www.nature.com/scientificreports

OPEN

Targeting the PI3K/Akt/mTOR signalling pathway in Cystic Fibrosis R. Reilly1, M. S. Mroz2, E. Dempsey6, K. Wynne1, S. J. Keely2, E. F. McKone1,4, C. Hiebel5, C. Behl5 & J. A. Coppinger1,3

Received: 6 February 2017 Accepted: 27 June 2017 Published: xx xx xxxx

Deletion of phenylalanine 508 of the cystic fibrosis transmembrane conductance regulator (ΔF508 CFTR) is a major cause of cystic fibrosis (CF), one of the most common inherited childhood diseases. ΔF508 CFTR is a trafficking mutant that is retained in the endoplasmic reticulum (ER) and unable to reach the plasma membrane. Efforts to enhance exit of ΔF508 CFTR from the ER and improve its trafficking are of utmost importance for the development of treatment strategies. Using protein interaction profiling and global bioinformatics analysis we revealed mammalian target of rapamycin (mTOR) signalling components to be associated with ∆F508 CFTR. Our results demonstrated upregulated mTOR activity in ΔF508 CF bronchial epithelial (CFBE41o-) cells. Inhibition of the Phosphatidylinositol 3-kinase/Akt/Mammalian Target of Rapamycin (PI3K/Akt/mTOR) pathway with 6 different inhibitors demonstrated an increase in CFTR stability and expression. Mechanistically, we discovered the most effective inhibitor, MK-2206 exerted a rescue effect by restoring autophagy in ΔF508 CFBE41o- cells. We identified Bcl-2-associated athanogene 3 (BAG3), a regulator of autophagy and aggresome clearance to be a potential mechanistic target of MK-2206. These data further link the CFTR defect to autophagy deficiency and demonstrate the potential of the PI3K/Akt/mTOR pathway for therapeutic targeting in CF. CF is caused by mutations in the CFTR gene. CFTR is a chloride channel primarily responsible for facilitating conductance of chloride and other ions across epithelial membranes. The loss of a functional CFTR channel disrupts ionic homeostasis resulting in tenacious mucus production and a descent into a vicious cycle of chronic infection inflammation, and progressive lung fibrosis1. There are almost 2,000 different variants in the CFTR gene2 and up to 70% of CF patients contain at least one allele with a mutation at position 508 (c.1521_1523delCTT; commonly known as ΔF508), which results in the loss of Phe508 and disruption of the folding pathway of CFTR in the ER3. CFTR is synthesised in the ER and transport of CFTR involves chaperones that facilitate folding and trafficking from the ER to the cell surface membrane. As ΔF508 CFTR fails to achieve a wild type fold, it does not engage with COPII ER export and is processed through ER-associated degradation (ERAD)4, 5. The inability of protein p.Phe508del (ΔF508 CFTR) to achieve a correct folded state disrupts cellular proteostasis networks which protect the cell from acute stress6. In situations of cellular stress where the 26S proteasome is compromised or overwhelmed, ubiquitinated ΔF508 CFTR is transported to a perinuclear location to form aggresomes7. Autophagy is important in clearing protein aggregates after overload of polyubiquitinated proteins. The accumulation of misfolded protein aggregates has been described in several human disorders and the PI3K/Akt/mTOR axis plays a central role in maintaining cellular proteostasis, with mTOR activation regulating autophagy8. ΔF508 CFTR is described as an aggresome prone protein and aggresome formation has been linked to defective autophagy in CF9. In order to rationally develop therapeutic strategies to stimulate CFTR trafficking from the ER to the surface and maintain cellular proteostasis, it is critical to understand the protein interactions regulating CFTR transport. State of the art approaches such as mass spectrometry-based proteomics have been successful in identifying novel effectors of ΔF508 CFTR rescue from ERAD10. Silencing of key proteostatic chaperones led to a partial rescue of ΔF508 CFTR cell surface channel activity10, 11. Proteostasis regulators, such as Cystamine, have also been shown to rescue and stabilize functional ΔF508 CFTR protein, restoring autophagy in vivo12. Clinically, progress 1

Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland. 2Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, 9 Ireland. 3Royal College of Surgeons in Ireland, 123 St. Stephens’s Green, Dublin, 2 Ireland. 4St Vincent’s University Hospital, Dublin, 4 Ireland. 5Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg- University Mainz, Frankfurt, Germany. 6School of Biology and Environmental Science, University College Dublin, Belfield, Dublin 4, Ireland. Correspondence and requests for materials should be addressed to J.A.C. (email: [email protected])

SCIentIfIC Reports | 7: 7642 | DOI:10.1038/s41598-017-06588-z

1

www.nature.com/scientificreports/ has been made in recent years identifying therapeutics that target CFTR dysfunction in patients with specific mutations. However, small molecules that directly target the most common misfolded CFTR mutant, ΔF508, and improve its intracellular trafficking in vitro, have shown modest effects13. This study aimed to identify new therapeutic targets that will help address the unmet clinical need for CF patients with the ΔF508 mutation. In this study, we describe a role for mTORC1/2 regulation in ΔF508 expressing CF cells. We observed increased mTOR activity in ΔF508 CF bronchial epithelial cells and discovered that inhibition of the PI3K/Akt/ mTOR pathway improved CFTR stability and expression. We also showed that selected inhibitors, exerted rescue effects by restoring autophagy in ΔF508 expressing cells.

Methods

General Materials.  Cystic Fibrosis Bronchial Epithelial cells (CFBE41o-) expressing ∆F508 CFTR mutation, HBE41o- cells possessing a functional WT CFTR allele, and isogenic CFTR null cells (CFBE41o-, null) were obtained from Prof ’s. E Sorscher/J Clancy (University of Alabama, Birmingham). All other materials crucial to experimentation are included in Supplementary Information. Cell culture.  WT HBE41o- and CFBE41o- (ΔF508 homozygous) cells were cultured in MEM (Minimum Essential Medium) supplemented with 10% FCS (Fetal Calf Serum), 2 mM glutamine and 1% penicillin/streptomycin. Cells were grown at 37 °C in 5% CO2. PI3K/Akt/mTOR inhibitors or DMSO (vehicle control) were incubated with cells for 48 hours at indicated concentrations. DMSO concentration never exceeded 0.1%. For Ussing chamber experiments, cells were plated onto Costar snapwell inserts (12 mm), as previously described14. Transepithelial resistance (RT) was monitored and when the cells achieved 500–1,000 Ω cm2, they were used for experimentation. Adequate RT was usually achieved after 7 days in culture. For autophagy experiments, ΔF508 CFBE41o- cells were incubated in serum depleted media for 24 hours and treated with 10 µM MG132 to detect aggresomes. siRNA Transfections.  ΔF508 CFBE41o- cells were transfected with 5 nM siRNA using 4 µl Lipofectamine (Invitrogen, Grand Island, NY) as described previously15. Briefly, lipid-siRNA complexes were prepared in serum free media and added to cell suspensions in culture medium with 10% FCS. After 24 h, the medium was replaced. The target sequence was Bag3: 5′-GCCUGAAAACAAACCAGAATT-3′. Co-interacting Protein Identification Technology (Co-PIT) and LC-MS/MS sample preparation.  This protocol was carried out using a method previously described with minor modifications16.

Anti-mouse CFTR 596 antibody was coupled to DynabeadsTM protein G (Thermo Scientific, Rockford, IL) and crosslinked with 25 mM dimethyl pimelimidate. In parallel, WT HBE41o- and ΔF508 CFBE41o- cells were grown to confluence, washed in PBS and lysed on ice using 1% NP-40 solution containing protease inhibitor cocktail, pH 7.4, and phosphatase inhibitor cocktail. After sonication and clarification at 1,000 rpm for 10 mins, the pre-cleared cell extracts (4 mg of protein for each condition) were incubated with crosslinked antibody-dynabead complex overnight at 4 °C. Proteins bound to the antibody were recovered by washing three times with immunoprecipitation (IP) lysis buffer and eluted by incubation at 37 °C in 8 M urea with agitation on two occasions. Endogenous immunoprecipitates were isolated by methanol-chloroform precipitation. The eluate was resuspended in 100 mM TRIS pH 8.5. 0.2% Rapigest (Waters, Milford, MA) was added to the eluted proteins and reduced with 5 mM dithiothreitol. The samples were digested overnight at 37 °C with 2 µg Sequencing Grade Modified Trypsin (Promega, Madison, WI). Proteolysis was acidified with 5% formic acid. The peptides were isolated using a stop and go extraction protocol (STAGE)17. Briefly, the samples were washed in buffer A (0.1% TFA), immobilized on a C18 column, washed in buffer B (50% ACN, 0.1% TFA) and eluted in buffer C (1% TFA). Non-specific proteins were accounted for by performing immunoprecipitations IPs with CFTR null CFBE41o- cells.

