Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Cell ; 84(13): 2553-2572.e19, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38917794

ABSTRACT

CRISPR-Cas technology has transformed functional genomics, yet understanding of how individual exons differentially shape cellular phenotypes remains limited. Here, we optimized and conducted massively parallel exon deletion and splice-site mutation screens in human cell lines to identify exons that regulate cellular fitness. Fitness-promoting exons are prevalent in essential and highly expressed genes and commonly overlap with protein domains and interaction interfaces. Conversely, fitness-suppressing exons are enriched in nonessential genes, exhibiting lower inclusion levels, and overlap with intrinsically disordered regions and disease-associated mutations. In-depth mechanistic investigation of the screen-hit TAF5 alternative exon-8 revealed that its inclusion is required for assembly of the TFIID general transcription initiation complex, thereby regulating global gene expression output. Collectively, our orthogonal exon perturbation screens established a comprehensive repository of phenotypically important exons and uncovered regulatory mechanisms governing cellular fitness and gene expression.


Subject(s)
Exons , Humans , Exons/genetics , CRISPR-Cas Systems , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Genetic Fitness , HEK293 Cells , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , RNA Splice Sites , Mutation , Gene Expression Regulation , Alternative Splicing
2.
Cell ; 163(6): 1515-26, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26627737

ABSTRACT

The ability to perturb genes in human cells is crucial for elucidating gene function and holds great potential for finding therapeutic targets for diseases such as cancer. To extend the catalog of human core and context-dependent fitness genes, we have developed a high-complexity second-generation genome-scale CRISPR-Cas9 gRNA library and applied it to fitness screens in five human cell lines. Using an improved Bayesian analytical approach, we consistently discover 5-fold more fitness genes than were previously observed. We present a list of 1,580 human core fitness genes and describe their general properties. Moreover, we demonstrate that context-dependent fitness genes accurately recapitulate pathway-specific genetic vulnerabilities induced by known oncogenes and reveal cell-type-specific dependencies for specific receptor tyrosine kinases, even in oncogenic KRAS backgrounds. Thus, rigorous identification of human cell line fitness genes using a high-complexity CRISPR-Cas9 library affords a high-resolution view of the genetic vulnerabilities of a cell.


Subject(s)
Genes, Essential , Bayes Theorem , CRISPR-Cas Systems , Cell Line, Tumor , Gene Knockout Techniques , Gene Library , Humans , Mutation
3.
Nature ; 586(7827): 120-126, 2020 10.
Article in English | MEDLINE | ID: mdl-32968282

ABSTRACT

The genetic circuits that allow cancer cells to evade destruction by the host immune system remain poorly understood1-3. Here, to identify a phenotypically robust core set of genes and pathways that enable cancer cells to evade killing mediated by cytotoxic T lymphocytes (CTLs), we performed genome-wide CRISPR screens across a panel of genetically diverse mouse cancer cell lines that were cultured in the presence of CTLs. We identify a core set of 182 genes across these mouse cancer models, the individual perturbation of which increases either the sensitivity or the resistance of cancer cells to CTL-mediated toxicity. Systematic exploration of our dataset using genetic co-similarity reveals the hierarchical and coordinated manner in which genes and pathways act in cancer cells to orchestrate their evasion of CTLs, and shows that discrete functional modules that control the interferon response and tumour necrosis factor (TNF)-induced cytotoxicity are dominant sub-phenotypes. Our data establish a central role for genes that were previously identified as negative regulators of the type-II interferon response (for example, Ptpn2, Socs1 and Adar1) in mediating CTL evasion, and show that the lipid-droplet-related gene Fitm2 is required for maintaining cell fitness after exposure to interferon-γ (IFNγ). In addition, we identify the autophagy pathway as a conserved mediator of the evasion of CTLs by cancer cells, and show that this pathway is required to resist cytotoxicity induced by the cytokines IFNγ and TNF. Through the mapping of cytokine- and CTL-based genetic interactions, together with in vivo CRISPR screens, we show how the pleiotropic effects of autophagy control cancer-cell-intrinsic evasion of killing by CTLs and we highlight the importance of these effects within the tumour microenvironment. Collectively, these data expand our knowledge of the genetic circuits that are involved in the evasion of the immune system by cancer cells, and highlight genetic interactions that contribute to phenotypes associated with escape from killing by CTLs.


