARDS model.
Collapse
Affiliation(s)
- Xiao-Yue Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ching Kao
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Syue-Wei Peng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
| | - Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- National Heart and Lung Institute, Imperial College London, London, UK.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Inhalation Toxicology Research Lab (ITRL), School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan.
| |
Collapse
|
7
|
Napodano C, Carnazzo V, Basile V, Pocino K, Stefanile A, Gallucci S, Natali P, Basile U, Marino M. NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases-A Lesson from Cytokine Storm Syndrome. Int J Mol Sci 2023; 24:16556. [PMID: 38068879 PMCID: PMC10706560 DOI: 10.3390/ijms242316556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Inflammation and inflammasomes have been proposed as important regulators of the host-microorganism interaction, playing a key role in morbidity and mortality due to the coronavirus disease 2019 (COVID-19) in subjects with chronic conditions and compromised immune system. The inflammasome consists of a multiprotein complex that finely regulates the activation of caspase-1 and the production and secretion of potent pro-inflammatory cytokines such as IL-1β and IL-18. The pyrin containing NOD (nucleotide-binding oligomerization domain) like receptor (NLRP) is a family of intracellular receptors, sensing patterns associated to pathogens or danger signals and NLRP3 inflammasome is the most deeply analyzed for its involvement in the innate and adaptive immune system as well as its contribution to several autoinflammatory and autoimmune diseases. It is highly expressed in leukocytes and up-regulated in sentinel cells upon inflammatory stimuli. NLRP3 expression has also been reported in B and T lymphocytes, in epithelial cells of oral and genital mucosa, in specific parenchymal cells as cardiomyocytes, and keratinocytes, and chondrocytes. It is well known that a dysregulated activation of the inflammasome is involved in the pathogenesis of different disorders that share the common red line of inflammation in their pathogenetic fingerprint. Here, we review the potential roles of the NLRP3 inflammasome in cardiovascular events, liver damage, pulmonary diseases, and in that wide range of systemic inflammatory syndromes named as a cytokine storm.
Collapse
Affiliation(s)
- Cecilia Napodano
- Department of Laboratory of Medicine and Pathology, S. Agostino Estense Hospital, 41126 Modena, Italy;
| | - Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, AUSL Latina, 04100 Latina, Italy; (V.C.); (U.B.)
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Krizia Pocino
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Annunziata Stefanile
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA;
| | - Patrizia Natali
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, 41124 Modena, Italy;
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, AUSL Latina, 04100 Latina, Italy; (V.C.); (U.B.)
| | - Mariapaola Marino
- Dipartimento di Medicina e Chirurgia Traslazionale, Sezione di Patologia Generale, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
8
|
Breder-Bonk C, Docter D, Barz M, Strieth S, Knauer SK, Gül D, Stauber RH. The Apoptosis Inhibitor Protein Survivin Is a Critical Cytoprotective Resistor against Silica-Based Nanotoxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2546. [PMID: 37764575 PMCID: PMC10535920 DOI: 10.3390/nano13182546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
Exposure to nanoparticles is inevitable as they become widely used in industry, cosmetics, and foods. However, knowledge of their (patho)physiological effects on biological entry routes of the human body and their underlying molecular mechanisms is still fragmented. Here, we examined the molecular effects of amorphous silica nanoparticles (aSiNPs) on cell lines mimicking the alveolar-capillary barrier of the lung. After state-of-the-art characterization of the used aSiNPs and the cell model, we performed cell viability-based assays and a protein analysis to determine the aSiNP-induced cell toxicity and underlying signaling mechanisms. We revealed that aSiNPs induce apoptosis in a dose-, time-, and size-dependent manner. aSiNP-induced toxicity involves the inhibition of pro-survival pathways, such as PI3K/AKT and ERK signaling, correlating with reduced expression of the anti-apoptotic protein Survivin on the protein and transcriptional levels. Furthermore, induced Survivin overexpression mediated resistance against aSiNP-toxicity. Thus, we present the first experimental evidence suggesting Survivin as a critical cytoprotective resistor against silica-based nanotoxicity, which may also play a role in responses to other NPs. Although Survivin's relevance as a biomarker for nanotoxicity needs to be demonstrated in vivo, our data give general impetus to investigate the pharmacological modulation of Survivin`s functions to attenuate the harmful effects of acute or chronic inhalative NP exposure.
Collapse
Affiliation(s)
- Christina Breder-Bonk
- Molecular and Cellular Oncology, University Medical Center Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany; (D.D.); (R.H.S.)
| | - Dominic Docter
- Molecular and Cellular Oncology, University Medical Center Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany; (D.D.); (R.H.S.)
| | - Matthias Barz
- Leiden Academic Center for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands;
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Sebastian Strieth
- Department of Otorhinolaryngology, University Medical Center Bonn, Venusberg-Campus 1, 53127 Bonn, Germany;
| | - Shirley K. Knauer
- Center for Medical Biotechnology (ZMB), Department of Molecular Biology II, University of Duisburg-Essen, Universitätsstrasse 5, 45141 Essen, Germany;
| | - Désirée Gül
- Molecular and Cellular Oncology, University Medical Center Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany; (D.D.); (R.H.S.)
| | - Roland H. Stauber
- Molecular and Cellular Oncology, University Medical Center Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany; (D.D.); (R.H.S.)
| |
Collapse
|
9
|
Zheng N, Liu X, Yang Y, Liu Y, Yan F, Zeng Y, Cheng Y, Wu D, Chen C, Wang X. Regulatory roles of NAT10 in airway epithelial cell function and metabolism in pathological conditions. Cell Biol Toxicol 2023; 39:1237-1256. [PMID: 35877022 DOI: 10.1007/s10565-022-09743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/20/2022] [Indexed: 12/01/2022]
Abstract
N-acetyltransferase 10 (NAT10), a nuclear acetyltransferase and a member of the GNAT family, plays critical roles in RNA stability and translation processes as well as cell proliferation. Little is known about regulatory effects of NAT10 in lung epithelial cell proliferation. We firstly investigated NTA10 mRNA expression in alveolar epithelial types I and II, basal, ciliated, club, and goblet/mucous epithelia from heathy and patients with chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, lung adenocarcinoma, para-tumor tissue, and systemic sclerosis, respectively. We selected A549 cells for representative of alveolar epithelia or H1299 and H460 cells as airway epithelia with different genetic backgrounds and studied dynamic responses of NAT10-down-regulated epithelia to high temperature, lipopolysaccharide, cigarette smoking extract (CSE), drugs, radiation, and phosphoinositide 3-kinase (PI3K) inhibitors at various doses. We also compared transcriptomic profiles between alveolar and airway epithelia, between cells with or without NAT10 down-regulation, between early and late stages, and between challenges. The present study demonstrated that NAT10 expression increased in human lung epithelia and varied among epithelial types, challenges, and diseases. Knockdown of NAT10 altered epithelial mitochondrial functions, dynamic responses to LPS, CSE, or PI3K inhibitors, and transcriptomic phenomes. NAT10 regulates biological phenomes, and behaviors are more complex and are dependent upon multiple signal pathways. Thus, NAT10-associated signal pathways can be a new alternative for understanding the disease and developing new biomarkers and targets.
Collapse
Affiliation(s)
- Nannan Zheng
- Department of Respiratory Medicine, The First Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xuanqi Liu
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China
| | - Ying Yang
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yifei Liu
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Furong Yan
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yiming Zeng
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Yunfeng Cheng
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China
| | - Duojiao Wu
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China.
| | - Chengshui Chen
- Department of Respiratory Medicine, The First Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Quzhou Hospital of Wenzhou Medical University, Quzhou, Zhejiang Province, China.
| | - Xiangdong Wang
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China.
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| |
Collapse
|
10
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Zhang J, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. Stem Cell Res Ther 2023; 14:185. [PMID: 37501095 PMCID: PMC10375781 DOI: 10.1186/s13287-023-03414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). METHODS Wild-type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony-forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. RESULTS A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of re-alveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. CONCLUSIONS Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA.
