1
|
Yue W, Yupeng G, Jun C, Kui J. Apatinib combined with olaparib induces ferroptosis via a p53-dependent manner in ovarian cancer. J Cancer Res Clin Oncol 2023; 149:8681-8689. [PMID: 37120435 DOI: 10.1007/s00432-023-04811-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
OBJECTIVE PARP inhibitors combined with antiangiogenic drugs have been reported to improve outcomes in BRCA wild-type ovarian cancer patients, the mechanism of the combination is unclear. In this study, we explored the mechanism of apatinib combined with olaparib in the treatment of ovarian cancer. METHODS In this study, human ovarian cancer cell lines A2780 and OVCAR3 were used as experimental objects, and the expression of ferroptosis-related protein GPX4 after treatment with apatinib and olaparib was detected by Western blot. The SuperPred database was used to predict the target of the combined action of apatinib and olaparib, and the predicted results were verified by Western blot experiment to explore the mechanism of ferroptosis induced by apatinib and olaparib. RESULTS Apatinib combined with olaparib-induced ferroptosis in p53 wild-type cells, and p53 mutant cells developed drug resistance. The p53 activator RITA sensitized drug-resistant cells to ferroptosis induced by apatinib combined with olaparib. Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. CONCLUSION This discovery revealed the specific mechanism of ferroptosis induced by apatinib combined with olaparib in p53 wild-type ovarian cancer cells and provided a theoretical basis for the clinical combined use of apatinib and olaparib in p53 wild-type ovarian cancer patients.
Collapse
Affiliation(s)
- Wang Yue
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Gu Yupeng
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Cao Jun
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| | - Jiang Kui
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
2
|
Retraction: Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2. PLoS One 2023; 18:e0290862. [PMID: 37624835 PMCID: PMC10456146 DOI: 10.1371/journal.pone.0290862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023] Open
|
3
|
Zhuang H, Li N, Chen S, Shen Y, Zhan W, Xu X, Zhuo S. Correlation between level of autophagy and frequency of CD8 + T cells in patients with chronic obstructive pulmonary disease. J Int Med Res 2020; 48:300060520952638. [PMID: 32910701 PMCID: PMC7488904 DOI: 10.1177/0300060520952638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective The pathogenesis of chronic obstructive pulmonary disease (COPD) remains elusive. Here, we assessed the correlation between CD8+ T cell frequencies and autophagy in COPD patients. Methods Subjects were divided into three groups (n = 30 patients/group): (1) COPD patients in the stable phase; (2) smokers with normal lung function; and (3) non-smokers with normal lung function. Flow cytometry was used to enumerate CD8+ T cell subsets (CD8+, CD8+ effector, and CD8+ memory T cells) and quantitate T-cell apoptosis. RT-PCR and western blotting were used to measure levels of LC3 and p62. Results Frequencies of CD8+ T cell subsets and expression of p62 and LC3 II/I were significantly higher in COPD patients compared with the other two groups, while the rate of apoptosis was lower. In COPD patients, LC3 II/I and p62 expression were positively correlated with CD8+ T cell subset frequencies. Moreover, a significant correlation was observed between LC3 II/I and p62 expression and T cell subset frequencies. Conclusion Autophagy level is positively correlated with the frequencies of CD8+ T cells, suggesting that autophagy might be involved in COPD pathogenesis.
Collapse
Affiliation(s)
- Hong Zhuang
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Na Li
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Sida Chen
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Yan Shen
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Wugen Zhan
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Xiangqun Xu
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Songming Zhuo
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| |
Collapse
|
4
|
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: 32] [Impact Index Per Article: 6.4] [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
|
5
|
Racanelli AC, Choi AMK, Choi ME. Autophagy in chronic lung disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:135-156. [PMID: 32620240 DOI: 10.1016/bs.pmbts.2020.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The development of chronic lung disease occurs as a consequence of multiple cellular events that involve an initial insult which often leads to the development of chronic inflammation, and the dysregulation of cellular proliferation and cell death mechanisms. Multiple cell types in the lung are key to the respiratory and protective/barrier functions necessary to manage the chronic exposures to environmental, mechanical, and oxidative stressors. Autophagy is essential to lung development and homeostasis, as well as the prevention and development of disease. The cellular process involves the collection and removal of unwanted organelles and proteins through lysosomal degradation. In recent years, investigations have addressed the roles of autophagy and selective autophagy in numerous chronic lung diseases. Here, we highlight recent advances on the role of autophagy in the pathogenesis of asthma, chronic obstructive pulmonary disease and emphysema, pulmonary arterial hypertension, and idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Alexandra C Racanelli
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States; NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States; NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Mary E Choi
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States; Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
6
|
Nrf2 positively regulates autophagy antioxidant response in human bronchial epithelial cells exposed to diesel exhaust particles. Sci Rep 2020; 10:3704. [PMID: 32111854 PMCID: PMC7048799 DOI: 10.1038/s41598-020-59930-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 02/04/2020] [Indexed: 01/06/2023] Open
Abstract
Diesel exhaust particles (DEP) are known to generate reactive oxygen species in the respiratory system, triggering cells to activate antioxidant defence mechanisms, such as Keap1-Nrf2 signalling and autophagy. The aim of this study was to investigate the relationship between the Keap1-Nrf2 signalling and autophagy pathways after DEP exposure. BEAS-2B cells were transfected with silencing RNA (siRNA) specific to Nrf2 and exposed to DEP. The relative levels of mRNA for Nrf2, NQO1, HO-1, LC3B, p62 and Atg5 were determined using RT-PCR, while the levels of LCB3, Nrf2, and p62 protein were determined using Western blotting. The autophagy inhibitor bafilomycin caused a significant decrease in the production of Nrf2, HO-1 and NQO1 compared to DEPs treatment, whereas the Nrf2 activator sulforaphane increased the LC3B (p = 0.020) levels. BEAS-2B cells exposed to DEP at a concentration of 50 μg/mL for 2 h showed a significant increase in the expression of LC3B (p = 0.001), p62 (p = 0.008), Nrf2 (p = 0.003), HO-1 (p = 0.001) and NQO1 (p = 0.015) genes compared to control. In siRNA-transfected cells, the LC3B (p < 0.001), p62 (p = 0.001) and Atg5 (p = 0.024) mRNA levels and the p62 and LC3II protein levels were decreased, indicating that Nrf2 modulated the expression of autophagy markers (R < 1). These results imply that, in bronchial cells exposed to DEP, the Nrf2 system positively regulates autophagy to maintain cellular homeostasis.
