scholarly journals CRISPR Activation Screen for HDAC Inhibitor Resistance

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3958-3958
Author(s):  
Huihui Tang ◽  
Ricardo De Matos Simoes ◽  
Ryosuke Shirasaki ◽  
Olga Dashevsky ◽  
Brian Glassner ◽  
...  

Abstract Panobinostat is an oral broad-spectrum histone deacetylase inhibitor that alters gene expression via epigenetic mechanisms and function of key proteins through changes in their protein acetylation state. Panobinostate was approved a few years ago by the FDA and EMA for use in combination with bortezomib and dexamethasone in patients with multiple myeloma (MM) who have received ≥2 prior regimens, including bortezomib and an immunomodulatory drug. To study the mechanisms which regulate the response vs. resistance of MM cells to panobinostat, we performed genome-scale CRISPR activation screen of MM1S cells. MM1S cells were transduced with dCas9 and pooled lentiviral particles of the Calabrese P65-HSF CRISPR activation library consisting of 2 pooled sgRNA sub-libraries. After selection for viral transduction, cells were continuously cultured with (2 different concentration of 12.5 and 20 nM) or without panobinostat for 5 weeks, before being harvested. At that time point, dose-response curves for panobinostat treatment confirmed that the drug-exposed MM.1S cell populations of our study had become significantly less sensitive to panobinostat than treatment- naive MM.1S cells. Genomic DNA was extracted and next generation sequencing was performed to quantify the abundance of the sgRNA "barcodes" within the tumor cell populations of our study, while rank aggregation algorithms were performed to rank genes according to the magnitude and concordance of enrichment for its different sgRNAs. In MM.1S cells which had developed resistance to the lower dose (12.5 nM) of panobinostat, we observed major sgRNA enrichment (at least 4 of 6 sgRNAs, enrichment p-value <0.05, average log2 fold change of enrichment >1.5) for sgRNAs for a variety of genes, of which the most prominently enriched ones encode for the cell surface ABC transporters ABCB1 (MDR1/p-glycoprotein), to a lesser extent ABCC4 (MRP4) and even less so for ABCG2. In addition, we observed sgRNA enrichment for transcription factors, such as IRF4, POU2AF1, AFF2, IKZF3, AFF3, and RELA, or the transcriptional coregulator MTA1; Bcl2 family members such as BCL2 and BCL2L1; and chromatin remodeling genes such as KAT6A. However, in MM.1S cells which had survived the treatment with higher concentration (25 nM) of panobinostat, the genes with significant and concordant sgRNA enrichment were restricted to ABCB1, ABCC4, and IRF4. These observations indicate that the most efficient mechanism for MM cells to develop resistance to both low and higher concentrations of panobinostat is by increasing its export from the cells, with ABCB1 as the primary, but not sole, transporter which can assume this role. When we transduced MM.1S cells, which already express high levels of IRF4 transcript and protein, with lentiviral construct for IRF4 cDNA, we observed a shift to the right for the panobinostat dose-response curve, further supporting the observation that modulation of IRF4 levels in MM cells can alter the degree of MM cell sensitivity to panobinostat. Proteasome inhibitors suppress the activity of RELA and NFkappaB more broadly, while thalidomide derivatives cause degradation of IKZF3 and can decrease the IRF4 transcript levels: these results may respectively explain, at least in part, the mechanistic basis for the enhanced combined effect of panobinostat with proteasome inhibitors and the favorable clinical results obtained with panobinostat and other broad spectrum HDAC inhibitors in combination with thalidomide derivatives. Our study provides unbiased genome-scale systematic characterization of the mechanisms regulating MM cell response vs. resistance to panobinostat and identify the pronounced and dose-dependent enrichment of these resistance mechanisms for genes contributing to panobinostat export from MM cells, as well as a role for IRF4 and (primarily at lower panobinostat concentrations) for other transcription factors, chromatin remodeling genes and anti-apoptotic BCL-2 family members. These observations also support the need for a more systematic characterization of the regulation of expression of ABC transporters in MM cells; and for development of novel strategies to disrupt more comprehensively IRF4 and other transcription factors for which gain-of-function is associated with decreased responsiveness to panobinostat, with the goal of improving the impact of this agent and potentially other broad spectrum HDAC inhibitors in MM. Disclosures Mitsiades: TEVA: Research Funding; Takeda: Other: employment of a relative; EMD Serono: Research Funding; Abbvie: Research Funding; Janssen/ Johnson & Johnson: Research Funding.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1915-1915
Author(s):  
Sondra L. Downey-Kopyscinski ◽  
Ricardo De Matos Simoes ◽  
Ryosuke Shirasaki ◽  
Olga Dashevsky ◽  
Brian Glassner ◽  
...  