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
11
|
Ghosh B, Loube J, Thapa S, Ryan H, Capodanno E, Chen D, Swaby C, Chen S, Mahmud S, Girgis M, Nishida K, Ying L, Chengala PP, Tieng E, Burnim M, Wally A, Bhowmik D, Zaykaner M, Yeung-Luk B, Mitzner W, Biswal S, Sidhaye VK. Loss of E-cadherin is causal to pathologic changes in chronic lung disease. Commun Biol 2022; 5:1149. [PMID: 36309587 PMCID: PMC9617938 DOI: 10.1038/s42003-022-04150-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022] Open
Abstract
Epithelial cells line the lung mucosal surface and are the first line of defense against toxic exposures to environmental insults, and their integrity is critical to lung health. An early finding in the lung epithelium of patients with chronic obstructive pulmonary disease (COPD) is the loss of a key component of the adherens junction protein called E-cadherin. The cause of this decrease is not known and could be due to luminal insults or structural changes in the small airways. Irrespective, it is unknown whether the loss of E-cadherin is a marker or a driver of disease. Here we report that loss of E-cadherin is causal to the development of chronic lung disease. Using cell-type-specific promoters, we find that knockout of E-cadherin in alveolar epithelial type II but not type 1 cells in adult mouse models results in airspace enlargement. Furthermore, the knockout of E-cadherin in airway ciliated cells, but not club cells, increase airway hyperreactivity. We demonstrate that strategies to upregulate E-cadherin rescue monolayer integrity and serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Baishakhi Ghosh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jeffrey Loube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shreeti Thapa
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hurley Ryan
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Daniel Chen
- Johns Hopkins University, Baltimore, MD, USA
| | | | - Si Chen
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Saborny Mahmud
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Kristine Nishida
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Linyan Ying
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Respiration, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Pratulya Pragadaraju Chengala
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ethan Tieng
- Johns Hopkins University, Baltimore, MD, USA
| | - Michael Burnim
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ara Wally
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Debarshi Bhowmik
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael Zaykaner
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Bonnie Yeung-Luk
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Venkataramana K Sidhaye
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Ahmed L, Bergot E, Prévot G, Cottin V. Ethnographic survey of patients and caregiver's life journey in idiopathic pulmonary fibrosis. Respir Med Res 2022; 83:100955. [PMID: 36634553 DOI: 10.1016/j.resmer.2022.100955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/30/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disease associated with high mortality. The IPF journey affects patients' and caregivers' quality of life, this should be taken into account as an important parameter for a better disease management. An ethnographic study was conducted between December 2019 and January 2020 to explore social disease representations, patients' and cargivers' experiences in the disease journey and consequences in their daily life, to identify the means of actions permitting a quality of life (QoL) improvement. PATIENTS/CAREGIVERS Twenty respondents, twelve patients aged 43 to 84 years old and eight caregivers in four French regions were met. Eleven patients were diagnosed with IPF between 2013 and 2017 and one patient in 1988 (at 12 years old). The lung function reported by patients ranged as follows: forced vital capacity from 112% to 40% and diffusing capacity of the lungs for carbon monoxide (DLCO) from 66% to <20%. RESULTS The survey included patients diagnosed at least three years ago. Collected data comprised disease representation, patients'/caregivers' experience of the disease, healthcare journey, and consequences for their daily life. The first signs identified by the patient or their caregiver may not have been taken seriously by the primary care physician. The pre-diagnosis period was considered particularly traumatic for most patients. The biopsy performed in 8 cases was experienced as violent by 4/8 patients, some of whom still feel pain. Patients/caregivers knew how to define their disease and spontaneously gave severe representations of the disease such as "Rare, incurable disease", "an organ being destroyed". DISCUSSION This study highlighted patients'/caregivers' common needs at each stage of the disease. The lack of disease knowledge from frontline practitioners (general physicians, community pulmonologists) can lead to significant diagnostic error. Patients require psychological support and more information on daily aspects in disease management, such as food good practices and importance of physical activity, along with information about disease progression. The fear caused by these shortages can be reduced through contact with patients' associations. CONCLUSIONS Numerous essential data were identified and should be considered for supporting actions that could allow to improve the QoL of patients with IPF.
Collapse
Affiliation(s)
| | | | | | - Vincent Cottin
- HCL UCBL - GH Est-Hôpital Louis Pradel, 69677 BRON, France.
| |
Collapse
|
13
|
Quan Y, Yin Z, Chen S, Lang J, Han L, Yi J, Zhang L, Yue Q, Tian W, Chen P, Du S, Wang J, Dai Y, Hua H, Zeng J, Li L, Zhao J. The gut-lung axis: Gut microbiota changes associated with pulmonary fibrosis in mouse models induced by bleomycin. Front Pharmacol 2022; 13:985223. [PMID: 36249808 PMCID: PMC9561135 DOI: 10.3389/fphar.2022.985223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
The main objective of this study was to investigate the alterations in the gut microbiota (GM) of pulmonary fibrosis (PF) mice induced by bleomycin (BLM) with its underlying mechanisms. BLM was docked with the targets of TGF-β/SMAD and caspase-3 pathways using the molecular docking technique. HE staining and Masson staining were applied to observe the histopathological changes in the pulmonary tissues. Detection of the apoptotic signals was conducted by flow cytometry and TUNEL staining. The mRNA expression of targets involved in the TGF-β/SMAD and caspase-3 signaling pathways in lungs was determined by qPCR. Immunohistochemistry (IHC) assay was used to detect the expression levels of cleaved caspase-3 and BAX proteins in mice lung tissues. 16S rDNA sequencing analysis was used to investigate the changes of GM in the fecal samples of mice in each group. The results showed that the apoptosis rate of pulmonary cells in the BLM group distinctly increased, with the expression levels of crucial target pro-apoptotic gene caspase-3, BAX with the corresponding protein, cleaved caspase-3, BAX were apparently elevated. This was accompanied by a significant increase in pro-fibrotic targets level such as TGF-β, fibronectin, collagen I, and collagen III. The mechanisms of PF induced by BLM were related to apoptosis of lung tissue cells such as alveolar epithelial cells and destroyed alveolar structure and excessive production of extracellular matrix (ECM), which may be bound up with activating TGF-β/SMAD and caspase-3 pathways. As for the GM, it was found that, after BLM induced PF in mice, the micro ecological balance of the GM was destroyed; the distance of PCo1 and Pco2 was significantly elongated, and the relative abundance of some intestinal probiotics like Catenibacterium and Lactobacillus (L. johnsonii and L. gasseri) dramatically lowered while the relative abundance of Verrucomicrobiales and Enterobacteriales substantially increased. Therefore, GM changes associated with PF in mouse models induced by BLM and the concept of "gut-lung axis" might provide an optional therapeutic strategy for PF.
Collapse
Affiliation(s)
- Yunyun Quan
- Department of Pharmacognosy, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Shilong Chen
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Jirui Lang
- Department of Pharmacognosy, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Liyang Han
- Department of Pharmacognosy, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jing Yi
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Lu Zhang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhua Yue
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Weiwei Tian
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Ping Chen
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Shenglin Du
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Jianbo Wang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Dai
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Jin Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Junning Zhao
- Department of Pharmacognosy, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Sehgal M, Jakhete SM, Manekar AG, Sasikumar S. Specific epigenetic regulators serve as potential therapeutic targets in idiopathic pulmonary fibrosis. Heliyon 2022; 8:e09773. [PMID: 36061031 PMCID: PMC9434059 DOI: 10.1016/j.heliyon.2022.e09773] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/27/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), a disorder observed mostly in older human beings, is characterised by chronic and progressive lung scarring leading to an irreversible decline in lung function. This health condition has a dismal prognosis and the currently available drugs only delay but fail to reverse the progression of lung damage. Consequently, it becomes imperative to discover improved therapeutic compounds and their cellular targets to cure IPF. In this regard, a number of recent studies have targeted the epigenetic regulation by histone deacetylases (HDACs) to develop and categorise antifibrotic drugs for lungs. Therefore, this review focuses on how aberrant expression or activity of Classes I, II and III HDACs alter TGF-β signalling to promote events such as epithelial-mesenchymal transition, differentiation of activated fibroblasts into myofibroblasts, and excess deposition of the extracellular matrix to propel lung fibrosis. Further, this study describes how certain chemical compounds or dietary changes modulate dysregulated HDACs to attenuate five faulty TGF-β-dependent profibrotic processes, both in animal models and cell lines replicating IPF, thereby identifying promising means to treat this lung disorder.
Collapse
Affiliation(s)
- Manas Sehgal
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Sharayu Manish Jakhete
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Amruta Ganesh Manekar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Satish Sasikumar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| |
Collapse
|
15
|
Nguyen N, Xu S, Lam TYW, Liao W, Wong WSF, Ge R. ISM1 suppresses LPS-induced acute lung injury and post-injury lung fibrosis in mice. Mol Med 2022; 28:72. [PMID: 35752760 PMCID: PMC9233842 DOI: 10.1186/s10020-022-00500-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/14/2022] [Indexed: 11/15/2022] Open
Abstract
Background Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are clinical syndromes characterized by acute lung inflammation, pulmonary edema and hypoxemia, with up to 50% mortality rate without effective pharmacological therapy. Following the acute inflammation, repair and remodeling occurs which in some cases resulting in lung fibrosis. The pathophysiology of ALI/ARDS remains incompletely understood. Lipopolysaccharide (LPS)-induced ALI in mice have been widely used as a model to study human ALI/ARDS. Isthmin 1 (ISM1) is a secreted protein highly abundant in mouse lung. We have previously reported that upon intratracheal LPS instillation, ISM1 expression in the lung is further upregulated. Recently, we also reported that ISM1 is an anti-inflammatory protein in the lung with Ism1-/- mice presenting spontaneous chronic low-grade lung inflammation and obvious emphysema at young adult stage. However, what role ISM1 plays in ALI/ARDS and lung fibrosis remain unclear. Methods Using Ism1-/- mice and intratracheal LPS-induced ALI, and local delivery of recombinant ISM1 (rISM1), we investigated the role ISM1 plays in ALI and post-ALI lung fibrosis using flow cytometry, Western blot, antibody array, immunohistochemistry (IHC), immunofluorescent and other histological staining. Results We reveal that ISM1 deficiency in mice led to an intensified acute lung inflammation upon intratracheal LPS challenge, with a heightened leukocyte infiltration including neutrophils and monocyte-derived alveolar macrophages, as well as upregulation of multiple pro-inflammatory cytokines/chemokines including tumor necrosis factor α (TNF-α). Although innate immune cells largely subsided to the baseline by day 7 post-LPS challenge in both wild-type and Ism1−/− mice, Ism1−/− lung showed increased post-ALI fibrosis from day 9 post-LPS treatment with increased myofibroblasts, excessive collagen accumulation and TGF-β upregulation. The heightened lung fibrosis remained on day 28 post-LPS. Moreover, intranasal delivered recombinant ISM1 (rISM1) effectively suppressed LPS-induced acute lung inflammation and ALI, and rISM1 suppressed LPS-induced NF-κB activation in cultured mouse alveolar macrophages. Conclusion Together with our previous report, this work further established ISM1 as an endogenous anti-inflammation protein in the lung, restraining excessive host inflammatory response to LPS-triggered ALI and suppressing post-ALI lung fibrosis likely through suppressing NF-κB activation and pro-inflammatory cytokine/chemokine production. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00500-w.