Collapse
|
7
|
Chen W, Wang Q, Xu X, Saxton B, Tessema M, Leng S, Choksi S, Belinsky SA, Liu ZG, Lin Y. Vasorin/ATIA Promotes Cigarette Smoke-Induced Transformation of Human Bronchial Epithelial Cells by Suppressing Autophagy-Mediated Apoptosis. Transl Oncol 2020; 13:32-41. [PMID: 31760267 PMCID: PMC6883318 DOI: 10.1016/j.tranon.2019.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Escaping cell death pathways is an important event during carcinogenesis. We previously identified anti-TNFα-induced apoptosis (ATIA, also known as vasorin) as an antiapoptotic factor that suppresses reactive oxygen species (ROS) production. However, the role of vasorin in lung carcinogenesis has not been investigated. METHODS Vasorin expression was examined in human lung cancer tissues with immunohistochemistry and database analysis. Genetic and pharmacological approaches were used to manipulate protein expression and autophagy activity in human bronchial epithelial cells (HBECs). ROS generation was measured with fluorescent indicator, apoptosis with release of lactate dehydrogenase, and cell transformation was assessed with colony formation in soft agar. RESULTS Vasorin expression was increased in human lung cancer tissues and cell lines, which was inversely associated with lung cancer patient survival. Cigarette smoke extract (CSE) and benzo[a]pyrene diol epoxide (BPDE)-induced vasorin expression in HBECs. Vasorin knockdown in HBECs significantly suppressed CSE-induced transformation in association with enhanced ROS accumulation and autophagy. Scavenging ROS attenuated autophagy and cytotoxicity in vasorin knockdown cells, suggesting that vasorin potentiates transformation by impeding ROS-mediated CSE cytotoxicity and improving survival of the premalignant cells. Suppression of autophagy effectively inhibited CSE-induced apoptosis, suggesting that autophagy was pro-apoptotic in CSE-treated cells. Importantly, blocking autophagy strongly potentiated CSE-induced transformation. CONCLUSION These results suggest that vasorin is a potential lung cancer-promoting factor that facilitates cigarette smoke-induced bronchial epithelial cell transformation by suppressing autophagy-mediated apoptosis, which could be exploited for lung cancer prevention.
Collapse
Affiliation(s)
- Wenshu Chen
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Qiong Wang
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Xiuling Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Bryanna Saxton
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Mathewos Tessema
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Shuguang Leng
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Swati Choksi
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bethesda, MD, 20892, USA
| | - Steven A Belinsky
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA
| | - Zheng-Gang Liu
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bethesda, MD, 20892, USA
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM, 87108, USA.
| |
Collapse
|
8
|
Mercado N, Colley T, Baker JR, Vuppussetty C, Kono Y, Clarke C, Tooze S, Johansen T, Barnes PJ. Bicaudal D1 impairs autophagosome maturation in chronic obstructive pulmonary disease. FASEB Bioadv 2019; 1:688-705. [PMID: 32123815 PMCID: PMC6996363 DOI: 10.1096/fba.2018-00055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 11/20/2018] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Bicaudal D1 (BICD1), an adaptor for the dynein-dynactin motor complex, has been identified as a susceptibility gene in chronic obstructive pulmonary disease (COPD). Autophagy, an essential cellular homeostasis process, is defective in COPD, in which oxidative stress-induced misfolded proteins accumulate into toxic aggregates dependent on the accumulation of the autophagic cargo receptor p62. Defective autophagy can be caused by mutations in the dynein and dynactin motor complex suggesting a possible link between BICD1 and defective autophagy in COPD. BICD1 levels were measured in peripheral lung tissue from COPD patients together with markers of autophagy and found to be increased in COPD together with autophagosomes, p62 and p62 oligomers. In vitro exposure of bronchial epithelial cells to cigarette smoke extracts (CSEs) revealed that high concentrations of CSE induced defective autophagosome maturation with accumulation of BICD1, p62 and ubiquitin-associated p62 oligomers. This was confirmed in vivo using CS-exposed mice. Furthermore, we identified that formation of CS-induced p62 oligomers required an interaction with Keap1. Overexpression and ablation of BICD1 confirmed that increased BICD1 negatively regulates autophagosome maturation inducing accumulation of p62 and p62 oligomers and that it can be reversed by cardiac glycosides. We conclude that defective autophagosome maturation in COPD is caused by oxidative stress-mediated BICD1 accumulation.
Collapse
Affiliation(s)
- Nicolas Mercado
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
- Novartis Institutes for BioMedical ResearchBaselSwitzerland
| | - Thomas Colley
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
- Pulmocide LtdLondonUK
| | - Jonathan R. Baker
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | | | - Yuta Kono
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Colin Clarke
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Sharon Tooze
- London Research InstituteCancer Research UKLondonUK
| | - Terje Johansen
- Molecular Cancer Research GroupInstitute of Medical BiologyUniversity of Tromsø – The Arctic University of NorwayTromsøNorway
| | - Peter J. Barnes
- Airway Disease SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| |
Collapse
|
9
|
Lee KH, Lee J, Woo J, Lee CH, Yoo CG. Proteasome Inhibitor-Induced IκB/NF-κB Activation is Mediated by Nrf2-Dependent Light Chain 3B Induction in Lung Cancer Cells. Mol Cells 2018; 41:1008-1015. [PMID: 30396235 PMCID: PMC6315323 DOI: 10.14348/molcells.2018.0277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/30/2018] [Accepted: 09/13/2018] [Indexed: 11/27/2022] Open
Abstract
IκB, a cytoplasmic inhibitor of nuclear factor-κB (NF-κB), is reportedly degraded via the proteasome. However, we recently found that long-term incubation with proteasome inhibitors (PIs) such as PS-341 or MG132 induces IκBα degradation via an alternative pathway, lysosome, which results in NF-κB activation and confers resistance to PI-induced lung cancer cell death. To enhance the anti-cancer efficacy of PIs, elucidation of the regulatory mechanism of PI-induced IκBα degradation is necessary. Here, we demonstrated that PI upregulates nuclear factor (erythroid-derived 2)-like 2 (Nrf2) via both de novo protein synthesis and Kelch-like ECH-associated protein 1 (KEAP1) degradation, which is responsible for IκBα degradation via macroautophagy activation. PIs increased the protein level of light chain 3B (LC3B, macroautophagy marker), but not lysosome-associated membrane protein 2a (Lamp2a, the receptor for chaperone-mediated autophagy) in NCI-H157 and A549 lung cancer cells. Pretreatment with macroautophagy inhibitor or knock-down of LC3B blocked PI-induced IκBα degradation. PIs up-regulated Nrf2 by increasing its transcription and mediating degradation of KEAP1 (cytoplasmic inhibitor of Nrf2). Overexpression of dominant-negative Nrf2, which lacks an N-terminal transactivating domain, or knock-down of Nrf2 suppressed PI-induced LC3B protein expression and subsequent IκBα degradation. Thus, blocking of the Nrf2 pathway enhanced PI-induced cell death. These findings suggest that Nrf2-driven induction of LC3B plays an essential role in PI-induced activation of the IκB/NF-κB pathway, which attenuates the anti-tumor efficacy of PIs.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Jungsil Lee
- 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 Hospital, Seoul,
Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, 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 Hospital, Seoul,
Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul,
Korea
| |
Collapse
|
10
|
Abstract
Mucociliary clearance is critically important in protecting the airways from infection and from the harmful effects of smoke and various inspired substances known to induce oxidative stress and persistent inflammation. An essential feature of the clearance mechanism involves regulation of the periciliary liquid layer on the surface of the airway epithelium, which is necessary for normal ciliary beating and maintenance of mucus hydration. The underlying ion transport processes associated with airway surface hydration include epithelial Na+ channel-dependent Na+ absorption occurring in parallel with CFTR and Ca2+-activated Cl- channel-dependent anion secretion, which are coordinately regulated to control the depth of the periciliary liquid layer. Oxidative stress is known to cause both acute and chronic effects on airway ion transport function, and an increasing number of studies in the past few years have identified an important role for autophagy as part of the physiological response to the damaging effects of oxidation. In this review, recent studies addressing the influence of oxidative stress and autophagy on airway ion transport pathways, along with results showing the potential of autophagy modulators in restoring the function of ion channels involved in transepithelial electrolyte transport necessary for effective mucociliary clearance, are presented.