Abstract As multiple myeloma (MM) remains incurable, development of more effective novel therapies will require a deeper understanding of which genes, pathways and molecular networks thereof that govern the biological behavior of MM cells. The emergence of new CRISPR-based functional genomics approaches, including gene editing for loss-of-function (LOF) studies, allowed our group to obtain insight into the role of individual genes for MM survival and cell proliferation in vitro, in vivo and in the context of treatment resistance (e.g. De Matos Simoes et al., Shirasaki et al., Gandolfi et al. ASH 2017). Through these studies, we identified 50+ genes with more pronounced sgRNA depletion in MM cells vs. non-MM lines, which we consider MM-preferential essential genes. These genes include many transcription factors (TFs) such as IRF4, PRDM1, NF-κB, and MAF. We hypothesized that, while these transcription factors are individually important for MM cell survival and proliferation, they cooperate to regulate MM cell behavior in a manner that cannot be fully captured by 1st-generation, single knockout, CRISPR studies. To address this hypothesis, we performed CRISPR/Cas9 dual knockout (DKO) screens against a collection of ~100 genes, which included ~50 MM preferential dependencies; additional genes with broad-spectrum as dependency in in MM and other neoplasias (e.g. KRAS, BRD4, MCL1, BCL2); tumor suppressors (TP53, PTEN); genes which are frequently expressed in MM cells, but not are major dependencies for them in single knockout studies with CRISPR (e.g. ZBP1); as well as control sgRNAs). For these studies, we applied an "orthogonal Cas9" system: MM.1S cells expressing 2 different Cas9 nucleases (from S. pyogenes and S. aureus, respectively) received a pooled lentiviral library of constructs containing 2 sgRNAs per construct, with each sgRNA operating under one of these 2 Cas9 versions, to avoid possible imbalance in editing due to 2 sgRNAs competing for a single Cas9. Our custom library contained all the 5,000 pairwise combinations of dual KOs of the 100 selected genes and their respective single-gene KOs; with at least 4 sgRNAs per gene for each type of cas9, and for a total of ~54,000 single or double KOs represented in the screen. The study contained multiple different types of controls, including the ability to compare the single gene KO data contained within this DKO study vs. our single gene KO genome-scale screens, which we observed to be very similar. In our DKO study, IRF4 was identified as the most common and strongest synergistic partner with synergy scores (SynDKO score) <-1.0 in >50% of its pairwise interactions with other genes of our study: the synergistic partners of IRF4 included MM-preferentially essential genes, broad-spectrum dependencies and non-essential genes for MM cells, indicating functional interactions of IRF4 with a broad spectrum of genes and its importance as a master regulator of MM cells. Other highly recurrent partners for synergistic interactions included MM-preferential dependencies such as TCF3, ZBTB38, PIM2, IKZF1 and EP300; other chromatin remodeling regulators such as CREBBP and ARID1A; or the anti-apoptotic Bcl-2 family members BCL2L1and MCL1. Interestingly, we observed that dual CRISPR knockout for both IKZF1 and IKZF3 did not produce a stronger anti-MM effect than the dual KO of either gene with IRF4 or single KO IRF4 alone. These results suggest that dual genetic depletion of IKZF1 and IKZF3, which simulates the pharmacological depletion of these transcription factors by thalidomide derivatives, induces an anti-MM effect that is quantitatively less pronounced than the knock-down of IRF4, alone or in combination with LOF of either IKZF1 or IKZF3, suggesting the value of further efforts to develop novel therapies to potently and comprehensively suppress the activity of IRF4. Interestingly, we noted that LOF of TP53 or PTEN attenuates the effect of LOF for several essential genes examined in our study, but not IRF4. To our knowledge this is the first study that leverages the power of CRISPR editing to systematically examine the functional interactions between pairs of MM genes. Our results point to several transcription factors, chromatin remodeling genes and anti-apoptotic regulators as "nodes" for recurrent synergistic pairwise-interactions with other genes and importantly identify IRF4 as central regulator in the hierarchy of these interactions in MM cells. Disclosures Mitsiades: EMD Serono: Research Funding; Janssen/ Johnson & Johnson: Research Funding; Abbvie: Research Funding; TEVA: Research Funding; Takeda: Other: employment of a relative.