Collapse
Affiliation(s)
- Ngan Nguyen
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Simin Xu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Terence Yin Weng Lam
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Republic of Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore, 138602, Republic of Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Republic of Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore, 138602, Republic of Singapore.,Drug Discovery and Optimization Platform, National University Health System, Singapore, 117600, Republic of Singapore
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore.
| |
Collapse
|
16
|
An Update on Advancements and Challenges in Inhalational Drug Delivery for Pulmonary Arterial Hypertension. Molecules 2022; 27:molecules27113490. [PMID: 35684428 PMCID: PMC9182169 DOI: 10.3390/molecules27113490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 12/17/2022] Open
Abstract
A lethal condition at the arterial–alveolar juncture caused the exhaustive remodeling of pulmonary arterioles and persistent vasoconstriction, followed by a cumulative augmentation of resistance at the pulmonary vascular and, consequently, right-heart collapse. The selective dilation of the pulmonary endothelium and remodeled vasculature can be achieved by using targeted drug delivery in PAH. Although 12 therapeutics were approved by the FDA for PAH, because of traditional non-specific targeting, they suffered from inconsistent drug release. Despite available inhalation delivery platforms, drug particle deposition into the microenvironment of the pulmonary vasculature and the consequent efficacy of molecules are influenced by pathophysiological conditions, the characteristics of aerosolized mist, and formulations. Uncertainty exists in peripheral hemodynamics outside the pulmonary vasculature and extra-pulmonary side effects, which may be further exacerbated by underlying disease states. The speedy improvement of arterial pressure is possible via the inhalation route because it has direct access to pulmonary arterioles. Additionally, closed particle deposition and accumulation in diseased tissues benefit the restoration of remolded arterioles by reducing fallacious drug deposition in other organs. This review is designed to decipher the pathological changes that should be taken into account when targeting the underlying pulmonary endothelial vasculature, especially with regard to inhaled particle deposition in the alveolar vasculature and characteristic formulations.
Collapse
|
17
|
Evaluation of Proteasome Inhibitors in the Treatment of Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11091543. [PMID: 35563849 PMCID: PMC9099509 DOI: 10.3390/cells11091543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common form of idiopathic interstitial pneumonia, and it has a worse prognosis than non-small cell lung cancer. The pathomechanism of IPF is not fully understood, but it has been suggested that repeated microinjuries of epithelial cells induce a wound healing response, during which fibroblasts differentiate into myofibroblasts. These activated myofibroblasts express α smooth muscle actin and release extracellular matrix to promote matrix deposition and tissue remodeling. Under physiological conditions, the remodeling process stops once wound healing is complete. However, in the lungs of IPF patients, myofibroblasts re-main active and deposit excess extracellular matrix. This leads to the destruction of alveolar tissue, the loss of lung elastic recoil, and a rapid decrease in lung function. Some evidence has indicated that proteasomal inhibition combats fibrosis by inhibiting the expressions of extracellular matrix proteins and metalloproteinases. However, the mechanisms by which proteasome inhibitors may protect against fibrosis are not known. This review summarizes the current research on proteasome inhibitors for pulmonary fibrosis, and provides a reference for whether proteasome inhibitors have the potential to become new drugs for the treatment of pulmonary fibrosis.
Collapse
|
18
|
Wyatt KD, Sarr D, Sakamoto K, Watford WT. Influenza-induced Tpl2 expression within alveolar epithelial cells is dispensable for host viral control and anti-viral immunity. PLoS One 2022; 17:e0262832. [PMID: 35051238 PMCID: PMC8775564 DOI: 10.1371/journal.pone.0262832] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/05/2022] [Indexed: 01/22/2023] Open
Abstract
Tumor progression locus 2 (Tpl2) is a serine/threonine kinase that regulates the expression of inflammatory mediators in response to Toll-like receptors (TLR) and cytokine receptors. Global ablation of Tpl2 leads to severe disease in response to influenza A virus (IAV) infection, characterized by respiratory distress, and studies in bone marrow chimeric mice implicated Tpl2 in non-hematopoietic cells. Lung epithelial cells are primary targets and replicative niches of influenza viruses; however, the specific regulation of antiviral responses by Tpl2 within lung epithelial cells has not been investigated. Herein, we show that Tpl2 is basally expressed in primary airway epithelial cells and that its expression increases in both type I and type II airway epithelial cells (AECI and AECII) in response to influenza infection. We used Nkx2.1-cre to drive Tpl2 deletion within pulmonary epithelial cells to delineate epithelial cell-specific functions of Tpl2 during influenza infection in mice. Although modest increases in morbidity and mortality were attributed to cre-dependent deletion in lung epithelial cells, no alterations in host cytokine production or lung pathology were observed. In vitro, Tpl2 inhibition within the type I airway epithelial cell line, LET1, as well as genetic ablation in primary airway epithelial cells did not alter cytokine production. Overall, these findings establish that Tpl2-dependent defects in cells other than AECs are primarily responsible for the morbidity and mortality seen in influenza-infected mice with global Tpl2 ablation.
Collapse
Affiliation(s)
- Kara D. Wyatt
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Demba Sarr
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Kaori Sakamoto
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Wendy T. Watford
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Respiratory flora: The potential biomarker of radiation-induced pulmonary injury. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Jiang J, Wang J, Li C, Mo L, Huang D. Hyperoxia induces alveolar epithelial cell apoptosis by regulating mitochondrial function through small mothers against decapentaplegic 3 (SMAD3) and extracellular signal-regulated kinase 1/2 (ERK1/2). Bioengineered 2021; 13:242-252. [PMID: 34898379 PMCID: PMC8805928 DOI: 10.1080/21655979.2021.2012953] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Oxygen therapy and mechanical ventilation are widely used to treat and manage neonatal emergencies in critically ill newborns. However, they are often associated with adverse effects and result in conditions such as chronic lung disease and bronchopulmonary dysplasia. Hence, aclear understanding of the mechanisms underlying hyperoxia-induced lung damage is crucial in order to mitigate the side effects of oxygen-based therapy. Here, we have established an in vitro model of hyperoxia-induced lung damage in type II alveolar epithelial cells (AECIIs) and delineated the molecular basis of oxygen therapy-induced impaired alveolar development. Thus, AECIIs were exposed to a hyperoxic environment and their cell viability, cell cycle progression, apoptosis, mitochondrial integrity and dynamics, and energy metabolism were assessed. The results showed that hyperoxia has no significant effect as an inhibitor of SMAD3 and ERK1/2 in AECIIs, but leads to significant inhibition of cell viability. Further, hyperoxia was found to promote AECII apoptosis and mitochondrial, whereas chemical inhibition of SMAD3 or ERK1/2 further exacerbated the detrimental effects of hyperoxia in AECIIs. Overall, these findings presented herein demonstrate the critical role of SMAD/ERK signaling in the regulation of AECII behavior in varying oxygen environments. Thus, this study offers novel insights for the prevention of neonatal lung dysfunction in premature infants.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Pediatric Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Juan Wang
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding, China
| | - Cen Li
- Department of Pediatric Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lianqin Mo
- Department of Pediatric Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dong Huang
- Department of Pediatric Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
21
|
Cardoso AM, Silvério MNO, de Oliveira Maciel SFV. Purinergic signaling as a new mechanism underlying physical exercise benefits: a narrative review. Purinergic Signal 2021; 17:649-679. [PMID: 34590239 PMCID: PMC8677870 DOI: 10.1007/s11302-021-09816-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/21/2021] [Indexed: 11/27/2022] Open
Abstract
In the last years, it has become evident that both acute and chronic physical exercise trigger responses/adaptations in the purinergic signaling and these adaptations can be considered one important mechanism related to the exercise benefits for health improvement. Purinergic system is composed of enzymes (ectonucleotidases), receptors (P1 and P2 families), and molecules (ATP, ADP, adenosine) that are able to activate these receptors. These components are widely distributed in almost all cell types, and they respond/act in a specific manner depending on the exercise types and/or intensities as well as the cell type (organ/tissue analyzed). For example, while acute intense exercise can be associated with tissue damage, inflammation, and platelet aggregation, chronic exercise exerts anti-inflammatory and anti-aggregant effects, promoting health and/or treating diseases. All of these effects are dependent on the purinergic signaling. Thus, this review was designed to cover the aspects related to the relationship between physical exercise and purinergic signaling, with emphasis on the modulation of ectonucleotidases and receptors. Here, we discuss the impact of different exercise protocols as well as the differences between acute and chronic effects of exercise on the extracellular signaling exerted by purinergic system components. We also reinforce the concept that purinergic signaling must be understood/considered as a mechanism by which exercise exerts its effects.
Collapse
Affiliation(s)
- Andréia Machado Cardoso
- Graduate Program in Biomedical Sciences and Medicine Course, Federal University of Fronteira Sul - UFFS, Campus Chapecó, Rodovia SC 484 - Km 02, Fronteira Sul, 89815-899, Brazil.
- Graduate Program in Physical Education, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil.
| | - Mauro Nicollas Oliveira Silvério
- Medicine Course, Federal University of Fronteira Sul - UFFS, Campus Chapecó, Rodovia SC 484 - Km 02, Fronteira Sul, 89815-899, Brazil
| | - Sarah Franco Vieira de Oliveira Maciel
- Graduate Program in Biomedical Sciences and Medicine Course, Federal University of Fronteira Sul - UFFS, Campus Chapecó, Rodovia SC 484 - Km 02, Fronteira Sul, 89815-899, Brazil
| |
Collapse
|
22
|
Gupta D, Kumar A, Mandloi A, Shenoy V. Renin angiotensin aldosterone system in pulmonary fibrosis: Pathogenesis to therapeutic possibilities. Pharmacol Res 2021; 174:105924. [PMID: 34607005 DOI: 10.1016/j.phrs.2021.105924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
Pulmonary fibrosis is a devastating lung disease with multifactorial etiology characterized by alveolar injury, fibroblast proliferation and excessive deposition of extracellular matrix proteins, which progressively results in respiratory failure and death. Accumulating evidence from experimental and clinical studies supports a central role of the renin angiotensin aldosterone system (RAAS) in the pathogenesis and progression of idiopathic pulmonary fibrosis. Angiotensin II (Ang II), a key vasoactive peptide of the RAAS mediates pro-inflammatory and pro-fibrotic effects on the lungs, adversely affecting organ function. Recent years have witnessed seminal discoveries in the field of RAAS. Identification of new enzymes, peptides and receptors has led to the development of several novel concepts. Of particular interest is the establishment of a protective axis of the RAAS comprising of Angiotensin converting enzyme 2 (ACE2), Angiotensin-(1-7) [Ang-(1-7)], and the Mas receptor (the ACE2/Ang-(1-7)/Mas axis), and the discovery of a functional role for the Angiotensin type 2 (AT2) receptor. Herein, we will review our current understanding of the role of RAAS in lung fibrogenesis, provide evidence on the anti-fibrotic actions of the newly recognized RAAS components (the ACE2/Ang-(1-7)/Mas axis and AT2 receptor), discuss potential strategies and translational efforts to convert this new knowledge into effective therapeutics for PF.
