1
|
Graeve FD, Debreuve E, Pushpalatha KV, Zhang X, Rahmoun S, Kozlowski D, Cedilnik N, Vijayakumar J, Cassini P, Schaub S, Descombes X, Besse F. An image-based RNAi screen identifies the EGFR signaling pathway as a regulator of Imp RNP granules. J Cell Sci 2024; 137:jcs262119. [PMID: 39479884 PMCID: PMC11698055 DOI: 10.1242/jcs.262119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
Biomolecular condensates have recently retained much attention given that they provide a fundamental mechanism of cellular organization. Among those, cytoplasmic ribonucleoprotein (RNP) granules selectively and reversibly concentrate RNA molecules and regulatory proteins, thus contributing to the spatiotemporal regulation of associated RNAs. Extensive in vitro work has unraveled the molecular and chemical bases of RNP granule assembly. The signaling pathways controlling this process in a cellular context are, however, still largely unknown. Here, we aimed at identifying regulators of cytoplasmic RNP granules characterized by the presence of the evolutionarily conserved Imp RNA-binding protein (a homolog of IGF2BP proteins). We performed a high-content image-based RNAi screen targeting all Drosophila genes encoding RNA-binding proteins, phosphatases and kinases. This led to the identification of dozens of genes regulating the number of Imp-positive RNP granules in S2R+ cells, among which were components of the MAPK pathway. Combining functional approaches, phospho-mapping and generation of phospho-variants, we further showed that EGFR signaling inhibits Imp-positive RNP granule assembly through activation of the MAPK-ERK pathway and downstream phosphorylation of Imp at the S15 residue. This work illustrates how signaling pathways can regulate cellular condensate assembly by post-translational modifications of specific components.
Collapse
Affiliation(s)
- Fabienne De Graeve
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| | - Eric Debreuve
- Université Côte D'Azur, CNRS, INRIA, I3S, 06902 Sophia Antipolis, France
| | | | - Xuchun Zhang
- Université Côte D'Azur, INRIA, CNRS, I3S, 06902 Sophia Antipolis, France
| | - Somia Rahmoun
- Université Côte D'Azur, INRIA, CNRS, I3S, 06902 Sophia Antipolis, France
| | - Djampa Kozlowski
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| | - Nicolas Cedilnik
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| | - Jeshlee Vijayakumar
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| | - Paul Cassini
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| | - Sebastien Schaub
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
- Université Sorbonne, CNRS, LBDV, 06230 Villefranche-sur-mer, France
| | - Xavier Descombes
- Université Côte D'Azur, INRIA, CNRS, I3S, 06902 Sophia Antipolis, France
| | - Florence Besse
- Université Côte D'Azur, CNRS, Inserm, Institut de Biologie Valrose, 06108 Nice, France
| |
Collapse
|
2
|
Yang C, Lei Y, Ren T, Yao M. The Current Situation and Development Prospect of Whole-Genome Screening. Int J Mol Sci 2024; 25:658. [PMID: 38203828 PMCID: PMC10779205 DOI: 10.3390/ijms25010658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
High-throughput genetic screening is useful for discovering critical genes or gene sequences that trigger specific cell functions and/or phenotypes. Loss-of-function genetic screening is mainly achieved through RNA interference (RNAi), CRISPR knock-out (CRISPRko), and CRISPR interference (CRISPRi) technologies. Gain-of-function genetic screening mainly depends on the overexpression of a cDNA library and CRISPR activation (CRISPRa). Base editing can perform both gain- and loss-of-function genetic screening. This review discusses genetic screening techniques based on Cas9 nuclease, including Cas9-mediated genome knock-out and dCas9-based gene activation and interference. We compare these methods with previous genetic screening techniques based on RNAi and cDNA library overexpression and propose future prospects and applications for CRISPR screening.
