Identification and characterization of small molecules that inhibit

Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
1
Identification and characterization of small molecules that inhibit nonsense mediated RNA
decay and suppress nonsense p53 mutations
Leenus Martin1, Arsen Grigoryan2, Ding Wang1, Jinhua Wang3, Laura Breda4, Stefano Rivella4,
Timothy Cardozo2, Lawrence B. Gardner1,2,3, #
Running title: Pharmacological inhibition of NMD
Key Words: nonsense mutations, nonsense mediated RNA decay, p53, tumor suppressor
genes, gene expression
Departments of Medicine1, Biochemistry and Molecular Pharmacology2, and the NYU Cancer
Institute3, New York University School of Medicine, 550 1st Avenue NYC, NY 10016,
Departments of Pediatrics and Cell and Biology Development, Weill Cornell School of Medicine,
New York, NY, 100214
#
Corresponding author: Lawrence B. Gardner, Division of Hematology and Medical Oncology,
550 1st Avenue, Smilow 1204, New York, NY 10016.
212-263-8038
[email protected]
Financial Support: NIH and the European Community and the Children's Cancer and Blood
Foundation
Word Count: 3799, 5 Figures.
The authors declare no conflicts of interest
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
2
Abstract
Many of the gene mutations found in genetic disorders, including cancer, result in premature
termination codons (PTCs) and the rapid degradation of their mRNAs by nonsense mediated
RNA decay (NMD). We used virtual library screening (VLS) targeting a pocket in the SMG7
protein, a key component of the NMD mechanism, to identify compounds that disrupt the
SMG7-UPF1 complex and inhibit NMD.
Several of these compounds upregulated NMD
targeted mRNAs at nanomolar concentrations with minimal toxicity in cell based assays. As
expected, pharmacological NMD inhibition disrupted SMG7-UPF1 interactions. When used in
cells with PTC mutated p53, pharmacological NMD inhibition combined with a PTC “readthrough” drug led to restoration of full-length p53 protein, upregulation of p53 downstream
transcripts, and cell death. These studies serve as proof-of-concept that pharmacological NMD
inhibitors can restore mRNA integrity in the presence of PTC and be used as part of a strategy
to restore full length protein in a variety of genetic diseases.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
3
Introduction
Human genetic disorders are caused by diverse types of mutations. Many of these mutations,
including nonsense mutations, frameshift mutations, and mutations that cause alternative
splicing events, result in premature termination codons (PTCs). For example, approximately
15% of the mutations leading to Duchenne muscular dystophy (DMD), 10% of the mutations
responsible for cystic fibrosis, and common β globin mutations responsible for thalassemia are
PTC mutations (1-3). It has been estimated that up to 30% of all mutations resulting in human
genetic disorders result in PTCs (4). In addition, many acquired mutations in cancer, including
those that disable p53 and BRCA1, ATM, VHL, and NF1 and NF2, result in PTCs (5).
Many transcripts carrying a PTC are targeted for rapid degradation before they can be
translated into protein through a multistep process termed nonsense mediated RNA decay
(NMD).
The molecular mechanism of NMD has not been fully delineated, though working
models have been proposed. During the processing of mammalian pre mRNA, introns are
excised and marked by a multi-protein exon junction complex (EJC) (6-8). When the translation
complex pauses at a PTC that is upstream of an EJC, eukaryotic release factors recruit the
RNA helicase UPF1, a vital component of the NMD mechanism (9). UPF1 then associates with
UPF2, a component of the EJC, and is phosphorylated at several sites by SMG1 (10).
Phosphorylated UPF1 recruits SMG-7 and SMG5, two 14-3-3 proteins which each bind to UPF1
and form a heterodimer, and results in the de-phosphorylation of UPF-1 by PP2A (11-12). The
phosphorylation and de-phosphorylation of UPF1 are necessary steps prior to the transcript’s
degradation by exonucleases (12-13).
One approach to treat genetic disorders and to restore tumor suppressor genes with PTC
mutations is to pharmacologically promote ribosomes to read-through a PTC and produce a full-
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
4
length protein (14). For example treating cells with the antibiotic gentamicin increases the full
length expression of both PTC mutated β globin and PTC mutated p53 (15-16). Gentamicin
treatment of cells with PTC mutations of the cystic fibrosis transmembrane conductance
regulator (CFTR) can increase cellular chloride transport (17-18) and patients with PTC
mutations treated with intranasal gentamicin increase CFTR protein expression and improve the
potential difference of nasal epithelial cells (19). While application of gentamicin or the closely
related geneticin (G418) is not practical clinically, a recent high throughput screen identified a
small molecule, Ataluren, which has similar properties but appears to be more potent and less
toxic (20), and the use of antisense oligonucleotides to promote exon skipping are also being
tested in muscular dystrophy (reviewed in (21)). The clinical activities of these approaches are
likely to be enhanced with higher cellular concentrations of PTC mutated mRNA. Indeed, in
vitro experiments have demonstrated that the expression of PTC mutated CFTR, as well as
CFTR mediated chloride transport, are improved in gentamicin treated cells when UPF1 or
UPF2 are also depleted (22).
Because translation is necessary for NMD (23), inhibitors of translation are effective inhibitors of
NMD (24).
Unfortunately these drugs are not candidates for clinical therapies because of
general toxicity and, as translation is inhibited, the ultimate goal of restoring protein expression
is not achieved. We have recently demonstrated that NMD inhibition can be achieved via other
mechanisms.