Collapse
Affiliation(s)
- Dipankar Gupta
- Congenital Heart Center, Department of Pediatrics, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Ashok Kumar
- Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS, USA
| | - Avinash Mandloi
- College of Pharmacy, VNS Group of Institutions, Bhopal, India
| | - Vinayak Shenoy
- College of Pharmacy, California Health Sciences University, Clovis, CA, USA.
| |
Collapse
|
23
|
Zeng X, Liu F, Liu K, Xin J, Chen J. HMGB1 could restrict 1,3-β-glucan induced mice lung inflammation by affecting Beclin1 and Bcl2 interaction and promoting the autophagy of epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 222:112460. [PMID: 34243113 DOI: 10.1016/j.ecoenv.2021.112460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Fungi were microorganisms that are ubiquitous in a variety of environments. Inhalation of fungi-contaminated organic dust led to hypersensitivity pneumonitis and might eventually cause irreversible pulmonary fibrosis. Studies showed that maintaining the homeostasis of epithelial cells was vital for defending the exogenous fungi invasion. HMGB1-dependent autophagy played a critical role in maintaining cell homeostasis in multiple inflammatory diseases. However, the actual role of HMGB1-dependent autophagy in hypersensitivity pneumonitis was unclear. In our study, mice were exposed to 0.3 mg/50 μL 1,3-β-glucan solution by intratracheal instillation to set up the lung inflammation model. To investigate the role of HMGB1-dependent autophagy in 1,3-β-glucan induced lung inflammation, AAV-sh-HMGB1 was intratracheally injected to silence HMGB1 in the lung. Our finding suggested that silencing HMGB1 could aggravate the 1,3-β-glucan induced lung inflammation by inhibiting the autophagy of epithelial cells. And ubiquitination of Beclin1 contributed to decreasing the interaction of Beclin1 and Bcl2, which might be a key regulatory mechanism of HMGB1 on 1,3-β-glucan induced autophagy.
Collapse
Affiliation(s)
- Xinning Zeng
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Kaiyue Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Jiaxuan Xin
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China.
| |
Collapse
|
24
|
Bluhmki T, Traub S, Müller AK, Bitzer S, Schruf E, Bammert MT, Leist M, Gantner F, Garnett JP, Heilker R. Functional human iPSC-derived alveolar-like cells cultured in a miniaturized 96‑Transwell air-liquid interface model. Sci Rep 2021; 11:17028. [PMID: 34426605 PMCID: PMC8382767 DOI: 10.1038/s41598-021-96565-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
In order to circumvent the limited access and donor variability of human primary alveolar cells, directed differentiation of human pluripotent stem cells (hiPSCs) into alveolar-like cells, provides a promising tool for respiratory disease modeling and drug discovery assays. In this work, a unique, miniaturized 96-Transwell microplate system is described where hiPSC-derived alveolar-like cells were cultured at an air-liquid interface (ALI). To this end, hiPSCs were differentiated into lung epithelial progenitor cells (LPCs) and subsequently matured into a functional alveolar type 2 (AT2)-like epithelium with monolayer-like morphology. AT2-like cells cultured at the physiological ALI conditions displayed characteristics of AT2 cells with classical alveolar surfactant protein expressions and lamellar-body like structures. The integrity of the epithelial barriers between the AT2-like cells was confirmed by applying a custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements. In order to generate an IPF disease-like phenotype in vitro, the functional AT2-like cells were stimulated with cytokines and growth factors present in the alveolar tissue of IPF patients. The cytokines stimulated the secretion of pro-fibrotic biomarker proteins both on the mRNA (messenger ribonucleic acid) and protein level. Thus, the hiPSC-derived and cellular model system enables the recapitulation of certain IPF hallmarks, while paving the route towards a miniaturized medium throughput approach of pharmaceutical drug discovery.
Collapse
Affiliation(s)
- Teresa Bluhmki
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany.
| | - Stefanie Traub
- Trenzyme GmbH, Byk-Gulden-Str. 2, 78467, Constance, Germany
| | | | - Sarah Bitzer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Eva Schruf
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marie-Therese Bammert
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marcel Leist
- In-vitro Toxicology and Biomedicine, University of Konstanz, 78457, Constance, Germany
| | - Florian Gantner
- Department of Translational Medicine and Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, 88397, Biberach an der Riss, Germany
| | - James P Garnett
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Ralf Heilker
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| |
Collapse
|
25
|
Oglesby IK, Schweikert A, Fox B, Redmond C, Donnelly SC, Hurley K. Lung organoids and other preclinical models of pulmonary fibrosis. QJM 2021; 114:167-173. [PMID: 33484260 DOI: 10.1093/qjmed/hcaa281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 11/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal disease affecting over 100 000 people in Europe with an increasing incidence. Available treatments offer only slowing of disease progression and are poorly tolerated by patients leading to cessation of therapy. Lung transplant remains the only cure. Therefore, alternative treatments are urgently required. The pathology of IPF is complex and poorly understood and thus creates a major obstacle to the discovery of novel treatments. Additionally, preclinical assessment of new treatments currently relies upon animal models where disparities with human lung biology often hamper drug development. At a cellular level, IPF is characterized by persistent and abnormal deposition of extracellular matrix by fibroblasts and alveolar epithelial cell injury which is seen as a key event in initiation of disease progression. In-depth investigation of the role of alveolar epithelial cells in health and disease has been impeded due to difficulties in primary cell isolation and culture ex vivo. Novel strategies employing patient-derived induced pluripotent stem cells engineered to produce type 2 alveolar epithelial cells (iAEC2) cultured as three-dimensional organoids have the potential to overcome these hurdles and inform new effective precision treatments for IPF leading to improved survival and quality of life for patients worldwide.
Collapse
Affiliation(s)
- I K Oglesby
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, D02 H903, Ireland
| | - A Schweikert
- Interfaculty Institute of Biochemistry, Eberhard Karls Universität Tübingen, Geschwister-Scholl-Platz 72074 Tübingen, Germany
| | - B Fox
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
| | - C Redmond
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
| | - S C Donnelly
- Department of Respiratory & Interstitial Lung Disease, Tallaght University Hospital Tallaght, Dublin D24 NR0A, Ireland
- School of Medicine, Trinity College Dublin, The University of Dublin, College Green, Dublin D02 PN40, Ireland
| | - K Hurley
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, D02 H903, Ireland
| |
Collapse
|
26
|
Kiener M, Roldan N, Machahua C, Sengupta A, Geiser T, Guenat OT, Funke-Chambour M, Hobi N, Kruithof-de Julio M. Human-Based Advanced in vitro Approaches to Investigate Lung Fibrosis and Pulmonary Effects of COVID-19. Front Med (Lausanne) 2021; 8:644678. [PMID: 34026781 PMCID: PMC8139419 DOI: 10.3389/fmed.2021.644678] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused considerable socio-economic burden, which fueled the development of treatment strategies and vaccines at an unprecedented speed. However, our knowledge on disease recovery is sparse and concerns about long-term pulmonary impairments are increasing. Causing a broad spectrum of symptoms, COVID-19 can manifest as acute respiratory distress syndrome (ARDS) in the most severely affected patients. Notably, pulmonary infection with Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), the causing agent of COVID-19, induces diffuse alveolar damage (DAD) followed by fibrotic remodeling and persistent reduced oxygenation in some patients. It is currently not known whether tissue scaring fully resolves or progresses to interstitial pulmonary fibrosis. The most aggressive form of pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF). IPF is a fatal disease that progressively destroys alveolar architecture by uncontrolled fibroblast proliferation and the deposition of collagen and extracellular matrix (ECM) proteins. It is assumed that micro-injuries to the alveolar epithelium may be induced by inhalation of micro-particles, pathophysiological mechanical stress or viral infections, which can result in abnormal wound healing response. However, the exact underlying causes and molecular mechanisms of lung fibrosis are poorly understood due to the limited availability of clinically relevant models. Recently, the emergence of SARS-CoV-2 with the urgent need to investigate its pathogenesis and address drug options, has led to the broad application of in vivo and in vitro models to study lung diseases. In particular, advanced in vitro models including precision-cut lung slices (PCLS), lung organoids, 3D in vitro tissues and lung-on-chip (LOC) models have been successfully employed for drug screens. In order to gain a deeper understanding of SARS-CoV-2 infection and ultimately alveolar tissue regeneration, it will be crucial to optimize the available models for SARS-CoV-2 infection in multicellular systems that recapitulate tissue regeneration and fibrotic remodeling. Current evidence for SARS-CoV-2 mediated pulmonary fibrosis and a selection of classical and novel lung models will be discussed in this review.
