1
|
Craparo EF, Cabibbo M, Scialabba C, Casula L, Lai F, Cavallaro G. Rapamycin-based inhaled therapy for potential treatment of COPD-related inflammation: production and characterization of aerosolizable nano into micro (NiM) particles. Biomater Sci 2024; 12:387-401. [PMID: 37997957 DOI: 10.1039/d3bm01210g] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Our paper describes the production and characterization of inhalable microparticles loaded with nanoparticles for the lung administration of rapamycin (Rapa). In detail, core-shell lipid/polymer hybrid nanoparticles loaded with Rapa (Rapa@Man-LPHNPs) were produced with mean size of about 128 nm and slightly negative ζ potential (-13.8 mV). A fluorescent graft polyaspartamide-poly(lactic-co-glycolic acid) copolymer (PHEA-g-RhB-g-PLGA) for use as the polymeric core was obtained by nanoprecipitation, while an appropriate mixture of DPPC and mannosylated phospholipid (DSPE-PEG2000-Man) was used to provide the macrophage-targeting lipid shell. The successful formation of Rapa@Man-LPHNPs was confirmed by TEM and DSC analyses. The loaded drug (4.3 wt% of the total weight) was slowly released from the polymeric core and protected from hydrolysis, with the amount of intact drug after 24 h of incubation in the medium being equal to 74 wt% (compared to 40% when the drug is freely incubated at the same concentration). To obtain a formulation administrable by inhalation, Rapa@Man-LPHNPs were entrapped inside PVA : LEU microparticles by using the nano into micro (NiM) strategy, specifically by spray drying (SD) in the presence of a pore-forming agent. In this way, NiM particles with geometric and theoretical aerodynamic diameters equal to 4.52 μm and 3.26 μm, respectively, were obtained. Furthermore, these particles showed optimal nebulization performance, having an FPF and an MMAD equal to 27.5% and 4.3 μm, respectively.
Collapse
Affiliation(s)
- Emanuela Fabiola Craparo
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy.
| | - Marta Cabibbo
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy.
| | - Cinzia Scialabba
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy.
| | - Luca Casula
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale, 72, 09124 Cagliari, Italy
| | - Francesco Lai
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale, 72, 09124 Cagliari, Italy
| | - Gennara Cavallaro
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy.
- Advanced Technology and Network Center (ATeN Center), University of Palermo, Palermo 90133, Italy
| |
Collapse
|
2
|
Levra S, Rosani U, Gnemmi I, Brun P, Leonardi A, Carriero V, Bertolini F, Balbi B, Profita M, Ricciardolo FLM, Di Stefano A. Impaired autophagy in the lower airways and lung parenchyma in stable COPD. ERJ Open Res 2023; 9:00423-2023. [PMID: 38111541 PMCID: PMC10726222 DOI: 10.1183/23120541.00423-2023] [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: 06/27/2023] [Accepted: 09/24/2023] [Indexed: 12/20/2023] Open
Abstract
Background There is increasing evidence of autophagy activation in COPD, but its role is complex and probably regulated through cell type-specific mechanisms. This study aims to investigate the autophagic process at multiple levels within the respiratory system, using different methods to clarify conflicting results reported so far. Methods This cross-sectional study was performed on bronchial biopsies and peripheral lung samples obtained from COPD patients (30 and 12 per sample type, respectively) and healthy controls (25 and 22 per sample type, respectively), divided by smoking history. Subjects were matched for age and smoking history. We analysed some of the most important proteins involved in autophagosome formation, such as LC3 and p62, as well as some molecules essential for lysosome function, such as lysosome-associated membrane protein 1 (LAMP1). Immunohistochemistry was used to assess the autophagic process in both sample types. ELISA and transcriptomic analysis were performed on lung samples. Results We found increased autophagic stimulus in smoking subjects, regardless of respiratory function. This was revealed by immunohistochemistry through a significant increase in LC3 (p<0.01) and LAMP1 (p<0.01) in small airway bronchiolar epithelium, alveolar septa and alveolar macrophages. Similar results were obtained in bronchial biopsy epithelium by evaluating LC3B (p<0.05), also increased in homogenate lung tissue using ELISA (p<0.05). Patients with COPD, unlike the others, showed an increase in p62 by ELISA (p<0.05). No differences were found in transcriptomics analysis. Conclusions Different techniques, applied at post-transcriptional level, confirm that cigarette smoke stimulates autophagy at multiple levels inside the respiratory system, and that autophagy failure may characterise COPD.
Collapse
Affiliation(s)
- Stefano Levra
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Umberto Rosani
- Department of Biology, University of Padova, Padua, Italy
| | - Isabella Gnemmi
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno (Novara), Italy
| | - Paola Brun
- Department of Molecular Medicine, Histology Unit, University of Padova, Padua, Italy
| | - Andrea Leonardi
- Department of Neuroscience, Ophthalmology Unit, University of Padova, Padua, Italy
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Mirella Profita
- Section of Palermo, Institute of Translational Pharmacology, National Research Council (IFT-CNR), Palermo, Italy
| | - Fabio Luigi Massimo Ricciardolo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Section of Palermo, Institute of Translational Pharmacology, National Research Council (IFT-CNR), Palermo, Italy
- Severe Asthma and Rare Lung Disease Unit, San Luigi Gonzaga University Hospital, Turin, Italy
- These authors contributed equally
| | - Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno (Novara), Italy
- These authors contributed equally
| |
Collapse
|
3
|
Albano GD, Montalbano AM, Gagliardo R, Profita M. Autophagy/Mitophagy in Airway Diseases: Impact of Oxidative Stress on Epithelial Cells. Biomolecules 2023; 13:1217. [PMID: 37627282 PMCID: PMC10452925 DOI: 10.3390/biom13081217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [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: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Autophagy is the key process by which the cell degrades parts of itself within the lysosomes. It maintains cell survival and homeostasis by removing molecules (particularly proteins), subcellular organelles, damaged cytoplasmic macromolecules, and by recycling the degradation products. The selective removal or degradation of mitochondria is a particular type of autophagy called mitophagy. Various forms of cellular stress (oxidative stress (OS), hypoxia, pathogen infections) affect autophagy by inducing free radicals and reactive oxygen species (ROS) formation to promote the antioxidant response. Dysfunctional mechanisms of autophagy have been found in different respiratory diseases such as chronic obstructive lung disease (COPD) and asthma, involving epithelial cells. Several existing clinically approved drugs may modulate autophagy to varying extents. However, these drugs are nonspecific and not currently utilized to manipulate autophagy in airway diseases. In this review, we provide an overview of different autophagic pathways with particular attention on the dysfunctional mechanisms of autophagy in the epithelial cells during asthma and COPD. Our aim is to further deepen and disclose the research in this direction to stimulate the develop of new and selective drugs to regulate autophagy for asthma and COPD treatment.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Section of Palermo, Via Ugo La Malfa 153, 90146 Palermo, Italy; (A.M.M.); (R.G.); (M.P.)