Specifically, we have shown that phosphorylation of the translation initiation
factor eIF2α by a variety of cellular stresses inhibits NMD (5, 25-26). We have also determined
that modest (80%) depletion of UPF1 or UPF2 can suppress NMD activity without diminishing
the proliferation or survival of cells (26). These observations suggest that the pharmacological
inhibition of NMD can be achieved with limited toxicity or adverse effects. We hypothesized that
the combination of a drug that can bypass a PTC and/or promote exon skipping in combination
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
5
with a drug that inhibits NMD would be a synergistic combination and serve as an effective
platform to treat genetic diseases. To pursue this approach, we performed a virtual screen to
identify commercially available compounds predicted to dock within a SMG7 pocket previously
shown to be vital in the binding of UPF1. We then tested whether these compounds interfere
with NMD, and using PTC mutated p53 as a proof of principle model, whether the
pharmacological inhibition of NMD would make ribosomal read-through strategies more
effective in producing full length proteins.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
6
Materials and Methods:
Virtual Library Screening: The crystallographic structure of SMG7 (PDB ID: 1ya0) was analyzed
using ICM PocketFinder (Molsoft, LLC, La Jolla, CA). An appropriately sized pocket (based on
criteria previously reported (27)) was selected as the target site for virtual library screening
using ICM-VLS software (Molsoft, LLC, La Jolla CA). ICM-VLS uses global optimization with a
biased probability Monte Carlo conformational search to rapidly dock fully flexible, full-atom
models of the ligands to a set of grid potential maps calculated from the coordinates of the
atoms in the protein receptor. Each ligand docking conformation is then evaluated with a scoring
function. The ICM scoring function integrates van der Waals energy, electrostatics, hydrogen
bonding, conformational entropy loss and solvation electrostatic energy change (28). ICM-VLS
was used to dock the entire set of 436,115 compounds from Chembridge Express Library using
default ICM-docking parameters on three 3.0-GHz Intel Xeon processors.
Cell lines and reagents: U2OS, Hela, 293, BJhTert, PBMC, MDA231, DU145, T98G and
HCT116 cells were grown as previously described (29-30). N417 cells were grown in RPMI
containing 10% FBS. Calu-6 (gift of E. Welch) and HDQP1 cell lines (DSMZ German Collection
of Microorganism and Cell Cultures) were grown in DMEM with 10% Fetal Bovine Serum. Pools
of β-Globin mRNA-expressing cells were generated as previously described (25). Compounds
were obtained from Chembridge. UPF1 shRNA (26) and SMG7 shRNA (TRCN0000127998)
were obtained from Sigma. Primer sequences for β-Globin, endogenous NMD targets, p53 and
p53 downstream targets are available upon request.
Immunoblots and RNA assessment: Immunoblots were prepared as previously described (29)
and membranes were probed with antibodies against p53 (Abcam Inc, ab26), SMG1 (Santa
Cruz Biotechnology, Inc. 6-RE13), UPF1/Rent1 (SC H-300), SMG7 (A302-170A, Bethyl
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
7
Laboratories, Inc). Immunoprecipitations were performed as described (10). Briefly, cells were
lysed and incubated 30 mins at 4°C with buffer containing 50mM Tris-HCL at pH 7.4, 50mM
NaCL, 0.05% Tween-20, 10mM tetrasodium pyrophosphate, 100mM NaF, 17.5 mM βGlycerophosphate, 1mM PMSF and protease inhibitor (Roche). Lysates were clarified and
incubated with 200µg/mL RNaseA (Qiagen) at room temperature for 15 mins with gentle rotation
prior to 4°C overnight immunoprecipitation with the appropriate antibodies. Isolation of RNA for
cDNA generation, real-time PCR, DRB treatment, RNA stability experiment, expression profiling
and expression array data analysis were as described previously (25-26). For unbiased gene
expression experiments, cells were treated with 50 μM NMDI14 for six hours, 100 μg/ml
emetine for three hrs, or depleted of UPF1 and RNA was harvested, prepared, hybridized to
Affymetrix HG-U133 Plus 2.0 GeneChips and analyzed as previously described (26).
Cell Viability Assay and Proliferation Assay: To assess viability cells were cultured in 6 well
dishes and incubated with DMSO, G418, NMDI alone or G418 with NMDI together for the
indicated hours. After incubations, cells and media were collected and cells viability was
measured as described (31). To assess cell proliferation U2OS, Hela and BJ-htert cells were
cultured in 6 well plates and, after 24hrs, treated with NMDI14 for 0, 24, 48 and 72hrs. The cells
were collected and viable cells were counted by using the Countess Automated Cell Counter
(Invitrogen).
Protein Synthesis (35S incorporation assay): U2OS, Hela and BJ-hTERT cells were incubated
with DMSO, NMDI14 or Emetine as indicated. The cells were labeled with 100 µCi/mL [35S]
Methionine for 30 min and the protein were precipitated using standard TCA precipitation
method as described previously (26). The precipitated protein was collected in glass fiber filters
and [35S]Methionine incorporation was measured by using liquid scintillation counter
(PerkinElmer, Inc.).
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
8
Results
Identification of potential nonsense mediated RNA decay inhibitors (NMDIs): Prior studies
have demonstrated that NMD activity is dependent on the formation of a UPF1-SMG7 complex
(Fig 1A), which has been crystallographically resolved (11). Though UPF1 and SMG7 may
have additional non-NMD roles, the interaction between the two appears to be unique for NMD.
Thus the disruption of this complex is an ideal target for NMD inhibition while minimimzing the
toxicity that might be seen with other known mechanism of NMD inhibitors, including translation
inhibitors and inducers of eIF2α phosphorylation (24-25).
