Identification of nonsense-mediated decay inhibitors that alter the tumor immune landscape

  1. Ashley L Cook
  2. Surojit Sur
  3. Laura Dobbyn
  4. Evangeline Watson
  5. Joshua D Cohen
  6. Blair Ptak
  7. Bum Seok Lee
  8. Suman Paul
  9. Emily Hsiue
  10. Maria Popoli
  11. Bert Vogelstein
  12. Nickolas Papadopoulos
  13. Chetan Bettegowda
  14. Kathy Gabrielson
  15. Shibin Zhou
  16. Kenneth W Kinzler  Is a corresponding author
  17. Nicolas Wyhs  Is a corresponding author
  1. Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of Medicine, United States
  2. Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, United States
  3. Department of Oncology, Johns Hopkins Medical Institutions, United States
  4. Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, United States
  5. Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, United States
  6. Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, United States
  7. Department of Neurosurgery, Johns Hopkins University School of Medicine, United States
  8. Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, United States
6 figures, 1 table and 7 additional files

Figures

Figure 1 with 8 supplements
LY3023414 is a small molecule capable of increasing transcription of nonsense-mediated decay (NMD) targets.

(A) Schematic of high-throughput screen (HTS) used to identify inhibitors of NMD. Mutant transcripts are represented by a smaller length in the cartoon for illustrative purposes only. All small molecules were tested at 10 μM. (B) Mutant RNA reads relative to wild-type reads for the top 8 hits from the HTS. The dotted line represents the minimum fraction required to be considered a hit (>5 standard deviations above dimethyl sulfoxide [DMSO] control). Full screen results are presented in Figure 1—figure supplement 4. (C) Targeted RNA-sequencing results of isogenic RPTec knockout clones treated with the eight best hits from the HTS at 10 µM. The dotted line represents a relative RNA expression level of 1, equal to that of DMSO-treated wells. Data for ceritinib, which did not validate on any line, are presented only in Figure 1—figure supplement 8. (D) TP53 western blot on RPE TP53 224, containing a homozygous TP53 mutation, using the four hit compounds that validated in RPTec isogenic lines at 10 μM. (E) Western blot analysis of full-length TP53α and isoform TP53β after treatment with two NMD inhibitor lead candidates at 10 μM. TP53β (expression known to be controlled by NMD) as well as mutant TP53 are prominently induced by LY3023414 whereas full length is not. Note that RPE TP53 223 is a heterozygous knockout clone with one near wild-type allele whereas RPTec TP53 588 contains a homozygous TP53 indel mutation. (F) Quantitative real-time PCR (qPCR) showing 10 μM LY3023414 treatment causes increased expression of the NMD controlled alternative transcript for TP53, TP53β, in parent cell lines for RPE1 and RPTec. Significance determined by Student’s t-test. Unless indicated otherwise cells were exposed to test compound for 16 hr.

Figure 1—figure supplement 1
Next-generation sequencing results depicting genomic mutations at the CRISPR target area in STAG2 (top left) and TP53 (top right) isogenic cell line clones in the RPE1 cell line which were used in subsequent experiments.

Western blot showing STAG2 protein loss in four independent clones of which only clones 2 and 8 were used in subsequent experiments (bottom left). Note that STAG2 clone 8 has two independent biallelic indel events. The mutation in exon 15 was used for scoring relative transcript levels in the screen and for subsequent work. Immunohistochemistry (IHC) results confirming p53 protein loss in RPE TP53 221 (bottom right).

Figure 1—figure supplement 2
Fold change in RNA expression levels from whole transcriptome RNA-sequencing data for STAG2 and TP53 knockout clones in the RPE1 cell line background.

RPE1 TP53 clone 221, RPE1 STAG2 clone 2, and RPE1 STAG2 clone 8 were used in the high-throughput screen (HTS). RPE1 TP53 clone 223 and RPE1 TP53 clone 224 are used in Figure 1—figure supplement 6. Note clone 223 has a 9-bp in-frame deletion in one allele and an out-of-frame deletion on the other allele, presumably accounting for the higher level of expression.

Figure 1—figure supplement 3
RNA transcript level changes based on quantitative real-time PCR (qPCR) in STAG2 and TP53 knockout clones treated with the known nonsense-mediated decay (NMD) inhibitor emetine at 12 mg/ml.