Subject(s)
Genome/genetics , Genomics , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/genetics , Tumor Escape/immunology , Animals , Autophagy , Cell Line, Tumor , Female , Genes, Neoplasm/genetics , Humans , Interferon-gamma/immunology , Male , Mice , NF-kappa B/metabolism , Reproducibility of Results , Signal Transduction
4.
Mol Cell ; 72(3): 510-524.e12, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388412

ABSTRACT

Alternative splicing is crucial for diverse cellular, developmental, and pathological processes. However, the full networks of factors that control individual splicing events are not known. Here, we describe a CRISPR-based strategy for the genome-wide elucidation of pathways that control splicing and apply it to microexons with important functions in nervous system development and that are commonly misregulated in autism. Approximately 200 genes associated with functionally diverse regulatory layers and enriched in genetic links to autism control neuronal microexons. Remarkably, the widely expressed RNA binding proteins Srsf11 and Rnps1 directly, preferentially, and frequently co-activate these microexons. These factors form critical interactions with the neuronal splicing regulator Srrm4 and a bi-partite intronic splicing enhancer element to promote spliceosome formation. Our study thus presents a versatile system for the identification of entire splicing regulatory pathways and further reveals a common mechanism for the definition of neuronal microexons that is disrupted in autism.


Subject(s)
Alternative Splicing/physiology , Genetic Engineering/methods , RNA Splice Sites/physiology , Animals , Autistic Disorder/genetics , CRISPR-Cas Systems/genetics , Cell Line , Exons/physiology , Humans , Mice , Nerve Tissue Proteins , Neurogenesis , Neurons , RNA Precursors/physiology , RNA Splicing/physiology , RNA-Binding Proteins , Ribonucleoproteins , Serine-Arginine Splicing Factors , Spliceosomes
5.
Nat Chem Biol ; 18(12): 1370-1379, 2022 12.
Article in English | MEDLINE | ID: mdl-35970996

ABSTRACT

Pyrvinium is a quinoline-derived cyanine dye and an approved anti-helminthic drug reported to inhibit WNT signaling and have anti-proliferative effects in various cancer cell lines. To further understand the mechanism by which pyrvinium is cytotoxic, we conducted a pooled genome-wide CRISPR loss-of-function screen in the human HAP1 cell model. The top drug-gene sensitizer interactions implicated the malate-aspartate and glycerol-3-phosphate shuttles as mediators of cytotoxicity to mitochondrial complex I inhibition including pyrvinium. By contrast, perturbation of the poorly characterized gene C1orf115/RDD1 resulted in strong resistance to the cytotoxic effects of pyrvinium through dysregulation of the major drug efflux pump ABCB1/MDR1. Interestingly, C1orf115/RDD1 was found to physically associate with ABCB1/MDR1 through proximity-labeling experiments and perturbation of C1orf115 led to mis-localization of ABCB1/MDR1. Our results are consistent with a model whereby C1orf115 modulates drug efflux through regulation of the major drug exporter ABCB1/MDR1.


Subject(s)
Antineoplastic Agents , Pyrvinium Compounds , Humans , Pyrvinium Compounds/pharmacology , Wnt Signaling Pathway , Antineoplastic Agents/pharmacology , Genomics
6.
Nature ; 559(7713): 285-289, 2018 07.
Article in English | MEDLINE | ID: mdl-29973717