| | | | | | | |
Collapse
|
4
|
Zhang P, Jiang Y, Ye X, Zhang C, Tang Y. PDK1 inhibition reduces autophagy and cell senescence through the PI3K/AKT signalling pathway in a cigarette smoke mouse emphysema model. Exp Ther Med 2023; 25:223. [PMID: 37123206 PMCID: PMC10133799 DOI: 10.3892/etm.2023.11922] [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: 03/28/2022] [Accepted: 11/03/2022] [Indexed: 04/03/2023] Open
Abstract
A number of previous studies have demonstrated the pivotal role of PI3K/AKT signalling in cigarette smoke (CS)-induced emphysema, where phosphoinositide dependent protein kinase 1 (PDK1) is a critical component of this pathway. Therefore, the present study aimed to investigate the effects of a PDK1 inhibitor (GSK-2334470) on the expression levels of PI3K, AKT, cyclin-dependent kinase inhibitor 2A (p16) and LC3B in a CS + CS extract (CSE)-induced mouse emphysema model. CS exposure and intraperitoneal injections of CSE were combined for 4 weeks to establish an emphysema model. Mice (n=35) were randomly divided into the normal control, emphysema (CS), PI3K inhibitor (CS3) and PDK1 inhibitor (CS1) groups. Immunohistochemistry staining of lung tissues was used to measure the expression of the PI3K, PDK1 and AKT proteins in airway epithelial tissues. Immunofluorescence staining was also used to measure the levels of p16 and LC3BII protein expression in the airway epithelial tissues. In addition, PI3K, PDK1, AKT, p16 and LC3B protein expression was semi-quantified using western blotting. The expression of PDK1, PI3K and AKT proteins in the airway epithelial tissues was significantly increased in the CS + CSE group compared with that in the control group. The expression levels of p16 and LC3B were also increased as well in the CS + CSE group compared with those in the control group. The expression levels of PI3K, PDK1, AKT, LC3B and p16 in the airway epithelial tissues of the CS3 group were lower compared with those in the CS + CSE group. A decrease in the expression levels of PDK1, AKT, p16 and LC3B in the airway epithelial tissues of the CS1 group compared with those in the CS + CSE group was also observed. However, there were no significant differences in the expression levels of PI3K between the CS1 and the CS groups. The present study concluded that the inhibition of PDK1 can potentially reduce autophagy and cell senescence by downregulating the expression of PI3K/AKT pathway related proteins in airway epithelial cells, thereby protecting against CS + CSE-induced emphysema in mice.
Collapse
Affiliation(s)
- Peibei Zhang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Youjun Jiang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xianwei Ye
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Cheng Zhang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yiling Tang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
5
|
Jeong YJ, Lee KH, Woo J, Kim JY, Lee CH, Yoo CG. Downregulation of Lysosome-Associated Membrane Protein-2A Contributes to the Pathogenesis of COPD. Int J Chron Obstruct Pulmon Dis 2023; 18:289-303. [PMID: 36942278 PMCID: PMC10024500 DOI: 10.2147/copd.s378386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
Background Macroautophagy plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD), but the role of chaperone-mediated autophagy (CMA) has not been investigated. We investigated if and how CMA is involved in the pathogenesis of COPD. Methods We measured the level of lysosome-associated membrane protein-2A (LAMP-2A), which is a critical component of CMA that functions as a receptor for cytosolic substrate proteins, in total lung tissues and primary human bronchial epithelial cells (HBECs) from healthy never smokers, smokers, and COPD patients. We assessed the effects of LAMP-2A knock-down on cigarette smoke extract (CSE)-induced aging, cell cycle arrest, and apoptosis in BEAS-2B cells and the expression levels of apoptosis hallmarks in primary HBECs and lung tissue sections. Results We found that the protein levels of LAMP-2A in lung homogenates and primary HBECs from smokers and COPD patients were lower than those from never smokers. In addition, its level in primary HBECs was negatively correlated with years of smoking. CSE caused degradation of LAMP-2A protein via the lysosomal pathway by activating macroautophagy. Knock-down of LAMP-2A markedly enhanced CSE-induced expression of senescence markers such as p16, p21, p27, and p53. G2/M cell cycle arrest, up-regulation of cyclin B1, and apoptosis in BEAS-2B cells. Apoptosis was increased in CSE-treated primary HBECs and in lung tissues from smokers and COPD patients. Conclusion Cigarette smoke-induced down-regulation of LAMP-2A is involved in acceleration of aging and apoptosis of lung epithelial cells, which might at least partially contribute to COPD pathogenesis.
