1
|
Wang D, Liu H, Bai S, Zheng X, Zhao L. The PAR6B-PRKCI-PAR3 complex influences alveolar regeneration in patients with the emphysema subtype of chronic obstructive pulmonary disease. Stem Cell Res Ther 2025; 16:97. [PMID: 40001200 PMCID: PMC11863855 DOI: 10.1186/s13287-025-04189-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is gaining increasing attention, with different subtypes being distinguished for separate research and treatment. The emphysema subtype is characterized by widespread alveolar destruction, which may be associated with aggravated alveolar damage and abnormal repair. Type II alveolar epithelial cells (AEC2s), known for their stem cell potential, have recently emerged as a promising target for COPD treatment. However, to date, few studies have elucidated the specific mechanisms by which AEC2s induce alveolar regeneration. METHODS Lung tissue samples from COPD patients were collected, and bioinformatics analysis was used to identify expression profiles affecting the emphysema phenotype and target genes regulating AEC2 proliferation. In vitro models of smoke-induced injury and viral transfection were established to clarify the role of the target gene PARD6B in regulating AEC2s proliferation and transdifferentiation potential. Co-immunoprecipitation and mass spectrometry were employed to elucidate the specific regulatory mechanisms. Primary mouse AEC2s were isolated for 3D spheroid formation experiments to further validate the role of the target gene. RESULTS We observed impaired self-proliferation and enhanced transdifferentiation of AEC2s into AEC1s in lung tissues from COPD patients with emphysema subtype, which was associated with reduced expression of PARD6B. Interestingly, PARD6B primarily functioned as part of a complex in AEC2s. Mechanistically, we found that reduced levels of the PAR3-PARD6B-PRKCI complex could arrest the cell cycle of AEC2s in the G0-G1 phase, thereby impairing their self-proliferation. CONCLUSIONS Our findings reveal a novel regulatory mechanism for alveolar regeneration, highlighting a potential therapeutic target for managing the emphysema subtype of COPD.
Collapse
Affiliation(s)
- Di Wang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongbo Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Bai
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuejian Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Zhao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Simborio H, Hayek H, Kosmider B, Elrod JW, Bolla S, Marchetti N, Criner GJ, Bahmed K. Mitochondrial dysfunction and impaired DNA damage repair through PICT1 dysregulation in alveolar type II cells in emphysema. Cell Commun Signal 2024; 22:562. [PMID: 39578839 PMCID: PMC11583753 DOI: 10.1186/s12964-024-01896-0] [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: 04/02/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Alveolar type II (ATII) cells have a stem cell potential in the adult lung and repair the epithelium after injury induced by harmful factors. Their damage contributes to emphysema development, characterized by alveolar wall destruction. Cigarette smoke is the main risk factor for this disease development. METHODS ATII cells were obtained from control non-smoker and smoker organ donors and emphysema patients. Isolated cells were used to study the role of PICT1 in this disease. Also, a cigarette smoke-induced murine model of emphysema was applied to define its function in disease progression further. RESULTS Decreased PICT1 expression was observed in human and murine ATII cells in emphysema. PICT1 was immunoprecipitated, followed by mass spectrometry analysis. We identified MRE11, which is involved in DNA damage repair, as its novel interactor. PICT1 and MRE11 protein levels were decreased in ATII cells in this disease. Moreover, cells with PICT1 deletion were exposed to cigarette smoke extract. This treatment induced cellular and mitochondrial ROS, cell cycle arrest, nuclear and mitochondrial DNA damage, decreased mitochondrial respiration, and impaired DNA damage repair. CONCLUSIONS This study indicates that PICT1 dysfunction can negatively affect genome stability and mitochondrial activity in ATII cells, contributing to emphysema development. Targeting PICT1 can lead to novel therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Hannah Simborio