We elected to take a 3D structure-activity approach to NMDI discovery by targeting this protein
interface. Only one pocket on the 3D structure of SMG7 was both of suitable size and volume
(between 150 and 550A3) and was lined with functionally sensitive amino acid side chains that
mediate the interaction with UPF1 (11) (Fig 1B). This pocket was selected as the target site for
virtual library screening of a collection of 315,102 compounds from the ChemBridge Express
Library (San Diego, CA, USA). The resulting compounds achieving a docking score of <32 (see
materials and methods) were further filtered by selecting those with extensive hydrogen bonds
and van der Waals contacts, followed by hierarchical clustering for a diversity of scaffolds,
resulting in the identification of 31 diverse compounds highly structurally and chemically
compatible with the target SMG7 pocket (Fig 1C, 1D).
Identified compounds inhibit NMD in a cell based assay.
To test these putative NMD
inhibitors (NMDIs), we utilized a common and straight-forward assay for NMD.
We first
generated fibroblasts stably expressing either a wild-type β globin construct or a PTC 39
mutated β globin gene construct well established to be degraded by NMD (32). In our initial
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
9
screen we treated cells expressing these genomic constructs with 50 μM of the 31 identified
compounds for 6 hours, a condition which led to no gross toxicity in these cells. RNA was then
harvested and expression of PTC β globin and wild-type β globin mRNA was determined using
real time PCR.
We identified ten compounds that selectively increased the expression level of PTC 39 β globin
greater than 2-fold, without affecting wild-type β globin expression, with a p value < 0.05 (Fig
2A). Because of rapid degradation by NMD, the steady-state mRNA expression of the PTC39 β
globin was approximately 3% of the wild-type β globin expression. Treating cells with NMDI14
for 6 hours, for example, led to an increase of PTC 39 β globin to 12%, a relative four-fold
increase that, if resulting in biologically active hemoglobin, would be sufficient to ameliorate the
clinical symptoms of thalassemia (Fig 2B).
When these ten compounds were tested at
decreasing concentrations, we identified several NMDIs active at nanomolar concentrations,
with some active over a wide range of concentrations and others exhibiting more narrow
effective concentration which peaked at micromolar concentrations and displayed even a
biphasic effect (Fig 2C and data not shown). These compounds, previously not reported to
affect NMD, represent a diverse group of chemical scaffolds (Fig 2D and data not shown).
We
chose to focus the majority of our further studies on one of these compounds, ethyl 2-{[(6,7 -dimethyl--3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetyl]amino}-4,5-dimethyl-3thiophenecarboxylate (NMDI14), which demonstrated dose dependent activity, with significant
activity at low concentrations.
Although the activity of our identified NMDIs does not depend on the mechanism of action,
because these drugs were screened in silico to fit into the SMG7 pocket necessary to bind
UPF1, we expected that NMDI14 would interfere with the interaction of UPF1 and SMG7. We
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
10
immunoprecipitated SMG7, in the presence of RNAse and confirmed that both over-expressed
(Fig 2E left) and endogenous (Fig 2E, right) SMG7 interacted with over-expressed and
endogenous UPF1, respectively, and observed that this interaction was disrupted when cells
were treated with NMDI14.
NMDI can be achieved with minimal cellular toxicity in multiple cell lines: NMD inhibition
can result in the upregulation of non-mutated, mutated and alternatively spliced mRNAs (5).
Thus NMD inhibition might be predicted to result in widespread biological effects. In fact, we
have recently reported that the genetic inhibition of NMD subtly but significantly increases
autophagy, presumably in an attempt to rid the cell of mutated and misfolded proteins that
accumulate with NMD suppression (31). To determine the cellular toxicity of select NMDIs, we
assessed viability in treated Hela, U2OS and Calu-6 cell lines, and in immortalized/nontransformed human fibroblasts. These cells were treated with 50 μM of NMDI14 and two other
effective NMDIs, as an assessment of any class toxicity effects, for 48 hours, and viability was
assessed. We noted almost no toxicity with NMDI14 and NMDI25, and minimal toxicity with
NMDI9, suggesting the pharmacological inhibition of NMD is not unduly toxic in contrast to 24
hours of treatment with emetine which blocks NMD by inhibiting translation (Fig 3A).
To determine whether the effective pharmacological inhibition of NMD exerted a sublethal effect
on proliferation, we assessed proliferation in NMDI14 treated transformed and non-transformed
cell lines (Fig 3B).
Three days of treatment resulted in no decrease in cell counts,
demonstrating that the pharmacological inhibition of NMD can be achieved without subtle
changes in proliferation.
Because suppression of translation is a potent mechanism to inhibit NMD, previously described
inhibitors of NMD also inhibit translation. Although we did not predict this mechanism of action
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
11
for our compounds, we tested protein synthesis after 6-24 hrs of NMDI14 treatment. During the
last hour of treatment cells were pulsed with S35 methionine, and protein was then precipitated
and the incorporated S35 assessed.
NMDI14 had no effect on S35 incorporation (Fig 3C).
Together these data suggest that the pharmacological inhibition of NMD can be achieved
without cellular toxicity.
