The Novel Small Molecule Inhibitor, OSU-T315, Suppresses Vestibular Schwannoma and Meningioma Growth by Inhibiting PDK Function in the AKT Pathway Activation

Research Article

Austin J Med Oncol. 016; 3(1): 105.

The Novel Small Molecule Inhibitor, OSU-T315, Suppresses Vestibular Schwannoma and Meningioma Growth by Inhibiting PDK Function in the AKT Pathway Activation

Mercado-Pimentel ME1,,3, Igarashi S1,, Dunn AM1,, Behbahani M1,, Miller C1,, Read CM1 and Jacob A1,,3,4*

¹Ear Institute, University of Arizona, USA

⊃Department of Otolaryngology, University of Arizona, USA

³Arizona Cancer Center, University of Arizona, USA

4BIO5 Institute, University of Arizona, USA

*Corresponding author: Jacob A, Department of Otolaryngology, University of Arizona, Tucson, USA

Received: April 05, 016; Accepted: April 18, 016; Published: April 21, 2016

Abstract

Activation of PKB/AKT signaling, which requires PDK1 and PDK function, drives Vestibular Schwannoma (VS) and meningioma growth. PDK function is defined as a molecule that phosphorylates AKT-Ser473. Integrin-Linked Kinase (ILK) functions as PDK in PKB/AKT activation in many cancers; therefore, we hypothesized that OSU-T315, a small molecule ILK inhibitor, will inhibit the ILK-PDK function in PKB/AKT signaling activation in VS and meningioma cell growth. OSU-T315 decreased cell viability at IC50 < μM in VS (HEI193) and meningioma (Ben-Men-1) cell lines, in primary cells at < 3.5μM, while in normal primary Schwann cells at 7.1μM. OSU-T315 inhibits AKT signaling by decreasing phosphorylation at AKT-Ser473, AKT-Thr308, ILK-Ser46 and ILKThr173. In addition, OSU-T315 affected the phosphorylation or expression levels of AKT downstream proliferation effectors as well as autophagy markers. Flow cytometry shows that OSU-T315 increased the percentage of cells arrested at G/M for both, HEI193 (39.99%) and Ben-Men-1 (6.96%) cells, compared to controls (1.54%, 8.47%). Two hours of OSU-T315 treatment increased cell death in both cell lines (34.3%, 9.1%) versus untreated (1.1%, 8.1%). Though longer exposure increased cell death in Ben-Men-1, TUNEL assays showed that OSU-T315 does not induce apoptosis. OSU-T315 was primarily cytotoxic for HEI193 and Ben-Men-1 inducing a dysregulated autophagy. Our studies suggest that OSU-T315 has translational potential as a chemotherapeutic agent against VS and meningioma.

Keywords: PDK; Integrin-linked kinase; AKT; OSU-T315; Vestibular schwannoma; Autophagy

Abbreviations

NF: Neurofibromatosis Type ; VS: Vestibular Schwannoma; PKB/AKT: Protein Kinase B/v-Akt Murine Thymoma Viral Oncogene Homolog; PI-3K: Phosphatidylinositol 3 Kinase; PIP: Phosphatidylinositol diphosphate; PIP3: Phosphatidylinositol Triphosphate; Thr: Threonine; Ser: Serine; PDK: Phosphoinositide- Dependent Kinase; mTORC: Mammalian Target of Rapamycin Complex ; ILK: Integrin-Linked Kinase; PAK1: P1-Activated Kinase 1; LC3: Light Chain 3; Atg: Autophagy-Related; GSK-3β: Glycogen Synthase Kinase -3 beta; OSU-T315: Ohio State University-T315; IC50: Inhibitory Concentration at 50%; PI: Propidium Iodide; si RNA: Small Interference Ribonucleic Acid

Introduction

Neurofibromatosis type (NF) is an autosomal-dominant familial syndrome caused by a loss-of-function mutation in the NF gene, which is localized on chromosome and encodes the tumor suppressor protein, merlin. NF syndrome occurs in 1:5,000 individuals. Presenting around 0 years of age, NF primarily affects the nervous system, eyes and skin [1]. Abnormalities in the nervous system include bilateral vestibular schwannomas (VS) and meningiomas, among others. VS are benign intracranial tumors that originate along the vestibulocochlear nerve and cause hearing loss, tinnitus and imbalance []. Present treatment options are limited to surgery or radiation. Unfortunately, the former poses a number of serious risks, including cerebrospinal fluid leaks, meningitis, intracranial hemorrhage, stroke, coma and death, while the latter raises concerns of near-term treatment failure, latent tumor growth, malignant transformation, and secondary skull-base malignancies [3,4]. There is currently no FDA approved chemotherapeutic agents to treat NF-associated tumors due to a poor understanding of the molecular mechanisms involved in this disease. However, recent studies have revealed key molecules that play a role in the growth and development of vestibular schwannoma tumors.