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hassan Hayek
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Aging & Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Sudhir Bolla
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
3
|
Wang Y, Wang L, Ma S, Cheng L, Yu G. Repair and regeneration of the alveolar epithelium in lung injury. FASEB J 2024; 38:e23612. [PMID: 38648494 DOI: 10.1096/fj.202400088r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/01/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024]
Abstract
Considerable progress has been made in understanding the function of alveolar epithelial cells in a quiescent state and regeneration mechanism after lung injury. Lung injury occurs commonly from severe viral and bacterial infections, inhalation lung injury, and indirect injury sepsis. A series of pathological mechanisms caused by excessive injury, such as apoptosis, autophagy, senescence, and ferroptosis, have been studied. Recovery from lung injury requires the integrity of the alveolar epithelial cell barrier and the realization of gas exchange function. Regeneration mechanisms include the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and proteins. While alveoli are damaged, alveolar type II (AT2) cells proliferate and differentiate into alveolar type I (AT1) cells to repair the damaged alveolar epithelial layer. Alveolar epithelial cells are surrounded by various cells, such as fibroblasts, endothelial cells, and various immune cells, which affect the proliferation and differentiation of AT2 cells through paracrine during alveolar regeneration. Besides, airway epithelial cells also contribute to the repair and regeneration process of alveolar epithelium. In this review, we mainly discuss the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and transcription factors.
Collapse
Affiliation(s)
- Yaxuan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang, China
| | - Shuaichen Ma
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang, China
| | - Lianhui Cheng
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang, China
| |
Collapse
|
4
|
Hayek H, Rehbini O, Kosmider B, Brandt T, Chatila W, Marchetti N, Criner GJ, Bolla S, Kishore R, Bowler RP, Bahmed K. The Regulation of Fatty Acid Synthase by Exosomal miR-143-5p and miR-342-5p in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2024; 70:259-282. [PMID: 38117249 PMCID: PMC11478129 DOI: 10.1165/rcmb.2023-0232oc] [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: 06/27/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive disease caused by an aberrant repair of injured alveolar epithelial cells. The maintenance of the alveolar epithelium and its regeneration after the damage is fueled by alveolar type II (ATII) cells. Injured cells release exosomes containing microRNAs (miRNAs), which can alter the recipient cells' function. Lung tissue, ATII cells, fibroblasts, plasma, and exosomes were obtained from naive patients with IPF, patients with IPF taking pirfenidone or nintedanib, and control organ donors. miRNA expression was analyzed to study their impact on exosome-mediated effects in IPF. High miR-143-5p and miR-342-5p levels were detected in ATII cells, lung tissue, plasma, and exosomes in naive patients with IPF. Decreased FASN (fatty acid synthase) and ACSL-4 (acyl-CoA-synthetase long-chain family member 4) expression was found in ATII cells. miR-143-5p and miR-342-5p overexpression or ATII cell treatment with IPF-derived exosomes containing these miRNAs lowered FASN and ACSL-4 levels. Also, this contributed to ATII cell injury and senescence. However, exosomes isolated from patients with IPF taking nintedanib or pirfenidone increased FASN expression in ATII cells compared with naive patients with IPF. Furthermore, fibroblast treatment with exosomes obtained from naive patients with IPF increased SMAD3, CTGF, COL3A1, and TGFβ1 expression. Our results suggest that IPF-derived exosomes containing miR-143-5p and miR-342-5p inhibited the de novo fatty acid synthesis pathway in ATII cells. They also induced the profibrotic response in fibroblasts. Pirfenidone and nintedanib improved ATII cell function and inhibited fibrogenesis. This study highlights the importance of exosomes in IPF pathophysiology.