Collapse
Affiliation(s)
- Mirjam Kiener
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Nuria Roldan
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Carlos Machahua
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Olivier Thierry Guenat
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nina Hobi
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
- Organoid Core, Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Sun X, Zhang X, Ren X, Sun H, Wu L, Wang C, Ye X, York P, Gao Z, Jiang H, Zhang J, Yin X. Multiscale Co-reconstruction of Lung Architectures and Inhalable Materials Spatial Distribution. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003941. [PMID: 33898181 PMCID: PMC8061354 DOI: 10.1002/advs.202003941] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/09/2020] [Indexed: 06/12/2023]
Abstract
The effective pulmonary deposition of inhaled particulate carriers loaded with drugs is a prerequisite for therapeutic effects of drug delivery via inhalation route. Revealing the sophisticated lung scaffold and intrapulmonary distribution of particles at three-dimensional (3D), in-situ, and single-particle level remains a fundamental and critical challenge for dry powder inhalation in pre-clinical research. Here, taking advantage of the micro optical sectioning tomography system, the high-precision cross-scale visualization of entire lung anatomy is obtained. Then, co-localized lung-wide datasets of both cyto-architectures and fluorescent particles are collected at full scale with the resolution down to individual particles. The precise spatial distribution pattern reveals the region-specific distribution and structure-associated deposition of the inhalable particles in lungs, which is undetected by previous methods. Overall, this research delivers comprehensive and high-resolution 3D detection of pulmonary drug delivery vectors and provides a novel strategy to evaluate materials distribution for drug delivery.
Collapse
Affiliation(s)
- Xian Sun
- Center for MOST and Image Fusion AnalysisShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xiaochuan Zhang
- School of PharmacyEast China University of Science and TechnologyShanghai200237China
- CAS Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Xiaohong Ren
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
| | - Hongyu Sun
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
| | - Li Wu
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
| | - Caifen Wang
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
| | - Xiaohui Ye
- School of Information Science and TechnologyUniversity of Science and Technology of ChinaHefei230027China
- Shanghai Institute for Advanced Immunochemical StudiesSchool of Life Science and TechnologyShanghaiTech UniversityShanghai200031China
| | - Peter York
- School of PharmacyUniversity of BradfordBradfordBD71DPUK
| | - Zhaobing Gao
- CAS Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Hualiang Jiang
- School of PharmacyEast China University of Science and TechnologyShanghai200237China
- CAS Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Information Science and TechnologyUniversity of Science and Technology of ChinaHefei230027China
- Shanghai Institute for Advanced Immunochemical StudiesSchool of Life Science and TechnologyShanghaiTech UniversityShanghai200031China
| | - Jiwen Zhang
- Center for Drug Delivery SystemsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
- University of Chinese Academy of SciencesBeijing100049China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical ExcipientsNational Institutes for Food and Drug ControlBeijing100050China
| | - Xianzhen Yin
- Center for MOST and Image Fusion AnalysisShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201210China
- CAS Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
28
|
Zhang ZQ, Tian HT, Liu H, Xie R. The role of macrophage-derived TGF-β1 on SiO 2-induced pulmonary fibrosis: A review. Toxicol Ind Health 2021; 37:240-250. [PMID: 33588701 DOI: 10.1177/0748233721989896] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Silicosis is an occupational fibrotic lung disease caused by inhaling large amounts of crystalline silica dust. Transforming growth factor-β1 (TGF-β1), which is secreted from macrophages, has an important role in the development of this disease. Macrophages can recognize and capture silicon dust, undergo M2 polarization, synthesize TGF-β1 precursors, and secrete them out of the cell where they are activated. Activated TGF-β1 induces cells from different sources, transforming them into myofibroblasts through autocrine and paracrine mechanisms, ultimately causing silicosis. These processes involve complex molecular events, which are not yet fully understood. This systematic summary may further elucidate the location and development of pulmonary fibrosis in the formation of silicosis. In this review, we discussed the proposed cellular and molecular mechanisms of production, secretion, activation of TGF-β1, as well as the mechanisms through which TGF-β1 induces cells from three different sources into myofibroblasts during the pathogenesis of silicosis. This study furthers the medical understanding of the pathogenesis and theoretical basis for diagnosing silicosis, thereby promoting silicosis prevention and treatment.
Collapse
Affiliation(s)
- Zhao-Qiang Zhang
- Department of Public Health, 74496Jining Medical University, Jining, China
| | - Hai-Tao Tian
- Department of Public Health, 74496Jining Medical University, Jining, China.,Jining No. 1 People's Hospital, Jining, China
| | - Hu Liu
- Department of Public Health, 74496Jining Medical University, Jining, China
| | - Ruining Xie
- Department of Public Health, 74496Jining Medical University, Jining, China
| |
Collapse
|
29
|
Tharayil A, Rajakumari R, Chirayil CJ, Thomas S, Kalarikkal N. A short review on nanotechnology interventions against COVID-19. EMERGENT MATERIALS 2021; 4:131-141. [PMID: 33554045 PMCID: PMC7856851 DOI: 10.1007/s42247-021-00163-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 05/02/2023]
Abstract
The COVID-19 has affected all major aspects of the society in a global perspective. The role of nanotechnology is much sought after in fighting this pandemic. Advanced materials based on nanotechnology are the basis of several technologies starting from masks and personal protection equipment to specific diagnostic tools that could diminish the impact of COVID-19. Development of nanotechnology-based products is therefore an absolute necessity for fight against COVID-19. We examine the fundamental concepts related to virology, histopathologic findings and how nanotechnology can help in fighting the disease. In this review we discuss the state of the art and ongoing nanotechnology-based strategies like antiviral coatings, 3D printing and therapeutics to fight against this deadly disease. The importance of using nanoparticles in point of care tests and biosensors is also highlighted.
Collapse
Affiliation(s)
- Abhimanyu Tharayil
- School of Energy Materials, Mahatma Gandhi University, Kottayam, Kerala 686560 India
| | - R. Rajakumari
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560 India
| | | | - Sabu Thomas
- School of Energy Materials, Mahatma Gandhi University, Kottayam, Kerala 686560 India
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560 India
- School of Chemical Sciences, Mahatma Gandhi University, Kottayam, Kerala 686560 India
| | - Nandakumar Kalarikkal
- International and Inter-University Centre for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560 India
- School of Pure and Applied Physics, Mahatma Gandhi University, Kottayam, Kerala 686560 India
| |
Collapse
|
30
|
Jarzebska N, Karetnikova ES, Markov AG, Kasper M, Rodionov RN, Spieth PM. Scarred Lung. An Update on Radiation-Induced Pulmonary Fibrosis. Front Med (Lausanne) 2021; 7:585756. [PMID: 33521012 PMCID: PMC7843914 DOI: 10.3389/fmed.2020.585756] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Radiation-induced pulmonary fibrosis is a common severe long-time complication of radiation therapy for tumors of the thorax. Current therapeutic options used in the clinic include only supportive managements strategies, such as anti-inflammatory treatment using steroids, their efficacy, however, is far from being satisfactory. Recent studies have demonstrated that the development of lung fibrosis is a dynamic and complex process, involving the release of reactive oxygen species, activation of Toll-like receptors, recruitment of inflammatory cells, excessive production of nitric oxide and production of collagen by activated myofibroblasts. In this review we summarized the current state of knowledge on the pathophysiological processes leading to the development of lung fibrosis and we also discussed the possible treatment options.
Collapse
Affiliation(s)
- Natalia Jarzebska
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | | | - Alexander G. Markov
- Department of General Physiology, Saint-Petersburg State University, Saint Petersburg, Russia
| | - Michael Kasper
- Institute of Anatomy, Technische Universität Dresden, Dresden, Germany
| | - Roman N. Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Peter M. Spieth
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
31
|
Khan T, Dasgupta S, Ghosh N, Chaudhury K. Proteomics in idiopathic pulmonary fibrosis: the quest for biomarkers. Mol Omics 2021; 17:43-58. [PMID: 33073811 DOI: 10.1039/d0mo00108b] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating chronic progressive and fibrosing lung disease that culminates in the destruction of alveolar integrity and dismal prognosis. Its etiology is unknown and pathophysiology remains unclear. While great advances have been made in elucidating the pathogenesis mechanism, considerable gaps related to information on pathogenetic pathways and key protein targets involved in the clinical course of the disease exist. These issues need to be addressed for better clinical management of this highly challenging disease. Omics approach has revolutionized the entire area of disease understanding and holds promise in its translation to clinical biomarker discovery. This review outlines the contribution of proteomics towards identification of important biomarkers in IPF in terms of their clinical utility, i.e. prognosis, differential diagnosis, disease progression and treatment monitoring. The major dysregulated pathways associated with IPF are also discussed. Based on numerous proteomics studies on human and animal models, it is proposed that IPF pathogenesis involves complex interactions of several pathways such as oxidative stress, endoplasmic reticulum stress, unfolded protein response, coagulation system, inflammation, abnormal wounding, fibroblast proliferation, fibrogenesis and deposition of extracellular matrix. These pathways and their key path-changing mediators need further validation in large well-planned multi-centric trials at various geographical locations for successful development of clinical biomarkers of this confounding disease.