The serine/threonine kinase Protein Kinase B (PKB)/AKT, modulates several downstream molecules involved in cell survival, growth and proliferation. We have previously shown that the PI-3K (Phosphatidylinositol 3 Kinase)/AKT signaling is activated in VS [5]. Activation of the PI-3K/AKT pathway requires PI-3K to catalyze the phosphorylation reaction of Phosphatidylinositol Diphosphate (PIP) to Phosphatidylinositol Triphosphate (PIP3), which recruits AKT to the lipid raft compartments in the cell membrane. This event facilitates the subsequent phosphorylation of AKT-Thr308 and AKT-Ser473 by Phosphoinositide-Dependent Kinase (PDK)1 and PDK, respectively [6]. Both AKT-Thr308 and AKT-Ser473 phosphorylation are highly induced by upstream signals [7,8]. PDK1 is a well-described single molecule in all cells; however, studies on AKT-Ser473 phosphorylation have implicated the Rictor-Mtor Complex (mTORC), Integrin-Linked Kinase (ILK) and rictor/ILK complex, among other kinases, to act as a “PDK” [9-14].

ILK interacts with the cytoplasmic beta1 subunit of integrins, to regulate adhesion-mediated migration, invasion, proliferation, anchorage independent growth, and survival [9]. Deregulation of ILK has been implicated in the pathogenesis of several human malignancies, including ovarian carcinoma, melanoma, and glioblastomas, among others [15]. Together, these studies indicate that ILK has autonomous function and is also an important upstream kinase for activation of AKT, thereby regulating cell processes required for cell survival, such as suppressing apoptosis and promoting cell cycle progression.

ILK is a 59 KDa cytoplasmic serine-threonine protein kinase made up of four ankyrin repeats at the N-terminus that facilitate protein interaction, a central pleckstrin homology-like domain (pH) that mediates phosphoinositide binding, and a C-terminal kinase domain, which acts to phosphorylate downstream targets such as AKT (at Ser473). Supporting this idea, studies using COS-1 and NIH 3T3 cells show that PAK serves as a scaffold to activate PDK1 and aid in the recruitment of AKT to the membrane [16]. Additional biochemical, co-immunoprecipitation, and mutation analyses show that the binding region of PIP3 in ILK is important for AKT phosphorylation [13]. These studies show that ILK complexes with AKT and PDK1, and that ILK can disrupt the PDK1/AKT association allowing phosphorylated ILK-Ser343 in the activation loop to phosphorylate AKT at Ser473.

A more relevant study in breast cancer cell lines has shown that P1-Activated Kinase 1 (PAK1) activates ILK by phosphorylating threonine-173 and serine-46, aiding in cell proliferation and motility [17]. Additional studies by Kissil et al. have shown that over expression of the NF protein, merlin, inhibits PAK activation, while the loss of merlin increases it [18]. This PAK activation may in turn be causing high activation of the ILK-signaling pathway in NF deficient tumors.

Though the role of autophagy in cancer progression is controversial, several studies have shown that autophagy and apoptosis mutually and negatively regulate each other [19-1]. While autophagy impacts the turnover of protein aggregates and damaged organelles, apoptosis eliminates unwanted cells entirely. Both processes appear to interlink in determining the fate of a given cell. Caspases are the main initiators of apoptosis and potentially mediate the complex crosstalk between autophagy and apoptosis. Caspase-9 is involved in processing apoptotic downstream effector caspases such as caspase-3, -6 and -7. Caspase-9 has also been shown to regulate autophagy-mediated cell survival in breast cancer cells []. Though several studies show molecules involved in the participation of caspase-9 in autophagy or apoptosis, [3] more work is needed to reveal the molecular mechanisms whereby caspase-9 participates in regulating the two processes. The autophagic marker, LC3 (light chain 3) is a homologue of the yeast Atg8 and it is found in humans in three isoforms, LC3 A, LC3 B and LC3 C. LC3 undergoes post-translational modification to yield LC3-I, which is lipidated by the Atg5-Atg1 conjugate to become associated as LC3-II with authophagic vesicles [4-6].