Collapse
Affiliation(s)
- Hassan Hayek
- Department of Microbiology, Immunology, and Inflammation
- Center for Inflammation and Lung Research
| | | | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation
- Center for Inflammation and Lung Research
- Department of Thoracic Medicine and Surgery
| | | | | | | | | | | | - Raj Kishore
- Center for Translational Medicine, and
- Department of Cardiovascular Sciences, Temple University, Philadelphia, Pennsylvania; and
| | | | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation
- Center for Inflammation and Lung Research
- Department of Thoracic Medicine and Surgery
| |
Collapse
|
5
|
Cui T, Wangpaichitr M, Schally AV, Griswold AJ, Vidaurre I, Sha W, Jackson RM. Alveolar epithelial cell growth hormone releasing hormone receptor in alveolar epithelial inflammation. Exp Lung Res 2023; 49:152-164. [PMID: 37584484 DOI: 10.1080/01902148.2023.2246074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
Purpose: Growth hormone-releasing hormone (GHRH) is a 44-amino acid peptide that regulates growth hormone (GH) secretion. We hypothesized that GHRH receptor (GHRH-R) in alveolar type 2 (AT2) cells could modulate pro-inflammatory and possibly subsequent pro-fibrotic effects of lipopolysaccharide (LPS) or cytokines, such that AT2 cells could participate in lung inflammation and fibrosis. Methods: We used human alveolar type 2 (iAT2) epithelial cells derived from induced pluripotent stem cells (iPSC) to investigate how GHRH-R modulates gene and protein expression. We tested iAT2 cells' gene expression in response to LPS or cytokines, seeking whether these mechanisms caused endogenous production of pro-inflammatory molecules or mesenchymal markers. Quantitative real-time PCR (RT-PCR) and Western blotting were used to investigate differential expression of epithelial and mesenchymal markers. Result: Incubation of iAT2 cells with LPS increased expression of IL1-β and TNF-α in addition to mesenchymal genes, including ACTA2, FN1 and COL1A1. Alveolar epithelial cell gene expression due to LPS was significantly inhibited by GHRH-R peptide antagonist MIA-602. Incubation of iAT2 cells with cytokines like those in fibrotic lungs similarly increased expression of genes for IL1-β, TNF-α, TGFβ-1, Wnt5a, smooth muscle actin, fibronectin and collagen. Expression of mesenchymal proteins, such as N-cadherin and vimentin, were also elevated after prolonged exposure to cytokines, confirming epithelial production of pro-inflammatory molecules as an important mechanism that might lead to subsequent fibrosis. Conclusion: iAT2 cells clearly expressed the GHRH-R. Exposure to LPS or cytokines increased iAT2 cell production of pro-inflammatory factors. GHRH-R antagonist MIA-602 inhibited pro-inflammatory gene expression, implicating iAT2 cell GHRH-R signaling in lung inflammation and potentially in fibrosis.
Collapse
Affiliation(s)
- Tengjiao Cui
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Andrew V Schally
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Pathology and Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anthony J Griswold
- Dr. John T. McDonald Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Wei Sha
- Research Service, Miami VAHS, Miami, Florida, USA
| | - Robert M Jackson
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
6
|
Lin CR, Bahmed K, Kosmider B. Impaired Alveolar Re-Epithelialization in Pulmonary Emphysema. Cells 2022; 11:2055. [PMID: 35805139 PMCID: PMC9265977 DOI: 10.3390/cells11132055] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/24/2023] Open
Abstract
Alveolar type II (ATII) cells are progenitors in alveoli and can repair the alveolar epithelium after injury. They are intertwined with the microenvironment for alveolar epithelial cell homeostasis and re-epithelialization. A variety of ATII cell niches, transcription factors, mediators, and signaling pathways constitute a specific environment to regulate ATII cell function. Particularly, WNT/β-catenin, YAP/TAZ, NOTCH, TGF-β, and P53 signaling pathways are dynamically involved in ATII cell proliferation and differentiation, although there are still plenty of unknowns regarding the mechanism. However, an imbalance of alveolar cell death and proliferation was observed in patients with pulmonary emphysema, contributing to alveolar wall destruction and impaired gas exchange. Cigarette smoking causes oxidative stress and is the primary cause of this disease development. Aberrant inflammatory and oxidative stress responses result in loss of cell homeostasis and ATII cell dysfunction in emphysema. Here, we discuss the current understanding of alveolar re-epithelialization and altered reparative responses in the pathophysiology of this disease. Current therapeutics and emerging treatments, including cell therapies in clinical trials, are addressed as well.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