ABSTRACT

The observation that BRCA1- and BRCA2-deficient cells are sensitive to inhibitors of poly(ADP-ribose) polymerase (PARP) has spurred the development of cancer therapies that use these inhibitors to target deficiencies in homologous recombination1. The cytotoxicity of PARP inhibitors depends on PARP trapping, the formation of non-covalent protein-DNA adducts composed of inhibited PARP1 bound to DNA lesions of unclear origins1-4. To address the nature of such lesions and the cellular consequences of PARP trapping, we undertook three CRISPR (clustered regularly interspersed palindromic repeats) screens to identify genes and pathways that mediate cellular resistance to olaparib, a clinically approved PARP inhibitor1. Here we present a high-confidence set of 73 genes, which when mutated cause increased sensitivity to PARP inhibitors. In addition to an expected enrichment for genes related to homologous recombination, we discovered that mutations in all three genes encoding ribonuclease H2 sensitized cells to PARP inhibition. We establish that the underlying cause of the PARP-inhibitor hypersensitivity of cells deficient in ribonuclease H2 is impaired ribonucleotide excision repair5. Embedded ribonucleotides, which are abundant in the genome of cells deficient in ribonucleotide excision repair, are substrates for cleavage by topoisomerase 1, resulting in PARP-trapping lesions that impede DNA replication and endanger genome integrity. We conclude that genomic ribonucleotides are a hitherto unappreciated source of PARP-trapping DNA lesions, and that the frequent deletion of RNASEH2B in metastatic prostate cancer and chronic lymphocytic leukaemia could provide an opportunity to exploit these findings therapeutically.


Subject(s)
CRISPR-Cas Systems , DNA Damage , Gene Editing , Neoplasms/genetics , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Ribonucleotides/genetics , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Cell Line , DNA Damage/drug effects , DNA Repair/genetics , DNA Replication , DNA Topoisomerases, Type I/metabolism , Female , Genes, BRCA1 , Genome/genetics , HeLa Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Ribonuclease H/deficiency , Ribonuclease H/genetics , Ribonuclease H/metabolism , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays
7.
Mol Cell ; 61(5): 734-746, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26942677

ABSTRACT

The creation of translation-competent mRNA is dependent on RNA polymerase II transcripts being modified by addition of the 7-methylguanosine (m7G) cap. The factors that mediate splicing, nuclear export, and translation initiation are recruited to the transcript via the cap. The cap structure is formed by several activities and completed by RNMT (RNA guanine-7 methyltransferase), which catalyzes N7 methylation of the cap guanosine. We report that CDK1-cyclin B1 phosphorylates the RNMT regulatory domain on T77 during G2/M phase of the cell cycle. RNMT T77 phosphorylation activates the enzyme both directly and indirectly by inhibiting interaction with KPNA2, an RNMT inhibitor. RNMT T77 phosphorylation results in elevated m7G cap methyltransferase activity at the beginning of G1 phase, coordinating mRNA capping with the burst of transcription that occurs following nuclear envelope reformation. RNMT T77 phosphorylation is required for the production of cohort of proteins, and inhibiting T77 phosphorylation reduces the cell proliferation rate.


Subject(s)
Cyclin B1/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase , Methyltransferases/metabolism , RNA Caps/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Transcription, Genetic , CDC2 Protein Kinase , Cell Proliferation , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Enzyme Activation , G2 Phase , HEK293 Cells , HeLa Cells , Humans , Methylation , Methyltransferases/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Caps/genetics , RNA Interference , RNA, Messenger/genetics , S Phase , Signal Transduction , Time Factors , Transfection , alpha Karyopherins/genetics , alpha Karyopherins/metabolism
8.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34083438

ABSTRACT

Regulation of cell volume is essential for tissue homeostasis and cell viability. In response to hypertonic stress, cells need rapid electrolyte influx to compensate water loss and to prevent cell death in a process known as regulatory volume increase (RVI). However, the molecular component able to trigger such a process was unknown to date. Using a genome-wide CRISPR/Cas9 screen, we identified LRRC8A, which encodes a chloride channel subunit, as the gene most associated with cell survival under hypertonic conditions. Hypertonicity activates the p38 stress-activated protein kinase pathway and its downstream MSK1 kinase, which phosphorylates and activates LRRC8A. LRRC8A-mediated Cl- efflux facilitates activation of the with-no-lysine (WNK) kinase pathway, which in turn, promotes electrolyte influx via Na+/K+/2Cl- cotransporter (NKCC) and RVI under hypertonic stress. LRRC8A-S217A mutation impairs channel activation by MSK1, resulting in reduced RVI and cell survival. In summary, LRRC8A is key to bidirectional osmotic stress responses and cell survival under hypertonic conditions.