Collapse
Affiliation(s)
- Scott M O'Grady
- Departments of Animal Science, Integrative Biology and Physiology, University of Minnesota , St. Paul, Minnesota
| |
Collapse
|
11
|
Vurusaner B, Gargiulo S, Testa G, Gamba P, Leonarduzzi G, Poli G, Basaga H. The role of autophagy in survival response induced by 27-hydroxycholesterol in human promonocytic cells. Redox Biol 2018; 17:400-410. [PMID: 29879549 PMCID: PMC5986166 DOI: 10.1016/j.redox.2018.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/16/2018] [Accepted: 05/21/2018] [Indexed: 11/25/2022] Open
Abstract
Autophagy has been shown to be stimulated in advanced atherosclerotic plaques by metabolic stress, inflammation and oxidized lipids. The lack of published studies addressing the potential stimulation of pro-survival autophagy by oxysterols, a family of cholesterol oxidation products, has prompted our study. Thus, the goal of the current study is to elucidate the molecular mechanism of the autophagy induced by 27-hydroxycholesterol (27-OH), that is one of the most abundant oxysterols in advanced atherosclerotic lesions, and to assess whether the pro-oxidant effect of the oxysterol is involved in the given response. Here we showed that 27-OH, in a low micromolar range, activates a pro-survival autophagic response in terms of increased LC3 II/LC3 I ratio and Beclin 1, that depends on the up-regulation of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt pathways as a potential result of an intracellular reactive oxygen species increase provoked by the oxysterol in human promonocytic U937 cells. Moreover, 27-OH induced autophagy is dependent on the relation between nuclear factor erythroid 2 p45-related factor 2 (Nrf2)-dependent antioxidant response and p62. The data obtained highlight the involvement of cholesterol oxidation products in the pathogenesis of oxidative stress related chronic diseases like atherosclerosis. Therefore, deeply understanding the complex mechanism and generating synthetic or natural molecules targeting this survival mechanism might be very promising tools in the prevention of such diseases.
Collapse
Affiliation(s)
- Beyza Vurusaner
- Biological Sciences and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Orhanli-Tuzla, 34956 Istanbul, Turkey.
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
| | - Huveyda Basaga
- Biological Sciences and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Orhanli-Tuzla, 34956 Istanbul, Turkey.
| |
Collapse
|
12
|
Ding S, Hou X, Wang F, Wang G, Tan X, Liu Y, Zhou Y, Qiu H, Sun E, Jiang N, Li Z, Song J, Feng L, Jia X. Regulation of Eclipta prostrata L. components on cigarette smoking-induced autophagy of bronchial epithelial cells via keap1-Nrf2 pathway. ENVIRONMENTAL TOXICOLOGY 2018; 33:811-820. [PMID: 29726624 DOI: 10.1002/tox.22567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/02/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Cigarette smoking extract (CSE)-induced autophagic injury has been regarded as an important contributor to the pathogenesis of lung cancer. We previously found that Eclipta prostrata L. component (CCE) reduced CSE-induced bronchial epithelial cells damage. However, the mechanism remains unknown. Human normal bronchial epithelial cells (NHBE) were exposed to CSE to establish stress model. Nrf2-siRNA and Keap1-siRNA transfection were performed. mRFP-GFP-LC3 dual fluorescence and transmission electron microscopy were used to observe the autophagic characteristics. CCE prevented CSE-induced Nrf2 transfer into cytoplasm and up-regulated Keap1 level of NHBE cells. Furthermore, CCE significantly increased p-p16, p-p21 and p-p53 phosphorylation levels in Nrf2-siRNA- or Keap1-siRNA-transfected cells. As demonstrated by transmission electron microscopy and mRFP-GFP-LC3 dual fluorescence assays, CCE mitigated autophagic injury, and also down-regulated autophagy-related Beclin-1, LC3II/LC3I ratio, Atg5 and ATF4 levels. Our findings showed the attenuation of CCE on CSE-induced NHBE cells injury was associated with Nrf-2-mediated oxidative signaling pathway.