7
|
Expression of SARS-CoV-2 Entry Factors in Human Alveolar Type II Cells in Aging and Emphysema. Biomedicines 2021; 9:biomedicines9070779. [PMID: 34356843 PMCID: PMC8301390 DOI: 10.3390/biomedicines9070779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/19/2023] Open
Abstract
Alveolar type II (ATII) cells proliferate and restore the injured epithelium. It has been described that SARS-CoV-2 infection causes diffuse alveolar damage in the lungs. However, host factors facilitating virus infection in ATII cells are not well known. We determined the SARS-CoV-2-related genes and protein expression using RT-PCR and Western blotting, respectively, in ATII cells isolated from young and elderly non-smokers, smokers, and ex-smokers. Cells were also obtained from lung transplants of emphysema patients. ACE2 has been identified as the receptor for SARS-CoV-2, and we found significantly increased levels in young and elderly smokers and emphysema patients. The viral entry depends on TMPRSS2 protease activity, and a higher expression was detected in elderly smokers and ex-smokers and emphysema patients. Both ACE2 and TMPRSS2 mRNA levels were higher in this disease in comparison with non-smokers. CD209L serves as a receptor for SARS-CoV-2, and we found increased levels in ATII cells obtained from smokers and in emphysema patients. Also, our data suggest CD209L regulation by miR142. Endoplasmic reticulum stress was detected in ATII cells in this disease. Our results suggest that upregulation of SARS-CoV-2 entry factors in ATII cells in aging, smokers, and emphysema patients may facilitate infection.
Collapse
|
8
|
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: 40] [Impact Index Per Article: 6.7] [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
|
9
|
Bahmed K, Lin CR, Simborio H, Karim L, Aksoy M, Kelsen S, Tomar D, Madesh M, Elrod J, Messier E, Mason R, Unterwald EM, Eisenstein TK, Criner GJ, Kosmider B. The role of DJ-1 in human primary alveolar type II cell injury induced by e-cigarette aerosol. Am J Physiol Lung Cell Mol Physiol 2019; 317:L475-L485. [PMID: 31313616 PMCID: PMC6842910 DOI: 10.1152/ajplung.00567.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 06/11/2019] [Accepted: 07/07/2019] [Indexed: 01/18/2023] Open
Abstract
The alveolus participates in gas exchange, which can be impaired by environmental factors and toxins. There is an increase in using electronic cigarettes (e-cigarettes); however, their effect on human primary alveolar epithelial cells is unknown. Human lungs were obtained from nonsmoker organ donors to isolate alveolar type II (ATII) cells. ATII cells produce and secrete pulmonary surfactant and restore the epithelium after damage, and mitochondrial function is important for their metabolism. Our data indicate that human ATII cell exposure to e-cigarette aerosol increased IL-8 levels and induced DNA damage and apoptosis. We also studied the cytoprotective effect of DJ-1 against ATII cell injury. DJ-1 knockdown in human primary ATII cells sensitized cells to mitochondrial dysfunction as detected by high mitochondrial superoxide production, decreased mitochondrial membrane potential, and calcium elevation. DJ-1 knockout (KO) mice were more susceptible to ATII cell apoptosis and lung injury induced by e-cigarette aerosol compared with wild-type mice. Regulation of the oxidative phosphorylation (OXPHOS) is important for mitochondrial function and protection against oxidative stress. Major subunits of the OXPHOS system are encoded by both nuclear and mitochondrial DNA. We found dysregulation of OXPHOS complexes in DJ-1 KO mice after exposure to e-cigarette aerosol, which could disrupt the nuclear/mitochondrial stoichiometry, resulting in mitochondrial dysfunction. Together, our results indicate that DJ-1 deficiency sensitizes ATII cells to damage induced by e-cigarette aerosol leading to lung injury.