Subject(s)
Cell Size , Chloride Channels/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Biological Transport , CRISPR-Cas Systems , Cell Death , Cell Survival , HeLa Cells , Humans , Osmotic Pressure , Phosphorylation , Potassium/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sodium/metabolism
9.
Mol Syst Biol ; 17(5): e10013, 2021 05.
Article in English | MEDLINE | ID: mdl-34018332

ABSTRACT

We present FLEX (Functional evaluation of experimental perturbations), a pipeline that leverages several functional annotation resources to establish reference standards for benchmarking human genome-wide CRISPR screen data and methods for analyzing them. FLEX provides a quantitative measurement of the functional information captured by a given gene-pair dataset and a means to explore the diversity of functions captured by the input dataset. We apply FLEX to analyze data from the diverse cell line screens generated by the DepMap project. We identify a predominant mitochondria-associated signal within co-essentiality networks derived from these data and explore the basis of this signal. Our analysis and time-resolved CRISPR screens in a single cell line suggest that the variable phenotypes associated with mitochondria genes across cells may reflect screen dynamics and protein stability effects rather than genetic dependencies. We characterize this functional bias and demonstrate its relevance for interpreting differential hits in any CRISPR screening context. More generally, we demonstrate the utility of the FLEX pipeline for performing robust comparative evaluations of CRISPR screens or methods for processing them.


Subject(s)
Gene Regulatory Networks , Genetic Testing/methods , Mitochondria/genetics , Systems Biology/methods , Algorithms , Benchmarking , Bias , CRISPR-Cas Systems , Cell Line , HEK293 Cells , Humans
11.
RNA Biol ; 14(1): 11-14, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27791484

ABSTRACT

The mRNA cap structure, which is added to nascent RNA pol II transcripts, recruits the protein complexes required for pre-mRNA transcript processing, mRNA export and translation initiation. The enzymes which catalyze mRNA cap synthesis are regulated by cellular signaling pathways which impact on their expression, localization and activity. Here we discuss the recent observation that the mRNA cap methyltransferase, RNMT, is phosphorylated on Thr-77 by CDK1-cyclin B1, which regulates its activity and the proteins with which it interacts. RNMT Thr-77 phosphorylation provides a burst of mRNA cap methyltransferase activity during early G1 phase at a time when transcription is reactivated following completion of the cell cycle. This co-ordination of transcription and mRNA capping makes an important contribution to gene expression in the cell; preventing RNMT Thr-77 phosphorylation inhibits cell proliferation. Here we discuss these findings and how mRNA cap synthesis may be regulated in other scenarios.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Humans , Methylation , Methyltransferases/metabolism , Phosphorylation , Protein Biosynthesis , Transcription, Genetic
12.
Mol Syst Biol ; 11(7): 821, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26208794

ABSTRACT

The application of the CRISPR­Cas9 system marks a major breakthrough for genetic screens, particularly in mammalian cells where high­throughput targeted gene editing has been lacking. Parnas et al (2015) apply this screening technology to mouse bone marrow­derived dendritic cells in order to study the regulation of the immune response triggered by PAMPs. Through integrated analysis of gene knockouts in conjunction with changes in protein and mRNA expression, CRISPR screens are facilitating dissection of immune regulatory networks at unprecedented resolution.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Dendritic Cells/immunology , Animals , Gene Knockdown Techniques , Gene Regulatory Networks , Immunity, Innate , Mice , RNA Editing
13.
PLoS Pathog ; 8(9): e1002941, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028332