NMDI14 increases the expression of endogenous NMD targets. The effectiveness of
NMDI14 in inhibiting NMD was originally determined by assessing the expression of a NMD
degraded reporter transcript which harbors a premature stop codon (Fig 2). While NMD has
been primarily appreciated as a mechanism to rapidly degrade mutated transcripts, we and
others have demonstrated that a wide variety of non-mutated transcripts are also degraded by
NMD which have several distinct motifs, as well as unknown features, which render them
sensitive to NMD (25-26, 33). We noted that the steady state mRNA expression, reflective of
both the rate of synthesis as well as the rate of decay, of seven non-mutated NMD targets which
we have validated (26) increased in U2OS and/or Hela cells after treatment with NMDI14,
whereas no such increase was seen in non-NMD targeted mRNAs (Fig 4A).
In order to assess potential off target effects of NMDI14, we used expression arrays to globally
assess gene expression in U2OS cells treated with NMDI14, emetine, or with UPF1 depletion
and compared this to gene expression in control U2OS cells. 941 genes were increased >1.5
fold with NMDI14 (Fig 4B). There was a significant overlap (22%, p=1.2E-25, Chitest) of genes
upregulated by NMDI with genes upregulated by emetine treatment (which inhibits NMD
potently but clearly has other effects), and with UPF1 depletion (11%). In comparison, 20% of
the 5290 genes upregulated by emetine were also upregulated in UPF1 depleted cells. The
overlap of genes regulated by NMDI14 and those regulated by either emetine or UPF1 depletion
is significantly higher than what would be expected by chance (~3%). Genes upregulated by
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
12
NMDI14 but not regulated by UPF1 or emetine, likely representative of off-target effects, were
not particularly enriched in any specific functional group , and only demonstrated a greater than
2 fold enrichment in ossification, actin binding, and cytoskeletal binding, suggesting that
NMDI14 may not have specific toxicities.
We next determined whether our NMDIs are effective in up-regulating mutated endogenously
expressed mRNAs. The small cell lung cancer cell line N417 has a p53 tumor suppressor gene
that is deleted on one allele and on the other allele carries a PTC mutation in the 298th codon
that renders it a target for NMD (34).
When treated with an NMDI we noted increased
expression of the mutated p53 mRNA (Fig 4C). Similar effects were seen in the breast cancer
HDQP-I cell line, which carries a PTC mutation in the 213th codon of the p53 gene (35), and the
lung adenocarcinoma cell line Calu-6 which carries a codon 196 PTC mutated p53 (36). The
treatment of N417 cells with NMDI14 for 6 hours led to a steady state expression of p53 similar
to that seen in U2OS cells (Fig 4D). No such effect was seen in cells expressing wild-type p53
in two other cell lines (Fig 4C). To directly assess p53 mRNA stability, N417 and U2OS cells
were treated with vehicle or NMDI14 for 6 hours, and the RNA polymerase II inhibitor 5,6Dichloro-1-β-D-ribofuranosylbenzimidazole (DRB) was added to stop mRNA synthesis. RNA
was collected at 0, 1.5, and 3 hrs after addition of DRB, and expression of p53 mRNA was
assessed by real time PCR. NMDI14 significantly increased the stability of PTC mutated p53
mRNA in N417 cells, without altering the stability of wild-type p53 in NMDI treated U2OS cells
(Fig 4E).
NMDI, along with a PTC “read-through” drug, increases the protein expression of
endogenous NMD targets: While NMDI14 increases the stability and expression of NMD
targeted mRNAs, a clinically effective strategy demands the expression of a full length,
biologically active protein. As noted previously, there are several drugs that can promote read-
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
13
through of PTCs. Although G418 has a modest effect of suppressing NMD, the most effective
agent at non-toxic concentrations, Ataluren, does not protect mRNAs from NMD (20). We
reasoned that if there were more mutated mRNA present, PTC read-through drugs might be
more effective.
We noted minimal expression of full length p53 in N417 cells treated with G418, but significantly
more in cells treated with both G418 and NMDI14 (Fig 5A). This increase was seen over a
variety of dose concentrations of G418, demonstrating that in the presence of NMDI14 higher
full length p53 expression can be achieved at lower and potentially less toxic G418
concentrations (Fig 5B). Similarly, the HDQP-I and Calu-6 cell lines which carry p53 PTC
mutations showed significant increased full length p53 expression with the combination of G418
and NMDI14 (Fig 5A). The expression of wild-type p53 in HCT116 cells was unaltered by these
treatments (Fig 5A). The effectiveness of the two drugs varied amongst cell lines, which could
either reflect variability in ribosomal read-through (which depends on the termination codon
sequence (20)), or cell-type dependent NMDI14 metabolism. Consistent with an increase in full
length/active p53 restoration, the combination of both drugs led to a synergistic increase in
multiple p53 target genes mRNAs including p21, BAX and PUMA in N417 and cells (Fig 5C).
This led to mRNAs levels of the pro-apoptotic BAX and PUMA similar to those seen in U2OS
cells (Fig 5D). As expected by the lack of p53 mRNA induction in non-mutated p53 cells with
NMDI14 (Fig 4C), in p53 wild-type cell lines HCT116 and U2OS NMDIs had no effect on p53
protein expression or p53 target gene expression over a wide range of concentrations (Fig 5A,
C and data not shwon). Together, these data indicate that NMDIs can increase endogenous
mutated mRNAs and, in combination with a PTC-bypass drug, can result in full-length protein
production.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
14
The restoration of full-length p53 with NMD inhibition and a “read-through” drug leads to
cell death in cells with a PTC mutated p53. The frequently mutated p53 gene is a tumor
suppressor which can induce apoptosis. The re-establishment of p53 in cancer cells, with or
without chemo/radiation, has been a therapeutic strategy pursued in a wide variety of cancer for
almost two decades, but this approach has been hampered by problems in delivery of wild-type
p53 (37-38). Although NMDIs are relatively non-toxic, we hypothesized that the combination of
NMDI with a “read through” drug would restore full-length p53 and result in a p53-mediated cell
death. We treated N417 and HDQP-I cells with a combination of read through drug and a dose
of NMDI14 that results in full length p53 expression. We noted a synergistic cell death only with
the combination of drugs that led to full-length p53 expression (Fig 5E). No such death was
seen when G418 and NMDI14 was used on cells with wild-type p53 including U2OS and
primary peripheral blood mononuclear cells (PBMCs) (Fig 5E).