Collapse
Affiliation(s)
- Karim Bahmed
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Chih-Ru Lin
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Hannah Simborio
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Loukmane Karim
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Mark Aksoy
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Steven Kelsen
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Dhanendra Tomar
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - John Elrod
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Elise Messier
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert Mason
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Ellen M Unterwald
- Department of Pharmacology, Center for Substance Abuse Research, Temple University, Philadelphia, Pennsylvania
| | - Toby K Eisenstein
- Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania
- Center for Substance Abuse Research, Temple University, Philadelphia, Pennsylvania
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Beata Kosmider
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
- Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
- Department of Medicine, National Jewish Health, Denver, Colorado
- Department of Physiology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
The effect of cysteine oxidation on DJ-1 cytoprotective function in human alveolar type II cells. Cell Death Dis 2019; 10:638. [PMID: 31474749 PMCID: PMC6717737 DOI: 10.1038/s41419-019-1833-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/24/2022]
Abstract
DJ-1 is a multifunctional protein with cytoprotective functions. It is localized in the cytoplasm, nucleus, and mitochondria. The conserved cysteine residue at position 106 (Cys106) within DJ-1 serves as a sensor of redox state and can be oxidized to both the sulfinate (-SO2−) and sulfonate (-SO3−) forms. DJ-1 with Cys106-SO2− has cytoprotective activity but high levels of reactive oxygen species can induce its overoxidation to Cys106-SO3−. We found increased oxidative stress in alveolar type II (ATII) cells isolated from emphysema patients as determined by 4-HNE expression. DJ-1 with Cys106-SO3− was detected in these cells by mass spectrometry analysis. Moreover, ubiquitination of Cys106-SO3− DJ-1 was identified, which suggests that this oxidized isoform is targeted for proteasomal destruction. Furthermore, we performed controlled oxidation using H2O2 in A549 cells with DJ-1 knockout generated using CRISPR-Cas9 strategy. Lack of DJ-1 sensitized cells to apoptosis induced by H2O2 as detected using Annexin V and propidium iodide by flow cytometry analysis. This treatment also decreased both mitochondrial DNA amount and mitochondrial ND1 (NADH dehydrogenase 1, subunit 1) gene expression, as well as increased mitochondrial DNA damage. Consistent with the decreased cytoprotective function of overoxidized DJ-1, recombinant Cys106-SO3− DJ-1 exhibited a loss of its thermal unfolding transition, mild diminution of secondary structure in CD spectroscopy, and an increase in picosecond–nanosecond timescale dynamics as determined using NMR. Altogether, our data indicate that very high oxidative stress in ATII cells in emphysema patients induces DJ-1 overoxidation to the Cys106-SO3− form, leading to increased protein flexibility and loss of its cytoprotective function, which may contribute to this disease pathogenesis.
Collapse
|
11
|
Kosmider B, Lin CR, Karim L, Tomar D, Vlasenko L, Marchetti N, Bolla S, Madesh M, Criner GJ, Bahmed K. Mitochondrial dysfunction in human primary alveolar type II cells in emphysema. EBioMedicine 2019; 46:305-316. [PMID: 31383554 PMCID: PMC6711885 DOI: 10.1016/j.ebiom.2019.07.063] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Cigarette smoke is the main risk factor of pulmonary emphysema development, which is characterized by alveolar wall destruction. Mitochondria are important for alveolar type II (ATII) cell metabolism due to ATP generation. METHODS We isolated ATII cells from control non-smoker and smoker organ donors, and after lung transplant of patients with emphysema to determine mitochondrial function, dynamics and mitochondrial (mt) DNA damage. FINDINGS We found high mitochondrial superoxide generation and mtDNA damage in ATII cells in emphysema. This correlated with decreased mtDNA amount. We also detected high TOP1-cc and low TDP1 levels in mitochondria in ATII cells in emphysema. This contributed to the decreased resolution of TOP1-cc leading to accumulation of mtDNA damage and mitochondrial dysfunction. Moreover, we used lung tissue obtained from areas with mild and severe emphysema from the same patients. We found a correlation between the impaired fusion and fission as indicated by low MFN1, OPA1, FIS1, and p-DRP1 levels and this disease severity. We detected lower TDP1 expression in severe compared to mild emphysema. INTERPRETATION We found high DNA damage and impairment of DNA damage repair in mitochondria in ATII cells isolated from emphysema patients, which contribute to abnormal mitochondrial dynamics. Our findings provide molecular mechanisms of mitochondrial dysfunction in this disease. FUND: This work was supported by National Institutes of Health (NIH) grant R01 HL118171 (B.K.) and the Catalyst Award from the American Lung Association (K.B.).