In recent years, ILK and AKT inhibition have been suggested as exciting targets for drug development against human cancers and vestibular schwannomas [7,8]. OSU-T315 has been reported as a novel ILK inhibitor that induces autophagy and apoptosis in prostate and breast cancer cell lines [9]. Here, we report initial validation studies of OSU-T315 as a potential treatment for vestibular schwannomas and meningiomas. Exposure of HEI193 schwannoma and Ben-Men-1 meningioma cells to OSU-T315 caused a significant decrease in cell viability at low micro-molar IC50 (inhibitory concentration 50%) concentrations, and this effect was correlated with the AKT pathway inhibition, cell cycle arrest at G/M, and cell death by deregulated autophagy. We report that OSU-T315 is an inhibitor of the cell survival PKB/AKT pathway via deactivation of PAK and ILK in schwannoma and meningioma cells, indicating that it is a promising new agent for preclinical drug development against NF-associated tumors.

Materials and Methods

Cells

Cell lines: HEI193 and Ben-Men-1 cell lines are immortalized human vestibular schwannoma and benign meningioma cells respectively. HEI193 cells have a mutation in the NF gene causing a splicing defect in the NF transcript but expressing moderately the active growth suppressive, merlin [30]. Ben-Men-1 cells lack one copy of chromosome and the other allele has a mutation in exon 7, which causes a premature stop codon and therefore do not express merlin [31].

Primary cells: Cells were retrieved from tumors of sporadic vestibular schwannoma and meningioma patients. Institutional Review Board protocols for the acquisition of surgically removed VS specimens are in place. VS and meningioma cells were identified with S100 and Epithelial Membrane Antigen (EMA) markers, respectively, and visualized with confocal microscope. Cell lines and primary cells were tested for contamination and authenticated by immunohistochemistry.

Cell culture

Cell lines were plated at 7.5X10³ cells/well in 96-well plates at 37o C, 5% CO overnight in complete medium (DMEM high glucose supplemented with 10% FBS) for Ben-Men-1 and HEI193 (plus 100 IU/ml penicillin-streptomycin) cells. Primary cells isolated from fresh VS tumors were grown in DMEM/10% FBS, 10 ng/mL β-heregulin (R&D Systems) and 0.μM Forskolin (sigma) under the same conditions as the cell lines.

Cell viability and cell cycle proliferation assays

MTT Assay: The treatment efficacy of OSU-T315 (OSUCCC Medicinal Chemistry Shared Resources) against HEI193, Ben- Men-1 and primary cells was measured by detecting reduction of the tetrazolium salt, MTT (3-(4,5- dimethylthiazole--yl)-,5-diphenyltetrazolium bromide), to formazan. Cell lines and primary cells were seeded at 7.5X103 and X104 cells/well in 96-well plates respectively, and treated with OSU-T315 in a dose dependent manner ranging from 0.5 – 5μM (0.5, 1, 1.5, .0, .5, 3, 3.5, 4, 4.5 and 5) versus untreated controls for 7 hours at 37oC, 5% CO. Experiments were repeated at least four times and each drug concentration was tested in quadruples. MTT assay was performed according to Porchia et al. [3]. Briefly, cells were incubated with 00 μl of 0.5 mg/ml of MTT (Alfa Aesar, # 98-93-1) in complete medium for 4 hours, followed with 00 μl of DMSO/well and incubated for 5 min. Colorimetric readings were performed in the BioTek Synergy HT plate reader at 540 nm, and the 50% inhibitory concentration (IC50) was calculated using linear regression.

Flow Cytometry/FACS (Fluorescent Activated Cell Sorter)

Cell cycle analysis: HEI193 cells were treated with .5 and 5μM of OSU-T315 for 4 hours while Ben-Men-1 cells were treated with 1, , 3 and 4 μM of OSU-T315 for 4 hours. After incubation, floating cells were harvested and combined with trypsinized adherent cells. Cells were washed with cold PBS, pelleted at 1,000 rpm for 10 min, resuspended in 1 ml ice-cold 70% ethanol and fixed overnight at -0oC. Fixed cells were washed with cold PBS and resuspended in 1 ml cold PBS. To ensure only DNA staining, 500μg/ml of RNase A (Sigma- Aldrich) and 40 μg/ml of Propidium Iodide (PI) (Sigma-Aldrich) were added. Cells were stained at 37° C, 5% CO for 30 min.