Although the combination of G418 and NMDI14 was not toxic in p53 wild-type cells, we
examined a number of additional cell lines as controls (Fig 5F). These included HCT116 p53
wild-type and isogenic HCT116 p53 null cells, to determine whether the combination of NMDI14
and G418 is toxic only to cells without a functional p53. We also tested the breast cancer cell
line MDA-231, the prostate cancer cell line DU145, and the glioblastoma cell line T98G, all of
which carry single nucleotide p53 mutations which do not result in PTCs and are thus not NMD
targets. As with other cell lines we examined, 48 hr treatment with NMDI14 alone was minimally
toxic (<5% cell death) in these cell lines. G418 alone showed variable toxicity. However,
similar to the p53 wild-type cells and in contrast to those cell lines with NMD provoking p53
mutations. In no case did the addition of NMDI14 and G418 lead to a synergistic cell death .
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
15
Discussion
We have used a virtual screening strategy to identify small compounds that inhibit NMD. The
inhibition of NMD augments the expression of PTC mutated mRNAs and non-mutated mRNAs
normally targeted by NMD. Several of these identified NMD inhibitors, representing diverse
chemical backbones, are active at the nanomolar level even without further chemical
modification
to
optimize
their
solubilities
or
pharmacokinetic
properties.
The
few
pharmacological inhibitors previously described have been useful in delineating the mechanism
of NMD, but either inhibit translation, interfere with mRNA splicing, have not been demonstrated
to upregulate enogenous NMD targets, have weak activity, do not synergize with read-through
drugs, were identified through functional screening assays, and/or have unknown mechanisms
of action (24, 39-41). The rational design of our screening makes it likely that our identified
compounds are more specific with less off-target effects than these. Although several of our
NMDIs do not appear excessively toxic in a variety of cell lines and have no effect on the
expression of mRNAs that are not targeted by NMD, further work is needed to document and
potentially improve the specificity, in vivo effectiveness, and off-target toxicity of these lead
compounds.
Because of the important role NMD plays in degrading many mutated, alternatively spliced, and
physiologically expressed mRNAs, even specific inhibition of NMD without any off-target effects
might be theorized to be toxic. Indeed, the complete depletion of UPF2 in mice results in nonviability (42). However, we and others have demonstrated that the more modest depletion of
UPF1 and UPF2, through shRNA, does not adversely affect cell proliferation or survival (26). In
addition,
inhibition of NMD by UPF2 deletion in murine hematopoietic cells results only in
diminished hematopoietic progenitor cells with almost no effect on differentiated hematopoietic
cells (42), and widespread constitutive expression of a dominant negative UPF1 only modestly
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
16
affects the differentiating mouse thymus and spleen (43). Although the inhibition of NMD by
these stresses normally occurs in growing tumors and in fact is necessary for the threedimensional growth of cells in soft agar and as explants, the depletion of UPF1 from cells with
shRNA does not increase their three dimensional growth, and thus we would not expect
inhibition of NMD to necessarily augment tumorigenesis (26). The future clinical use of these or
other NMD inhibitors, however, will obviously require further studies to determine the short and
long-term effects of potent NMD inhibition.
Although NMD requires the pausing of a ribosome at a PTC, the use of the PTC read through
drug Ataluren does not inhibit NMD, potentially because only a small percentage of mutated
mRNAs are read through by Ataluren. Thus we have predicted that Ataluren, as well as other
strategies, would be more effective in the presence of more substrate mRNA.
We have
validated this approach by demonstrating that the combination of a read through drug and NMD
inhibitor results in the increased protein expression of a PTC mutated p53, resulting in improved
p53 function.
Many genetic disorders have mutations that either directly (non-sense) or
indirectly (e.g. splicing mutations, additions or deletions that alter reading frame, alternations in
poly-A processing) can render a mRNA sensitive to NMD. These include cancer (e.g. p53 and
BRCA1). Because truncated proteins may be able to replicate the function of the full length
protein, may be completely non-functional, or may act as dominant negatives against the full
normal allele, the effects of inhibiting NMD will have to be carefully considered, particularly
without a simultaneous consideration of restoring a full length protein with ribosome read
though.
However, based on our data the concomitant pharmacological inhibition of NMD
remains an attractive therapeutic strategy for many genetic diseases, including cancer.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
17
Acknowledgements
Supported by RO1DK081641 (L.B.G), 1DP2OD004631 (TC), UL1 TR000038 from the National
Center for the Advancement of Translational Science (NCATS), National Institutes of Health
(L.B.G and T.C), R01HL102449
and FP7-HEALTH-2012-INNOVATION from the European
Community and the Children's Cancer and Blood Foundation (S.R.). L.B.G. is the Saul Farber
Associate Professor of Medicine
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
18
References
1.
Bobadilla JL, Macek M, Jr., Fine JP, Farrell PM. Cystic fibrosis: a worldwide analysis of
CFTR mutations--correlation with incidence data and application to screening. Hum Mutat.
2002;19:575-606.