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 1001
Author(s):  
Katharina Körholz ◽  
Johannes Ridinger ◽  
Damir Krunic ◽  
Sara Najafi ◽  
Xenia F. Gerloff ◽  
...  

Depending on context and tumor stage, deregulation of autophagy can either suppress tumorigenesis or promote chemoresistance and tumor survival. Histone deacetylases (HDACs) can modulate autophagy; however, the exact mechanisms are not fully understood. Here, we analyze the effects of the broad-spectrum HDAC inhibitors (HDACi) panobinostat and vorinostat on the transcriptional regulation of autophagy with respect to autophagy transcription factor activity (Transcription factor EB—TFEB, forkhead boxO—FOXO) and autophagic flux in neuroblastoma cells. In combination with the late-stage autophagic flux inhibitor bafilomycin A1, HDACis increase the number of autophagic vesicles, indicating an increase in autophagic flux. Both HDACi induce nuclear translocation of the transcription factors FOXO1 and FOXO3a, but not TFEB and promote the expression of pro-autophagic FOXO1/3a target genes. Moreover, FOXO1/3a knockdown experiments impaired HDACi treatment mediated expression of autophagy related genes. Combination of panobinostat with the lysosomal inhibitor chloroquine, which blocks autophagic flux, enhances neuroblastoma cell death in culture and hampers tumor growth in vivo in a neuroblastoma zebrafish xenograft model. In conclusion, our results indicate that pan-HDACi treatment induces autophagy in neuroblastoma at a transcriptional level. Combining HDACis with autophagy modulating drugs suppresses tumor growth of high-risk neuroblastoma cells. These experimental data provide novel insights for optimization of treatment strategies in neuroblastoma.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3648-3648
Author(s):  
Marilynn Chow ◽  
Jason D. MacManiman ◽  
Vincent T. Bicocca ◽  
Lina Gao ◽  
Joshi A. Alumkal ◽  
...  

Abstract Introduction: Acute lymphoblastic leukemia is the most common form of pediatric cancer, and a translocation between chromosomes 1 and 19 represents one of the most frequent cytogenetic abnormalities, accounting for ~5% of ALL cases. The 1;19 rearrangement results in a fusion between the E2A and PBX1 genes, and pre B-ALL cells carrying the E2A-PBX1 fusion are consistently arrested at an intermediate stage of B cell lineage development in which the cells express a functional pre B-cell receptor (pre-BCR). We have previously shown that receptor orphan tyrosine kinase receptor 1 (ROR1) is highly expressed on t(1;19) pre B-ALL blasts and required for their viability, even though ROR1 is not directly regulated by either the E2A-PBX1 fusion or the pre-BCR. Moreover, ROR1 exhibits compensatory signaling cross-talk with the pre-BCR such that maximal killing of t(1;19) pre B-ALL cells is achieved by combining silencing of ROR1 with siRNA and inhibition of the pre-BCR with kinase inhibitors such as dasatinib. However, clinical employment of this strategy is hampered as ROR1 is a pseudokinase whose biology is poorly understood and no ROR1 small-molecule inhibitors currently exist. Therefore, it is critical to advance our understanding of the upstream regulatory mechanisms responsible for ROR1 expression to uncover new therapeutic strategies for t(1;19) pre B-cell ALL. Methods: To identify potential regulators of ROR1 expression, we designed two siRNA panels targeting transcription factors predicted to bind the ROR1 locus or an array of chromatin remodeling factors and epigenetic regulators. Pre B-ALL cell lines that are positive or negative for the 1;19 rearrangement were screened with these siRNA panels and any gene targets required for the viability of t(1;19) pre B-cell ALL cells but not control cells lines were