Red arrows indicate the cell line containing the truncating mutation in the gene being assessed. Error bars show standard deviation of three biological replicates. All changes are statistically significant by Student’s t-test (p < 0.05).

Figure 1—figure supplement 4
Primary screen results from high-throughput assay.

The x-axis represents all 2658 compounds screened at 10 µM, y-axis shows ratio of mutant to wild-type reads for each of the three isogenic cell lines. Higher values indicate more mutant RNA reads, representing inhibition of nonsense-mediated decay (NMD). The dotted line at 0.46 is the cutoff for a hit to be called (5 standard deviations above dimethyl sulfoxide [DMSO]-treated samples). Colored data points demarcate the eight hit compounds in the three screened cell lines.

Figure 1—figure supplement 5
Emetine and dimethyl sulfoxide (DMSO) control sample data from the high-throughput screen (HTS) for each of the three clones used as measured by deep-targeted RNA-sequencing.

Six DMSO and two emetine (12 mg/ml) samples were included in each dosing plate for a total of 198 DMSO and 66 emetine measurements in each boxplot.

Figure 1—figure supplement 6
Knockout status for isogenic cell lines used in this study.

(A) Immunohistochemistry (IHC) staining of TP53 in RPTec TP53 knockout clones. (B) Western blot against STAG2 on RPTec STAG2 knockout clones demonstrating successful knockout at the protein level of all 10 clones. Out-of-frame indels were confirmed in all clones by next-generation sequencing (NGS) (data not shown). The arrow indicates the expected size for full-length STAG2 protein. (C) IHC staining of TP53 protein on RPE1 TP53 knockout clones. Clone 223 contains the same truncating mutation found in clone 224 on allele 1 and has a 9-bp in-frame deletion that preserves some full-length TP53α and TP53β isoform expression on allele 2 (see Figure 1E). Note that the IHC image shown for RPE1 parent (control) in part C is the same image shown in Figure 1—figure supplement 1.

Figure 1—figure supplement 7
Protein schematic cartoons showing indel mutation site and expected size of various TP53 knockout clones used in this study.

Note: RPE TP53 223 has an in-frame deletion in the DNA-binding domain.

Figure 1—figure supplement 8
Fold change in mutant RNA transcription levels for STAG2 and TP53 in three knockout cell lines from the RPtec background containing out-of-frame indels targeted by nonsense-mediated decay (NMD).

Each line was treated with each of the eight hit compounds from the screen. The 10 µM dose is also shown in Figure 1C.

Inhibiting nonsense-mediated decay (NMD) in cancer cells increases broad expression of truncated gene messenger RNA (mRNA) and protein.

(A) Mutant transcript recovery rates for genes containing heterozygous indel mutations based on RNA-sequencing results in cell lines treated with 5 µM LY3023414 for 16 hr. Strict inclusion criteria were used, such that only mutations with sufficient sequencing coverage are shown (see methods). Recovery is defined as at least two-fold increase over dimethyl sulfoxide (DMSO) treatment. (B) Targeted high coverage RNA-sequencing confirms recovery of mutant transcript levels in NCI-H358 and LS180 cancer cell lines treated with 5 µM LY3023414. RNF43 and DROSHA contain common heterozygous single-nucleotide polymorphisms (SNPs) and the mutant allele refers to the non-reference genome allele. Error bars indicate 95% confidence limits. (C) Western blot analyses of NCI-H358 cells showing mutant and wild-type protein levels in EXOC1 and (D) SPTAN1 with and without 5 µM LY3023414 treatment. The black arrow indicates the expected size of the mutant protein. The C-terminal SPTAN1 antibody is downstream of the out-of-frame indel mutation and is not expected to identify the mutant allele. (E) Fold change in the number of mutant RNA transcripts from deep-targeted RNA-sequencing of heterozygous mutated genes in NCI-H358 and LS180 xenografts treated by oral gavage with 60 mg/kg LY3023414 assayed 16 hr post-treatment. Student’s t-test for target genes are all p < 0.05, while the null hypothesis holds for RNF43 (common SNP).

Figure 3 with 7 supplements
Novel nonsense-mediated decay (NMD) inhibitor KVS0001 is SMG1 specific and induces expression of NMD-targeted genes in vitro and in vivo.