Collapse
Affiliation(s)
- Tila Khan
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| |
Collapse
|
32
|
Wang J, Zhao X, Feng W, Li Y, Peng C. Inhibiting TGF-[Formula: see text] 1-Mediated Cellular Processes as an Effective Strategy for the Treatment of Pulmonary Fibrosis with Chinese Herbal Medicines. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1965-1999. [PMID: 34961416 DOI: 10.1142/s0192415x21500932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic and irreversible interstitial lung disease that even threatens the lives of some patients infected with COVID-19. PF is a multicellular pathological process, including the initial injuries of epithelial cells, recruitment of inflammatory cells, epithelial-mesenchymal transition, activation and differentiation of fibroblasts, etc. TGF-[Formula: see text]1 acts as a key effect factor that participates in these cellular processes of PF. Recently, much attention was paid to inhibiting TGF-[Formula: see text]1 mediated cell processes in the treatment of PF with Chinese herbal medicines (CHM), an important part of traditional Chinese medicine. Here, this review first summarized the effects of TGF-[Formula: see text]1 in different cellular processes of PF. Then, this review summarized the recent research on CHM (compounds, multi-components, single medicines and prescriptions) to directly and/or indirectly inhibit TGF-[Formula: see text]1 signaling (TLRs, PPARs, micrRNA, etc.) in PF. Most of the research focused on CHM natural compounds, including but not limited to alkaloids, flavonoids, phenols and terpenes. After review, the research perspectives of CHM on TGF-[Formula: see text]1 inhibition in PF were further discussed. This review hopes that revealing the inhibiting effects of CHM on TGF-[Formula: see text]1-mediated cellular processes of PF can promote CHM to be better understood and utilized, thus transforming the therapeutic activities of CHM into practice.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xingtao Zhao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Wuwen Feng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Yunxia Li
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| |
Collapse
|
33
|
Miao H, Wu XQ, Zhang DD, Wang YN, Guo Y, Li P, Xiong Q, Zhao YY. Deciphering the cellular mechanisms underlying fibrosis-associated diseases and therapeutic avenues. Pharmacol Res 2020; 163:105316. [PMID: 33248198 DOI: 10.1016/j.phrs.2020.105316] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Fibrosis is the excessive deposition of extracellular matrix components, which results in disruption of tissue architecture and loss of organ function. Fibrosis leads to high morbidity and mortality worldwide, mainly due to the lack of effective therapeutic strategies against fibrosis. It is generally accepted that fibrosis occurs during an aberrant wound healing process and shares a common pathogenesis across different organs such as the heart, liver, kidney, and lung. A better understanding of the fibrosis-related cellular and molecular mechanisms will be helpful for development of targeted drug therapies. Extensive studies revealed that numerous mediators contributed to fibrogenesis, suggesting that targeting these mediators may be an effective therapeutic strategy for antifibrosis. In this review, we describe a number of mediators involved in tissue fibrosis, including aryl hydrocarbon receptor, Yes-associated protein, cannabinoid receptors, angiopoietin-like protein 2, high mobility group box 1, angiotensin-converting enzyme 2, sphingosine 1-phosphate receptor-1, SH2 domain-containing phosphatase-2, and long non-coding RNAs, with the goal that drugs targeting these important mediators might exhibit a beneficial effect on antifibrosis. In addition, these mediators show profibrotic effects on multiple tissues, suggesting that targeting these mediators will exert antifibrotic effects on different organs. Furthermore, we present a variety of compounds that exhibit therapeutic effects against fibrosis. This review suggests therapeutic avenues for targeting organ fibrosis and concurrently identifies challenges and opportunities for designing new therapeutic strategies against fibrosis.
Collapse
Affiliation(s)
- Hua Miao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Dan-Dan Zhang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Yan-Ni Wang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, 1700 Lomas Blvd NE, Albuquerque, 87131, USA
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, Department of Nephrology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, 223003, Jiangsu, China.
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
34
|
Rai M, Bonde S, Yadav A, Plekhanova Y, Reshetilov A, Gupta I, Golińska P, Pandit R, Ingle AP. Nanotechnology-based promising strategies for the management of COVID-19: current development and constraints. Expert Rev Anti Infect Ther 2020; 20:1299-1308. [PMID: 33164589 DOI: 10.1080/14787210.2021.1836961] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION COVID-19 pandemic has been declared as a global emergency by the World Health Organization which has mounted global pressure on the healthcare system. The design and development of rapid tests for the precise and early detection of infection are urgently needed to detect the disease and also for bulk screening of infected persons. The traditional drugs moderately control the symptoms, but so far, no specific drug has been discovered. The prime concern is to device novel tools for rapid and precise diagnosis, drug delivery, and effective therapies for coronavirus. In this context, nanotechnology offers novel ways to fight against COVID-19. AREA COVERED This review includes the use of nanomaterials for the control of COVID-19. The tools for diagnosis of coronavirus, nano-based vaccines, and nanoparticles as a drug delivery system for the treatment of virus infection have been discussed. The toxicity issues related to nanoparticles have also been addressed. EXPERT OPINION The research on nanotechnology-based diagnosis, drug delivery, and antiviral therapies is at a preliminary stage. The antiviral nanomedicine therapies are cost-effective and with high quality. Nanoparticles are a promising tool for prevention, diagnosis, antiviral drug delivery, and therapeutics, which may open up new avenues in the treatment of COVID-19.
Collapse
Affiliation(s)
- Mahendra Rai
- Nanobiotechnology Laboratory, Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, MH, India.,Department of Microbiology, Nicolaus Copernicus University, Torun, Poland
| | - Shital Bonde
- Nanobiotechnology Laboratory, Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, MH, India
| | - Alka Yadav
- Nanobiotechnology Laboratory, Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, MH, India
| | - Yulia Plekhanova
- Laboratory of Biosensors, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly Reshetilov
- Laboratory of Biosensors, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, Moscow, Russia
| | - Indarchand Gupta
- Department of Biotechnology, Institute of Science, Aurangabad, MH, India
| | - Patrycja Golińska
- Department of Microbiology, Nicolaus Copernicus University, Torun, Poland
| | - Raksha Pandit
- Nanobiotechnology Laboratory, Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, MH, India
| | - Avinash P Ingle
- Department of Biotechnology, Engineering School of Lorena, University of Sao Paulo, Lorena, SP, Brazil
| |
Collapse
|
35
|
van Moorsel CHM. Desmoplakin: An Important Player in Aging Lung Disease. Am J Respir Crit Care Med 2020; 202:1201-1202. [PMID: 32716632 PMCID: PMC7605188 DOI: 10.1164/rccm.202006-2457ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
36
|
Yadav E, Yadav N, Hus A, Yadav JS. Aquaporins in lung health and disease: Emerging roles, regulation, and clinical implications. Respir Med 2020; 174:106193. [PMID: 33096317 DOI: 10.1016/j.rmed.2020.106193] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/17/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Aquaporins (AQPs) aka water channels are a family of conserved transmembrane proteins (~30 kDa monomers) expressed in various organ systems. Of the 13 AQPs (AQP0 through AQP12) in the human body, four (AQPs 1, 3, 4, and 5) are expressed in the respiratory system. These channels are conventionally known for mediating transcellular fluid movements. Certain AQPs (aquaglyceroporins) have the capability to transport glycerol and potentially other solutes. There is an emerging body of literature unveiling the non-conventional roles of AQPs such as in cell proliferation and migration, gas permeation, signal potentiation, etc. Initial gene knock-out studies established a physiological role for lung AQPs, particularly AQP5, in maintaining homeostasis, by mediating fluid secretion from submucosal glands onto the airway surface liquid (ASL) lining. Subsequent studies have highlighted the functional significance of AQPs, particularly AQP1 and AQP5 in lung pathophysiology and diseases, including but not limited to chronic and acute lung injury, chronic obstructive pulmonary disease (COPD), other inflammatory lung conditions, and lung cancer. AQP1 has been suggested as a potential prognostic marker for malignant mesothelioma. Recent efforts are directed toward exploiting AQPs as targets for diagnosis, prevention, intervention, and/or treatment of various lung conditions. Emerging information on regulatory pathways and directed mechanistic research are posited to unravel novel strategies for these clinical implications. Future considerations should focus on development of AQP inhibitors, blockers, and modulators for therapeutic needs, and better understanding the role of lung-specific AQPs in inter-individual susceptibility to chronic lung diseases such as COPD and cancer.
Collapse
Affiliation(s)
- Ekta Yadav
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Niket Yadav
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, 22908-0738, USA
| | - Ariel Hus
- Department of Biology, University of Miami, Coral Gables, Florida, 33146, USA
| | - Jagjit S Yadav
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
37
|
Nemeth J, Schundner A, Quast K, Winkelmann VE, Frick M. A Novel Fibroblast Reporter Cell Line for in vitro Studies of Pulmonary Fibrosis. Front Physiol 2020; 11:567675. [PMID: 33162897 PMCID: PMC7582034 DOI: 10.3389/fphys.2020.567675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease of the lower respiratory tract with restricted therapeutic options. Repetitive injury of the bronchoalveolar epithelium leads to activation of pulmonary fibroblasts, differentiation into myofibroblasts and excessive extracellular matrix (ECM) deposition resulting in aberrant wound repair. However, detailed molecular and cellular mechanisms underlying initiation and progression of fibrotic changes are still elusive. Here, we report the generation of a representative fibroblast reporter cell line (10-4A BFP ) to study pathophysiological mechanisms of IPF in high throughput or high resolution in vitro live cell assays. To this end, we immortalized primary fibroblasts isolated from the distal lung of Sprague-Dawley rats. Molecular and transcriptomic characterization identified clone 10-4A as a matrix fibroblast subpopulation. Mechanical or chemical stimulation induced a reversible fibrotic state comparable to effects observed in primary isolated fibroblasts. Finally, we generated a reporter cell line (10-4A BFP ) to express nuclear blue fluorescent protein (BFP) under the promotor of the myofibroblast marker alpha smooth muscle actin (Acta2) using CRISPR/Cas9 technology. We evaluated the suitability of 10-4A BFP as reporter tool in plate reader assays. In summary, the 10-4A BFP cell line provides a novel tool to study fibrotic processes in vitro to gain new insights into the cellular and molecular processes involved in fibrosis formation and propagation.