considered candidate regulators of ROR1 expression. Candidates were validated for the capacity to regulate ROR1 expression by qRT-PCR and immunoblot. Results: Screening of the t(1;19) pre B-cell ALL cell line, RCH-ACV, with our siRNA panels targeting transcription factors and chromatin remodeling/epigenetic regulators identified four potential regulators of ROR1 that were required for viability of RCH-ACV cells but not our negative control cell line, REH (t(12;21)-positive). Among these four candidates was UHRF1, which interacts with histone and DNA methyltransferases. Interestingly, silencing of UHRF1 by siRNA led to reduced ROR1 protein levels, but had no significant impact on ROR1 mRNA expression. Since one primary function of UHRF1 is to regulate DNA methylation, we hypothesized that UHRF1 regulates ROR1 protein expression through an intermediate gene whose transcription is impacted by UHRF1-mediated DNA methylation. To test this hypothesis, RCH-ACV cells were treated with the RNA synthesis inhibitor actinomycin D 48 hours after silencing UHRF1. This resulted in a transient rescue of ROR1 protein levels, indicating that UHRF1 regulation of ROR1 protein depends on the transcription of an intermediate gene whose gene product subsequently regulates ROR1 protein levels. Conclusions: Data from siRNA screens and subsequent validation experiments suggest that UHRF1 functions to regulate ROR1 protein levels, but not mRNA expression. This is intriguing due to the primary role of UHRF1 as a modulator of DNA and histone methylation. The observed rescue of ROR1 protein with actinomycin D suggests that UHRF1 regulation of ROR1 protein occurs through an undefined intermediary whose locus is methylated in a UHRF1-dependent manner and whose gene product modulates ROR1 protein levels. Inhibitors of additional stages of gene transcription, translation, and stabilization are currently being tested to continue characterizing UHRF1-dependent regulation of ROR1. Cumulatively, this work has identified a novel regulatory mechanism of ROR1 protein expression by UHRF1 in t(1;19) pre B-cell ALL. In addition to advancing our understanding of ROR1 biology, this regulatory mechanism suggests a new therapeutic strategy to target ROR1-expressing cancers. Disclosures Tyner: Incyte: Research Funding; Aptose Biosciences: Research Funding; Janssen Pharmaceuticals: Research Funding; Constellation Pharmaceuticals: Research Funding; Array Biopharma: Research Funding.


Genomics ◽  
2010 ◽  
Vol 95 (1) ◽  
pp. 56-65 ◽  
Author(s):  
Sang-Gyu Kim ◽  
Sangmin Lee ◽  
Pil Joon Seo ◽  
Soon-Kap Kim ◽  
Jeong-Kook Kim ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 18-19
Author(s):  
Ricardo De Matos Simoes ◽  
Ryosuke Shirasaki ◽  
Huihui Tang ◽  
Shizuka Yamano ◽  
Benjamin G Barwick ◽  
...  

Background: Functional genomics studies based on CRISPR and shRNA have documented that multiple myeloma (MM) cells are preferentially dependent (compared to other neoplasias) on a series of TFs, including IKZF1 and IKZF3 (which are targeted by thalidomide derivatives) and others that are not amenable to degradation or small molecule inhibition. Transcriptional co-factors have been therapeutically targeted, for example, inhibitors of BRD4, a co-factor for pTEFB, can be used to down-regulate c-myc. Aim: To identify new transcriptional vulnerabilities in MM with an emphasis on transcriptional co-factors Methods: We integrated results from genome-scale studies using the AVANA library for loss-of-function by gene editing (in 19 MM lines) and the Calabrese library for CRISPR-mediated gene activation (in 5 MM lines) to identify critical transcriptional co-factors (co-TFs). RNA-Seq analysis was used to identify critical pathways affected by POU2AF1 