2.
Maquat LE, Kinniburgh AJ, Rachmilewitz EA, Ross J. Unstable beta-globin mRNA in
mRNA-deficient beta o thalassemia. Cell. 1981;27:543-53.
3.
Kan YW, Lee KY, Furbetta M, Angius A, Cao A. Polymorphism of DNA sequence in the
beta-globin gene region. Application to prenatal diagnosis of beta 0 thalassemia in Sardinia. N
Engl J Med. 1980;302:185-8.
4.
Frischmeyer PA, Dietz HC. Nonsense-mediated mRNA decay in health and disease. Hum
Mol Genet. 1999;8:1893-900.
5.
Gardner LB. Nonsense-mediated RNA decay regulation by cellular stress: implications
for tumorigenesis. Mol Cancer Res. 2010;8:295-308.
6.
Lykke-Andersen J, Shu MD, Steitz JA. Human Upf proteins target an mRNA for
nonsense-mediated decay when bound downstream of a termination codon. Cell. 2000;103:112131.
7.
Lykke-Andersen J, Shu MD, Steitz JA. Communication of the position of exon-exon
junctions to the mRNA surveillance machinery by the protein RNPS1. Science. 2001;293:18369.
8.
Modrek B, Lee C. A genomic view of alternative splicing. Nat Genet. 2002;30:13-9.
9.
Czaplinski K, Ruiz-Echevarria MJ, Paushkin SV, Han X, Weng Y, Perlick HA, et al. The
surveillance complex interacts with the translation release factors to enhance termination and
degrade aberrant mRNAs. Genes Dev. 1998;12:1665-77.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
19
10.
Okada-Katsuhata Y, Yamashita A, Kutsuzawa K, Izumi N, Hirahara F, Ohno S. N- and
C-terminal Upf1 phosphorylations create binding platforms for SMG-6 and SMG-5:SMG-7
during NMD. Nucleic Acids Res. 2012;40:1251-66.
11.
Fukuhara N, Ebert J, Unterholzner L, Lindner D, Izaurralde E, Conti E. SMG7 is a 14-3-
3-like adaptor in the nonsense-mediated mRNA decay pathway. Mol Cell. 2005;17:537-47.
12.
Ohnishi T, Yamashita A, Kashima I, Schell T, Anders KR, Grimson A, et al.
Phosphorylation of hUPF1 induces formation of mRNA surveillance complexes containing
hSMG-5 and hSMG-7. Mol Cell. 2003;12:1187-200.
13.
Kashima I, Yamashita A, Izumi N, Kataoka N, Morishita R, Hoshino S, et al. Binding of
a novel SMG-1-Upf1-eRF1-eRF3 complex (SURF) to the exon junction complex triggers Upf1
phosphorylation and nonsense-mediated mRNA decay. Genes Dev. 2006;20:355-67.
14.
Burke JF, Mogg AE. Suppression of a nonsense mutation in mammalian cells in vivo by
the aminoglycoside antibiotics G-418 and paromomycin. Nucleic Acids Res. 1985;13:6265-72.
15.
Salvatori F, Breveglieri G, Zuccato C, Finotti A, Bianchi N, Borgatti M, et al. Production
of beta-globin and adult hemoglobin following G418 treatment of erythroid precursor cells from
homozygous beta(0)39 thalassemia patients. Am J Hematol. 2009;84:720-8.
16.
Floquet C, Deforges J, Rousset JP, Bidou L. Rescue of non-sense mutated p53 tumor
suppressor gene by aminoglycosides. Nucleic Acids Res. 2011;39:3350-62.
17.
Howard M, Frizzell RA, Bedwell DM. Aminoglycoside antibiotics restore CFTR
function by overcoming premature stop mutations. Nat Med. 1996;2:467-9.
18.
Bedwell DM, Kaenjak A, Benos DJ, Bebok Z, Bubien JK, Hong J, et al. Suppression of a
CFTR premature stop mutation in a bronchial epithelial cell line. Nat Med. 1997;3:1280-4.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
20
19.
Wilschanski M, Yahav Y, Yaacov Y, Blau H, Bentur L, Rivlin J, et al. Gentamicin-
induced correction of CFTR function in patients with cystic fibrosis and CFTR stop mutations. N
Engl J Med. 2003;349:1433-41.
20.
Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, et al. PTC124
targets genetic disorders caused by nonsense mutations. Nature. 2007;447:87-91.
21.
Nelson SF, Crosbie RH, Miceli MC, Spencer MJ. Emerging genetic therapies to treat
Duchenne muscular dystrophy. Curr Opin Neurol. 2009;22:532-8.
22.
Linde L, Boelz S, Nissim-Rafinia M, Oren YS, Wilschanski M, Yaacov Y, et al.
Nonsense-mediated mRNA decay affects nonsense transcript levels and governs response of
cystic fibrosis patients to gentamicin. J Clin Invest. 2007;117:683-92.
23.
Ishigaki Y, Li X, Serin G, Maquat LE. Evidence for a pioneer round of mRNA
translation: mRNAs subject to nonsense-mediated decay in mammalian cells are bound by
CBP80 and CBP20. Cell. 2001;106:607-17.
24.
Dang Y, Low WK, Xu J, Gehring NH, Dietz HC, Romo D, et al. Inhibition of nonsense-
mediated mRNA decay by the natural product pateamine A through eukaryotic initiation factor
4AIII. J Biol Chem. 2009.
25.
Gardner LB. Hypoxic inhibition of nonsense-mediated RNA decay regulates gene
expression and the integrated stress response. Mol Cell Biol. 2008;28:3729-41.