ABSTRACT

In plants, RNA silencing-based antiviral defense is mediated by Dicer-like (DCL) proteins producing short interfering (si)RNAs. In Arabidopsis infected with the bipartite circular DNA geminivirus Cabbage leaf curl virus (CaLCuV), four distinct DCLs produce 21, 22 and 24 nt viral siRNAs. Using deep sequencing and blot hybridization, we found that viral siRNAs of each size-class densely cover the entire viral genome sequences in both polarities, but highly abundant siRNAs correspond primarily to the leftward and rightward transcription units. Double-stranded RNA precursors of viral siRNAs can potentially be generated by host RDR-dependent RNA polymerase (RDR). However, genetic evidence revealed that CaLCuV siRNA biogenesis does not require RDR1, RDR2, or RDR6. By contrast, CaLCuV derivatives engineered to target 30 nt sequences of a GFP transgene by primary viral siRNAs trigger RDR6-dependent production of secondary siRNAs. Viral siRNAs targeting upstream of the GFP stop codon induce secondary siRNAs almost exclusively from sequences downstream of the target site. Conversely, viral siRNAs targeting the GFP 3'-untranslated region (UTR) induce secondary siRNAs mostly upstream of the target site. RDR6-dependent siRNA production is not necessary for robust GFP silencing, except when viral siRNAs targeted GFP 5'-UTR. Furthermore, viral siRNAs targeting the transgene enhancer region cause GFP silencing without secondary siRNA production. We conclude that the majority of viral siRNAs accumulating during geminiviral infection are RDR1/2/6-independent primary siRNAs. Double-stranded RNA precursors of these siRNAs are likely generated by bidirectional readthrough transcription of circular viral DNA by RNA polymerase II. Unlike transgenic mRNA, geminiviral mRNAs appear to be poor templates for RDR-dependent production of secondary siRNAs.


Subject(s)
Arabidopsis/virology , Geminiviridae/genetics , RNA Interference , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , 3' Untranslated Regions , 5' Untranslated Regions/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Gene Expression Regulation, Plant , Green Fluorescent Proteins/genetics , High-Throughput Nucleotide Sequencing , Plant Diseases/genetics , Plant Diseases/virology , RNA Polymerase II/metabolism , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
14.
Biochem J ; 455(1): 67-73, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23863084

ABSTRACT

Gene expression in eukaryotes is dependent on the mRNA methyl cap which mediates mRNA processing and translation initiation. Synthesis of the methyl cap initiates with the addition of 7-methylguanosine to the initiating nucleotide of RNA pol II (polymerase II) transcripts, which occurs predominantly during transcription and in mammals is catalysed by RNGTT (RNA guanylyltransferase and 5' phosphatase) and RNMT (RNA guanine-7 methyltransferase). RNMT has a methyltransferase domain and an N-terminal domain whose function is unclear; it is conserved in mammals, but not required for cap methyltransferase activity. In the present study we report that the N-terminal domain is necessary and sufficient for RNMT recruitment to transcription initiation sites and that recruitment occurs in a DRB (5,6-dichloro-1-ß-D-ribofuranosylbenzimidazole)-dependent manner. The RNMT-activating subunit, RAM (RNMT-activating miniprotein), is also recruited to transcription initiation sites via an interaction with RNMT. The RNMT N-terminal domain is required for transcript expression, translation and cell proliferation.


Subject(s)
Gene Expression Regulation/drug effects , Methyltransferases/metabolism , Peptide Chain Initiation, Translational/drug effects , RNA-Binding Proteins/metabolism , Transcription Initiation Site , Transcription, Genetic/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Methyltransferases/genetics , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , Signal Transduction
15.
Nucleic Acids Res ; 40(13): 6241-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22434877

ABSTRACT

Biogenesis of trans-acting siRNAs (tasiRNAs) is initiated by miRNA-directed cleavage of TAS gene transcripts and requires RNA-dependent RNA polymerase 6 (RDR6) and Dicer-like 4 (DCL4). Here, we show that following miR173 cleavage the entire polyadenylated parts of Arabidopsis TAS1a/b/c and TAS2 transcripts are converted by RDR6 to double-stranded (ds)RNAs. Additionally, shorter dsRNAs are produced following a second cleavage directed by a TAS1c-derived siRNA. This tasiRNA and miR173 guide Argonaute 1 complexes to excise the segments from TAS2 and three TAS1 transcripts including TAS1c itself to be converted to dsRNAs, which restricts siRNA production to a region between the two cleavage sites. TAS1c is also feedback regulated by a cis-acting siRNA. We conclude that TAS1c generates a master siRNA that controls a complex network of TAS1/TAS2 siRNA biogenesis and gene regulation. TAS1/TAS2 short dsRNAs produced in this network are processed by DCL4 from both ends in distinct registers, which increases repertoires of tasiRNAs.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Gene Expression Regulation, Plant , RNA Processing, Post-Transcriptional , RNA, Double-Stranded/metabolism , RNA, Small Interfering/metabolism , RNA-Dependent RNA Polymerase/metabolism , Arabidopsis/metabolism , Base Sequence , Gene Knockdown Techniques , Genes, Plant , MicroRNAs/metabolism , Molecular Sequence Data , Polyadenylation , RNA Cleavage , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Double-Stranded/chemistry , RNA, Plant/chemistry , RNA, Plant/genetics , RNA, Plant/metabolism , RNA, Small Interfering/genetics
16.
Neuro Oncol ; 26(6): 1083-1096, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38134889