activation and existing ChIP-Seq tracks in MM cells were reanalyzed. Results: POU2AF1 (OCA-B) was the most preferentially essential TF co-factor in MM cell lines vs. non-MM and one of top genes which, upon CRISPR activation in genome-scale studies, increased MM cell fitness in vitro. We further confirmed the role of this gene using focused libraries of sgRNAs against POU2AF1 in vitro and in an in vivo model of MM cell growth in bone marrow-like scaffolds "functionalized" with humanized mesenchymal bone marrow stromal cells to simulate the human BM. CRISPR activation of POU2AF1 is associated with increased MM cell growth. RNA-Seq of POU2AF1 activation in LP1 cells a transcriptional program characterized by upregulation of other genes that are preferentially essential for MM including PRDM1, SUPT7L, UBE2G2 and TSC1; broad-spectrum oncogenic dependencies (e.g KRAS) and genes known or proposed to be involved in the pathophysiology of MM or other neoplasias (e.g. RUNX2, FGFR3, SMO, CREB5, TNFRSF13B, MEF2D, PCGF2). POU2AF1 overexpression was also associated with down-regulation of CDKN1C; of MHC class II molecules and their transcriptional activator CIITA, suggesting that POU2AF1 activation could also contribute to increased MM growth in vivo by allowing escape from immune surveillance. ATAC-Seq data and genome-wide ChIPseq for H3K27Ac in MM cell lines indicate that chromatin surrounding the POU2AF1 locus was highly accessible, concordant with the consistent expression of this TF in MM cell lines and patient-derived cells. CoMMpass data showed that POU2AF1 expression was enhanced in a subset of MM patients at relapse compared to diagnosis. Motif analysis of ChIP-seq data for POU2AF1 identified significant overlap with motifs for TFs relevant to the POU family (e.g. Oct11, Oct2, Oct4); members of the ETS family (e.g. ELF1, Elf4, GABPA); and other TFs with roles in MM including c-myc; IRF4; NF-kappaB, PRDM1, RUNX2 and the POU2AF1 target CREB5. These data suggest a functional interaction between POU2AF1 and other MM-relevant TFs. The transcriptional signature of POU2AF1 activation is enriched for genes downregulated by suppression/inhibition of MM-preferential TFs or epigenetic regulators including IRF4, PRDM1, IKZF1/3 and DOT1L. POU2AF1 binding motifs are also enriched in the promoter regions of MM-preferential dependencies including several MM-preferential TFs. Conclusions: POU2AF1 is essential for MM cells in vitro and in vivo; has a significantly more pronounced and recurrent role as a dependency in MM compared to most other neoplasias; and can further drive MM cell growth, through its ability to interact with several TFs critical for MM, forming multi-protein functional complexes. These results establish POU2AF1 as a central component in the regulatory network of oncogenic TFs in MM and highlight the value of further exploring POU2AF1 as a therapeutic target in MM. Disclosures Downey-Kopyscinski: Rancho BioSciences, LLC: Current Employment. Tsherniak:Cedilla Therapeutics: Consultancy; Tango Therapeutics: Consultancy. Boise:AstraZeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genetech: Membership on an entity's Board of Directors or advisory committees. Mitsiades:FIMECS: Consultancy, Honoraria; Ionis Pharmaceuticals, Inc.: Consultancy, Honoraria; Arch Oncology: Research Funding; Janssen/Johnson & Johnson: Research Funding; Karyopharm: Research Funding; TEVA: Research Funding; Takeda: Other: employment of a relative; Fate Therapeutics: Consultancy, Honoraria; Sanofi: Research Funding; Abbvie: Research Funding; EMD Serono: Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3197-3197
Author(s):  
Huihui Tang ◽  
Ricardo De Matos Simoes ◽  
Ryosuke Shirasaki ◽  
Olga Dashevsky ◽  
Brian Glassner ◽  
...  