Collapse
Affiliation(s)
| | | | | | - Mingze Yao
- Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education and Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (Y.L.); (T.R.)
| |
Collapse
|
3
|
Singh M, Ye B, Kim JH. Dual Leucine Zipper Kinase Regulates Dscam Expression through a Noncanonical Function of the Cytoplasmic Poly(A)-Binding Protein. J Neurosci 2022; 42:6007-6019. [PMID: 35764381 PMCID: PMC9351639 DOI: 10.1523/jneurosci.0543-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Dual leucine zipper kinase (DLK) plays a pivotal role in the development, degeneration, and regeneration of neurons. DLK can regulate gene expression post-transcriptionally, but the underlying mechanism remains poorly understood. The Drosophila DLK, Wallenda (Wnd), regulates the expression of Down syndrome cell adhesion molecule (Dscam) to control presynaptic arbor growth. This regulation is mediated by the 3' untranslated region (3'UTR) of Dscam mRNA, which suggests that RNA binding proteins (RBPs) mediate DLK function. We performed a genome-wide cell-based RNAi screen of RBPs and identified the cytoplasmic poly(A)-binding protein, pAbp, as an RBP that mediates Wnd-induced increase in Dscam expression. Genetic analysis shows that Wnd requires pAbp for promoting presynaptic arbor growth and for enhancing Dscam expression. Our analysis revealed that Dscam mRNAs harbor short poly(A) tails. We identified a region in Dscam 3'UTR that specifically interacts with pAbp. Removing this region significantly reduced Wnd-induced increase in Dscam expression. These suggest that a noncanonical interaction of PABP with the 3'UTR of target transcripts is essential for DLK functions.SIGNIFICANCE STATEMENT The kinase DLK plays key roles in a multitude of neuronal responses, including axon development, neurodegeneration, and nerve injury. Previous studies show that DLK acts via mRNAs to regulate protein synthesis, but how DLK does so is poorly understood. This study demonstrates that DLK regulates the synthesis of Dscam through the poly(A)-binding protein PABP-C. Whereas PABP-C is known as a general translational activator, our study shows that DLK-mediated Dscam expression involves a noncanonical interaction between PABP-C and the Dscam mRNA, which leads to a selective regulation of Dscam translation by PABP-C. Thus, our study provides novel insights into the mechanisms that underlie the function of DLK and regulation of gene expression of PABP-C.
Collapse
Affiliation(s)
- Monika Singh
- Department of Biology, University of Nevada, Reno, Nevada 89557
| | - Bing Ye
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Jung Hwan Kim
- Department of Biology, University of Nevada, Reno, Nevada 89557,
| |
Collapse
|
4
|
Kotian N, Troike KM, Curran KN, Lathia JD, McDonald JA. A Drosophila RNAi screen reveals conserved glioblastoma-related adhesion genes that regulate collective cell migration. G3 GENES|GENOMES|GENETICS 2022; 12:6388037. [PMID: 34849760 PMCID: PMC8728034 DOI: 10.1093/g3journal/jkab356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/06/2021] [Indexed: 11/14/2022]
Abstract
Abstract
Migrating cell collectives are key to embryonic development but also contribute to invasion and metastasis of a variety of cancers. Cell collectives can invade deep into tissues, leading to tumor progression and resistance to therapies. Collective cell invasion is also observed in the lethal brain tumor glioblastoma (GBM), which infiltrates the surrounding brain parenchyma leading to tumor growth and poor patient outcomes. Drosophila border cells, which migrate as a small cell cluster in the developing ovary, are a well-studied and genetically accessible model used to identify general mechanisms that control collective cell migration within native tissue environments. Most cell collectives remain cohesive through a variety of cell–cell adhesion proteins during their migration through tissues and organs. In this study, we first identified cell adhesion, cell matrix, cell junction, and associated regulatory genes that are expressed in human brain tumors. We performed RNAi knockdown of the Drosophila orthologs in border cells to evaluate if migration and/or cohesion of the cluster was impaired. From this screen, we identified eight adhesion-related genes that disrupted border cell collective migration upon RNAi knockdown. Bioinformatics analyses further demonstrated that subsets of the orthologous genes were elevated in the margin and invasive edge of human GBM patient tumors. These data together show that conserved cell adhesion and adhesion regulatory proteins with potential roles in tumor invasion also modulate collective cell migration. This dual screening approach for adhesion genes linked to GBM and border cell migration thus may reveal conserved mechanisms that drive collective tumor cell invasion.