ABSTRACT

BACKGROUND: The cell cycle is tightly regulated by checkpoints, which play a vital role in controlling its progression and timing. Cancer cells exploit the G2/M checkpoint, which serves as a resistance mechanism against genotoxic anticancer treatments, allowing for DNA repair prior to cell division. Manipulating cell cycle timing has emerged as a potential strategy to augment the effectiveness of DNA damage-based therapies. METHODS: In this study, we conducted a forward genome-wide CRISPR/Cas9 screening with repeated exposure to the alkylating agent temozolomide (TMZ) to investigate the mechanisms underlying tumor cell survival under genotoxic stress. RESULTS: Our findings revealed that canonical DNA repair pathways, including the Ataxia-telangiectasia mutated (ATM)/Fanconi and mismatch repair, determine cell fate under genotoxic stress. Notably, we identified the critical role of PKMYT1, in ensuring cell survival. Depletion of PKMYT1 led to overwhelming TMZ-induced cytotoxicity in cancer cells. Isobologram analysis demonstrated potent drug synergy between alkylating agents and a Myt1 kinase inhibitor, RP-6306. Mechanistically, inhibiting Myt1 forced G2/M-arrested cells into an unscheduled transition to the mitotic phase without complete resolution of DNA damage. This forced entry into mitosis, along with persistent DNA damage, resulted in severe mitotic abnormalities. Ultimately, these aberrations led to mitotic exit with substantial apoptosis. Preclinical animal studies demonstrated that the combination regimen involving TMZ and RP-6306 prolonged the overall survival of glioma-bearing mice. CONCLUSIONS: Collectively, our findings highlight the potential of targeting cell cycle timing through Myt1 inhibition as an effective strategy to enhance the efficacy of current standard cancer therapies, potentially leading to improved disease outcomes.


Subject(s)
Antineoplastic Agents, Alkylating , DNA Damage , G2 Phase Cell Cycle Checkpoints , Temozolomide , Xenograft Model Antitumor Assays , Humans , Animals , Temozolomide/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Mice , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , DNA Damage/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , CRISPR-Cas Systems , Mice, Nude , Cell Line, Tumor , Tumor Cells, Cultured , Drug Resistance, Neoplasm/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , DNA Repair/drug effects
17.
Nucleic Acids Res ; 39(12): 5003-14, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21378120

ABSTRACT

To successfully infect plants, viruses must counteract small RNA-based host defense responses. During infection of Arabidopsis, Cauliflower mosaic pararetrovirus (CaMV) is transcribed into pregenomic 35S and subgenomic 19S RNAs. The 35S RNA is both reverse transcribed and also used as an mRNA with highly structured 600 nt leader. We found that this leader region is transcribed into long sense- and antisense-RNAs and spawns a massive quantity of 21, 22 and 24 nt viral small RNAs (vsRNAs), comparable to the entire complement of host-encoded small-interfering RNAs and microRNAs. Leader-derived vsRNAs were detected bound to the Argonaute 1 (AGO1) effector protein, unlike vsRNAs from other viral regions. Only negligible amounts of leader-derived vsRNAs were bound to AGO4. Genetic evidence showed that all four Dicer-like (DCL) proteins mediate vsRNA biogenesis, whereas the RNA polymerases Pol IV, Pol V, RDR1, RDR2 and RDR6 are not required for this process. Surprisingly, CaMV titers were not increased in dcl1/2/3/4 quadruple mutants that accumulate only residual amounts of vsRNAs. Ectopic expression of CaMV leader vsRNAs from an attenuated geminivirus led to increased accumulation of this chimeric virus. Thus, massive production of leader-derived vsRNAs does not restrict viral replication but may serve as a decoy diverting the silencing machinery from viral promoter and coding regions.