Collapse
Affiliation(s)
- Yun-Jeong Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongguk University Ilsan Hospital, Ilsan Dong-gu, Goyang-si, Gyeonggi-do, Korea
| | - Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jisu Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Yeon Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Correspondence: Chul-Gyu Yoo, 101 Daehakno, Jongno-gu, Seoul, 03080, Korea, Tel +82-2-2072-3760, Fax +82-2-762-9662, Email
| |
Collapse
|
6
|
Fang L, Zhang M, Li J, Zhou L, Tamm M, Roth M. Airway Smooth Muscle Cell Mitochondria Damage and Mitophagy in COPD via ERK1/2 MAPK. Int J Mol Sci 2022; 23:ijms232213987. [PMID: 36430467 PMCID: PMC9694999 DOI: 10.3390/ijms232213987] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by irreversible deterioration of the airway wall. Cigarette smoking is the major trigger, and in vitro studies showed that cigarette smoke extract (CSE) induced mitophagy in airway epithelial cells via oxidative stress, but this mechanism was not studied in airway smooth muscle cells (ASMCs). Primary ASMCs isolated from COPD patients or non-disease donors were investigated for CSE-induced remodeling and mitochondria structure. Proteins were assessed by Western blots for remodeling: collagen type-I, α-smooth muscle actin (α-SMA) and fibronectin; autophagy: beclin-1, protein62 (p62), light chain (LC)3A/B; mitochondria activity: mitochondrially encoded cytochrome c oxidase II & -IV (MTCO2, MTCO4), peroxisome proliferator activated receptor gamma coactivator 1α (PGC-1α); lysosomes: early endosome antigen 1, lysosome activated membrane protein 1; and cell signaling: extracellular signal regulated kinase (ERK1/2). Lysotracker and Mitotracker were used to monitor mitochondria morphology and organelle co-localization. Compared with controls, untreated COPD ASMCs showed lower collagen type-I and α-SMA expressions, but increased fibronectin levels. CSE further downregulated collagen type-I and α-SMA expression, but upregulated fibronectin. CSE decreased PGC-1α, MTCO2, and MTCO4, but increased beclin-1, p62, and LC3. CSE upregulated mitophagy and lysosomes activity via ERK1/2 phosphorylation. In vitro, cigarette smoke induced the deterioration of ASMCs, which might explain the tissue loss and structural remodeling in COPD bronchi. The results suggest that preventing exceeded mitophagy in ASMCs might present a novel therapeutic target for COPD.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Clinic of Respiratory Medicine, Department of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Ming Zhang
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710049, China
| | - Junling Li
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan 523000, China
| | - Liang Zhou
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Clinic of Respiratory Medicine, Department of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Michael Tamm
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Clinic of Respiratory Medicine, Department of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Clinic of Respiratory Medicine, Department of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
7
|
Chen J, Yao Y, Wang Y, Wang X, Peng X, Li T, Liu Y, Du J. Autophagy triggered by the ROS/ERK signaling pathway protects mouse embryonic palatal cells from apoptosis induced by nicotine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:81909-81922. [PMID: 35739442 DOI: 10.1007/s11356-022-21496-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/12/2022] [Indexed: 06/15/2023]
Abstract
Maternal cigarette smoking during pregnancy is a known high-risk factor for having a child with a cleft lip and/or palate (CLP), a common congenital malformation. Nicotine is the major teratogen component of cigarettes and e-cigarettes, and nicotine plays an important role in the development of CLP. However, the mechanism underlying nicotine's effect on CLP remains unclear. Here, we aimed to determine the role and molecular mechanisms of nicotine-induced autophagy, an important process involved in regulating the cellular stress response in mouse embryonic palatal cells (MEPCs). First, we found that nicotine promoted MEPCs proliferation and inhibited their apoptosis from 0 to 12 h. After 12 h, the proliferation was inhibited, and apoptosis was promoted. The migration of MEPCs was also inhibited by nicotine. Simultaneously, long-term nicotine stimulation inhibited the osteogenic differentiation of MEPCs. We then found that nicotine significantly increased autophagy flux in MEPCs at 12 h by increasing the expression of microtubule-associated protein light chain 3 (LC3) and reducing P62 expression levels. After nicotine exposure, intracellular reactive oxygen species (ROS) and extracellular signal-regulated kinase-1/2 (ERK1/2) expression significantly increased, and the expression of ERK1/2 was reversed by the ROS scavenging agent N-acetylcysteine (NAC). Moreover, the autophagy induced by nicotine was reversed by SCH772984, a specific inhibitor of ERK1/2, and the autophagy inhibitor chloroquine (CQ). These results suggest that in the early stage of nicotine exposure, MEPCs may trigger autophagy through the ROS/ERK1/2 signaling pathway to avoid cell damage caused by nicotine.
Collapse
Affiliation(s)
- Jing Chen
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yaxia Yao
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yijia Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Xiaotong Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Xia Peng
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Tianli Li
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Ying Liu
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Juan Du
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.
| |
Collapse
|
8
|
Environmental Exposures and Lung Aging: Molecular Mechanisms and Implications for Improving Respiratory Health. Curr Environ Health Rep 2021; 8:281-293. [PMID: 34735706 PMCID: PMC8567983 DOI: 10.1007/s40572-021-00328-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Inhaled environmental exposures cause over 12 million deaths per year worldwide. Despite localized efforts to reduce environmental exposures, tobacco smoking and air pollution remain the urgent public health challenges that are contributing to the growing prevalence of respiratory diseases. The purpose of this review is to describe the mechanisms through which inhaled environmental exposures accelerate lung aging and cause overt lung disease. RECENT FINDINGS Environmental exposures related to fossil fuel and tobacco combustion and occupational exposures related to silica and coal mining generate oxidative stress and inflammation in the lungs. Sustained oxidative stress causes DNA damage, epigenetic instability, mitochondrial dysfunction, and cell cycle arrest in key progenitor cells in the lung. As a result, critical repair mechanisms are impaired, leading to premature destruction of the lung parenchyma. Inhaled environmental exposures accelerate lung aging by injuring the lungs and damaging the cells responsible for wound healing. Interventions that minimize exposure to noxious antigens are critical to improve lung health, and novel research is required to expand our knowledge of therapies that may slow or prevent premature lung aging.