(A) Fraction of mutant allele transcripts in genes with heterozygous indels previously established in this study as sensitive to NMD inhibition. Results show mutant levels after siRNA treatment targeting kinases inhibited by LY3023414. RNF43 and DROSHA are common heterozygous single-nucleotide polymorphisms (SNPs) (shaded gray) and serve as negative controls. (B) Fraction of mutant allele transcripts in genes with truncating mutations known to be sensitive to NMD inhibition after siRNA treatment with siUPF1 or non-targeting siRNA. Data from deep-targeted RNA-sequencing. (C) Structure of novel NMD inhibitor KVS0001. (D) Targeted RNA-sequencing on three genes with heterozygous, out-of-frame, indel mutations in LS180 cancer cells treated in a dose–response with KVS0001 or SMG1i-11. RNF43 serves as a control (common heterozygous SNP) and the mutant allele refers to the non-reference genome allele. (E) Western blot of EXOC1 protein in NCI-H358 cells treated with 5 µM novel inhibitor KVS0001, LY3023414, or SMG1i-11 for 24 hr. (F) Western blot of phosphorylated UPF1 on three cell lines treated with 5 µM KVS0001, SMG1i-11, or dimethyl sulfoxide (DMSO). Note that total UPF1 and p-UPF1 were run on different gels, loading controls correspond to indicated gel. (G) Fold change in the number of mutant allele transcripts measured by targeted RNA-seq in genes containing heterozygous out-of-frame indel mutations in NCI-H358 or (H) LS180 subcutaneous xenografts in bilateral flanks of nude mice. Mice were treated once with intraperitoneal (IP) injection of vehicle or 30 mg/kg KVS0001 and tumors harvested 16 hr post IP treatment. All genes shown contain heterozygous out-of-frame truncating mutations except RNF43 and DROSHA which serve as controls (contain heterozygous SNPs).

Figure 3—figure supplement 1
RNA expression levels of kinases post siRNA targeting in NCI-H358 and LS180 cells by quantitative real-time PCR (qPCR).

Cells are treated with siRNAs targeting genes known to be inhibited by LY3023414. Error bars represent 95% confidence intervals.

Figure 3—figure supplement 2
Slider plots showing mutant allele fraction relative to total reads measured by deep-targeted RNA-sequencing for in vitro treated LS180 (top in red) or NCI-H358 (bottom in blue) cells with 5 µM LY3023414 (labeled LYO) or a previously described SMG1 inhibitor SMG1i-11 at 1 µM.

Gene names are shown in the boxes above each slider plot, genes highlighted in gray are common single-nucleotide polymorphisms (SNPs) and serve as a negative control (not expected to change). In the case of the control SNPs, the mutant allele refers to the non-reference genome allele. TRIM21 did not show a large change in expression with either small molecule, while ANLN did not show a difference with LY3023414 but did respond to SMG1i-11. The remaining genes responded to nonsense-mediated decay (NMD) inhibition by both LY3023414 and SMG1i-11.

Figure 3—figure supplement 3
Mutant protein expression only occurs in the presence of nonsense-mediated decay (NMD) inhibtion.

(A) Western blot showing SPTAN1 expression after treatment with dimethyl sulfoxide (DMSO), 5 µM LY3023414, or 1 µM SMG1 inhibitor SMG1i-11 (lanes 1, 2, and 3, respectively, for each antibody). The arrow indicates the expected size of the mutant NMD-targeted protein. Antibody ab75755 (Abcam) binds C-terminal to the out-of-frame indel and does not show mutant protein as expected. Antibody A301-249 (Bethyl) is polyclonal and also did not bind mutant protein. Antibody ab11755 (Abcam) is located N-terminal to the indel and does display mutant protein expression. (B) Western blot showing EXOC1 expression after treatment with DMSO, 5 µM LY3023414, or 1 µM SMG1i-11 (lanes 1, 2, and 3, respectively). The arrow indicates the expected size of the mutant (NMD-targeted) protein.

Figure 3—figure supplement 4
Biophysical properties for novel SMG1 inhibitor KVS0001.
Figure 3—figure supplement 5
Kinase specifity for KVS0001.

(A) Kinativ assay results for KVS0001 at 100 nM and (B) 1 µM run with biological replicates showing KVS0001 specificity against the known kinome. Results are based on the average between two unique peptides for each kinase.

Figure 3—figure supplement 6
(A) Slider plots showing mutant allele fraction measured by deep-targeted RNA-sequencing for genes from NCI-H358 (top in red) and (B) LS180 (bottom in blue) treated in a dose–response in vitro with novel nonsense-mediated decay (NMD) inhibitor KVS0001.