Collapse
Affiliation(s)
- Shumin Ding
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, People's Republic of China
| | - Xuefeng Hou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Fujing Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Gang Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Xiaobin Tan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Ying Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Yuanli Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Huihui Qiu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - E Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Nan Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
| | - Zihao Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Jie Song
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Liang Feng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- Chinese Materia Medica Department, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaobin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, People's Republic of China
- Chinese Materia Medica Department, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Wang Y, Liu J, Zhou JS, Huang HQ, Li ZY, Xu XC, Lai TW, Hu Y, Zhou HB, Chen HP, Ying SM, Li W, Shen HH, Chen ZH. MTOR Suppresses Cigarette Smoke-Induced Epithelial Cell Death and Airway Inflammation in Chronic Obstructive Pulmonary Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2571-2580. [PMID: 29507104 DOI: 10.4049/jimmunol.1701681] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/12/2018] [Indexed: 12/17/2023]
Abstract
Airway epithelial cell death and inflammation are pathological features of chronic obstructive pulmonary disease (COPD). Mechanistic target of rapamycin (MTOR) is involved in inflammation and multiple cellular processes, e.g., autophagy and apoptosis, but little is known about its function in COPD pathogenesis. In this article, we illustrate how MTOR regulates cigarette smoke (CS)-induced cell death, airway inflammation, and emphysema. Expression of MTOR was significantly decreased and its suppressive signaling protein, tuberous sclerosis 2 (TSC2), was increased in the airway epithelium of human COPD and in mouse lungs with chronic CS exposure. In human bronchial epithelial cells, CS extract (CSE) activated TSC2, inhibited MTOR, and induced autophagy. The TSC2-MTOR axis orchestrated CSE-induced autophagy, apoptosis, and necroptosis in human bronchial epithelial cells; all of which cooperatively regulated CSE-induced inflammatory cytokines IL-6 and IL-8 through the NF-κB pathway. Mice with a specific knockdown of Mtor in bronchial or alveolar epithelial cells exhibited significantly augmented airway inflammation and airspace enlargement in response to CS exposure, accompanied with enhanced levels of autophagy, apoptosis, and necroptosis in the lungs. Taken together, these data demonstrate that MTOR suppresses CS-induced inflammation and emphysema-likely through modulation of autophagy, apoptosis, and necroptosis-and thus suggest that activation of MTOR may represent a novel therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Yong Wang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Juan Liu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Jie-Sen Zhou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Hua-Qiong Huang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Zhou-Yang Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Xu-Chen Xu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Tian-Wen Lai
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Yue Hu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Hong-Bin Zhou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Hai-Pin Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Song-Min Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| | - Hua-Hao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Zhi-Hua Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; and
| |
Collapse
|
14
|
Racanelli AC, Kikkers SA, Choi AM, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Autophagy 2018; 14:221-232. [PMID: 29130366 PMCID: PMC5902194 DOI: 10.1080/15548627.2017.1389823] [Citation(s) in RCA: 378] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022] Open
Abstract
Persistent inflammation within the respiratory tract underlies the pathogenesis of numerous chronic pulmonary diseases including chronic obstructive pulmonary disease, asthma and pulmonary fibrosis. Chronic inflammation in the lung may arise from a combination of genetic susceptibility and environmental influences, including exposure to microbes, particles from the atmosphere, irritants, pollutants, allergens, and toxic molecules. To this end, an immediate, strong, and highly regulated inflammatory defense mechanism is needed for the successful maintenance of homeostasis within the respiratory system. Macroautophagy/autophagy plays an essential role in the inflammatory response of the lung to infection and stress. At baseline, autophagy may be critical for inhibiting spontaneous pulmonary inflammation and fundamental for the response of pulmonary leukocytes to infection; however, when not regulated, persistent or inefficient autophagy may be detrimental to lung epithelial cells, promoting lung injury. This perspective will discuss the role of autophagy in driving and regulating inflammatory responses of the lung in chronic lung diseases with a focus on potential avenues for therapeutic targeting. Abbreviations AR allergic rhinitis AM alveolar macrophage ATG autophagy-related CF cystic fibrosis CFTR cystic fibrosis transmembrane conductance regulator COPD chronic obstructive pulmonary disease CS cigarette smoke CSE cigarette smoke extract DC dendritic cell IH intermittent hypoxia IPF idiopathic pulmonary fibrosis ILD interstitial lung disease MAP1LC3B microtubule associated protein 1 light chain 3 beta MTB Mycobacterium tuberculosis MTOR mechanistic target of rapamycin kinase NET neutrophil extracellular traps OSA obstructive sleep apnea PAH pulmonary arterial hypertension PH pulmonary hypertension ROS reactive oxygen species TGFB1 transforming growth factor beta 1 TNF tumor necrosis factor.
Collapse
Affiliation(s)
- Alexandra C. Racanelli
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- New York-Presbyterian Hospital, New York, NY, USA
| | - Sarah Ann Kikkers
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- New York-Presbyterian Hospital, New York, NY, USA
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
15
|
Sukkar MB, Harris J. Potential impact of oxidative stress induced growth inhibitor 1 (OSGIN1) on airway epithelial cell autophagy in chronic obstructive pulmonary disease (COPD). J Thorac Dis 2017; 9:4825-4827. [PMID: 29312667 DOI: 10.21037/jtd.2017.10.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Maria B Sukkar
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Ultimo, New South Wales, Australia
| | - James Harris
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
16
|
Kota A, Deshpande DA, Haghi M, Oliver B, Sharma P. Autophagy and airway fibrosis: Is there a link? F1000Res 2017; 6:409. [PMID: 28815017 PMCID: PMC5416906 DOI: 10.12688/f1000research.11236.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2018] [Indexed: 12/14/2022] Open
Abstract
In the past decade, an emerging process named “autophagy” has generated intense interest in many chronic lung diseases. Tissue remodeling and fibrosis is a common feature of many airway diseases, and current therapies do not prevent or reverse these structural changes. Autophagy has evolved as a conserved process for bulk degradation and recycling of cytoplasmic components to maintain basal cellular homeostasis and healthy organelle populations in the cell. Furthermore, autophagy serves as a cell survival mechanism and can also be induced by chemical and physical stress to the cell. Accumulating evidence demonstrates that autophagy plays an essential role in vital cellular processes, including tissue remodeling. This review will discuss some of the recent advancements made in understanding the role of this fundamental process in airway fibrosis with emphasis on airway remodeling, and how autophagy can be exploited as a target for airway remodeling in asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Anudeep Kota
