Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition

Abstract Background Pancreatic ductal adenocarcinoma (PDAC) patients suffer poor outcomes, including a five-year survival of below 10%. Poor outcomes result in part from therapeutic resistance that limits the impact of cytotoxic first-line therapy. Novel therapeutic approaches are needed, but curren...

Full description

Bibliographic Details
Main Authors: Ryne C. Ramaker, Andrew A. Hardigan, Emily R. Gordon, Carter A. Wright, Richard M. Myers, Sara J. Cooper
Format: Article
Language:English
Published: BMC 2021-05-01
Series:BMC Cancer
Subjects:
Online Access:https://doi.org/10.1186/s12885-021-08388-1
id doaj-71384a5ac1ba4f2b8773d05276b07ff9
record_format Article
spelling doaj-71384a5ac1ba4f2b8773d05276b07ff92021-05-30T11:49:08ZengBMCBMC Cancer1471-24072021-05-0121111310.1186/s12885-021-08388-1Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transitionRyne C. Ramaker0Andrew A. Hardigan1Emily R. Gordon2Carter A. Wright3Richard M. Myers4Sara J. Cooper5University of Alabama-BirminghamUniversity of Alabama-BirminghamHudsonAlpha Institute for BiotechnologyHudsonAlpha Institute for BiotechnologyHudsonAlpha Institute for BiotechnologyHudsonAlpha Institute for BiotechnologyAbstract Background Pancreatic ductal adenocarcinoma (PDAC) patients suffer poor outcomes, including a five-year survival of below 10%. Poor outcomes result in part from therapeutic resistance that limits the impact of cytotoxic first-line therapy. Novel therapeutic approaches are needed, but currently no targeted therapies exist to treat PDAC. Methods To assess cellular resistance mechanisms common to four cytotoxic chemotherapies (gemcitabine, 5-fluorouracil, irinotecan, and oxaliplatin) used to treat PDAC patients, we performed four genome-wide CRISPR activation (CRISPRact) and CRISPR knock-out (CRISPRko) screens in two common PDAC cell lines (Panc-1 and BxPC3). We used pathway analysis to identify gene sets enriched among our hits and conducted RNA-sequencing and chromatin immunoprecipitation-sequencing (ChIP-seq) to characterize top hits from our screen. We used scratch assays to assess changes in cellular migration with HDAC1 overexpression. Results Our data revealed activation of ABCG2, a well-described efflux pump, as the most consistent mediator of resistance in each of our screens. CRISPR-mediated activation of genes involved in transcriptional co-repressor complexes also conferred resistance to multiple drugs. Expression of many of these genes, including HDAC1, is associated with reduced survival in PDAC patients. Up-regulation of HDAC1 in vitro increased promoter occupancy and expression of several genes involved in the epithelial-to-mesenchymal transition (EMT). These cells also displayed phenotypic changes in cellular migration consistent with activation of the EMT pathway. The expression changes resulting from HDAC1 activation were also observed with activation of several other co-repressor complex members. Finally, we developed a publicly available analysis tool, PancDS, which integrates gene expression profiles with our screen results to predict drug sensitivity in resected PDAC tumors and cell lines. Conclusion Our results provide a comprehensive resource for identifying cellular mechanisms of drug resistance in PDAC, mechanistically implicate HDAC1, and co-repressor complex members broadly, in multi-drug resistance, and provide an analytical tool for predicting treatment response in PDAC tumors and cell lines.https://doi.org/10.1186/s12885-021-08388-1Genome-wide screenCRISPRPancreatic cancerDrug resistanceABCG2HDAC1
collection DOAJ
language English
format Article
sources DOAJ
author Ryne C. Ramaker
Andrew A. Hardigan
Emily R. Gordon
Carter A. Wright
Richard M. Myers
Sara J. Cooper
spellingShingle Ryne C. Ramaker
Andrew A. Hardigan
Emily R. Gordon
Carter A. Wright
Richard M. Myers
Sara J. Cooper
Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
BMC Cancer
Genome-wide screen
CRISPR
Pancreatic cancer
Drug resistance
ABCG2
HDAC1
author_facet Ryne C. Ramaker
Andrew A. Hardigan
Emily R. Gordon
Carter A. Wright
Richard M. Myers
Sara J. Cooper
author_sort Ryne C. Ramaker
title Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
title_short Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
title_full Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
title_fullStr Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
title_full_unstemmed Pooled CRISPR screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
title_sort pooled crispr screening in pancreatic cancer cells implicates co-repressor complexes as a cause of multiple drug resistance via regulation of epithelial-to-mesenchymal transition
publisher BMC
series BMC Cancer
issn 1471-2407
publishDate 2021-05-01
description Abstract Background Pancreatic ductal adenocarcinoma (PDAC) patients suffer poor outcomes, including a five-year survival of below 10%. Poor outcomes result in part from therapeutic resistance that limits the impact of cytotoxic first-line therapy. Novel therapeutic approaches are needed, but currently no targeted therapies exist to treat PDAC. Methods To assess cellular resistance mechanisms common to four cytotoxic chemotherapies (gemcitabine, 5-fluorouracil, irinotecan, and oxaliplatin) used to treat PDAC patients, we performed four genome-wide CRISPR activation (CRISPRact) and CRISPR knock-out (CRISPRko) screens in two common PDAC cell lines (Panc-1 and BxPC3). We used pathway analysis to identify gene sets enriched among our hits and conducted RNA-sequencing and chromatin immunoprecipitation-sequencing (ChIP-seq) to characterize top hits from our screen. We used scratch assays to assess changes in cellular migration with HDAC1 overexpression. Results Our data revealed activation of ABCG2, a well-described efflux pump, as the most consistent mediator of resistance in each of our screens. CRISPR-mediated activation of genes involved in transcriptional co-repressor complexes also conferred resistance to multiple drugs. Expression of many of these genes, including HDAC1, is associated with reduced survival in PDAC patients. Up-regulation of HDAC1 in vitro increased promoter occupancy and expression of several genes involved in the epithelial-to-mesenchymal transition (EMT). These cells also displayed phenotypic changes in cellular migration consistent with activation of the EMT pathway. The expression changes resulting from HDAC1 activation were also observed with activation of several other co-repressor complex members. Finally, we developed a publicly available analysis tool, PancDS, which integrates gene expression profiles with our screen results to predict drug sensitivity in resected PDAC tumors and cell lines. Conclusion Our results provide a comprehensive resource for identifying cellular mechanisms of drug resistance in PDAC, mechanistically implicate HDAC1, and co-repressor complex members broadly, in multi-drug resistance, and provide an analytical tool for predicting treatment response in PDAC tumors and cell lines.
topic Genome-wide screen
CRISPR
Pancreatic cancer
Drug resistance
ABCG2
HDAC1
url https://doi.org/10.1186/s12885-021-08388-1
work_keys_str_mv AT rynecramaker pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
AT andrewahardigan pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
AT emilyrgordon pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
AT carterawright pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
AT richardmmyers pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
AT sarajcooper pooledcrisprscreeninginpancreaticcancercellsimplicatescorepressorcomplexesasacauseofmultipledrugresistanceviaregulationofepithelialtomesenchymaltransition
_version_ 1721419928812126208