DROSHA and RNF43 are common heterozygous single-nucleotide polymorphisms (SNPs) and serve as a negative control. In the case of the control SNPs, the mutant allele refers to the non-reference genome allele. Only the highest two concentrations were tested on LY3023414 which served as a positive control in this experiment. The dotted line indicates the mutant expression with dimethyl sulfoxide (DMSO) treatment and the solid line is a reference for equal expression of both the wild-type and mutant alleles.

Figure 3—figure supplement 7
Mutant protein expression in presence of nonsense-mediated decay (NMD) inhibtion.

(A) Western blot showing expression of LMAN1 in LS180 cells treated with dimethyl sulfoxide (DMSO) (lane 1), KVS0001 at 5 µM (lane 2), or SMG1i-11 at 1 µM (lane 3). Arrows indicate expected size of wild-type and mutant LMAN1 protein. (B) Stain free loading control image for gel.

Figure 4 with 4 supplements
KVS0001 treatment induces targetable cell surface presentation of peptides known to be downregulated by nonsense-mediated decay (NMD).

(A) MHC class I HLA presentation of mutant specific peptide sequences from NCI-H358 and (B) LS180 cells by quantitative HPLC–mass spectrometry treated with dimethyl sulfoxide (DMSO) or 5 µM KVS0001. The gene name, type of mutation (in parenthesis), and presented peptide are shown on the y-axis for each gene. Colors indicate different ions. (C) TP53 gene structure and mutant DNA sequence for NCI-H716 and NCI-H2228 cancer cell lines, both contain a homozygous splice site mutation in TP53. Capital letters represent exonic sequence; lowercase letters represent intronic sequence. DNA mutation reflected by gold bases. (D) Western blot against TP53 in the presence or absence of 5 µM NMD inhibitor in NCI-H716_A24 and NCI-H2228 cell lines. NCI-H2228 has an expected size of 46.6 kDa and NCI-H716 of 34.7 kDa. (E) Interferon (IFN-γ) levels over baseline based on enzyme-linked immunosorbent assay (ELISA) in a co-culture assay with NCI-H716_A24 and NCI-H2228 cells, 1.25 µM NMD inhibitor, human CD3+ T-cells, and bispecific antibody for TP53 and CD3. Chemotherapy (5-fluorouracil) is shown as a control. (F) Cell killing based on luciferase levels in a co-culture assay in NCI-H716 cells with and without A24 expression, treated with TP53-CD3 bispecific antibody, 1.25 µM NMD inhibitor and human CD3+ T-cells.

Figure 4—figure supplement 1
DNA and protein sequences for the wild-type and mutant alleles of (A) EXOC1, (B) RAB14, and (C) ZDHHC16 genes.

The mutant protein sequences caused by the out-of-frame indel are highlighted in red and the boxes indicate the peptides presented on the cell surface and identified by mass spectrometry in cells treated with 5 µM of KVS0001 (see Figure 4A and B).

Figure 4—figure supplement 2
Heavy peptide loading controls and endogenous (light) peptide presentation of genes in LS180 and NCI-H358 treated with dimethyl sulfoxide (DMSO) or 5 µM KVS0001.

Data are from quantitative HPLC–mass spectrometry. Note: y-axis scale changes between samples. Tables on the right show relative increase in peptide presentation with KVS0001.

Figure 4—figure supplement 3
Waterfall plot of publicly available TP53 RNA expression (as shown by FPKM) for 675 cancer cell lines.

The two cell lines used in this study are highlighted in red and were in the bottom quartile of TP53 expression.

Figure 4—figure supplement 4
Western blot of TP53 on NCI-H716 and NCI-H2228 cells treated with 5 or 7.5 µM of nonsense-mediated decay (NMD) inhibitor, 1 µM SMG1i-11, or 200 mg/ml chemotherapy, showing controls related to Figure 4D.

LYO is LY3023414 and 5-FU is 5-fluorouracil. HEK293 parent cells are shown as a control (wild-type TP53 protein).

Figure 5 with 2 supplements
In vivo treatment of murine tumors with KVS0001 yield differential tumor growth compared with vehicle treatment.