Subject(s)
Arabidopsis/virology , Caulimovirus/genetics , RNA, Small Untranslated/biosynthesis , RNA, Viral/biosynthesis , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Argonaute Proteins , Caulimovirus/physiology , DNA, Viral/biosynthesis , Mutation , Plant Diseases/virology , Ribonuclease III/genetics , Virus Replication
18.
Cell Syst ; 14(5): 418-422.e2, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37201508

ABSTRACT

CRISPR screens are used extensively to systematically interrogate the phenotype-to-genotype problem. In contrast to early CRISPR screens, which defined core cell fitness genes, most current efforts now aim to identify context-specific phenotypes that differentiate a cell line, genetic background, or condition of interest, such as a drug treatment. While CRISPR-related technologies have shown great promise and a fast pace of innovation, a better understanding of standards and methods for quality assessment of CRISPR screen results is crucial to guide technology development and application. Specifically, many commonly used metrics for quantifying screen quality do not accurately measure the reproducibility of context-specific hits. We highlight the importance of reporting reproducibility statistics that directly relate to the purpose of the screen and suggest the use of metrics that are sensitive to context-specific signal. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Reproducibility of Results , Phenotype , Cell Line
19.
Methods Mol Biol ; 2377: 1-27, 2022.
Article in English | MEDLINE | ID: mdl-34709608

ABSTRACT

Human pluripotent stem cells (hPSCs) have the capacity for self-renewal and differentiation into most cell types and, in contrast to widely used cell lines, are karyotypically normal and non-transformed. Hence, hPSCs are considered the gold-standard system for modelling diseases, especially in the field of regenerative medicine. Despite widespread research use of hPSCs and induced pluripotent stem cells (iPSCs), the systematic understanding of pluripotency and lineage differentiation mechanisms are still incomplete. Before tackling the complexities of lineage differentiation with genetic screens, it is critical to catalogue the general genetic requirements for cell fitness and proliferation in the pluripotent state and assess their plasticity under commonly used culture conditions.We describe a method to map essential genetic determinants of hPSC fitness and pluripotency, herein defined as cell reproduction, by genome-scale loss-of-function CRISPR screens in an inducible S. pyogenes Cas9 H1 hPSC line. To address questions of context-dependent gene essentiality, we include protocols for screening hPSCs cultured on feeder cells and laminin, two commonly used growth substrates. This method establishes parameters for genome-wide screens in hPSCs, making human stem cells amenable for functional genomics approaches to facilitate investigation of hPSC biology.


Subject(s)
Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Feeder Cells , Genes, Essential , Humans
20.
Nat Protoc ; 16(10): 4722-4765, 2021 10.
Article in English | MEDLINE | ID: mdl-34508260

ABSTRACT

CRISPR-based forward genetic screening represents a powerful approach for the systematic characterization of gene function. Recent efforts have been directed toward establishing CRISPR-based tools for the programmable delivery of combinatorial genetic perturbations, most of which are mediated by a single nuclease and the expression of structurally identical guide backbones from two promoters. In contrast, we have developed CHyMErA (Cas hybrid for multiplexed editing and screening applications), which is based on the co-expression of Cas9 and Cas12a nucleases in conjunction with a hybrid guide RNA (hgRNA) engineered by the fusion of Cas9 and Cas12a guides and expressed from a single U6 promoter. CHyMErA is suitable for the high-throughput deletion of genetic segments including the excision of individual exons. Furthermore, CHyMErA enables the concomitant targeting of two or more genes and can thus be used for the systematic mapping of genetic interactions in mammalian cells. CHyMErA can also be applied for the perturbation of paralogous gene pairs, thereby allowing the capturing of phenotypic roles that would otherwise be masked because of genetic redundancy. Here, we provide instructions for the cloning of hgRNA screening libraries and individual hgRNA constructs and offer guidelines for designing and performing combinatorial pooled genetic screens using CHyMErA. Starting with the generation of Cas9- and Cas12a-expressing cell lines, CHyMErA screening can be implemented within 15-20 weeks.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Cell Line , Gene Deletion , Humans
SELECTION OF CITATIONS
SEARCH DETAIL