26.
Wang D, Zavadil J, Martin L, Parisi F, Friedman E, Levy D, et al. Inhibition of nonsense-
mediated RNA decay by the tumor microenvironment promotes tumorigenesis. Mol Cell Biol.
2011;31:3670-80.
27.
Cardozo T, Abagyan R. Druggability of SCF ubiquitin ligase-protein interfaces. Methods
Enzymol. 2005;399:634-53.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
21
28.
Abagyan R, Totrov M. Biased probability Monte Carlo conformational searches and
electrostatic calculations for peptides and proteins. J Mol Biol. 1994;235:983-1002.
29.
Martin L, Kimball SR, Gardner LB. Regulation of the unfolded protein response by
eif2bdelta isoforms. J Biol Chem. 2010;285:31944-53.
30.
Martin L, Rainey M, Santocanale C, Gardner LB. Hypoxic activation of ATR and the
suppression of the initiation of DNA replication through cdc6 degradation. Oncogene.
2012;31:4076-84.
31.
Wengrod J, Martin L, Wang D, Frischmeyer-Guerrerio P, Dietz HC, Gardner LB. The
inhibition of nonsense mediated RNA decay activates autophagy. Mol Cell Biol. 2013.
32.
Chang JC, Kan YW. beta 0 thalassemia, a nonsense mutation in man. Proc Natl Acad Sci
U S A. 1979;76:2886-9.
33.
Mendell JT, Sharifi NA, Meyers JL, Martinez-Murillo F, Dietz HC. Nonsense
surveillance regulates expression of diverse classes of mammalian transcripts and mutes genomic
noise. Nat Genet. 2004;36:1073-8.
34.
Mitsudomi T, Steinberg SM, Nau MM, Carbone D, D'Amico D, Bodner S, et al. p53
gene mutations in non-small-cell lung cancer cell lines and their correlation with the presence of
ras mutations and clinical features. Oncogene. 1992;7:171-80.
35.
Wang CS, Goulet F, Lavoie J, Drouin R, Auger F, Champetier S, et al. Establishment and
characterization of a new cell line derived from a human primary breast carcinoma. Cancer
Genet Cytogenet. 2000;120:58-72.
36.
Lehman TA, Bennett WP, Metcalf RA, Welsh JA, Ecker J, Modali RV, et al. p53
mutations, ras mutations, and p53-heat shock 70 protein complexes in human lung carcinoma
cell lines. Cancer Res. 1991;51:4090-6.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
22
37.
Fujiwara T, Grimm EA, Mukhopadhyay T, Zhang WW, Owen-Schaub LB, Roth JA.
Induction of chemosensitivity in human lung cancer cells in vivo by adenovirus-mediated
transfer of the wild-type p53 gene. Cancer Res. 1994;54:2287-91.
38.
Gabrilovich DI. INGN 201 (Advexin): adenoviral p53 gene therapy for cancer. Expert
Opin Biol Ther. 2006;6:823-32.
39.
Keeling KM, Wang D, Dai Y, Murugesan S, Chenna B, Clark J, et al. Attenuation of
nonsense-mediated mRNA decay enhances in vivo nonsense suppression. PLoS ONE.
2013;8:e60478.
40.
Durand S, Cougot N, Mahuteau-Betzer F, Nguyen CH, Grierson DS, Bertrand E, et al.
Inhibition of nonsense-mediated mRNA decay (NMD) by a new chemical molecule reveals the
dynamic of NMD factors in P-bodies. J Cell Biol. 2007;178:1145-60.
41.
Gonzalez-Hilarion S, Beghyn T, Jia J, Debreuck N, Berte G, Mamchaoui K, et al. Rescue
of nonsense mutations by amlexanox in human cells. Orphanet J Rare Dis. 2012;7:58.
42.
Weischenfeldt J, Damgaard I, Bryder D, Theilgaard-Monch K, Thoren LA, Nielsen FC,
et al. NMD is essential for hematopoietic stem and progenitor cells and for eliminating byproducts of programmed DNA rearrangements. Genes Dev. 2008;22:1381-96.
43.
Frischmeyer-Guerrerio PA, Montgomery RA, Warren DS, Cooke SK, Lutz J, Sonnenday
CJ, et al. Perturbation of thymocyte development in nonsense-mediated decay (NMD)-deficient
mice. Proc Natl Acad Sci U S A. 2011;108:10638-43.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
23
Figure Legends
Fig 1. Virtual library screening of SMG7
highlighting the interaction of SMG7 with
UPF1. A. A model of the NMD. UPF1 interactions with the EJC results in recruitment of
SMG7 and dephosphorylation of UPF1. B. Plot of SMG7 pocket sizes. 12 drug-binding
pockets on the 3D structure of SMG7 were identified using the ICM-PocketFinder
algorithm. The volume/areas of all twelve pockets are plotted with volume in cubedangstroms (Å3) on the X-axis and pocket surface area in squared-angstroms (Å2) on
the Y-axis. 15,000 drugs in clinical use have a range of volumes/areas represented
approximately by the hashed blue square area, so a pocket with a volume/area within
this region is suitable for VLS. The target pocket selected on SMG7 is outlined with a
red box and falls within the suitable area. C. Image of the target pocket location on
SMG7. The pocket is displayed as a gold-colored solid geometric object. The peptide
backbone of SMG7 is depicted in ribbon diagram. D. Superimposition of the 31
compounds identified by VLS in their docked conformations within the target pocket on
SMG7, which is at its interface with Upf1. The electrostatic molecular surface of SMG7
is depicted as a smooth contoured surface colored red for negative charge, blue for
positive charge and white for hydrophobic areas.