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia.,Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Deepak A Deshpande
- Centre for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mehra Haghi
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Brian Oliver
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia
| | - Pawan Sharma
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia
| |
Collapse
|
17
|
Kota A, Deshpande DA, Haghi M, Oliver B, Sharma P. Autophagy and airway fibrosis: Is there a link? F1000Res 2017; 6:409. [PMID: 28815017 PMCID: PMC5416906 DOI: 10.12688/f1000research.11236.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2018] [Indexed: 09/15/2023] Open
Abstract
In the past decade, an emerging process named "autophagy" has generated intense interest in many chronic lung diseases. Tissue remodeling and fibrosis is a common feature of many airway diseases, and current therapies do not prevent or reverse these structural changes. Autophagy has evolved as a conserved process for bulk degradation and recycling of cytoplasmic components to maintain basal cellular homeostasis and healthy organelle populations in the cell. Furthermore, autophagy serves as a cell survival mechanism and can also be induced by chemical and physical stress to the cell. Accumulating evidence demonstrates that autophagy plays an essential role in vital cellular processes, including tissue remodeling. This review will discuss some of the recent advancements made in understanding the role of this fundamental process in airway fibrosis with emphasis on airway remodeling, and how autophagy can be exploited as a target for airway remodeling in asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Anudeep Kota
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Deepak A. Deshpande
- Centre for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mehra Haghi
- Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Brian Oliver
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia
| | - Pawan Sharma
- Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, 2037, Australia
| |
Collapse
|
18
|
He Y, Li S, Zhang W, Dai W, Cui T, Wang G, Gao T, Li C. Dysregulated autophagy increased melanocyte sensitivity to H 2O 2-induced oxidative stress in vitiligo. Sci Rep 2017; 7:42394. [PMID: 28186139 PMCID: PMC5301258 DOI: 10.1038/srep42394] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/10/2017] [Indexed: 11/09/2022] Open
Abstract
In vitiligo, melanocytes are particularly vulnerable to oxidative stress owing to the pro-oxidant state generated during melanin synthesis and to the genetic antioxidant defects. Autophagy is a controlled self-digestion process which can protect cells against oxidative damage. However, the exact role of autophagy in vitiligo melanocytes in response to oxidative stress and the mechanism involved are still not clear. To determine the implications of autophagy for melanocyte survival in response to oxidative stress, we first detected the autophagic flux in normal melanocytes exposure to H2O2, and found that autophagy was significantly enhanced in normal melanocytes, for protecting cells against H2O2-induced oxidative damage. Nevertheless, vitiligo melanocytes exhibited dysregulated autophagy and hypersensitivity to H2O2-induced oxidative injury. In addition, we confirmed that the impairment of Nrf2-p62 pathway is responsible for the defects of autophagy in vitiligo melanocytes. Noteworthily, upregulation of the Nrf2-p62 pathway or p62 reduced H2O2-induced oxidative damage of vitiligo melanocytes. Therefore, our data demonstrated that dysregulated autophagy owing to the impairment of Nrf2-p62 pathway increase the sensitivity of vitiligo melanocytes to oxidative stress, thus promote the development of vitiligo. Upregulation of p62-dependent autophagy may be applied to vitiligo treatment in the future.
Collapse
Affiliation(s)
- Yuanmin He
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuli Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weigang Zhang
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Dai
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tingting Cui
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Gang Wang
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
19
|
Mannam P, Rauniyar N, Lam TT, Luo R, Lee PJ, Srivastava A. MKK3 influences mitophagy and is involved in cigarette smoke-induced inflammation. Free Radic Biol Med 2016; 101:102-115. [PMID: 27717867 DOI: 10.1016/j.freeradbiomed.2016.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/16/2016] [Accepted: 10/02/2016] [Indexed: 11/26/2022]
Abstract
Cigarette smoking is the primary risk factor for COPD which is characterized by excessive inflammation and airflow obstruction of the lung. While inflammation is causally related to initiation and progression of COPD, the mitochondrial mechanisms that underlie the associated inflammatory responses are poorly understood. In this context, we have studied the role played by Mitogen activated protein (MAP) kinase kinase 3 (MKK3), a dual-specificity protein kinase, in cigarette smoke induced-inflammation and mitochondrial dysfunction. Serum pro-inflammatory cytokines were significantly elevated in WT but not in MKK3-/- mice exposed to Cigarette smoke (CS) for 2 months. To study the cellular mechanisms of inflammation, bone marrow derived macrophages (BMDMs), wild type (WT) and MKK3-/-, were exposed to cigarette smoke extract (CSE) and inflammatory cytokine production and mitochondrial function assessed. The levels of IL-1β, IL-6, and TNFα were increased along with higher reactive oxygen species (ROS) and P-NFκB after CSE treatment in WT but not in MKK3-/- BMDMs. CSE treatment adversely affected basal mitochondrial respiration, ATP production, maximum respiratory capacity, and spare respiratory capacity in WT BMDMs only. Mitophagy, clearance of dysfunctional mitochondria, was up regulated in CS exposed WT mice lung tissue and CSE exposed WT BMDMs, respectively. The proteomic analysis of BMDMs by iTRAQ (isobaric tags for relative and absolute quantitation) showed up regulation of mitochondrial dysfunction associated proteins in WT and higher OXPHOS (Oxidative phosphorylation) and IL-10 signaling proteins in MKK3-/- BMDMs after CSE exposure, confirming the critical role of mitochondrial homeostasis. Interestingly, we found increased levels of p-MKK3 by immunohistochemistry in COPD patient lung tissues that could be responsible for insufficient mitophagy and disease progression. This study identifies MKK3 as a negative regulator of mitochondrial function and inflammatory responses to CS and suggests that MKK3 could be a therapeutic target.
Collapse
Affiliation(s)
- Praveen Mannam
- Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA.
| | - Navin Rauniyar
- MS & Proteomics Resource at Yale University, WM Keck Foundation Biotechnology Resource Laboratory, Department of Molecular Biophysics and Biochemistry, New Haven, CT 06520-8057, USA
| | - TuKiet T Lam
- MS & Proteomics Resource at Yale University, WM Keck Foundation Biotechnology Resource Laboratory, Department of Molecular Biophysics and Biochemistry, New Haven, CT 06520-8057, USA
| | - Ruiyan Luo
- Department of Epidemiology & Biostatistics, School of Public Health, Georgia State University, Atlanta, GA, USA
| | - Patty J Lee
- Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA
| | - Anup Srivastava
- Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA.