(A) Fold change in RNA transcript levels in LLC or (B) RENCA cells treated in vitro with 5 µM of nonsense-mediated decay (NMD) inhibitor KVS0001 or dimethyl sulfoxide (DMSO). Orange bars indicate genes with homozygous indel mutations potentially targeted by NMD. Purple bars show genes with no mutations but that are known to have their normal transcription levels controlled by NMD. Green bar is a control gene that should not change with treatment. The dotted line shows relative expression of DMSO treatment (equal to 1). * indicates significantly different from untreated by Student’s T-test. (C) Treatment schedule for syngeneic tumor mouse experiments. (D) Average tumor size of LLC (left) and RENCA (right) syngeneic tumors in immune-competent mice (n = 8) treated with 30 mg/kg KVS0001 or vehicle control IP. Difference is statistically significant after day 10 based on one-way analysis of variance (ANOVA) with Dunnett’s test p < 0.001 (p < 0.05 for day 23 RENCA data point) for both tumors tested. (E) Average tumor size of LLC (left) and RENCA (right) in immunodeficient mice (n = 8) treated with 30 mg/kg KVS0001 or vehicle control. Error bars show 95% confidence intervals in all plots.

Figure 5—figure supplement 1
Mouse weights for (A) C57BL/6N and (B) BALB/c tumor-bearing mice treated with 30 mg/kg KVS0001 or vehicle control intraperitoneal (IP) daily.

N = 8 for all arms.

Figure 5—figure supplement 2
Mouse tumor size as measured by calipers for mammary fat pad placed syngeneic tumor models treated daily with 30 mg/kg KVS0001 or vehicle IP.

(A) Tumors with low/moderate and (B) high indel mutational loads are shown. No results presented in this figure supplement are statistically significant. Error bars show 95% confidence intervals.

Chemical structure 1
Flowchart describing synthesis for KVS0001.