Abstract CRISPR/Cas9-based gene editing has become a powerful tool for loss-of-function (LOF) studies and has allowed us to systematically interrogate the function of genes regulating the survival and proliferation of multiple myeloma (MM) cells in vitro, in vivo and in the context of treatment resistance (e.g. De Matos Simoes et al., Shirasaki et al., and Gandolfi et al. ASH 2017). We reasoned, however, that important additional information can be obtained from CRISPR-based gain-of-function (GOF) approaches which can achieve transcriptional activation at endogenous genomic loci. We thus performed genome-scale CRISPR activation studies using the dCas9-P65-HSF transcriptional activation system (in which a Cas9 variant lacking nuclease activity [dCas9] confers P65-HSF-mediated activation of genes recognized by sgRNAs against their promoter regions). Specifically, MM.1S cells were transduced with the dCas9-P65-HSF system and pooled lentiviral particles of the Calabrese CRISPR activation sgRNA library, consisting of 2 pooled sgRNA sub-libraries (total of ~110,000 sgRNAs targeting ~18000 genes, at initial coverage of 800 cells/sgRNA). Cells were cultured for 12 weeks and harvested at baseline and various intervals, e.g. 4 and 12 weeks of culture. Next generation sequencing of genomic DNA quantified the abundance of sgRNAs in the tumor cell population and genes were ranked (with rank aggregation algorithms) in terms of their sgRNA enrichment or depletion. These analyses allowed us to observe a series of genes with statistically significant sgRNA enrichment and known or presumed roles in MM biology, including key MM transcription factors such as IRF4, the thalidomide derivative targets IKZF3 and IKZF1, and the co-factor POU2AF1; known oncogenes, e.g. KRAS and MYC; NF-kappaB pathway members, e.g. RELA; and signal transduction regulators, e.g. IGF1R and its downstream effectors IRS1 and AKT2. These results are consistent with the major depletion of sgRNAs for these genes in loss-of-function (LOF) CRISPR knockout studies. However, several other genes with significant sgRNA enrichment in CRISPR activation studies did not exhibit major sgRNA depletion in CRISPR knockout studies, including the B/plasma cell transcription factor POU2F2 (Oct2), for which high protein expression correlates with reduced survival in MM (Toman I. et al 2011); the transcription factor PAX2, the TRAF interacting protein TIFA, or the Toll-like receptor TLR4. Interestingly, significant depletion of sgRNAs was observed for several genes with known or proposed tumor suppressive properties including YAP1 (an oncogene for solid tumors, but reported as tumor suppressor for MM and other blood cancers); the pro-death TNFRSF10A (TRAIL receptor DR4), TP73, CDKN1A, the negative regulator of c-Myc MXI1, or the pro-apoptotic Bcl2 family member BAK1. Depletion or enrichment of sgRNAs for most of the aforementioned genes was detectable by 4 weeks of culture, while more pronounced changes and detection of additional statistically-significant hits was observed in later time-points. For genes with significant sgRNA enrichment in our CRISPR activation study, we examined a series of molecular alterations, including transcript overexpression in MM cell lines or patient-derived samples vs. normal plasma cells, or relapsed/refractory MM vs. earlier disease MM stages; mutational status; correlation of transcript levels with clinical outcome in MM; and extent of open chromatin (based on H3K27Ac chromatin marks) within or proximal to each gene in MM cell lines. Some "hits" from our screen exhibited at least one of these molecular alterations, but most genes harbored no such alterations or their magnitude or frequency ranked outside the top 50-100 genes. These results suggest that CRISPR activation studies can identify important putative regulators of MM biology, which may not be readily detectable based on known annotations of the MM cell genome, transcriptome, or epigenome. Genome-scale CRISPR-based transcriptional activation are an important gain-of-function system to uncover genes which induce vs. suppress tumor cell survival and proliferation, and provide information orthogonal to those yielded by other CRISPR-based approaches that involve loss-of-function interventions. Our use of CRISPR activation allowed us to both validate previously known genes and identify promising new candidate regulators of MM cell biology. Disclosures Mitsiades: EMD Serono: Research Funding; Abbvie: Research Funding; Takeda: Other: employment of a relative; TEVA: Research Funding; Janssen/ Johnson & Johnson: Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-4
Author(s):  
Ryosuke Shirasaki ◽  
Ricardo De Matos Simoes ◽  
Shizuka Yamano ◽  
Sondra L. Downey-Kopyscinski ◽  
Brian Glassner ◽  
...  