Collapse
Affiliation(s)
- Beata Kosmider
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America; Department of Physiology, Temple University, Philadelphia, PA 19140, United States of America.
| | - Chih-Ru Lin
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Loukmane Karim
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Dhanendra Tomar
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, United States of America
| | - Liudmila Vlasenko
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Sudhir Bolla
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America
| | - Muniswamy Madesh
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, United States of America
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America
| | - Karim Bahmed
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, United States of America; Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, PA 19140, United States of America.
| |
Collapse
|
12
|
Lin CR, Bahmed K, Tomar D, Marchetti N, Criner GJ, Bolla S, Wilson MA, Madesh M, Kosmider B. The relationship between DJ-1 and S100A8 in human primary alveolar type II cells in emphysema. Am J Physiol Lung Cell Mol Physiol 2019; 317:L791-L804. [PMID: 31313618 DOI: 10.1152/ajplung.00494.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary emphysema is characterized by alveolar type II (ATII) cell death, destruction of alveolar wall septa, and irreversible airflow limitation. Cigarette smoke induces oxidative stress and is the main risk factor for this disease development. ATII cells isolated from nonsmokers, smokers, and patients with emphysema were used for this study. ATII cell apoptosis in individuals with this disease was detected. DJ-1 and S100A8 have cytoprotective functions against oxidative stress-induced cell injury. Reduced DJ-1 and S100A8 interaction was found in ATII cells in patients with emphysema. The molecular function of S100A8 was determined by an analysis of the oxidation status of its cysteine residues using chemoselective probes. Decreased S100A8 sulfination was observed in emphysema patients. In addition, its lower levels correlated with higher cell apoptosis induced by cigarette smoke extract in vitro. Cysteine at position 106 within DJ-1 is a central redox-sensitive residue. DJ-1 C106A mutant construct abolished the cytoprotective activity of DJ-1 against cell injury induced by cigarette smoke extract. Furthermore, a molecular and complementary relationship between DJ-1 and S100A8 was detected using gain- and loss-of-function studies. DJ-1 knockdown sensitized cells to apoptosis induced by cigarette smoke extract, and S100A8 overexpression provided cytoprotection in the absence of DJ-1. DJ-1 knockout mice were more susceptible to ATII cell apoptosis induced by cigarette smoke compared with wild-type mice. Our results indicate that the impairment of DJ-1 and S100A8 function may contribute to cigarette smoke-induced ATII cell injury and emphysema pathogenesis.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Karim Bahmed
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Dhanendra Tomar
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | - Sudhir Bolla
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
| | - Mark A Wilson
- Redox Biology Center and Department of Biochemistry, University of Nebraska, Lincoln, Nebraska
| | - Muniswamy Madesh
- Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, Pennsylvania
| | - Beata Kosmider
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania.,Department of Physiology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
13
|
Ross JT, Nesseler N, Lee JW, Ware LB, Matthay MA. The ex vivo human lung: research value for translational science. JCI Insight 2019; 4:128833. [PMID: 31167972 DOI: 10.1172/jci.insight.128833] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Respiratory diseases are among the leading causes of death and disability worldwide. However, the pathogenesis of both acute and chronic lung diseases remains incompletely understood. As a result, therapeutic options for important clinical problems, including acute respiratory distress syndrome and chronic obstructive pulmonary disease, are limited. Research efforts have been held back in part by the difficulty of modeling lung injury in animals. Donor human lungs that have been rejected for transplantation offer a valuable alternative for understanding these diseases. In 2007, our group developed a simple preparation of an ex vivo-perfused single human lung. In this Review, we discuss the availability of donor human lungs for research, describe the ex vivo-perfused lung preparation, and highlight how this preparation can be used to study the mechanisms of lung injury, to isolate primary cells, and to test novel therapeutics.