Fig 2. NMDIs increase expression and stability of NMD reporters. A. Wild type and PTC 39 β
globin expressing U2OS cells were treated with 50 μM NMDIs for 6 hrs, and RNA was
harvested and assessed for β globin expression by quantitative PCR. Black bars indicate the
ratio of PTC to WT expression. * = p<0.05 by students t test. Experiments were biologically
replicated five times (average + SE displayed). B. Relative mRNA expression of wild-type β
globin and PTC39 β globin in cells treated with DMSO, and PTC39 β globin in cells treated with
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
24
NMDI14 for 6 hours. Experiments were repeated three times and average + SE displayed. C.
Induction of PTC 39 β globin mRNA after 6 hrs of treatment with various concentrations of
selected drugs, ranging from 50 μM to 5 X 10-4 μM, normalized to expression with DMSO.
Experiments were biologically replicated five times (average + SE displayed). D. Structure of
three NMDIs. E. UPF1 and SMG7 over-expressed 293T or control 293T cells were treated with
DMSO or 50 μM of NMDI14 for 6 hours and, interaction of UPF1 and SMG7 was determined.
Experiment is representative of three biological replicates, with fold change reflective of UPF1
immunoprecipitated/total UPF1 loaded.
Fig 3. NMDIs are not unduly toxic. A. U2OS, Hela, Calu-6 and BJ-hTERT cells were treated
with three NMDIs at 50 μM for 48 hours, and viability was assessed.
Toxicity with
translation/NMD inhibitor emetine is shown as control. Average of three readings shown. B.
U2OS, Hela and BJ-hTERT cells were treated with NMDI14 at 5 μM. The cell numbers were
determined at 0, 24, 48 and 72hrs. C. U2OS, Hela and BJ-hTERT cells were treated with 5 μM
of NMDI14 for 6 and 24 hours. The cells were pulsed with S35 methionine (100 μCi/mL) for last
one hour and protein synthesis was measured by S35 incorporation. The protein synthesis with
translation/NMD inhibitor emetine is shown as control
Fig 4. NMDIs stabilize endogenous mRNA. A. U2OS Hela cells were treated with 50 μM of
NMDI14 for 6 hrs, and endogenous non-mutated NMD target expression was assessed by real
time PCR. Actin and eIF2B4 serve as negative controls. * = p<0.05 by student’s T test vs
control. B. U2OS cells treated with NMDI14 at 50 μM for 6 hours. The whole gene expression
was determined and compared with UPF1 knockdown and emetine treated U2OS cells. C.
N417 cells, with a PTC mutated p53, were treated with 5 μM of NMDI14 for 24 hrs and p53
mRNA expression was assessed by real time PCR. Experiments were repeated three times
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
25
and average + standard error is displayed. * = p<0.05 by student’s T test vs DMSO. D. Relative
p53 mRNA expression in U2OS cells and N417 cells treated with DMSO, and N417 cells treated
with NMDI14 for 6 hours. Experiments were repeated three times and average + SE displayed.
E. p53 mRNA stability was determined, as described in text, in N417 and U2OS cells treated
with DMSO or NMDI14 for six hours. Experiments were repeated twice, with average + SE for
each time point shown.
Fig 5. Combination of read-through drug and NMDI restores full length PTC mutated p53 and
leads to cell death. A. Cells were treated with DMSO, G418 (200 μg/mL), NMDI14 (5 μM) or
both for 48 hrs, and, full length p53 was assessed in N417, HDQP-1 and Calu-6 cells or, as a
control HCT116 cells. B. N417 cells were treated with DMSO, 5 μM of NMDI14 and, 150, 175
and 400 μg/mL of G418 or both for 48 hours. The full length p53 was determined and U2OS
serves as a control. * = p<0.05 by student’s T test vs DMSO, # = p< 0.05 vs G418 alone. C.
mRNA expression of p53 downstream targets were assessed in N417 cells, U2OS cells, or HCT
116 cells with 48 hrs treatment of DMSO, G418 (200 μg/mL), NMDI (5 μM), or both. D. mRNA
expression of Bax and Puma were detected in DMSO and NMDI14 treated cells by real time
PCR. Experiments were repeated three times and average + standard error is displayed. E.
N417, HDQP-1 and U2OS cell were treated with DMSO, 200 μg/mL of G418, 5 μM of NMDI or
both for 48 hrs and viability was assessed. Experiments were repeated twice in duplicates, with
average + standard error displayed. * = p<0.05 by student’s T test vs DMSO, # = p< 0.05 vs
G418 alone. F. Additional cell lines with p53 deletion or missense mutations were treated with
DMSO, G418 (200 μg/mL), NMDI (5 μM) and viability was assessed.
Experiments were
repeated twice in duplicates, with average + standard error displayed.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Re
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Re
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Ca
Author Manuscript Published OnlineFirst on March 24, 2014; DOI: 10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Identification and characterization of small molecules that
inhibit nonsense mediated RNA decay and suppress nonsense
p53 mutations
Leenus Martin, Arsen Grigoryan, Ding Wang, et al.
Cancer Res Published OnlineFirst March 24, 2014.
Updated version
Author
Manuscript
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
doi:10.1158/0008-5472.CAN-13-2235
Author manuscripts have been peer reviewed and accepted for publication but have not yet been
edited.
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from cancerres.aacrjournals.org on June 17, 2017. © 2014 American Association for Cancer Research.