Collapse
|
9
|
Zhang Q, Li W, Ayidaerhan N, Han W, Chen Y, Song W, Yue Y. IP 3 R attenuates oxidative stress and inflammation damage in smoking-induced COPD by promoting autophagy. J Cell Mol Med 2021; 25:6174-6187. [PMID: 34060199 PMCID: PMC8256356 DOI: 10.1111/jcmm.16546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/14/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Tobacco smoking is one of the most important risk factors for chronic obstructive pulmonary disease (COPD). However, the most critical genes and proteins remain poorly understood. Therefore, we aimed to investigate these hub genes and proteins in tobacco smoke-induced COPD, together with the potential mechanism(s). Differentially expressed genes (DEGs) were analysed between smokers and patients with COPD. mRNA expression and protein expression of IP3 R were confirmed in patients with COPD and extracted smoke solution (ESS)-treated human bronchial epithelial (HBE) cells. Moreover, expression of oxidative stress, inflammatory cytokines and/or autophagy-related protein was tested when IP3 R was silenced or overexpressed in ESS-treated and/or 3-MA-treated cells. A total of 30 DEGs were obtained between patients with COPD and smoker samples. IP3 R was identified as one of the key targets in tobacco smoke-induced COPD. In addition, IP3 R was significantly decreased in patients with COPD and ESS-treated cells. Loss of IP3 R statistically increased expression of oxidative stress and inflammatory cytokines in ESS-treated HBE cells, and overexpression of IP3 R reversed the above functions. Furthermore, the autophagy-related proteins (Atg5, LC3 and Beclin1) were statistically decreased, and p62 was increased by silencing of IP3 R cells, while overexpression of IP3 R showed contrary results. Additionally, we detected that administration of 3-MA significantly reversed the protective effects of IP3 R overexpression on ESS-induced oxidative stress and inflammatory injury. Our results suggest that IP3 R might exert a protective role against ESS-induced oxidative stress and inflammation damage in HBE cells. These protective effects might be associated with promoting autophagy.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Wei Li
- Key Laboratory of Intelligent Computing in Medical ImageMinistry of EducationNortheastern UniversityShenyangChina
| | - Nahemuguli Ayidaerhan
- Department of Pulmonary and Critical Care MedicineTarbagatay Prefecture People’s HospitalTachengChina
| | - Wuxin Han
- Department of Clinical LaboratoryTarbagatay Prefecture People’s HospitalTachengChina
| | - Yingying Chen
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Wei Song
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Yuanyi Yue
- Department of Gastroenterology MedicineShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
10
|
Schneider JL, Rowe JH, Garcia-de-Alba C, Kim CF, Sharpe AH, Haigis MC. The aging lung: Physiology, disease, and immunity. Cell 2021; 184:1990-2019. [PMID: 33811810 PMCID: PMC8052295 DOI: 10.1016/j.cell.2021.03.005] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/01/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
The population is aging at a rate never seen before in human history. As the number of elderly adults grows, it is imperative we expand our understanding of the underpinnings of aging biology. Human lungs are composed of a unique panoply of cell types that face ongoing chemical, mechanical, biological, immunological, and xenobiotic stress over a lifetime. Yet, we do not fully appreciate the mechanistic drivers of lung aging and why age increases the risk of parenchymal lung disease, fatal respiratory infection, and primary lung cancer. Here, we review the molecular and cellular aspects of lung aging, local stress response pathways, and how the aging process predisposes to the pathogenesis of pulmonary disease. We place these insights into context of the COVID-19 pandemic and discuss how innate and adaptive immunity within the lung is altered with age.
Collapse
Affiliation(s)
- Jaime L Schneider
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Boston, MA 02115, USA; Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Jared H Rowe
- Division of Hematology Boston Children's Hospital and Division of Pediatric Oncology Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Carolina Garcia-de-Alba
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Carla F Kim
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Disease, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
EVs from BALF-Mediators of Inflammation and Potential Biomarkers in Lung Diseases. Int J Mol Sci 2021; 22:ijms22073651. [PMID: 33915715 PMCID: PMC8036254 DOI: 10.3390/ijms22073651] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have been identified as key messengers of intracellular communication in health and disease, including the lung. EVs that can be found in bronchoalveolar lavage fluid (BALF) are released by multiple cells of the airways including bronchial epithelial cells, endothelial cells, alveolar macrophages, and other immune cells, and they have been shown to mediate proinflammatory signals in many inflammatory lung diseases. They transfer complex molecular cargo, including proteins, cytokines, lipids, and nucleic acids such as microRNA, between structural cells such as pulmonary epithelial cells and innate immune cells such as alveolar macrophages, shaping mutually their functions and affecting the alveolar microenvironment homeostasis. Here, we discuss this distinct molecular cargo of BALF-EVs in the context of inducing and propagating inflammatory responses in particular acute and chronic lung disorders. We present different identified cellular interactions in the inflammatory lung via EVs and their role in lung pathogenesis. We also summarize the latest studies on the potential use of BALF-EVs as diagnostic and prognostic biomarkers of lung diseases, especially of lung cancer.
Collapse
|
12
|
Buyuan decoction inhibits autophagy in a rat model of chronic obstructive pulmonary disease. ARCH BIOL SCI 2021. [DOI: 10.2298/abs211104047h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Efforts have been made to find a better therapeutic approach with fewer side
effects in treating chronic obstructive pulmonary disease (COPD). This study
investigated the effect of Buyuan decoction (BYD) on autophagy in COPD rats.
An experimental model with Sprague-Dawley rats was established by
lipopolysaccharide (LPS) injection and cigarette smoke exposure. Rats were
randomly allocated into blank control (normal control), experimental model,
low-dose BYD (8.0 g/kg/day), medium-dose BYD (16.0 g/kg/day), high-dose BYD
(32.0 g/kg/day) and 3-MA (methyladenine) groups (6 rats/group). Cell and
tissue morphology were observed using hematoxylin and eosin staining.
Autophagic vesicles were examined with a transmission electron microscope.