Collapse
Affiliation(s)
- Julia Nemeth
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Karsten Quast
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
38
|
Pathological effects of nano-sized particles on the respiratory system. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102242. [DOI: 10.1016/j.nano.2020.102242] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/26/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
|
39
|
Zuo WL, Rostami MR, LeBlanc M, Kaner RJ, O’Beirne SL, Mezey JG, Leopold PL, Quast K, Visvanathan S, Fine JS, Thomas MJ, Crystal RG. Dysregulation of club cell biology in idiopathic pulmonary fibrosis. PLoS One 2020; 15:e0237529. [PMID: 32941426 PMCID: PMC7498242 DOI: 10.1371/journal.pone.0237529] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/28/2020] [Indexed: 11/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, chronic fibrotic lung disease with an irreversible decline of lung function. "Bronchiolization", characterized by ectopic appearance of airway epithelial cells in the alveolar regions, is one of the characteristic features in the IPF lung. Based on the knowledge that club cells are the major epithelial secretory cells in human small airways, and their major secretory product uteroglobin (SCGB1A1) is significantly increased in both serum and epithelial lining fluid of IPF lung, we hypothesize that human airway club cells contribute to the pathogenesis of IPF. By assessing the transcriptomes of the single cells from human lung of control donors and IPF patients, we identified two SCGB1A1+ club cell subpopulations, highly expressing MUC5B, a significant genetic risk factor strongly associated with IPF, and SCGB3A2, a marker heterogeneously expressed in the club cells, respectively. Interestingly, the cellular proportion of SCGB1A1+MUC5B+ club cells was significantly increased in IPF patients, and this club cell subpopulation highly expressed genes related to mucous production and immune cell chemotaxis. In contrast, though the cellular proportion did not change, the molecular phenotype of the SCGB1A1+SCGB3A2high club cell subpopulation was significantly altered in IPF lung, with increased expression of mucins, cytokine and extracellular matrix genes. The single cell transcriptomic analysis reveals the cellular and molecular heterogeneity of club cells, and provide novel insights into the biological functions of club cells in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Wu-Lin Zuo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Mahboubeh R. Rostami
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Michelle LeBlanc
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Robert J. Kaner
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Sarah L. O’Beirne
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Jason G. Mezey
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Philip L. Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Karsten Quast
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sudha Visvanathan
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Jay S. Fine
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, United States of America
| | - Matthew J. Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Medicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
40
|
Agudelo CW, Samaha G, Garcia-Arcos I. Alveolar lipids in pulmonary disease. A review. Lipids Health Dis 2020; 19:122. [PMID: 32493486 PMCID: PMC7268969 DOI: 10.1186/s12944-020-01278-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Lung lipid metabolism participates both in infant and adult pulmonary disease. The lung is composed by multiple cell types with specialized functions and coordinately acting to meet specific physiologic requirements. The alveoli are the niche of the most active lipid metabolic cell in the lung, the type 2 cell (T2C). T2C synthesize surfactant lipids that are an absolute requirement for respiration, including dipalmitoylphosphatidylcholine. After its synthesis and secretion into the alveoli, surfactant is recycled by the T2C or degraded by the alveolar macrophages (AM). Surfactant biosynthesis and recycling is tightly regulated, and dysregulation of this pathway occurs in many pulmonary disease processes. Alveolar lipids can participate in the development of pulmonary disease from their extracellular location in the lumen of the alveoli, and from their intracellular location in T2C or AM. External insults like smoke and pollution can disturb surfactant homeostasis and result in either surfactant insufficiency or accumulation. But disruption of surfactant homeostasis is also observed in many chronic adult diseases, including chronic obstructive pulmonary disease (COPD), and others. Sustained damage to the T2C is one of the postulated causes of idiopathic pulmonary fibrosis (IPF), and surfactant homeostasis is disrupted during fibrotic conditions. Similarly, surfactant homeostasis is impacted during acute respiratory distress syndrome (ARDS) and infections. Bioactive lipids like eicosanoids and sphingolipids also participate in chronic lung disease and in respiratory infections. We review the most recent knowledge on alveolar lipids and their essential metabolic and signaling functions during homeostasis and during some of the most commonly observed pulmonary diseases.
Collapse
Affiliation(s)
- Christina W Agudelo
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Ghassan Samaha
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Itsaso Garcia-Arcos
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA.
| |
Collapse
|
41
|
Bueno M, Calyeca J, Rojas M, Mora AL. Mitochondria dysfunction and metabolic reprogramming as drivers of idiopathic pulmonary fibrosis. Redox Biol 2020; 33:101509. [PMID: 32234292 PMCID: PMC7251240 DOI: 10.1016/j.redox.2020.101509] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease of unknown etiology. It is characterized by deposition of extracellular matrix proteins, like collagen and fibronectin in the lung interstitium leading to respiratory failure. Our understanding of the pathobiology underlying IPF is still incomplete; however, it is accepted that aging is a major risk factor in the disease while growing evidence suggests that the mitochondria plays an important role in the initiation and progression of pulmonary fibrosis. Mitochondria dysfunction and metabolic reprogramming had been identified in different IPF lung cells (alveolar epithelial cells, fibroblasts, and macrophages) promoting low resilience and increasing susceptibility to activation of profibrotic responses. Here we summarize changes in mitochondrial numbers, biogenesis, turnover and associated metabolic adaptations that promote disrepair and fibrosis in the lung. Finally, we highlight new possible therapeutic approaches focused on ameliorate mitochondrial dysfunction.
Collapse
Affiliation(s)
- Marta Bueno
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jazmin Calyeca
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Dorothy and Richard Simmons Center for Interstitial Lung Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana L Mora
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Diagnostic and Prognostic Biomarkers for Chronic Fibrosing Interstitial Lung Diseases With a Progressive Phenotype. Chest 2020; 158:646-659. [PMID: 32268131 DOI: 10.1016/j.chest.2020.03.037] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/05/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Biomarkers have the potential to become central to the clinical evaluation and monitoring of patients with chronic fibrosing interstitial lung diseases (ILDs) with a progressive phenotype. Here we summarize the current understanding of putative serum, BAL fluid, and genetic biomarkers in this setting, according to their hypothesized pathobiologic mechanisms: evidence of epithelial cell dysfunction (eg, Krebs von den Lungen-6 antigen), fibroblast proliferation and extracellular matrix production or turnover (eg, matrix metalloproteinase-1), or immune dysregulation (eg, CC chemokine ligand 18). While most of the available data come from idiopathic pulmonary fibrosis (IPF), the prototypic progressive fibrosing ILD, data are available in the broader patient population of chronic fibrosing ILDs. A number of these biomarkers show promise, however, none have been validated. In this review article, we assess both the status of proposed biomarkers for chronic fibrosing lung diseases with a progressive phenotype in predicting disease risk or predisposition, diagnosis, prognosis, and treatment response and provide a direct comparison between IPF and other chronic fibrotic ILDs. We also reflect on the current clinical usefulness and future direction of research for biomarkers in the setting of chronic fibrosing ILDs with a progressive phenotype.
Collapse
|
43
|
Schmidt M, Cattani-Cavalieri I, Nuñez FJ, Ostrom RS. Phosphodiesterase isoforms and cAMP compartments in the development of new therapies for obstructive pulmonary diseases. Curr Opin Pharmacol 2020; 51:34-42. [PMID: 32622335 PMCID: PMC7529846 DOI: 10.1016/j.coph.2020.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/17/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022]
Abstract
The second messenger molecule 3'5'-cyclic adenosine monophosphate (cAMP) imparts several beneficial effects in lung diseases such as asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). While cAMP is bronchodilatory in asthma and COPD, it also displays anti-fibrotic properties that limit fibrosis. Phosphodiesterases (PDEs) metabolize cAMP and thus regulate cAMP signaling. While some existing therapies inhibit PDEs, there are only broad family specific inhibitors. The understanding of cAMP signaling compartments, some centered around lipid rafts/caveolae, has led to interest in defining how specific PDE isoforms maintain these signaling microdomains. The possible altered expression of PDEs, and thus abnormal cAMP signaling, in obstructive lung diseases has been poorly explored. We propose that inhibition of specific PDE isoforms can improve therapy of obstructive lung diseases by amplifying specific cAMP signals in discreet microdomains.
Collapse
Affiliation(s)
- Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Francisco J Nuñez
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA.
| |
Collapse
|
44
|
Abstract
The respiratory system plays an essential role for human life. This system (like all others) undergoes physiological regeneration due to many types of stem cells found both in the respiratory tract itself and in the alveoli. The stem cell hierarchy is very extensive due to their variety in the lungs and is still not completely understood.The best described lung stem cells are alveolar type II cells, which as progenitor lung stem cells are precursors of alveolar type I cells, i.e., cells that perform gas exchange in the lungs. These progenitor stem cells, which reside in alveoli corners, express high levels of surfactant protein C (SFTPC). Despite the fact that type II pneumocytes occupy only 7-10% of the lung surface, there are almost twice as many as alveolar type I cells occupying almost 95% of the surface.Other stem cells making up the lung regenerative potential have also been identified in the lungs. Both endothelial, mesodermal, and epithelial stem cells are necessary for the lungs to function properly and perform their physiological functions.The lungs, like all other organs, undergo an aging process. As a result of this process, not only the total number of cells changes, the percentage of particular types of cells, but also their efficiency is reduced. With age, the proliferative potential of lung stem cells also decreases, not just their number. This brings about the need to increase the intensity of research in the field of regenerative medicine.
Collapse
Affiliation(s)
- Andrzej Ciechanowicz
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical Univeristy of Warsaw, Warsaw, Poland.
| |
Collapse
|
45
|
Schneider JP, Wrede C, Mühlfeld C. The Three-Dimensional Ultrastructure of the Human Alveolar Epithelium Revealed by Focused Ion Beam Electron Microscopy. Int J Mol Sci 2020; 21:ijms21031089. [PMID: 32041332 PMCID: PMC7038159 DOI: 10.3390/ijms21031089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Thin type 1 alveolar epithelial (AE1) and surfactant producing type 2 alveolar epithelial (AE2) cells line the alveoli in the lung and are essential for normal lung function. Function is intimately interrelated to structure, so that detailed knowledge of the epithelial ultrastructure can significantly enhance our understanding of its function. The basolateral surface of the cells or the epithelial contact sites are of special interest, because they play an important role in intercellular communication or stabilizing the epithelium. The latter is in particular important for the lung with its variable volume. The aim of the present study was to investigate the three-dimensional (3D) ultrastructure of the human alveolar epithelium focusing on contact sites and the basolateral cell membrane of AE2 cells using focused ion beam electron microscopy and subsequent 3D reconstructions. The study provides detailed surface reconstructions of two AE1 cell domains and two AE2 cells, showing AE1/AE1, AE1/AE2 and AE2/AE2 contact sites, basolateral microvilli pits at AE2 cells and small AE1 processes beneath AE2 cells. Furthermore, we show reconstructions of a surfactant secretion pore, enlargements of the apical AE1 cell surface and long folds bordering grooves on the basal AE1 cell surface. The functional implications of our findings are discussed. These findings may lay the structural basis for further molecular investigations.