Background: Endoplasmic reticulum (ER)-associated degradation (ERAD) for unfolded proteins represents an important biological vulnerability for multiple myeloma (MM) cells, given the high proteostatic stress in these cells due to immunoglobulin secretion. This helps explain the efficacy of proteasome inhibitors in MM compared to most other neoplasms. Aim: To systematically investigate ER-associated genes that mediate the essential role of ERAD in MM. Methods: We integrated results from (i) genome-scale studies for CRISPR-gene knockout in 19 MM cell lines (Avana library) and CRISPR-activation (Calabrese library) in 5 MM lines, to define ER-associated dependencies and drivers of increased MM cell growth; (ii) publicly-available CRISPR-based studies for genes regulating ERAD of substrates located in the luminal (ERAD-L), membrane (ERAD-M) or cytosolic (ERAD-C) side of the ER in non-MM models; (iii) functional characterization of candidate genes in MM models in vitro and in vivo. Results: While ER function in general is globally required for all normal and malignant cells, a surprisingly large proportion of ER-related genes are not "pan-essential" genes (e.g. CERES scores &lt;-0.4 are for &lt;80% of human cell lines): in fact CRISPR-mediated disruption of many ER genes elicits no growth disadvantage for most non-MM tumor cell lines. Notably, though, several of these latter non-"pan- essential" genes are recurrently essential for MM cell lines, or even preferential dependencies for MM compared to other cancers. By overlaying the patterns of CRISPR essentiality in MM lines vs. the functional modules of ER function, we observe that recurrent or preferential ER-associated dependencies for MM cells include genes involved in (a) ER membrane protein complexes mediating dislocation of misfolded ER proteins to the cytosol (e.g. HERPUD1, SEL1L, AUP1 for ERAD-L substrates; and AMFR and RNF139 for ERAD-M and -C substrates,) and associated ER-specific E2/E3 enzymes (SYVN1, UBE2J1, UBE2J2, and UBE2G2); (b) upstream of the ER-to-cytosol dislocation complexes, several enzymes (e.g. DPM1, DPM3, ALG3, ALG9, ALG12, ALG6, ALG8) required for N-glycan-dependent surveillance of quality control for luminal ER glycoproteins; (c) chaperones (e.g. DNAJB11, DNAJBC3) for BiP complexes with misfolded proteins; (d) the known ER stress-sensor IRE1a (ERN1) and its downstream transcription factor XBP1. CRISPR-activation of nearly all these recurrent/MM-preferential ER-associated genes led to no significant growth advantage for MM cell lines. Several of the MM-preferential ER-associated genes (HERPUD1, SEL1L, SYVN1, UBE2J1) are expressed at significantly higher levels in MM vs. non-MM cell lines (CCLE) or patient derived samples (CoMMpass study vs. TCGA) and are proximal to binding sites of key transcription factors for plasma cell identity such as IRF4 and PRDM1. By contrast, CRISPR-mediated LOF of such genes in MM cells decreases their growth in vitro (e.g. competition assays vs. CRISPR LOF of non-essential genes not expressed in MM cells) and in vivo for MM cells growing in a bone marrow-like scaffold system with a "humanized" mesenchymal bone marrow stromal cell compartment. Knockout of MM-preferential ER-associated genes induces markers of ER stress/unfolded protein response (e.g. BiP induction by UBE2J1 or HERPUD1 LOF); and sensitizes MM cells to proteasome inhibition. Accordingly, pharmacological inhibition (LS-102) of SYVN1 (Synovolin-1) an E3 ligase which helps remove unfolded proteins from the cytosol into the ER was more effective at a lower drug concentration in MM compared to non-MM-cell lines. Conclusions: Our results provide functional insights into the role of ERAD in MM cells and nominate specific mediators which are not broadly essential across all cancers, but are recurrent or even preferentially essential for MM. Our study indicates a previously underappreciated role for the ER-to-cytosol dislocon complex that includes HERPUD1, SEL1L, SYVN1 and UBE2J1; distinct enzymes of the N-glycan-dependent ER glycoproteins quality control system; and ER chaperones in MM. These findings provide a rationale for development of agents selectively targeting these key regulators of ER protein folding processes in MM. Disclosures Downey-Kopyscinski: Rancho BioSciences, LLC: Current Employment. Tsherniak:Tango Therapeutics: Consultancy; Cedilla Therapeutics: Consultancy. Boise:AstraZeneca: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genetech: Membership on an entity's Board of Directors or advisory committees. Mitsiades:EMD Serono: Research Funding; Abbvie: Research Funding; Karyopharm: Research Funding; Janssen/Johnson & Johnson: Research Funding; TEVA: Research Funding; FIMECS: Consultancy, Honoraria; Takeda: Other: employment of a relative; Ionis Pharmaceuticals, Inc.: Consultancy, Honoraria; Fate Therapeutics: Consultancy, Honoraria; Sanofi: Research Funding; Arch Oncology: Research Funding.


Cell Reports ◽  
2014 ◽  
Vol 8 (2) ◽  
pp. 622-632 ◽  
Author(s):  
Jose L. Pruneda-Paz ◽  
Ghislain Breton ◽  
Dawn H. Nagel ◽  
S. Earl Kang ◽  
Katia Bonaldi ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 313-313
Author(s):  
Huihui Tang ◽  
Ryosuke Shirasaki ◽  
Ricardo De Matos Simoes ◽  
Sara Gandolfi ◽  
Olga Dashevsky ◽  
...  

