p66Shc Mediates Mitochondrial Dysfunction Dependent on PKC Activation in Airway Epithelial Cells Induced by Cigarette Smoke

Airway epithelial mitochondrial injury plays a critical role in the pathogenesis of chronic obstructive pulmonary disease (COPD). The p66Shc adaptor protein is a newly recognized mediator of mitochondrial dysfunction. However, little is known about the effect of p66Shc on airway epithelial damage in the development of COPD. The aim of the present study is to investigate the roles of p66Shc and its upstream regulators in the mitochondrial injury of airway epithelial cells (Beas-2b) induced by cigarette smoke extract (CSE). Our present study revealed that CSE increased p66Shc expression and its mitochondrial translocation in concentration and time-dependent manners in airway epithelial cells. And p66Shc siRNA significantly attenuated mitochondrial dysfunction and cell injury when airway epithelial cells were stimulated with 7.5% CSE. The total and phosphorylated expression of PKCβ and PKCδ was significantly increased associated with mitochondrial dysfunction and cell injury when airway epithelial cells were exposed to 7.5% CSE. The pretreatments with pharmacological inhibitors of PKCβ and PKCδ could notably suppress p66Shc phosphorylation and its mitochondrial translocation and protect the mitochondria and cells against oxidative damage when airway epithelial cells were incubated with 7.5% CSE. These data suggest that a novel PKCβ/δ-p66Shc signaling pathway may be involved in the pathogenesis of COPD and other oxidative stress-associated pulmonary diseases and provide a potential therapeutic target for these diseases.

[1]  Min Chen,et al.  Cigarette smoke extract induces placental growth factor release from human bronchial epithelial cells via ROS/MAPK (ERK-1/2)/Egr-1 axis , 2016, International journal of chronic obstructive pulmonary disease.

[2]  R. Yan,et al.  Curcumin ameliorates alveolar epithelial injury in a rat model of chronic obstructive pulmonary disease. , 2016, Life sciences.

[3]  M. Hermann,et al.  Novel Insights into the PKCβ-dependent Regulation of the Oxidoreductase p66Shc* , 2016, The Journal of Biological Chemistry.

[4]  M. Hermann,et al.  cJun N-terminal kinase (JNK) phosphorylation of serine 36 is critical for p66Shc activation , 2016, Scientific Reports.

[5]  W. Zou,et al.  Nicotine reduces the levels of surfactant proteins A and D via Wnt/β-catenin and PKC signaling in human airway epithelial cells , 2016, Respiratory Physiology & Neurobiology.

[6]  E. Vagdatli,et al.  Effects of acute cigarette smoking on total blood count and markers of oxidative stress in active and passive smokers. , 2015, Hippokratia.

[7]  T. Soldati,et al.  Reactive oxygen species and mitochondria: A nexus of cellular homeostasis , 2015, Redox biology.

[8]  Cuk-Seong Kim,et al.  Trichostatin A Modulates Angiotensin II-induced Vasoconstriction and Blood Pressure Via Inhibition of p66shc Activation , 2015, The Korean journal of physiology & pharmacology : official journal of the Korean Physiological Society and the Korean Society of Pharmacology.

[9]  Wei Yang,et al.  High-dose alcohol induces reactive oxygen species-mediated apoptosis via PKC-β/p66Shc in mouse primary cardiomyocytes. , 2015, Biochemical and biophysical research communications.

[10]  Yi-Xiong Tan,et al.  The expression of p66shc in peripheral blood monocytes is increased in patients with coronary heart disease and correlated with endothelium-dependent vasodilatation , 2015, Heart and Vessels.

[11]  Lin Sun,et al.  PKCδ Promotes High Glucose Induced Renal Tubular Oxidative Damage via Regulating Activation and Translocation of p66Shc , 2014, Oxidative medicine and cellular longevity.

[12]  H. Tse,et al.  Mitochondrial transfer of induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial cells attenuates cigarette smoke-induced damage. , 2014, American journal of respiratory cell and molecular biology.

[13]  Shui-juan Zhang,et al.  Cigarette smoke extract-induced BEAS-2B cell apoptosis and anti-oxidative Nrf-2 up-regulation are mediated by ROS-stimulated p38 activation , 2014, Toxicology mechanisms and methods.

[14]  G. Borchard,et al.  Toll-like receptor 2 regulates the barrier function of human bronchial epithelial monolayers through atypical protein kinase C zeta, and an increase in expression of claudin-1 , 2014, Tissue barriers.

[15]  Jianping Zhou,et al.  The Expression of p66Shc Protein in Benign, Premalignant, and Malignant Gastrointestinal Lesions , 2014, Pathology & Oncology Research.

[16]  R. Foronjy,et al.  Cigarette smoke activates the proto-oncogene c-src to promote airway inflammation and lung tissue destruction. , 2014, American journal of respiratory cell and molecular biology.