Protein expression of LC3-II/I, BNIP-1, ATG7, p62, PI3K and p-PI3K in lung
tissue was detected by Western blotting. Compared with the experimental
model group, the inflammatory infiltrate in lung tissue was reduced, the
nuclei of the pulmonary epithelial cells were restored to normal, and the
expression of LC3, BNIP1, ATG7 and p-PI3K was significantly downregulated,
while p62 expression was significantly upregulated after treatment with the
BYD. The effect was most significant in the lowdose BYD group (P<0.05, all
groups). These findings suggest that the BYD inhibits the occurrence of
autophagy in the pathogenesis of COPD and that it can be a potential
treatment.
Collapse
|
13
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
14
|
Wang SY, Ni X, Hu KQ, Meng FL, Li M, Ma XL, Meng TT, Wu HH, Ge D, Zhao J, Li Y, Su GH. Cilostazol alleviate nicotine induced cardiomyocytes hypertrophy through modulation of autophagy by CTSB/ROS/p38MAPK/JNK feedback loop. Int J Biol Sci 2020; 16:2001-2013. [PMID: 32398966 PMCID: PMC7211170 DOI: 10.7150/ijbs.43825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022] Open
Abstract
Nicotine is proved to be an important factor for cardiac hypertrophy. Autophagy is important cell recycling system involved in the regulation of cardiac hypertrophy. Cilostazol, which is often used in the management of peripheral vascular disease. However, the effects of cilostazol on nicotine induced autophagy and cardiac hypertrophy are unclear. Here, we aim to determine the role and molecular mechanism of cilostazol in alleviating nicotine-induced cardiomyocytes hypertrophy through modulating autophagy and the underlying mechanisms. Our results clarified that nicotine stimulation caused cardiomyocytes hypertrophy and autophagy flux impairment significantly in neonatal rat ventricular myocytes (NRVMs), which were evidenced by augments of LC3-II and p62 levels, and impaired autophagosomes clearance. Interestingly, cathepsin B (CTSB) activity decreased dramatically after stimulation with nicotine in NRVMs, which was crucial for substrate degradation in the late stage of autophagy process, and cilostazol could reverse this effect dramatically. Intracellular ROS levels were increased significantly after nicotine exposure. Meanwhile, p38MAPK and JNK were activated after nicotine treatment. By using ROS scavenger N-acetyl-cysteine (NAC) could reverse the effects of nicotine by down-regulation the phosphorylation of p38MAPK and JNK pathways, and pretreatment of specific inhibitors of p38MAPK and JNK could restore the autophagy impairment and cardiomyocytes hypertrophy induced by nicotine. Moreover, CTSB activity of lysosome regained after the treatment with cilostazol. Cilostazol also inhibited the ROS accumulation and the activation of p38MAPK and JNK, which providing novel connection between lysosome CTSB and ROS/p38MAPK/JNK related oxidative stress pathway. This is the first demonstration that cilostazol could alleviate nicotine induced cardiomyocytes hypertrophy through restoration of autophagy flux by activation of CTSB and inhibiting ROS/p38/JNK pathway, exhibiting a feedback loop on regulation of autophagy and cardiomyocytes hypertrophy.
Collapse
Affiliation(s)
- Shu-Ya Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xi Ni
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ke-Qing Hu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fan-Liang Meng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xiao-Li Ma
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ting-Ting Meng
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Hui-Hui Wu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, China
| | - Jing Zhao
- Development Biology, School of Life Science, Shandong University, Jinan, China
| | - Ying Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guo-Hai Su
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
15
|
Morsch ALBC, Wisniewski E, Luciano TF, Comin VH, Silveira GDB, Marques SDO, Thirupathi A, Silveira Lock PC, De Souza CT. Cigarette smoke exposure induces ROS-mediated autophagy by regulating sestrin, AMPK, and mTOR level in mice. Redox Rep 2020; 24:27-33. [PMID: 30957679 PMCID: PMC6748578 DOI: 10.1080/13510002.2019.1601448] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many pathological conditions linked to cigarette smoking are caused by the
production of reactive oxygen species (ROS). The present study was conducted to
analyze the effect of ROS on the lungs of Swiss mice exposed to cigarette
smoking, focusing on autophagy-mediated mechanisms, and investigate the
involvement of SESN2, AMPK, and mTOR signaling. Mice were exposed to cigarette
smoke (CS) for 7, 15, 30, 45, and 60 days; the control group was not exposed to
CS. Only mice exposed to CS for 45 days were selected for subsequent
N-acetylcysteine (NAC) supplementation and smoke cessation
analyses. Exposure to CS increased the production of ROS and induced molecular
changes in the autophagy pathway, including an increase in phosphorylated AMPK
and ULK1, reduction in phosphorylated mTOR, and increases in SESN2, ATG12, and
LC3B levels. NAC supplementation reduced ROS levels and reversed all molecular
changes observed upon CS treatment, suggesting the involvement of oxidative
stress in inducing autophagy upon CS exposure. When exposure to CS was stopped,
there were decreases in the levels of oxidative stress, AMPK and ULK1
phosphorylation, and autophagy-initiating molecules and increase in mTOR
phosphorylation. In conclusion, these results suggest the involvement of ROS,
SESN2, AMPK, and mTOR in the CS-induced autophagic process in the lung.