Collapse
Affiliation(s)
- Nirupama Kotian
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Katie M Troike
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kristen N Curran
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
5
|
Umer Z, Akhtar J, Khan MHF, Shaheen N, Haseeb MA, Mazhar K, Mithani A, Anwar S, Tariq M. Genome-wide RNAi screen in Drosophila reveals Enok as a novel trithorax group regulator. Epigenetics Chromatin 2019; 12:55. [PMID: 31547845 PMCID: PMC6757429 DOI: 10.1186/s13072-019-0301-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
Background Polycomb group (PcG) and trithorax group (trxG) proteins contribute to the specialization of cell types by maintaining differential gene expression patterns. Initially discovered as positive regulators of HOX genes in forward genetic screens, trxG counteracts PcG-mediated repression of cell type-specific genes. Despite decades of extensive analysis, molecular understanding of trxG action and regulation are still punctuated by many unknowns. This study aimed at discovering novel factors that elicit an anti-silencing effect to facilitate trxG-mediated gene activation. Results We have developed a cell-based reporter system and performed a genome-wide RNAi screen to discover novel factors involved in trxG-mediated gene regulation in Drosophila. We identified more than 200 genes affecting the reporter in a manner similar to trxG genes. From the list of top candidates, we have characterized Enoki mushroom (Enok), a known histone acetyltransferase, as an important regulator of trxG in Drosophila. Mutants of enok strongly suppressed extra sex comb phenotype of Pc mutants and enhanced homeotic transformations associated with trx mutations. Enok colocalizes with both TRX and PC at chromatin. Moreover, depletion of Enok specifically resulted in an increased enrichment of PC and consequently silencing of trxG targets. This downregulation of trxG targets was also accompanied by a decreased occupancy of RNA-Pol-II in the gene body, correlating with an increased stalling at the transcription start sites of these genes. We propose that Enok facilitates trxG-mediated maintenance of gene activation by specifically counteracting PcG-mediated repression. Conclusion Our ex vivo approach led to identification of new trxG candidate genes that warrant further investigation. Presence of chromatin modifiers as well as known members of trxG and their interactors in the genome-wide RNAi screen validated our reverse genetics approach. Genetic and molecular characterization of Enok revealed a hitherto unknown interplay between Enok and PcG/trxG system. We conclude that histone acetylation by Enok positively impacts the maintenance of trxG-regulated gene activation by inhibiting PRC1-mediated transcriptional repression.
Collapse
Affiliation(s)
- Zain Umer
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Jawad Akhtar
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Haider Farooq Khan
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Najma Shaheen
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Abdul Haseeb
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Khalida Mazhar
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Aziz Mithani
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Saima Anwar
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.,Biomedical Engineering Centre, University of Engineering and Technology Lahore, KSK Campus, Lahore, Pakistan
| | - Muhammad Tariq
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| |
Collapse
|
6
|
Genome-scale CRISPR knockout screen identifies TIGAR as a modifier of PARP inhibitor sensitivity. Commun Biol 2019; 2:335. [PMID: 31508509 PMCID: PMC6733792 DOI: 10.1038/s42003-019-0580-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Treatment of cancer with poly (ADP-ribose) polymerase (PARP) inhibitors is currently limited to cells defective in the homologous recombination (HR) pathway. Identification of genetic targets that induce or mimic HR deficiencies will extend the clinical utility of PARP inhibitors. Here we perform a CRISPR/Cas9-based genome-scale loss-of-function screen, using the sensitivity of PARP inhibitor olaparib as a surrogate. We identify C12orf5, encoding TP53 induced glycolysis and apoptosis regulator (TIGAR), as a modifier of PARP inhibitor response. We show that TIGAR is amplified in several cancer types, and higher expression of TIGAR associates with poor overall survival in ovarian cancer. TIGAR knockdown enhances sensitivity to olaparib in cancer cells via downregulation of BRCA1 and the Fanconi anemia pathway and increases senescence of these cells by affecting metabolic pathways and increasing the cytotoxic effects of olaparib. Our results indicate TIGAR should be explored as a therapeutic target for treating cancer and extending the use of PARP inhibitors. Pingping Fang et al. report a CRISPR/Cas9-based genome-scale loss-of-function screen identifying TIGAR as a modifier of response to PARP inhibition. The authors find that knockdown of TIGAR increases intracellular reactive oxygen species levels, enhances more DNA damage after olaparib treatment, and induces a state of “BRCAness”, suggesting that TIGAR is a potential therapeutic target in ovarian cancer patients.