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
AntibodyDonkey anti goat IgG 680RD monoclonalLicor926-68074,
RRID:AB_10956736
Used for western blot at 1:10 K
AntibodyDonkey anti mouse 680RD monoclonalLicor926-68072,
RRID:AB_10953628
Used for western blot at 1:15 K
AntibodyGoat anti mouse HRP monoclonalJackson Immuno Research115-035-006,
RRID:AB_2338500
Used for western blot at 1:2500
AntibodyDonkey anti rabbit 800CW monoclonalLicor926-32213,
RRID:AB_621848
Used for western blot at 1:10 K
AntibodyGoat anti rabbit HRP monoclonalJackson Immuno Research111-035-006,
RRID:AB_2337936
Used for western blot at 1:2500
AntibodyMouse anti Beta-actin monoclonalCell Signaling Technology3700SUsed for western blot at 1:5000
AntibodyRabbit anti EXOC1 polyclonalAbcamab251853Used for western blot at 0.4 µg/ml
AntibodyMouse anti LMAN1 monoclonalThermo Fisher ScientificCF502137Used for western blot at 1:200
AntibodyMouse anti OKT-3 monoclonalBiolegend317347Used for T-cell culturing at 15 ng/ml
AntibodyRabbit anti p21/WAF1 monoclonalCell Signaling Technology2947S,
RRID:AB_823586
Used for western blot at 1:1000
AntibodyMouse anti TP53 monoclonalCell Signaling Technology18032S,
RRID:AB_2798793
Used for western blot at 1:1000
AntibodyRabbit anti Phospho-(Ser/Thr) ATM/ATR polyclonalCell Signaling Technology2851S,
RRID:AB_330318
Used for western blot at 1:1000
AntibodyRabbit anti Sodium Potassium Pump (Na-K) polyclonalCell Signaling Technology3010S,
RRID:AB_2060983
Used for western blot at 1:500
AntibodyRabbit anti SPTAN1 (polyclonal)BethylA301-249A,
RRID:AB_890655
Used for western blot at 0.04 µg/ml
AntibodyMouse anti SPTAN1 (N-Terminal) monoclonalAbcamab11755,
RRID:AB_298540
Used for western blot at 1:1000
AntibodyRabbit anti SPTAN1 (C-Terminal) monoclonalAbcamab75755,
RRID:AB_1309947
Used for western blot at 1:1000
AntibodyRabbit anti STAG2 monoclonalCell Signaling Technology5882S,
RRID:AB_10834529
Used for western blot at 1:1000
AntibodyGoat anti UPF1 polyclonalAbcamab10510,
RRID:AB_297251
Used for western blot at 1:2500
AntibodyMouse α-HLA-A24 monoclonalMBL Life ScienceCat #K0208-A64,
RRID:AB_1953030
Used for flow cytometry at 10 µg/ml
AntibodyTP53-CD3 bispecific antibodyThis studyN/ASee ‘bispecific scFv construction’ in methods, anti CD-3 and anti TP53 sequence. Used for co-culture at 12.5 pg/ml
OtherHLA-A24 RetrovirusHsiue et al., 2021N/ARetrovirus which introduces and HLA-A24 expression vector
OtherLuciferase lentivirusOriGeneCat #PS100071Lentivirus that introduces luciferase into cells
Chemical compound, drug1% penicillin–streptomycinThermo Fisher ScientificCat #15140122
Other4–15% Mini-PROTEAN TGX Precast Protein GelsBio-RadCat #456-1086Used for western blotting
Chemical compound, drug5-FluoruracilSigma-AldrichCat #F6627
OtherAMPure beadsBeckman CoulterCat #A63880Used to purify DNA before sequencing
Chemical compound, drugCremaphorSigma-AldrichCat #C5135
Chemical compound, drugDharmaFECT1 transfection reagentHorizonCat #T-2001-02
Chemical compound, drugDimethyl sulfoxide (DMSO)Sigma-AldrichCat #C6295
Chemical compound, drugDMEM mediumGibcoCat #11995065
Chemical compound, drugEMEM mediumATCCCat #30-2003
Chemical compound, drugEmetineSigma-AldrichCat #7083-71-8
Chemical compound, drugEPITHELIAL CELL MEDIUM-Complete KitScience Cell ResearchCat #4101
Chemical compound, drugFetal bovine serum (FBS)HyCloneCat #16777-006
Chemical compound, drugGlycerolSigma-AldrichCat #G5516
Chemical compound, drugKVS0001This studyN/ANovel small molecule targeting SMG1 kinase. See ‘Resource availability’
Chemical compound, drugLY3023414SelleckchemCat #S8322
Chemical compound, drugMatrigel Phenol Red Free Standard FormulationCorningCat #356237
Chemical compound, drugMEGM Mammary Epithelial Cell Growth Medium BulletKitLonzaCat #CC3150
Chemical compound, drugMethylcelluloseSigma-AldrichCat #M6385
Chemical compound, drugPhosphate-buffered saline (PBS)Thermo FisherCat #J60465.K2
OtherPhusion Flash High-Fidelity PCR Master MixThermo FisherCat #F548SPCR reagent
OtherPierce ECL Western Blotting SubstrateThermo FisherCat #32106Western blot reagent
OtherPRIMETIME Gene Expression master mixIntegrated DNA TechnologiesCat #1055770Real-time PCR reagent
Chemical compound, drugProtease inhibitorMillipore SigmaCat #4693159001
OtherQIAshredderQIAGENCat #79656Western blot reagent
Recombinant proteinRecombinant IL-2 proteinPrometheus Therapeutics and DiagnosticsCat #aldesleukin
Recombinant proteinRecombinant IL-7 proteinBioLegendCat #581908
Chemical compound, drugRediJect D-Luciferin Ultra Bioluminescent SubstratePerkinElmerCat #770505
Chemical compound, drugRIPA Lysis and Extraction bufferThermo FisherCat #89901Western blot and real-time PCR reagent
Chemical compound, drugRNA LaterInvitrogenCat #AM7020Real-time PCR reagent
Chemical compound, drugRPMI 1640 mediumGibcoCat #11875-119
Chemical compound, drugSelleckchem Bioactive Compound library (Supplementary file 2)SelleckchemCat #L1700
Chemical compound, drugSMG1-specific inhibitor: 11jAscendex LLCN/A
OtherSsoAdvanced Universal SYBR Green SupermixBio-RadCat #1725270Real-time PCR reagent
Chemical compound, drugTrypsinGibcoCat #25300054
Commercial assay or kitAllPrep DNA/RNA Mini KitQIAGENCat #80204
Commercial assay or kitAgilent RNA ScreenTapeAgilentCat #5067-5576
Commercial assay or kitAgilent RNA ScreenTape Sample BufferAgilentCat #5067-5577