Loss-of-function (LOF) studies (e.g. using RNAi or CRISPR) have historically been the main functional approaches to identify and study genes which drive the biology of Multiple Myeloma(MM) or other neoplasias. We hypothesized though that substantial complementary data can be derived from systematically examining the impact of transcriptional activation of endogenous genomic loci of different genes. To perform such gain-of-function (GOF) studies in an open-ended genome-scale manner, we applied CRISPR activation approaches in 4 MM lines (MM1S, KMS11, LP1, L363) which were transduced with the dCas9-P65-HSF transcriptional activation system and the Calabrese genome-scale sgRNA library (2 pooled sub-libraries; total of ~110,000 sgRNAs targeting promoters of ~18000 genes; initial coverage ~800-1000 cells/sgRNA). Next generation sequencing quantified the sgRNA abundance in MM cells at baseline and various time points (e.g. 2-12 weeks of culture), and rank aggregation algorithms identified genes with statistically significant enrichment or depletion of their sgRNAs, reflecting positive vs. negative, respectively, effects of GOF of these genes on MM cell survival/proliferation. These studies identified distinct groups of genes which serve as positive regulators of MM cell growth in at least 3 of the cell lines tested, including critical transcription factors (TFs)/cofactors, such as POU2F2, POU2AF1, IRF4, MYC; growth factor signaling mediators, such as IGF1R and IRS1; Ras family members (e.g. KRAS in MM1S cells); diverse members of the solute carrier family of transporters for amino acids and other bioactive small molecules. We also observed positive regulators of MM cell growth with distinct roles in certain cell lines, e.g. induction of IKZF3, IKZF1, RELA, CD48, MBNL1, PAX2 exhibited significantly more pronounced role as positive regulators of MM1S cells compared to the other cell lines tested. Notably, several positive regulators of MM growth identified in these CRISPR activation studies are not essential for MM cell survival/proliferation in CRISPR knockout studies in the same MM cell lines or many others. Examples of such genes include the TFs POU2F2 (Oct2) and PAX2, the TRAF interacting protein TIFA or the Toll-like receptor TLR4. We went on to validate these results for several such genes (e.g. POU2F2, POU2AF1) with individual sgRNAs for CRISPR activation and/or cDNA overexpression (vs. isogenic controls) in competition experiments and cell cycle analyses. We focused on further probing the mechanistic basis for the role of POU2AF1 as one of the top positive regulators of MM cell growth and performed RNA sequencing analyses of LP1 cells harboring induction of POU2AF1 expression using CRISPR activation. We observed upregulation of a distinct cluster of &gt;50 genes that are themselves positive regulators of LP1 cell growth in our genome-scale CRISPR activation studies. While some of these are known drivers of tumor cell growth (e.g. AURKB, E2F1, FGFR3), the large majority of these POU2AF1-driven positive regulators of MM cell growth have not been previously studied in MM; and exhibit a distinct enrichment for transcriptional regulators and other genes which are not required for LP1 cell survival/proliferation in CRISPR KO studies of LP1 cells. These results indicate that CRISPR-based activation of endogenous gene expression provides data which complementary to those derived from CRISPR LOF studies. Indeed, we not only validated through our GOF studies the relevance of genes which with prominent essentiality in CRISPR LOF studies (e.g. IRF4, POU2AF1, etc.), but also identified promising new candidate regulators of MM cell biology which are not essential for baseline survival/growth of MM cells, but can induce MM cell growth when further activated. These results provide novel insights into the biology of MM cells and may have therapeutic implications towards suppressing the ability of MM cells to transition to states of advanced biological aggressiveness. Disclosures Mitsiades: Takeda: Other: employment of a relative ; Ionis Pharmaceuticals: Honoraria; Fate Therapeutics: Honoraria; Arch Oncology: Research Funding; Sanofi: Research Funding; Karyopharm: Research Funding; Abbvie: Research Funding; TEVA: Research Funding; EMD Serono: Research Funding; Janssen/Johnson & Johnson: Research Funding.


Author(s):  
Araceli García-Martínez ◽  
Sandra Silva-Ortega ◽  
Beatriz López-Muñoz ◽  
Óscar Moreno-Pérez ◽  
Irene Monjas ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document