Collapse
Affiliation(s)
- Jan Philipp Schneider
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (C.W.); (C.M.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Correspondence:
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (C.W.); (C.M.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (C.W.); (C.M.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
46
|
Idiopathic Pulmonary Fibrosis: Pathogenesis and the Emerging Role of Long Non-Coding RNAs. Int J Mol Sci 2020; 21:ijms21020524. [PMID: 31947693 PMCID: PMC7013390 DOI: 10.3390/ijms21020524] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive chronic disease characterized by excessing scarring of the lungs leading to irreversible decline in lung function. The aetiology and pathogenesis of the disease are still unclear, although lung fibroblast and epithelial cell activation, as well as the secretion of fibrotic and inflammatory mediators, have been strongly associated with the development and progression of IPF. Significantly, long non-coding RNAs (lncRNAs) are emerging as modulators of multiple biological processes, although their function and mechanism of action in IPF is poorly understood. LncRNAs have been shown to be important regulators of several diseases and their aberrant expression has been linked to the pathophysiology of fibrosis including IPF. This review will provide an overview of this emerging role of lncRNAs in the development of IPF.
Collapse
|
47
|
NOVEL NON-INVASIVE SEVERITY MARKERS IN IDIOPATHIC PULMONARY FIBROSIS. WORLD OF MEDICINE AND BIOLOGY 2020. [DOI: 10.26724/2079-8334-2020-3-73-91-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
48
|
Li C, Sun X, Li A, Mo M, Zhao Z. S-Allylmercaptocysteine attenuates Bleomycin-induced pulmonary fibrosis in mice via suppressing TGF-β1/Smad and oxidative stress pathways. Int Immunopharmacol 2019; 79:106110. [PMID: 31874367 DOI: 10.1016/j.intimp.2019.106110] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/14/2019] [Accepted: 11/30/2019] [Indexed: 12/23/2022]
Abstract
Pulmonary fibrosis (PF) is a disease characterized by diffusing alveolar inflammation and alveolar structural disorders that ultimately lead to pulmonary interstitial fibrosis. S-allylmercaptocysteine (SAMC) as a water-soluble organosulfur garlic derivative exhibits efficient anti-inflammatory and anti-oxidative activities. In this study, we attempted to explore the function of SAMC in inhibiting bleomycin (BLM)-induced pulmonary fibrosis in mice. 0.035 U/g of BLM was intraperitoneally injected into mice twice per week for 4 weeks to induce fibrosis. SAMC (25 and 50 mg/kg) and N-acetylcysteine (NAC, 600 mg/kg) were given to mice for 28 days. The results indicate that SAMC could significantly ameliorate the pathological structure, and decrease inflammatory cell infiltration and pro-inflammatory cytokines in bronchoalveolar lavage fluid (BALF) in BLM-induced pulmonary fibrosis mice. SAMC showed an anti-fibrosis effect by increasing anti-oxidants like HO-1, GSH and SOD as well as decreasing hydroxyproline (HYP) in BLM-induced mice. Mechanistic studies suggested that SAMC alleviated oxidative stress probably by impacting the Nox4/Nrf2 pathways, and played an anti-fibrosis role with decreasing the expression of α-SMA, collagen III, collagen I by suppressing the TGF-β1/Smad pathway. These findings indicate that SAMC may be partially responsible for the therapeutic effect on PF patients.
Collapse
Affiliation(s)
- Chunyan Li
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Xiao Sun
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Ang Li
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Min Mo
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhongxi Zhao
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
49
|
Moimas S, Salton F, Kosmider B, Ring N, Volpe MC, Bahmed K, Braga L, Rehman M, Vodret S, Graziani ML, Wolfson MR, Marchetti N, Rogers TJ, Giacca M, Criner GJ, Zacchigna S, Confalonieri M. miR-200 family members reduce senescence and restore idiopathic pulmonary fibrosis type II alveolar epithelial cell transdifferentiation. ERJ Open Res 2019; 5:00138-2019. [PMID: 31857992 PMCID: PMC6911923 DOI: 10.1183/23120541.00138-2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
Rationale Alveolar type II (ATII) cells act as adult stem cells contributing to alveolar type I (ATI) cell renewal and play a major role in idiopathic pulmonary fibrosis (IPF), as supported by familial cases harbouring mutations in genes specifically expressed by these cells. During IPF, ATII cells lose their regenerative potential and aberrantly express pathways contributing to epithelial–mesenchymal transition (EMT). The microRNA miR-200 family is downregulated in IPF, but its effect on human IPF ATII cells remains unproven. We wanted to 1) evaluate the characteristics and transdifferentiating ability of IPF ATII cells, and 2) test whether miR-200 family members can rescue the regenerative potential of fibrotic ATII cells. Methods ATII cells were isolated from control or IPF lungs and cultured in conditions promoting their transdifferentiation into ATI cells. Cells were either phenotypically monitored over time or transfected with miR-200 family members to evaluate the microRNA effect on the expression of transdifferentiation, senescence and EMT markers. Results IPF ATII cells show a senescent phenotype (p16 and p21), overexpression of EMT (ZEB1/2) and impaired expression of ATI cell markers (AQP5 and HOPX) after 6 days of culture in differentiating medium. Transfection with certain miR-200 family members (particularly miR-200b-3p and miR-200c-3p) reduced senescence marker expression and restored the ability to transdifferentiate into ATI cells. Conclusions We demonstrated that ATII cells from IPF patients express senescence and EMT markers, and display a reduced ability to transdifferentiate into ATI cells. Transfection with certain miR-200 family members rescues this phenotype, reducing senescence and restoring transdifferentiation marker expression. Idiopathic pulmonary fibrosis alveolar epithelial type II cells show senescence and EMT features, but miR-200b and miR-200c can restore the ability of type II cells to transdifferentiate in vitro into type I alveolar epithelial cellshttp://bit.ly/359tlit
Collapse
Affiliation(s)
- Silvia Moimas
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,These authors contributed equally to this work (co-first authors)
| | - Francesco Salton
- Pulmonology Dept, University Hospital of Cattinara, Trieste, Italy.,These authors contributed equally to this work (co-first authors)
| | - Beata Kosmider
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Physiology, Temple University, Philadelphia, PA, USA.,These authors contributed equally to this work (co-first authors)
| | - Nadja Ring
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Maria C Volpe
- Pulmonology Dept, University Hospital of Cattinara, Trieste, Italy.,Dept of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Karim Bahmed
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Luca Braga
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Michael Rehman
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Simone Vodret
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | | | - Marla R Wolfson
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Physiology, Temple University, Philadelphia, PA, USA.,CENTRe: Collaborative for Environmental and Neonatal Therapeutics, Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Nathaniel Marchetti
- Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Dept of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gerard J Criner
- Dept of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,These authors contributed equally to this work (co-last authors)
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Dept of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.,These authors contributed equally to this work (co-last authors)
| | - Marco Confalonieri
- Pulmonology Dept, University Hospital of Cattinara, Trieste, Italy.,Dept of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.,These authors contributed equally to this work (co-last authors)
| |
Collapse
|
50
|
Veith C, Boots AW, Idris M, van Schooten FJ, van der Vliet A. Redox Imbalance in Idiopathic Pulmonary Fibrosis: A Role for Oxidant Cross-Talk Between NADPH Oxidase Enzymes and Mitochondria. Antioxid Redox Signal 2019; 31:1092-1115. [PMID: 30793932 PMCID: PMC6767863 DOI: 10.1089/ars.2019.7742] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Significance: Idiopathic pulmonary fibrosis (IPF) is a progressive age-related lung disease with a median survival of only 3 years after diagnosis. The pathogenic mechanisms behind IPF are not clearly understood, and current therapeutic approaches have not been successful in improving disease outcomes. Recent Advances: IPF is characterized by increased production of reactive oxygen species (ROS), primarily by NADPH oxidases (NOXes) and mitochondria, as well as altered antioxidant defenses. Recent studies have identified the NOX isoform NOX4 as a key player in various important aspects of IPF pathology. In addition, mitochondrial dysfunction is thought to enhance pathological features of IPF, in part by increasing mitochondrial ROS (mtROS) production and altering cellular metabolism. Recent findings indicate reciprocal interactions between NOX enzymes and mitochondria, which affect regulation of NOX activity as well as mitochondrial function and mtROS production, and collectively promote epithelial injury and profibrotic signaling. Critical Issues and Future Directions: The precise molecular mechanisms by which ROS from NOX or mitochondria contribute to IPF pathology are not known. This review summarizes the current knowledge with respect to the various aspects of ROS imbalance in the context of IPF and its proposed roles in disease development, with specific emphasis on the importance of inappropriate NOX activation, mitochondrial dysfunction, and the emerging evidence of NOX-mitochondria cross-talk as important drivers in IPF pathobiology.
Collapse
Affiliation(s)
- Carmen Veith
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Agnes W. Boots
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Musa Idris
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Sciences, NUTRIM School of Nutrition, Translational Research and Metabolism, University of Maastricht, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Address correspondence to: Dr. Albert van der Vliet, Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, HSRF 216, 149 Beaumont Avenue, Burlington, VT 05405
| |
Collapse
|