| |
Collapse
|
20
|
Aggarwal S, Mannam P, Zhang J. Differential regulation of autophagy and mitophagy in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2016; 311:L433-52. [PMID: 27402690 PMCID: PMC5504426 DOI: 10.1152/ajplung.00128.2016] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/01/2016] [Indexed: 12/26/2022] Open
Abstract
Lysosomal-mediated degradation of intracellular lipids, proteins and organelles, known as autophagy, represents a inducible adaptive response to lung injury resulting from exposure to insults, such as hypoxia, microbes, inflammation, ischemia-reperfusion, pharmaceuticals (e.g., bleomycin), or inhaled xenobiotics (i.e., air pollution, cigarette smoke). This process clears damaged or toxic cellular constituents and facilitates cell survival in stressful environments. Autophagic degradation of dysfunctional or damaged mitochondria is termed mitophagy. Enhanced mitophagy is usually an early response to promote survival. However, overwhelming or prolonged mitochondrial damage can induce excessive/pathological levels of mitophagy, thereby promoting cell death and tissue injury. Autophagy/mitophagy is therefore an important modulator in human pulmonary diseases and a potential therapeutic target. This review article will summarize the most recent studies highlighting the role of autophagy/mitophagy and its molecular pathways involved in stress response in pulmonary pathologies.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Praveen Mannam
- Department of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
21
|
Tao S, Rojo de la Vega M, Quijada H, Wondrak GT, Wang T, Garcia JGN, Zhang DD. Bixin protects mice against ventilation-induced lung injury in an NRF2-dependent manner. Sci Rep 2016; 6:18760. [PMID: 26729554 PMCID: PMC4700431 DOI: 10.1038/srep18760] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 08/26/2015] [Indexed: 12/19/2022] Open
Abstract
Mechanical ventilation (MV) is a therapeutic intervention widely used in the clinic to assist patients that have difficulty breathing due to lung edema, trauma, or general anesthesia. However, MV causes ventilator-induced lung injury (VILI), a condition characterized by increased permeability of the alveolar-capillary barrier that results in edema, hemorrhage, and neutrophil infiltration, leading to exacerbated lung inflammation and oxidative stress. This study explored the feasibility of using bixin, a canonical NRF2 inducer identified during the current study, to ameliorate lung damage in a murine VILI model. In vitro, bixin was found to activate the NRF2 signaling pathway through blockage of ubiquitylation and degradation of NRF2 in a KEAP1-C151 dependent manner; intraperitoneal (IP) injection of bixin led to pulmonary upregulation of the NRF2 response in vivo. Remarkably, IP administration of bixin restored normal lung morphology and attenuated inflammatory response and oxidative DNA damage following MV. This observed beneficial effect of bixin derived from induction of the NRF2 cytoprotective response since it was only observed in Nrf2+/+ but not in Nrf2−/− mice. This is the first study providing proof-of-concept that NRF2 activators can be developed into pharmacological agents for clinical use to prevent patients from lung injury during MV treatment.
Collapse
Affiliation(s)
- Shasha Tao
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | | | - Hector Quijada
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, AZ 85721
| | - Georg T Wondrak
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, 1515 North Campbell Avenue, Tucson, AZ 85724, USA
| | - Ting Wang
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, AZ 85721
| | - Joe G N Garcia
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, AZ 85721
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, 1515 North Campbell Avenue, Tucson, AZ 85724, USA
| |
Collapse
|
22
|
Cui T, Lai Y, Janicki JS, Wang X. Nuclear factor erythroid-2 related factor 2 (Nrf2)-mediated protein quality control in cardiomyocytes. Front Biosci (Landmark Ed) 2016; 21:192-202. [PMID: 26709769 DOI: 10.2741/4384] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein quality control (PQC) acts to minimize the level and toxicity of malfolded proteins in the cell. It is performed by an elaborate network of molecular chaperones and targeted protein degradation pathways. PQC monitors and maintains protein homeostasis or proteostasis in the cells. Whilst chaperones may actively promote refolding of malfolded proteins, the malfolded proteins which cannot be correctly refolded are degraded by the ubiquitin proteasome system (UPS) and the autophagic-lysosome pathway (ALP). The UPS degrades individual misfolded protein molecules, whereas the ALP removes large and less soluble protein aggregates and organelles. Emerging evidence indicates that dysregulated and inadequate PQC play an important role in the pathogenesis of not only classic conformational disease but more common forms of cardiac pathology such as cardiac pathological hypertrophy and heart failure. Nuclear factor erythroid 2-related factor 2 (Nrf2), a master transcription factor of cellular defense, appears to regulate the USP and the ALP by directly controlling the expression of UPS- and ALP- related genes. This article highlights an emerging role of Nrf2 in the regulation of intracellular PQC as well as its potential involvement in cardiac pathology.
Collapse
Affiliation(s)
| | | | - Jospeh S Janicki
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Xuejun Wang
- Division of Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, South Dakota, USA.,
| |
Collapse
|
23
|
Tomasovic A, Kurrle N, Sürün D, Heidler J, Husnjak K, Poser I, Schnütgen F, Scheibe S, Seimetz M, Jaksch P, Hyman A, Weissmann N, von Melchner H. Sestrin 2 protein regulates platelet-derived growth factor receptor β (Pdgfrβ) expression by modulating proteasomal and Nrf2 transcription factor functions. J Biol Chem 2015; 290:9738-52. [PMID: 25716320 DOI: 10.1074/jbc.m114.632133] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Indexed: 01/12/2023] Open
Abstract
We recently identified the antioxidant protein Sestrin 2 (Sesn2) as a suppressor of platelet-derived growth factor receptor β (Pdgfrβ) signaling and Pdgfrβ signaling as an inducer of lung regeneration and injury repair. Here, we identified Sesn2 and the antioxidant gene inducer nuclear factor erythroid 2-related factor 2 (Nrf2) as positive regulators of proteasomal function. Inactivation of Sesn2 or Nrf2 induced reactive oxygen species-mediated proteasomal inhibition and Pdgfrβ accumulation. Using bacterial artificial chromosome (BAC) transgenic HeLa and mouse embryonic stem cells stably expressing enhanced green fluorescent protein-tagged Sesn2 at nearly endogenous levels, we also showed that Sesn2 physically interacts with 2-Cys peroxiredoxins and Nrf2 albeit under different reductive conditions. Overall, we characterized a novel, redox-sensitive Sesn2/Pdgfrβ suppressor pathway that negatively interferes with lung regeneration and is up-regulated in the emphysematous lungs of patients with chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Ana Tomasovic
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Nina Kurrle
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Duran Sürün
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Juliana Heidler
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Koraljka Husnjak
- Institute of Biochemistry II, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Frank Schnütgen
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - Susan Scheibe
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-University Giessen, Department of Internal Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), 35392 Giessen, Germany, and
| | - Michael Seimetz
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-University Giessen, Department of Internal Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), 35392 Giessen, Germany, and
| | - Peter Jaksch
- Department of Thoracic Surgery, University Hospital of Vienna, A-1090 Vienna, Austria