Collapse
Affiliation(s)
- Ana Lucia Bernardo Carvalho Morsch
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Elvis Wisniewski
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Thais Fernandes Luciano
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Vitor Hugo Comin
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Gustavo de Bem Silveira
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Scherolin de Oliveira Marques
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Anand Thirupathi
- b Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology , Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University , Shijiazhuang , People's Republic of China
| | - Paulo Cesar Silveira Lock
- a Laboratory of Exercise Biochemistry and Physiology , Graduate Program in Health Sciences, Extremo Sul Catarinense University , Criciúma , Brazil
| | - Claudio Teodoro De Souza
- c Department of Internal Medicine , Medicine School, Federal University of Juiz de Fora , Juiz de Fora , Brazil
| |
Collapse
|
16
|
Zhang L, Luo B, Ting Y, He S, Xie L, Sun S. SIRT1 attenuates endoplasmic reticulum stress and apoptosis in rat models of COPD. Growth Factors 2020; 38:94-104. [PMID: 32819170 DOI: 10.1080/08977194.2020.1810029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The present study aimed to investigate the protective role of sirtuin 1 (SIRT1) and oxygen regulated protein 150 (ORP150) in a rat COPD model by inducing changes in ER stress and apoptosis. We separated 48 Sprague Dawley (SD) rats into four groups randomly: the control group, resveratrol group, COPD group and the resveratrol intervention group. Rats were challenged with cigarette smoke and lipopolysaccharide with resveratrol (a selective activator of SIRT1). The lung functions of the rats were measured and recorded. The expression levels of SIRT1 and ORP150 in lung tissues were examined by western blot and RTq PCR. The expression levels of the ER stress apoptosis-associated protein were determined .The apoptotic level of lung tissues was analyzed. The results suggest that SIRT1 attenuated apoptosis and ER stress in the lung tissues of rats with COPD. During this process, a positive correlation was identified between SIRT1 and ORP150.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Bailing Luo
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Yuan Ting
- Department of Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Shengyang He
- Department of Respiratory Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Lihua Xie
- Department of Respiratory Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Shenghua Sun
- Department of Respiratory Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| |
Collapse
|
17
|
Zhang L, Huang J, Dong R, Feng Y, Zhou M. Therapeutic potential of BLT1 antagonist for COPD: involvement of inducing autophagy and ameliorating inflammation. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3105-3116. [PMID: 31564828 PMCID: PMC6732561 DOI: 10.2147/dddt.s215433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/10/2019] [Indexed: 12/22/2022]
Abstract
Purpose Leukotriene B4 (LTB4) is a major pro-inflammatory mediator that leads to the persistence of chronic inflammation in chronic obstructive pulmonary disease (COPD). The purpose of this study was to evaluate therapeutic potential of BLT1 antagonist for cigarette smoke (CS)-induced COPD and to explore the underlying mechanism. Materials and methods In vitro, autophagy proteins were determined by Western blotting in RAW264.7 macrophages treated with U75302 (BLT1 antagonist) or autophagy inhibitor in cigarette smoke extract-induced inflammation. In vivo, C57BL/6J mice were randomly divided into three groups: Control group, CS group and CS+U75302 group. After 12-week exposure, histological analysis and lung function tests were performed to evaluate the inflammatory infiltration and emphysema. The expression of inflammatory cytokines was measured by real-time PCR and enzyme-linked immunosorbent assay. Immunohistochemical analysis and Western blotting detected the expression of autophagy-related proteins. Transmission electron microscopy (TEM) showed the alterations of autophagosomes and lysosomes. Results Lower levels of inflammatory factors and autophagy markers were detected in U75302-treated cells and mice after CS exposure than control. In vitro, LC3 mRNA expression was elevated when treated with U75302. Autophagy inhibition resulted in augmented inflammatory response and autophagy proteins even with U75302 treatment. Furthermore, BLT1 antagonist decreased the number of lysosomes and autophagosomes in alveolar macrophages of mice and potentially enhanced the expression of transcriptional activation of transcription factor-EB (TFEB) in vitro and vivo. Conclusion Insufficient autophagy of macrophages was associated with LTB4-mediated inflammation in CS-exposure models. BLT1 antagonist ameliorated inflammatory response through inducing autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jingwen Huang
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ran Dong
- Department of Respiratory Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yun Feng
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Jiang S, Sun J, Mohammadtursun N, Hu Z, Li Q, Zhao Z, Zhang H, Dong J. Dual role of autophagy/mitophagy in chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2019; 56:116-125. [DOI: 10.1016/j.pupt.2019.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/18/2019] [Accepted: 04/07/2019] [Indexed: 02/06/2023]
|
19
|
Yoon YS, Jin M, Sin DD. Accelerated lung aging and chronic obstructive pulmonary disease. Expert Rev Respir Med 2019; 13:369-380. [PMID: 30735057 DOI: 10.1080/17476348.2019.1580576] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The prevalence of chronic obstructive pulmonary disease (COPD) increases exponentially with aging. Its pathogenesis, however, is not well known and aside from smoking cessation, there are no disease-modifying treatments for this disease. Areas covered: COPD is associated with accelerating aging and aging-related diseases. In this review, we will discuss the hallmarks of aging including genomic instability, telomere attrition, epigenetic alteration, loss of proteostasis, mitochondrial dysfunction, deregulated nutrient sensing, cellular senescence, stem cell exhaustion, and altered intercellular communication, which may be involved in COPD pathogenesis. Expert commentary: COPD and the aging process share similar molecular and cellular changes. Aging-related molecular pathways may represent novel therapeutic targets and biomarkers for COPD.