Apoptosis analysis: HEI193 and Ben-Men-1 cells were seeded at 7X105 cells/100 mm culture plates. Cells were treated with 1, , 3, 4 μM of OSU-T315 versus non-treated cells in triplicates for , 4, 48 and 7 hours at 37oC, 5% CO. Cells were trypsinized and resuspended in 10ml of cold PBS. Pelleted cells were stained with 100μl of 1μg/ml of PI in Annexin binding buffer. Staining of the cells with Annexin V conjugates for flow cytometry analysis was carried out according to the Alexa Fluor 488 Annexin V Dead Cell Apoptosis Kit (Molecular Probes #A1301). Flow cytometry was performed using the BD FACScan and the Modfit software.

TUNEL apoptosis assay

HEI193 and Ben-Men-1 cells were seeded at 7X104 cells/cover slips for 4 hours and treated with and 4 μM of OSU-T315 versus untreated negative controls. Positive controls were treated with DNase I for 0 minutes. TUNEL staining was performed according to ClickiT ® TUNEL Alexa Fluor® Imaging Assay (Invitrogen) instructions. Briefly, cells were fixed with 3.7% paraformaldehyde, permeabilized with 0.5% Triton-X 100 in PBS1X, and probed with the EdUTP nucleotide mixture. Fixed and stained cells were then examined using a fluorescence deconvolution light microscope. Gray scale images were captured due to improved contrast/visualization of individual cells. Images were processed and assembled using Adobe Photoshop CS6.1. Auto Contrast tool was applied to the negative control images to better visualize the cells.

Western blots

Sub-confluent cells were treated with 0, .5 and 5μM of OSU-T315 for 48 and 7 hours for cell lines, and 7 hours for primary cells. Cells were then harvested and homogenized in lysis buffer (1% SDS, 10mM EDTA, 50mM Tris, pH 8.1) supplemented with protease/phosphatase inhibitor cocktail (Thermo Scientific, Waltham MA). Electrophoresis was performed with 5μg or 50μg of protein/lane in a 4-0% gradient SDS- polyacrylamide gel (Thermo Scientific, Waltham MA). Proteins were transferred to PVDF membranes (Millipore, Billerica MA), and probed with specific antibodies of interest and horseradish peroxidase-conjugated secondary antibodies. The chemiluminescent signals were detected in X-ray films.

Antibodies

Antibodies were obtained from: 1) Cell Signaling Technology: Akt Antibody (#97), Phospho-Akt (ser473) (D9E)

XP Rabbit mAb (#4060), Phospho-Akt (Thr308) Antibody (#975), Cleaved Caspase-9 Antibody (#9509), GSK-3 β

(7C10) Rabbit mAb (#9315), Phospho-GSK-3β (Ser9) Antibody (#9336), ILK1 Antibody (#386), LC3A (D50G8) XP Rabbit mAb (#4599) and LC3B (D11) XP Rabbit mAb (#3868). ) Sigma-Aldrich: Monoclonal Anti- β–Actin (A1978). 3) Thermo Scientific: phospho- ILK pThr173 Antibody (PA5-1917), Phospho-ILK pSer46 Polyclonal Antibody (PA5-1943), Aurora-B Polyclonal Antibody (PA5-14076). Dilutions of all primary and secondary antibodies were 1:1000 and 1:10,000 respectively.

Results OSU-T315 suppresses schwannoma and meningioma proliferation at low micromolar concentrations

OSU-T315 inhibited proliferation of vestibular schwannoma HEI193 cells, and meningioma Ben-Men-1 cells in a dose dependent manner, with IC50 values of 1.8 and 1.58 μM respectively (Figure 1A). OSU-T315 treatment of primary vestibular schwannoma and meningioma cells decreased cell viability at IC50, 3.41 μM and .5 μM respectively (Figure 1B and 1C), while in normal Schwann cells it decreased cell viability at 7 μM (Figure 1D). These data indicate that OSU-T315 is a putative selective drug for VS and meningioma treatment.