Collapse
Affiliation(s)
| | - Nicolas Nesseler
- Cardiovascular Research Institute, UCSF, San Francisco, California, USA.,Department of Anesthesia and Critical Care, Pontchaillou, University Hospital of Rennes, Rennes, France.,Univ Rennes, CHU de Rennes, Inra, INSERM, Institut Nutrition, Métabolismes, Cancer- UMR_A 1341, UMR_S 1241, Rennes, France.,Univ Rennes, CHU Rennes, INSERM, Centre d'Investigation Clinique de Rennes 1414, Rennes, France
| | - Jae-Woo Lee
- Department of Anesthesiology, Cardiovascular Research Institute, UCSF, San Francisco, California
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michael A Matthay
- Department of Anesthesiology, Cardiovascular Research Institute, UCSF, San Francisco, California.,Department of Medicine, Cardiovascular Research Institute, UCSF, San Francisco, California, USA
| |
Collapse
|
14
|
Everman JL, Sajuthi S, Saef B, Rios C, Stoner AM, Numata M, Hu D, Eng C, Oh S, Rodriguez-Santana J, Vladar EK, Voelker DR, Burchard EG, Seibold MA. Functional genomics of CDHR3 confirms its role in HRV-C infection and childhood asthma exacerbations. J Allergy Clin Immunol 2019; 144:962-971. [PMID: 30930175 DOI: 10.1016/j.jaci.2019.01.052] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/30/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Research in transformed immortalized cell lines indicates the cadherin-related family member 3 (CDHR3) protein serves as a receptor for human rhinovirus (HRV)-C. Similar experiments indicate that the CDHR3 coding variant rs6967330 increases CDHR3 protein surface expression. OBJECTIVE We sought to determine whether CDHR3 is necessary for HRV-C infection of primary airway epithelial cells (AECs) and to identify molecular mechanisms by which CDHR3 variants confer risk for asthma exacerbations. METHODS CDHR3 function and influence on HRV-C infection were investigated by using single-cell transcriptomics, CRISPR-Cas9 gene knockout, and genotype-specific donor experiments performed in primary AECs. Nasal airway epithelium cis-expression quantitative trait locus (eQTL) analysis of CDHR3 was performed, followed by association testing for asthma hospitalization in minority children. RESULTS CDHR3 lung expression is exclusive to ciliated AECs and associated with basal bodies during and after motile ciliogenesis. Knockout of CDHR3 in human AECs did not prevent ciliated cell differentiation but was associated with a decrease in transepithelial resistance and an 80% decrease in HRV-C infection of the mucociliary epithelium. AECs from subjects homozygous for the risk-associated rs6967330 single nucleotide polymorphism (SNP) exhibited greater HRV-C infection compared with cells homozygous for the nonrisk allele. AEC cis-eQTL analysis indicated that rs6967330 and other SNPs are eQTLs for CDHR3. Only the eQTL block containing the rs6967330 SNP showed a significant association with childhood asthma hospitalization. CONCLUSIONS Genetic deletion and genotype-specific studies in primary AECs indicate CDHR3 is critical to HRV-C infection of ciliated cells. The rs6967330 SNP confers risk of severe childhood asthma exacerbations, likely through increasing HRV-C infection levels and protein surface localization.