Commercial assay or kitAgilent RNA LadderAgilentCat #5067-5578
Commercial assay or kitBCA Protein Assay KitThermo FisherCat #23227
Commercial assay or kitBio-Rad SingleShot Cell Lysis kitBio-RadCat #1725080
Commercial assay or kitELISAR&D SystemsCat #SIF50C
Commercial assay or kitHigh-Capacity cDNA Reverse Transcription KitAdvanced BiosystemsCat #4368814
Commercial assay or kitIllumina RNA library prep kitIlluminaCat #RS-122-2001
Commercial assay or kitKinase kinativ assayActivX BiosciencesN/A
Commercial assay or kitLuciferase Assay SystemPromegaCat #E1501
Commercial assay or kitRNeasyQIAGENCat #74104
OtherWhole transcriptome RNA-seq: NCI-H358This studyFASTQ files for RNA-seq. Available on Dryad
OtherWhole transcriptome RNA-seq: LS180This studyFASTQ files for RNA-seq. Available on Dryad
OtherWhole transcriptome RNA-seq: Isogenic knockout cell linesCook et al., 2022EGAD00001008559FASTQ files for RNA-seq. Available on EGA database
Cell Line (murine)B16-F10ATCCCat #CRL-6475,
RRID:CVCL_0159
Cell Line (Human)HEK293TATCCCat #CRL-3216,
RRID:CVCL_0063
Cell Line (murine)Lewis lung carcinoma (LLC)ATCCCat #CRL-1642,
RRID:CVCL_4358
Cell Line (Human)LS180ATCCCat #CL-187,
RRID:CVCL_0397
Cell Line (Human)MCF10aATCCCat #CRL-10317,
RRID:CVCL_0598
Cell Line (Human)NCI-H716ATCCCat #CCL-251,
RRID:CVCL_1581
Cell Line (Human)NCI-H2228ATCCCat #CRL-5935,
RRID:CVCL_1543
Cell Line (Human)NCI-H358ATCCCat #CRL-5807,
RRID:CVCL_1559
Cell Line (murine)RencaATCCCat #CRL-2947,
RRID:CVCL_2174
Cell Line (Human)RPE1ATCCCat #CRL-4000,
RRID:CVCL_4388
Cell Line (Human)RPE1 STAG2 2Cook et al., 2022N/A
Cell Line (Human)RPE1 STAG2 5Cook et al., 2022N/A
Cell Line (Human)RPE1 STAG2 6Cook et al., 2022N/A
Cell Line (Human)RPE1 STAG2 8Cook et al., 2022N/A
Cell Line (Human)RPE1 TP53 221Cook et al., 2022N/A
Cell Line (Human)RPE1 TP53 223Cook et al., 2022N/A
Cell Line (Human)RPE1 TP53 224Cook et al., 2022N/A
Cell Line (Human)RPTecATCCCat #CRL-4031,
RRID:CVCL_K278
Cell Line (Human)RPTec STAG2 3Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 943Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 943ACook et al., 2022N/A
Cell Line (Human)RPTec STAG2 944Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 945Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 946Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 947Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 951Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 952Cook et al., 2022N/A
Cell Line (Human)RPTec STAG2 953Cook et al., 2022N/A
Cell Line (Human)RPTec TP53 544Cook et al., 2022N/A
Cell Line (Human)RPTec TP53 588Cook et al., 2022N/A
Strain, strain background (Mus musculus, female)BALB/cAnAHsdHarlan Laboratories
Strain, strain background (M. musculus, female)C57BL/6NCrlCharles River GmbH
Strain, strain background (M. musculus, female)BALB/cAnNCrlCharles River GmbH
Strain, strain background (M. musculus, female)Hsd:Athymic Nude-Foxn1nuHarlan Laboratories
Strain, strain background (M. musculus, female)NOD.Cg-Prkdcscid
Il2rgtm1Wjl/SzJ (NSG)
Jackson Laboratories
Recombinant DNA reagentfor Primers see Supplementary file 6This studyN/A
Recombinant DNA reagentHTS Screen sequencing primer:
STAG2
Seq Forward: AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNNNGAAT
TTCTCTACAAAAAGCTCTTCA
This studyN/AUsed in high-throughput screen to amplify STAG2 CRISPR mutation site
Recombinant DNA reagentHTS Screen sequencing primer: STAG2
Seq Reverse: CAAGCAGAAGACGGCATACGAGATNNNNNNNNNNTTCATCATTCCATCCTCCTC
This studyN/AUsed in high-throughput screen to amplify STAG2 CRISPR mutation site
Recombinant DNA reagentHTS Screen sequencing primer:
TP53
Seq Forward: AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNNN
GAAACTACTTCCTG
AAAACAACGT
This studyN/AUsed in high-throughput screen to amplify TP53 CRISPR mutation site
Recombinant DNA reagentHTS Screen sequencing primer:
TP53 Seq Reverse: CAAGCAGAAGACGGCATACGAGATN
NNNNNNNNNGCTTCATC
TGGACCTGGGTC
This studyN/AUsed in high-throughput screen to amplify TP53 CRISPR mutation site
Recombinant DNA reagentON-TARGETplus Human UPF1
siRNA Smartpool 10 nmol
Horizon DiscoveryCat #L-011763-00-0010
Recombinant DNA reagentON-TARGETplus Human mTOR
siRNA Smartpool 10 nmol
Horizon DiscoveryCat #L-003008-00-0010
Recombinant DNA reagentON-TARGETplus Human ATM
siRNA Smartpool 10 nmol
Horizon DiscoveryCat #L-003201-00-0010
Recombinant DNA reagentON-TARGETplus Human ATR
siRNA Smartpool 10 nmol
Horizon DiscoveryCat #L-003202-00-0010
Recombinant DNA reagentSMARTpool: ON-TARGETplus
SMG1 siRNA
Horizon DiscoveryCat #L-005033-00-0020
Recombinant DNA reagentSMARTpool: ON-TARGETplus
PRKDC siRNA
Horizon DiscoveryCat #L-005030-00-0010
Recombinant DNA reagentSMARTpool: ON-TARGETplus
PIK3CA siRNA
Horizon DiscoveryCat #L-003018-00-0010
Recombinant DNA reagentON-TARGETplus Non-targeting PoolHorizon DiscoveryCat #D-001810-10
Recombinant DNA reagentPhage display libraryGeneArtN/A
Software, algorithmHISAT2 (version 2.0.5)Kim et al., 2019N/A
Software, algorithmStringTie (version 1.3.3)Pertea et al., 2016N/A
Software, algorithmBallgown (version 2.6.0)Pertea et al., 2016N/A
Software, algorithmR (version 4.0.3)R Development Core Team, 2022N/A
Software, algorithmggplot (version 3.4.1)Wickham, 2016N/A
Software, algorithmPipeline for analyzing mutant and normal transcript abundanceMSSQL was used for initial data processingN/A