| | - Anthony Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-University Giessen, Department of Internal Medicine, Universities of Giessen and Marburg Lung Centre (UGMLC), 35392 Giessen, Germany, and
| | - Harald von Melchner
- From the Department of Molecular Hematology, Goethe University Medical School, 60590 Frankfurt am Main, Germany,
| |
Collapse
|
24
|
Yoshizaki K, Brito JM, Moriya HT, Toledo AC, Ferzilan S, Ligeiro de Oliveira AP, Machado ID, Farsky SHP, Silva LFF, Martins MA, Saldiva PHN, Mauad T, Macchione M. Chronic exposure of diesel exhaust particles induces alveolar enlargement in mice. Respir Res 2015; 16:18. [PMID: 25848680 PMCID: PMC4345004 DOI: 10.1186/s12931-015-0172-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 01/17/2015] [Indexed: 12/21/2022] Open
Abstract
Background Diesel exhaust particles (DEPs) are deposited into the respiratory tract and are thought to be a risk factor for the development of diseases of the respiratory system. In healthy individuals, the timing and mechanisms of respiratory tract injuries caused by chronic exposure to air pollution remain to be clarified. Methods We evaluated the effects of chronic exposure to DEP at doses below those found in a typical bus corridor in Sao Paulo (150 μg/m3). Male BALB/c mice were divided into mice receiving a nasal instillation: saline (saline; n = 30) and 30 μg/10 μL of DEP (DEP; n = 30). Nasal instillations were performed five days a week, over a period of 90 days. Bronchoalveolar lavage (BAL) was performed, and the concentrations of interleukin (IL)-4, IL-10, IL-13 and interferon-gamma (INF-γ) were determined by ELISA-immunoassay. Assessment of respiratory mechanics was performed. The gene expression of Muc5ac in lung was evaluated by RT-PCR. The presence of IL-13, MAC2+ macrophages, CD3+, CD4+, CD8+ T cells and CD20+ B cells in tissues was analysed by immunohistochemistry. Bronchial thickness and the collagen/elastic fibers density were evaluated by morphometry. We measured the mean linear intercept (Lm), a measure of alveolar distension, and the mean airspace diameter (D0) and statistical distribution (D2). Results DEP decreased IFN-γ levels in BAL (p = 0.03), but did not significantly alter IL-4, IL-10 and IL-13 levels. MAC2+ macrophage, CD4+ T cell and CD20+ B cell numbers were not altered; however, numbers of CD3+ T cells (p ≤ 0.001) and CD8+ T cells (p ≤ 0.001) increased in the parenchyma. Although IL-13 (p = 0.008) expression decreased in the bronchiolar epithelium, Muc5ac gene expression was not altered in the lung of DEP-exposed animals. Although respiratory mechanics, elastic and collagen density were not modified, the mean linear intercept (Lm) was increased in the DEP-exposed animals (p ≤ 0.001), and the index D2 was statistically different (p = 0.038) from the control animals. Conclusion Our data suggest that nasal instillation of low doses of DEP over a period of 90 days results in alveolar enlargement in the pulmonary parenchyma of healthy mice. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0172-z) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Yin J, Duan J, Cui Z, Ren W, Li T, Yin Y. Hydrogen peroxide-induced oxidative stress activates NF-κB and Nrf2/Keap1 signals and triggers autophagy in piglets. RSC Adv 2015. [DOI: 10.1039/c4ra13557a] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
H2O2 induces autophagy and activates NF-κB and Nrf2/Keap1 signals in a piglet model.
Collapse
Affiliation(s)
- Jie Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| | - Jielin Duan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| | - Zhijie Cui
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| | - Wenkai Ren
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| | - Tiejun Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| | - Yulong Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central
- Ministry of Agriculture
- Hunan Provincial Engineering Research Center of Healthy Livestock
- Key Laboratory of Agro-ecological Processes in Subtropical Region
- Institute of Subtropical Agriculture
| |
Collapse
|
26
|
Systems toxicology approaches enable mechanistic comparison of spontaneous and cigarette smoke-related lung tumor development in the A/J mouse model. Interdiscip Toxicol 2014; 7:73-84. [PMID: 26109882 PMCID: PMC4427718 DOI: 10.2478/intox-2014-0010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 01/09/2023] Open
Abstract
The A/J mouse is highly susceptible to lung tumor induction and has been widely used as a screening model in carcinogenicity testing and chemoprevention studies. However, the A/J mouse model has several disadvantages. Most notably, it develops lung tumors spontaneously. Moreover, there is a considerable gap in our understanding of the underlying mechanisms of pulmonary chemical carcinogenesis in the A/J mouse. Therefore, we examined the differences between spontaneous and cigarette smoke-related lung tumors in the A/J mouse model using mRNA and microRNA (miRNA) profiling. Male A/J mice were exposed whole-body to mainstream cigarette smoke (MS) for 18 months. Gene expression interaction term analysis of lung tumors and surrounding non-tumorous parenchyma samples from animals that were exposed to either 300 mg/m3 MS or sham-exposed to fresh air indicated significant differential expression of 296 genes. Ingenuity Pathway Analysis® (IPA®) indicated an overall suppression of the humoral immune response, which was accompanied by a disruption of sphingolipid and glycosaminoglycan metabolism and a deregulation of potentially oncogenic miRNA in tumors of MS-exposed A/J mice. Thus, we propose that MS exposure leads to severe perturbations in pathways essential for tumor recognition by the immune system, thereby potentiating the ability of tumor cells to escape from immune surveillance. Further, exposure to MS appeared to affect expression of miRNA, which have previously been implicated in carcinogenesis and are thought to contribute to tumor progression. Finally, we identified a 50-gene expression signature and show its utility in distinguishing between cigarette smoke-related and spontaneous lung tumors.
Collapse
|
27
|
Bao LJ, Jaramillo MC, Zhang ZB, Zheng YX, Yao M, Zhang DD, Yi XF. Nrf2 induces cisplatin resistance through activation of autophagy in ovarian carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1502-1513. [PMID: 24817946 PMCID: PMC4014230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 03/01/2014] [Indexed: 06/03/2023]
Abstract
UNLABELLED Cisplatin resistance is a major problem affecting ovarian carcinoma treatment. NF-E2-related factor 2 (Nrf2), a nuclear transcription factor, plays an important role in chemotherapy resistance. However, the underlying mechanism by which Nrf2 mediates cisplatin chemoresistance is unclear. METHODS The human ovarian carcinoma cell line, A2780, and its cisplatin-resistant variant, A2780cp were cultivated. Cell viability was determined with WST-8 assay. Western blot was applied to detect the expression of Nrf2, Nrf2 target genes, and autophagy-related proteins. RNA interference was used to knock down target genes. Annexin V and propidium iodide (PI) staining was utilized to quantify apoptosis. The ultrastructural analysis of autophagosomes was performed by transmission electron microscopy (TEM). RESULTS Nrf2 and its targeting genes, NQO1 and HO-1, are overexpressed in A2780cp cells compared with A2780 cells. Knocking down Nrf2 sensitized A2780cp cells to cisplatin treatment and decreased autophagy-related genes, Atg3, Atg6, Atg12 and p62 in both mRNA and protein levels. Furthermore, we demonstrated that in both cell lines cisplatin could induce the formation of autophagosomes and upregulate the expression of autophagy-related genes Atg3, Atg6 and Atg12. Treatment with an autophagy inhibitor, 3-Methyladenine (3-MA), or beclin 1 siRNA enhanced cisplatin-induced cell death in A2780cp cells, suggesting that inhibition of autophagy renders resistant cells to be more sensitive to cisplatin. Taken together, Nrf2 signaling may regulate cisplatin resistance by activating autophagy. CONCLUSIONS Nrf2-activated autophagy may function as a novel mechanism causing cisplatin-resistance.