Collapse
Affiliation(s)
- Jamie L Everman
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo
| | - Satria Sajuthi
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo
| | - Benjamin Saef
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo
| | - Cydney Rios
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo
| | - Ari M Stoner
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, Colo
| | - Donglei Hu
- Department of Medicine, University of California-San Francisco, San Francisco, Calif
| | - Celeste Eng
- Department of Medicine, University of California-San Francisco, San Francisco, Calif
| | - Sam Oh
- Department of Medicine, University of California-San Francisco, San Francisco, Calif
| | | | - Eszter K Vladar
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colo; Department of Medicine, University of Colorado School of Medicine, Aurora, Colo; Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colo
| | | | - Esteban G Burchard
- Department of Medicine, University of California-San Francisco, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, Calif
| | - Max A Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colo; Department of Pediatrics, National Jewish Health, Denver, Colo.
| |
Collapse
|
15
|
Leach SM, Finigan J, Vasu VT, Mishra R, Ghosh M, Foster D, Mason R, Kosmider B, Farias Hesson E, Kern JA. The Kinome of Human Alveolar Type II and Basal Cells, and Its Reprogramming in Lung Cancer. Am J Respir Cell Mol Biol 2019; 61:481-491. [PMID: 30917006 DOI: 10.1165/rcmb.2018-0283oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The discovery of mutant tyrosine kinases as oncogenic drivers of lung adenocarcinomas has changed the basic understanding of lung cancer development and therapy. Yet, expressed kinases (kinome) in lung cancer progenitor cells, as well as whether kinase expression and the overall kinome changes or is reprogrammed upon transformation, is incompletely understood. We hypothesized that the kinome differs between lung cancer progenitor cells, alveolar type II cells (ATII), and basal cells (BC) and that their respective kinomes undergo distinct lineage-specific reprogramming to adenocarcinomas and squamous cell carcinomas upon transformation. We performed RNA sequencing on freshly isolated human ATII, BC, and lung cancer cell lines to define the kinome in nontransformed cells and transformed cells. Our studies identified a unique kinome for ATII and BC and changes in their kinome upon transformation to their respective carcinomas.
Collapse
Affiliation(s)
- Sonia M Leach
- Department of Biomedical Research.,Center for Genes, Environment and Health, and
| | - Jay Finigan
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Vihas T Vasu
- Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; and
| | - Rangnath Mishra
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Moumita Ghosh
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Daniel Foster
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert Mason
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Beata Kosmider
- Department of Physiology.,Department of Thoracic Medicine and Surgery, and.,Center for Inflammation, Translational and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania
| | | | - Jeffrey A Kern
- Department of Medicine, National Jewish Health, Denver, Colorado
| |
Collapse
|
16
|
Impaired non-homologous end joining in human primary alveolar type II cells in emphysema. Sci Rep 2019; 9:920. [PMID: 30696938 PMCID: PMC6351635 DOI: 10.1038/s41598-018-37000-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Emphysema is characterized by alveolar wall destruction induced mainly by cigarette smoke. Oxidative damage of DNA may contribute to the pathophysiology of this disease. We studied the impairment of the non-homologous end joining (NHEJ) repair pathway and DNA damage in alveolar type II (ATII) cells and emphysema development. We isolated primary ATII cells from control smokers, nonsmokers, and patients with emphysema to determine DNA damage and repair. We found higher reactive oxygen species generation and DNA damage in ATII cells obtained from individuals with this disease in comparison with controls. We also observed low phosphorylation of H2AX, which activates DSBs repair signaling, in emphysema. Our results indicate the impairement of NHEJ, as detected by low XLF expression. We also analyzed the role of DJ-1, which has a cytoprotective activity. We detected DJ-1 and XLF interaction in ATII cells in emphysema, which suggests the impairment of their function. Moreover, we found that DJ-1 KO mice are more susceptible to DNA damage induced by cigarette smoke. Our results suggest that oxidative DNA damage and ineffective the DSBs repair via the impaired NHEJ may contribute to ATII cell death in emphysema.
Collapse
|