Additional files

Supplementary file 1

Summary of cell lines used in this manuscript and their origin.

https://cdn.elifesciences.org/articles/95952/elife-95952-supp1-v1.xlsx
Supplementary file 2

Selleck Chem Library compounds used for the nonsense-mediated decay (NMD) high-throughput screen (HTS screen).

https://cdn.elifesciences.org/articles/95952/elife-95952-supp2-v1.xlsx
Supplementary file 3

Structures of novel nonsense-mediated decay (NMD; SMG1) inhibitors designed for this study.

https://cdn.elifesciences.org/articles/95952/elife-95952-supp3-v1.xlsx
Supplementary file 4

List of kiNativ assay results for all kinase proteins tested at three concentrations of KVS0001.

https://cdn.elifesciences.org/articles/95952/elife-95952-supp4-v1.xlsx
Supplementary file 5

Mutations found in LLC and RENCA murine tumors.

https://cdn.elifesciences.org/articles/95952/elife-95952-supp5-v1.xlsx
Supplementary file 6

List of primers used in this study with sequences.

https://cdn.elifesciences.org/articles/95952/elife-95952-supp6-v1.xlsx
MDAR checklist
https://cdn.elifesciences.org/articles/95952/elife-95952-mdarchecklist1-v1.pdf

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Ashley L Cook
  2. Surojit Sur
  3. Laura Dobbyn
  4. Evangeline Watson
  5. Joshua D Cohen
  6. Blair Ptak
  7. Bum Seok Lee
  8. Suman Paul
  9. Emily Hsiue
  10. Maria Popoli
  11. Bert Vogelstein
  12. Nickolas Papadopoulos
  13. Chetan Bettegowda
  14. Kathy Gabrielson
  15. Shibin Zhou
  16. Kenneth W Kinzler
  17. Nicolas Wyhs
(2025)
Identification of nonsense-mediated decay inhibitors that alter the tumor immune landscape
eLife 13:RP95952.
https://doi.org/10.7554/eLife.95952.3