Collapse
Affiliation(s)
- Ling-Jie Bao
- Obstetrics and Gynecology Hospital, Fudan University Shanghai 200011, China ; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University Shanghai 200011, China ; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai 200011, China
| | - Melba C Jaramillo
- Department of Pharmacology and Toxicology, University of Arizona Arizona, USA
| | - Zhen-Bo Zhang
- Department of Obstetrics and Gynecology, Shanghai First Peoples' Hospital, Jiaotong University Shanghai 200080, China
| | - Yun-Xi Zheng
- Obstetrics and Gynecology Hospital, Fudan University Shanghai 200011, China ; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University Shanghai 200011, China ; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai 200011, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200032, China
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona Arizona, USA
| | - Xiao-Fang Yi
- Obstetrics and Gynecology Hospital, Fudan University Shanghai 200011, China ; Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University Shanghai 200011, China ; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases Shanghai 200011, China
| |
Collapse
|
28
|
Darvekar SR, Elvenes J, Brenne HB, Johansen T, Sjøttem E. SPBP is a sulforaphane induced transcriptional coactivator of NRF2 regulating expression of the autophagy receptor p62/SQSTM1. PLoS One 2014; 9:e85262. [PMID: 24416372 PMCID: PMC3887019 DOI: 10.1371/journal.pone.0085262] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 11/24/2013] [Indexed: 12/14/2022] Open
Abstract
Organisms exposed to oxidative stress respond by orchestrating a stress response to prevent further damage. Intracellular levels of antioxidant agents increase, and damaged components are removed by autophagy induction. The KEAP1-NRF2 signaling pathway is the main pathway responsible for cell defense against oxidative stress and for maintaining the cellular redox balance at physiological levels. Sulforaphane, an isothiocyanate derived from cruciferous vegetables, is a potent inducer of KEAP1-NRF2 signaling and antioxidant response element driven gene expression. In this study, we show that sulforaphane enhances the expression of the transcriptional coregulator SPBP. The expression curve peaks 6–8 hours post stimulation, and parallels the sulforaphane-induced expression of NRF2 and the autophagy receptor protein p62/SQSTM1. Reporter gene assays show that SPBP stimulates the expression of p62/SQSTM1 via ARE elements in the promoter region, and siRNA mediated knock down of SPBP significantly decreases the expression of p62/SQSTM1 and the formation of p62/SQSTM1 bodies in HeLa cells. Furthermore, SPBP siRNA reduces the sulforaphane induced expression of NRF2, and the expression of the autophagy marker protein LC3B. Both these proteins contain ARE-like elements in their promoter regions. Over-expressed SPBP and NRF2 acts synergistically on the p62/SQSTM1 promoter and colocalize in nuclear speckles in HeLa cells. Collectively, these results suggest that SPBP is a coactivator of NRF2, and hence may be important for securing enhanced and sustained expression of NRF2 induced genes such as proteins involved in selective autophagy.
Collapse
Affiliation(s)
- Sagar Ramesh Darvekar
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Julianne Elvenes
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Hanne Britt Brenne
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Eva Sjøttem
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| |
Collapse
|
29
|
Malaviya R, Laskin JD, Laskin DL. Oxidative stress-induced autophagy: role in pulmonary toxicity. Toxicol Appl Pharmacol 2014; 275:145-51. [PMID: 24398106 DOI: 10.1016/j.taap.2013.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/28/2013] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily conserved catabolic process important in regulating the turnover of essential proteins and in elimination of damaged organelles and protein aggregates. Autophagy is observed in the lung in response to oxidative stress generated as a consequence of exposure to environmental toxicants. Whether autophagy plays role in promoting cell survival or cytotoxicity is unclear. In this article recent findings on oxidative stress-induced autophagy in the lung are reviewed; potential mechanisms initiating autophagy are also discussed. A better understanding of autophagy and its role in pulmonary toxicity may lead to the development of new strategies to treat lung injury associated with oxidative stress.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
30
|
Lam HC, Cloonan SM, Bhashyam AR, Haspel JA, Singh A, Sathirapongsasuti JF, Cervo M, Yao H, Chung AL, Mizumura K, An CH, Shan B, Franks JM, Haley KJ, Owen CA, Tesfaigzi Y, Washko GR, Quackenbush J, Silverman EK, Rahman I, Kim HP, Mahmood A, Biswal SS, Ryter SW, Choi AMK. Histone deacetylase 6-mediated selective autophagy regulates COPD-associated cilia dysfunction. J Clin Invest 2013; 123:5212-30. [PMID: 24200693 DOI: 10.1172/jci69636] [Citation(s) in RCA: 247] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 08/30/2013] [Indexed: 01/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) involves aberrant airway inflammatory responses to cigarette smoke (CS) that are associated with epithelial cell dysfunction, cilia shortening, and mucociliary clearance disruption. Exposure to CS reduced cilia length and induced autophagy in vivo and in differentiated mouse tracheal epithelial cells (MTECs). Autophagy-impaired (Becn1+/- or Map1lc3B-/-) mice and MTECs resisted CS-induced cilia shortening. Furthermore, CS increased the autophagic turnover of ciliary proteins, indicating that autophagy may regulate cilia homeostasis. We identified cytosolic deacetylase HDAC6 as a critical regulator of autophagy-mediated cilia shortening during CS exposure. Mice bearing an X chromosome deletion of Hdac6 (Hdac6-/Y) and MTECs from these mice had reduced autophagy and were protected from CS-induced cilia shortening. Autophagy-impaired Becn1-/-, Map1lc3B-/-, and Hdac6-/Y mice or mice injected with an HDAC6 inhibitor were protected from CS-induced mucociliary clearance (MCC) disruption. MCC was preserved in mice given the chemical chaperone 4-phenylbutyric acid, but was disrupted in mice lacking the transcription factor NRF2, suggesting that oxidative stress and altered proteostasis contribute to the disruption of MCC. Analysis of human COPD specimens revealed epigenetic deregulation of HDAC6 by hypomethylation and increased protein expression in the airways. We conclude that an autophagy-dependent pathway regulates cilia length during CS exposure and has potential as a therapeutic target for COPD.
Collapse
|