Collapse
Affiliation(s)
- Young Soon Yoon
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , BC , Canada.,b Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine , Dongguk University Ilsan Hospital , Goyang , South Korea
| | - Minhee Jin
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , BC , Canada
| | - Don D Sin
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , BC , Canada.,c Division of Respiratory Medicine (Department of Medicine) , University of British Columbia , Vancouver , BC , Canada
| |
Collapse
|
20
|
Bodas M, Vij N. Adapting Proteostasis and Autophagy for Controlling the Pathogenesis of Cystic Fibrosis Lung Disease. Front Pharmacol 2019; 10:20. [PMID: 30774592 PMCID: PMC6367269 DOI: 10.3389/fphar.2019.00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis (CF), a fatal genetic disorder predominant in the Caucasian population, is caused by mutations in the cystic fibrosis transmembrane conductance regulator (Cftr) gene. The most common mutation is the deletion of phenylalanine from the position-508 (F508del-CFTR), resulting in a misfolded-CFTR protein, which is unable to fold, traffic and retain its plasma membrane (PM) localization. The resulting CFTR dysfunction, dysregulates variety of key cellular mechanisms such as chloride ion transport, airway surface liquid (ASL) homeostasis, mucociliary-clearance, inflammatory-oxidative signaling, and proteostasis that includes ubiquitin-proteasome system (UPS) and autophagy. A collective dysregulation of these key homoeostatic mechanisms contributes to the development of chronic obstructive cystic fibrosis lung disease, instead of the classical belief focused exclusively on ion-transport defect. Hence, therapeutic intervention(s) aimed at rescuing chronic CF lung disease needs to correct underlying defect that mediates homeostatic dysfunctions and not just chloride ion transport. Since targeting all the myriad defects individually could be quite challenging, it will be prudent to identify a process which controls almost all disease-promoting processes in the CF airways including underlying CFTR dysfunction. There is emerging experimental and clinical evidence that supports the notion that impaired cellular proteostasis and autophagy plays a central role in regulating pathogenesis of chronic CF lung disease. Thus, correcting the underlying proteostasis and autophagy defect in controlling CF pulmonary disease, primarily via correcting the protein processing defect of F508del-CFTR protein has emerged as a novel intervention strategy. Hence, we discuss here both the rationale and significant therapeutic utility of emerging proteostasis and autophagy modulating drugs/compounds in controlling chronic CF lung disease, where targeted delivery is a critical factor-influencing efficacy.
Collapse
Affiliation(s)
- Manish Bodas
- Department of Medicine, University of Oklahoma, Oklahoma City, OK, United States
| | - Neeraj Vij
- Department of Pediatric Pulmonary Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- 4Dx Limited, Los Angeles, CA, United States
- VIJ Biotech LLC, Baltimore, MD, United States
| |
Collapse
|
21
|
Bodas M, Pehote G, Silverberg D, Gulbins E, Vij N. Autophagy augmentation alleviates cigarette smoke-induced CFTR-dysfunction, ceramide-accumulation and COPD-emphysema pathogenesis. Free Radic Biol Med 2019; 131:81-97. [PMID: 30500419 DOI: 10.1016/j.freeradbiomed.2018.11.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 01/27/2023]
Abstract
In this study, we aimed to investigate precise mechanism(s) of sphingolipid-imbalance and resulting ceramide-accumulation in COPD-emphysema. Where, human and murine emphysema lung tissues or human bronchial epithelial cells (Beas2b) were used for experimental analysis. We found that lungs of smokers and COPD-subjects with increasing emphysema severity demonstrate sphingolipid-imbalance, resulting in significant ceramide-accumulation and increased ceramide/sphingosine ratio, as compared to non-emphysema/non-smoker controls. Next, we found a substantial increase in emphysema chronicity-related ceramide-accumulation in murine (C57BL/6) lungs, while sphingosine levels only slightly increased. In accordance, the expression of the acid ceramidase decreased after CS-exposure. Moreover, CS-induced (sub-chronic) ceramide-accumulation was significantly (p < 0.05) reduced by treatment with TFEB/autophagy-inducing drug, gemfibrozil (GEM), suggesting that autophagy regulates CS-induced ceramide-accumulation. Next, we validated experimentally that autophagy/lipophagy-induction using an anti-oxidant, cysteamine, significantly (p < 0.05) reduces CS-extract (CSE)-mediated intracellular-ceramide-accumulation in p62 + aggresome-bodies. In addition to intracellular-accumulation, we found that CSE also induces membrane-ceramide-accumulation by ROS-dependent acid-sphingomyelinase (ASM) activation and plasma-membrane translocation, which was significantly controlled (p < 0.05) by cysteamine (an anti-oxidant) and amitriptyline (AMT, an inhibitor of ASM). Cysteamine-mediated and CSE-induced membrane-ceramide regulation was nullified by CFTR-inhibitor-172, demonstrating that CFTR controls redox impaired-autophagy dependent membrane-ceramide accumulation. In summary, our data shows that CS-mediated autophagy/lipophagy-dysfunction results in intracellular-ceramide-accumulation, while acquired CFTR-dysfunction-induced ASM causes membrane ceramide-accumulation. Thus, CS-exposure alters the sphingolipid-rheostat leading to the increased membrane- and intracellular- ceramide-accumulation inducing COPD-emphysema pathogenesis that is alleviated by treatment with cysteamine, a potent anti-oxidant with CFTR/autophagy-augmenting properties.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - Garrett Pehote
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - David Silverberg
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - Erich Gulbins
- Dept. of Molecular Biology, University of Duisburg-Essen, Germany and Dept. of Surgery, University of Cincinnati, OH, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA; The Johns Hopkins University SOM University, Baltimore, MD, USA; VIJ Biotech LLC, Baltimore, MD, USA and 4Dx Ltd, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Stincardini C, Renga G, Villella V, Pariano M, Oikonomou V, Borghi M, Bellet MM, Sforna L, Costantini C, Goldstein AL, Garaci E, Romani L. Cellular proteostasis: a new twist in the action of thymosin α1. Expert Opin Biol Ther 2018; 18:43-48. [DOI: 10.1080/14712598.2018.1484103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Valeria Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M. Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Allan L. Goldstein
- Department of Biochemistry and Molecular Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
Wu Y, Xu B, He X, Wu B, Li Y, Yu G, Tan C, Wang H. Correlation between autophagy levels in peripheral blood mononuclear cells and clinical parameters in patients with chronic obstructive pulmonary disease. Mol Med Rep 2018; 17:8003-8009. [PMID: 29620199 DOI: 10.3892/mmr.2018.8831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 03/02/2018] [Indexed: 11/05/2022] Open
Abstract
Autophagy serves a role in the pathogenesis of chronic inflammatory diseases. The aim of the present study was to compare the autophagy levels in the peripheral blood mononuclear cells (PBMCs) of patients with chronic obstructive pulmonary disease (COPD) and healthy individuals and to assess the association between autophagy and the clinical parameters of COPD. Samples of peripheral blood from 20 patients with stable COPD and 20 healthy controls were collected. PBMCs were harvested using Ficoll density gradient centrifugation. Levels of the autophagy‑associated proteins ubiquitin‑binding protein p62 (p62), microtubule‑associated proteins 1A/1B light chain 3A (LC3I/II) and beclin‑1 in PBMCs were detected by western blotting. Enzyme‑linked immunosorbent assay kits were used to detect the serum concentrations of interleukin (IL)‑6, IL‑8 and tumor necrosis factor (TNF)‑α. Associations between the levels of autophagy and forced expiratory volume in 1 sec % predicted (FEV1%) and pro‑inflammatory factors were assessed. Western blotting demonstrated that the protein expression of p62 was decreased, but LC3II/I and beclin‑1 levels increased in patients with COPD compared with healthy controls. Serum levels of IL‑6, IL‑8 and TNF‑α were increased in patients with COPD. The extent of PBMC autophagy was negatively correlated with FEV1% predicted, but positively correlated with levels of pro‑inflammatory cytokines. The levels of autophagy in PBMCs in patients with COPD were increased and were negatively correlated with FEV1% predicted and positively correlated with circulating levels of pro‑inflammatory cytokines. Autophagy may serve a role as a biomarker of the severity of COPD or as a therapeutic target for treatment of COPD.