[17]  Y. Tesfaigzi,et al.  Molecular processes that drive cigarette smoke-induced epithelial cell fate of the lung. , 2014, American journal of respiratory cell and molecular biology.

[18]  Sina Zarrintan,et al.  Prolonged cigarette smoke exposure alters mitochondrial structure and function in airway epithelial cells , 2013, Respiratory Research.

[19]  Michael A Thompson,et al.  Mitochondria in lung diseases , 2013, Expert Review of Respiratory Medicine.

[20]  R. Yan,et al.  Calreticulin-STAT3 Signaling Pathway Modulates Mitochondrial Function in a Rat Model of Furazolidone-Induced Dilated Cardiomyopathy , 2013, PloS one.

[21]  J. Schauer,et al.  Mitochondrial Genetic Background Modifies the Relationship between Traffic-Related Air Pollution Exposure and Systemic Biomarkers of Inflammation , 2013, PloS one.

[22]  P. Pelicci,et al.  The P66Shc/Mitochondrial Permeability Transition Pore Pathway Determines Neurodegeneration , 2013, Oxidative medicine and cellular longevity.

[23]  Y. Katayama,et al.  Protein kinase C (PKC) isozyme-specific substrates and their design. , 2012, Biotechnology advances.

[24]  T. Hibi,et al.  p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice. , 2012, Journal of hepatology.

[25]  A. Azghani,et al.  Inhibition of protein kinase C attenuates Pseudomonas aeruginosa elastase-induced epithelial barrier disruption. , 2011, American journal of respiratory cell and molecular biology.

[26]  Wen-bin Tang,et al.  p66Shc mediates high-glucose and angiotensin II-induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway. , 2010, American journal of physiology. Renal physiology.

[27]  John D. Taylor COPD and the response of the lung to tobacco smoke exposure. , 2010, Pulmonary pharmacology & therapeutics.

[28]  C. Steegborn,et al.  The Lifespan-regulator p66Shc in mitochondria: redox enzyme or redox sensor? , 2010, Antioxidants & redox signaling.

[29]  Yi Na Wang,et al.  Genistein protects against UVB-induced senescence-like characteristics in human dermal fibroblast by p66Shc down-regulation. , 2010, Journal of dermatological science.

[30]  M. Profita,et al.  Chronic obstructive pulmonary disease and neutrophil infiltration: role of cigarette smoke and cyclooxygenase products. , 2010, American journal of physiology. Lung cellular and molecular physiology.

[31]  M. Rajendran,et al.  p66Shc—a longevity redox protein in human prostate cancer progression and metastasis , 2010, Cancer and Metastasis Reviews.

[32]  H. Matsuzaki,et al.  Epidermal growth factor receptor phosphorylates protein kinase C {delta} at Tyr332 to form a trimeric complex with p66Shc in the H2O2-stimulated cells. , 2007, Journal of biochemistry.

[33]  C. Cool,et al.  Lung disease and PKCs. , 2007, Pharmacological research.

[34]  S. Minucci,et al.  Protein Kinase C ß and Prolyl Isomerase 1 Regulate Mitochondrial Effects of the Life-Span Determinant p66Shc , 2007, Science.

[35]  S. Menini,et al.  Deletion of p66Shc Longevity Gene Protects Against Experimental Diabetic Glomerulopathy by Preventing Diabetes-Induced Oxidative Stress , 2006, Diabetes.

[36]  B. Chorley,et al.  Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C{delta}-mediated mechanism. , 2005, The American journal of pathology.

[37]  S. Minucci,et al.  Electron Transfer between Cytochrome c and p66Shc Generates Reactive Oxygen Species that Trigger Mitochondrial Apoptosis , 2005, Cell.

[38]  S. Shen,et al.  Mitochondrial-dependent, reactive oxygen species-independent apoptosis by myricetin: roles of protein kinase C, cytochrome c, and caspase cascade. , 2005, Biochemical pharmacology.

[39]  James D Crapo,et al.  The Role of Oxidative Stress in Chronic Obstructive Pulmonary Disease , 2004, COPD.

[40]  H. Gottlieb,et al.  Superoxide organic chemistry within the liposomal bilayer, part II: a correlation between location and chemistry. , 2002, Free radical biology & medicine.

[41]  S. Alkan,et al.  Caspase activation and disruption of mitochondrial membrane potential during UV radiation-induced apoptosis of human keratinocytes requires activation of protein kinase C , 2002, Cell Death and Differentiation.

[42]  B S Polla,et al.  Effects of tobacco smoke and benzo[a]pyrene on human endothelial cell and monocyte stress responses. , 2001, American journal of physiology. Heart and circulatory physiology.

[43]  Pier Paolo Pandolfi,et al.  The p66shc adaptor protein controls oxidative stress response and life span in mammals , 1999, Nature.