Liquid Chromatography Mass Spectrometry Analysis.  Chromatography.  Tryptic digested peptide mixtures from the Endogenous IP samples were run on a Thermo Scientific Q Exactive mass spectrometer connected to a Dionex Ultimate 3000 (RSLC nano) chromatography system. Tryptic peptides were re-suspended in 0.1% formic acid. Each sample was loaded onto Biobasic Picotip Emitter (120 mm length, 75 μm ID) packed with Reprocil Pur C18 (1.9 μm) reverse phase media and was separated by an increasing acetonitrile gradient over 59.5 min at a flow rate of 250 nL/min. From 0–15 minutes the sample is loaded on to the column at 500 nl/min in 100% Buffer A (97% water, 2.5% acetonitrile, 0.5% acetic acid). From 15–16 minutes Buffer B (97% acetonitrile, 2.5% water, 0.5% acetic acid) increased from 1–2% and the flow rate decreased from 500–250 nl/min, from 16–64 minutes Buffer B increased from 2–35% at a flow rate of 250 nl/min, from 64–66 minutes Buffer B increased from 35–90% and the flow rate increased from 250–600 nl/min, from 66–70 minutes Buffer B remained at 90% and a flow rate of 600 nl/min, from 70–71 minutes Buffer B decreased from 90–1% at a flow rate of 600 nl/min, from 71–71.5 minutes Buffer B remained at 1%, at a flow rate of 600 nl/min. Ionisation and Mass Spectrometry.  The mass spectrometer was operated in positive ion mode with a capillary temperature of 220 °C, and with a potential of 2.1 kV applied to the frit. The heated capillary was kept at 150 °C. The capillary voltage was set at 45 V, and the tube lens was offset at 100 V. The mass spectrometer was operated in data-dependent positive ion-mode A top 12 method was used. Full MS scans were acquired in the Orbitrap mass analyzer over the range m/z 300–1600 with a mass resolution of 70000 (at m/z 200). The target value was 3.00 E+06. The twelve most intense peaks were fragmented in the high energy collision dissociation cell with a normalized collision energy of 27%, and tandem mass spectra were acquired in the Orbitrap mass analyzer with a mass resolution of 17500 at m/z 200. SCIentIfIC Reports | 7: 7642 | DOI:10.1038/s41598-017-06588-z

2

www.nature.com/scientificreports/ Analysis of Tandem Mass Spectra.  The Q-Exactive MS raw data files were de novo sequenced and searched against a Human UniProtKB database Release 2013_07, 20,266 entries using the search engine PEAKS Studio 6, for peptides cleaved with trypsin. Each peptide used for protein identification met specific Peaks parameters, for example; only peptide scores that corresponded to a false discovery rate (FDR) of ≤1% were accepted from the Peaks database search. The database searching parameters included up to two missed cleavages allowed for full tryptic digestion, and a precursor ion mass tolerance of 20 ppm. A fragment ion error tolerance of 0.03 Da was also included in the search parameters. A fixed modification of cysteine (57.02146) and up to 669 variable modifications were included in the Peaks PTM search.

Ingenuity and GO Analysis.  Proteins identified by mass spectrometry were displayed in Venn Diagrams using Venny Software (http://bioinfogp.cnb.csic.es/tools/venny). Proteins identified by mass spectrometry were overlaid onto a global molecular network developed from information contained in the ingenuity knowledge base (Ingenuity Systems , http://www.ingenuity.com, content version 12402621, release date: 2012-03-09). The top ten canonical pathways significantly (p 

mTOR signalling pathway in Cystic Fibrosis.

Deletion of phenylalanine 508 of the cystic fibrosis transmembrane conductance regulator (ΔF508 CFTR) is a major cause of cystic fibrosis (CF), one of...
NAN Sizes 1 Downloads 13 Views