Collapse
Affiliation(s)
- Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Bo Xu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xin He
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Bo Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yunxiao Li
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Chunting Tan
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
24
|
Schlemmer F, Boyer L, Soumagne T, Ridoux A, Chouaid C, Maitre B, Lanone S, Adnot S, Audureau E, Boczkowski J. Beclin1 circulating levels and accelerated aging markers in COPD. Cell Death Dis 2018; 9:156. [PMID: 29402890 PMCID: PMC5833829 DOI: 10.1038/s41419-017-0178-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Frédéric Schlemmer
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0001 2292 1474grid.412116.1Unité de Pneumologie, APHP, Hôpital Henri Mondor, DHU-ATVB, Créteil, France
| | - Laurent Boyer
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0001 2292 1474grid.412116.1Département de Physiologie-Explorations Fonctionnelles, APHP, Hôpital Henri Mondor, DHU-ATVB, Créteil, France
| | - Thibaud Soumagne
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France
| | - Audrey Ridoux
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France
| | - Christos Chouaid
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0004 1765 2136grid.414145.1Département de Pneumologie et Pathologie Professionnelle, Centre Hospitalier Intercommunal, DHU-ATVB, Créteil, France
| | - Bernard Maitre
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0001 2292 1474grid.412116.1Unité de Pneumologie, APHP, Hôpital Henri Mondor, DHU-ATVB, Créteil, France ,0000 0004 1765 2136grid.414145.1Département de Pneumologie et Pathologie Professionnelle, Centre Hospitalier Intercommunal, DHU-ATVB, Créteil, France
| | - Sophie Lanone
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France
| | - Serge Adnot
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0001 2292 1474grid.412116.1Département de Physiologie-Explorations Fonctionnelles, APHP, Hôpital Henri Mondor, DHU-ATVB, Créteil, France
| | - Etienne Audureau
- 0000 0001 2292 1474grid.412116.1Département de Santé Publique, Unité de Recherche Clinique (URC-Mondor), APHP, Hôpital Henri Mondor, IMRB EA7376, Clinical Epidemiology and Aging (CEpiA), Créteil, France
| | - Jorge Boczkowski
- 0000 0004 0386 3258grid.462410.5INSERM U955, Institut Mondor de Recherche Biomédicale and Université Paris Est-Créteil (UPEC), Faculté de Médecine, Créteil, France ,0000 0001 2292 1474grid.412116.1Unité de Pneumologie, APHP, Hôpital Henri Mondor, DHU-ATVB, Créteil, France
| |
Collapse
|
25
|
Pehote G, Bodas M, Brucia K, Vij N. Cigarette Smoke Exposure Inhibits Bacterial Killing via TFEB-Mediated Autophagy Impairment and Resulting Phagocytosis Defect. Mediators Inflamm 2017; 2017:3028082. [PMID: 29445254 PMCID: PMC5763241 DOI: 10.1155/2017/3028082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/03/2017] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Cigarette smoke (CS) exposure is the leading risk factor for COPD-emphysema pathogenesis. A common characteristic of COPD is impaired phagocytosis that causes frequent exacerbations in patients leading to increased morbidity. However, the underlying mechanism is unclear. Hence, we investigated if CS exposure causes autophagy impairment as a mechanism for diminished bacterial clearance via phagocytosis by utilizing murine macrophages (RAW264.7 cells) and Pseudomonas aeruginosa (PA01-GFP) as an experimental model. METHODS Briefly, RAW cells were treated with cigarette smoke extract (CSE), chloroquine (autophagy inhibitor), TFEB-shRNA, CFTR(inh)-172, and/or fisetin prior to bacterial infection for functional analysis. RESULTS Bacterial clearance of PA01-GFP was significantly impaired while its survival was promoted by CSE (p < 0.01), autophagy inhibition (p < 0.05; p < 0.01), TFEB knockdown (p < 0.01; p < 0.001), and inhibition of CFTR function (p < 0.001; p < 0.01) in comparison to the control group(s) that was significantly recovered by autophagy-inducing antioxidant drug, fisetin, treatment (p < 0.05; p < 0.01; and p < 0.001). Moreover, investigations into other pharmacological properties of fisetin show that it has significant mucolytic and bactericidal activities (p < 0.01; p < 0.001), which warrants further investigation. CONCLUSIONS Our data suggests that CS-mediated autophagy impairment as a critical mechanism involved in the resulting phagocytic defect, as well as the therapeutic potential of autophagy-inducing drugs in restoring is CS-impaired phagocytosis.
Collapse
Affiliation(s)
- Garrett Pehote
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Kathryn Brucia
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|