Collapse
|
7
|
Meltzer H, Marom E, Alyagor I, Mayseless O, Berkun V, Segal-Gilboa N, Unger T, Luginbuhl D, Schuldiner O. Tissue-specific (ts)CRISPR as an efficient strategy for in vivo screening in Drosophila. Nat Commun 2019; 10:2113. [PMID: 31068592 PMCID: PMC6506539 DOI: 10.1038/s41467-019-10140-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022] Open
Abstract
Gene editing by CRISPR/Cas9 is commonly used to generate germline mutations or perform in vitro screens, but applicability for in vivo screening has so far been limited. Recently, it was shown that in Drosophila, Cas9 expression could be limited to a desired group of cells, allowing tissue-specific mutagenesis. Here, we thoroughly characterize tissue-specific (ts)CRISPR within the complex neuronal system of the Drosophila mushroom body. We report the generation of a library of gRNA-expressing plasmids and fly lines using optimized tools, which provides a valuable resource to the fly community. We demonstrate the application of our library in a large-scale in vivo screen, which reveals insights into developmental neuronal remodeling.
Collapse
Affiliation(s)
- Hagar Meltzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Efrat Marom
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Idan Alyagor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oded Mayseless
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Victoria Berkun
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Netta Segal-Gilboa
- Structural Proteomics Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Unger
- Structural Proteomics Unit, Weizmann Institute of Science, Rehovot, Israel
| | - David Luginbuhl
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, USA
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
Mintz RL, Gao MA, Lo K, Lao YH, Li M, Leong KW. CRISPR Technology for Breast Cancer: Diagnostics, Modeling, and Therapy. ADVANCED BIOSYSTEMS 2018; 2:1800132. [PMID: 32832592 PMCID: PMC7437870 DOI: 10.1002/adbi.201800132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Indexed: 12/17/2022]
Abstract
Molecularly, breast cancer represents a highly heterogenous family of neoplastic disorders, with substantial interpatient variations regarding genetic mutations, cell composition, transcriptional profiles, and treatment response. Consequently, there is an increasing demand for alternative diagnostic approaches aimed at the molecular annotation of the disease on a patient-by-patient basis and the design of more personalized treatments. The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) technology enables the development of such novel approaches. For instance, in diagnostics, the use of the RNA-specific C2c2 system allows ultrasensitive nucleic acid detection and could be used to characterize the mutational repertoire and transcriptional breast cancer signatures. In disease modeling, CRISPR/Cas9 technology can be applied to selectively engineer oncogenes and tumor-suppressor genes involved in disease pathogenesis. In treatment, CRISPR/Cas9 can be used to develop gene-therapy, while its catalytically-dead variant (dCas9) can be applied to reprogram the epigenetic landscape of malignant cells. As immunotherapy becomes increasingly prominent in cancer treatment, CRISPR/Cas9 can engineer the immune cells to redirect them against cancer cells and potentiate antitumor immune responses. In this review, CRISPR strategies for the advancement of breast cancer diagnostics, modeling, and treatment are highlighted, culminating in a perspective on developing a precision medicine-based approach against breast cancer.
Collapse
Affiliation(s)
- Rachel L. Mintz
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Madeleine A. Gao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Kahmun Lo
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Mingqiang Li
- Guangdong Provincial Key Laboratory of Liver Disease The Third Affiliated Hospital of Sun Yat-Sen University Guangzhou, Guangdong 510630, China
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Kam W. Leong
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
9
|
An efficient and multiple target transgenic RNAi technique with low toxicity in Drosophila. Nat Commun 2018; 9:4160. [PMID: 30297884 PMCID: PMC6175926 DOI: 10.1038/s41467-018-06537-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/12/2018] [Indexed: 01/19/2023] Open
Abstract
Being relatively simple and practical, Drosophila transgenic RNAi is the technique of top priority choice to quickly study genes with pleiotropic functions. However, drawbacks have emerged over time, such as high level of false positive and negative results. To overcome these shortcomings and increase efficiency, specificity and versatility, we develop a next generation transgenic RNAi system. With this system, the leaky expression of the basal promoter is significantly reduced, as well as the heterozygous ratio of transgenic RNAi flies. In addition, it has been first achieved to precisely and efficiently modulate highly expressed genes. Furthermore, we increase versatility which can simultaneously knock down multiple genes in one step. A case illustration is provided of how this system can be used to study the synthetic developmental effect of histone acetyltransferases. Finally, we have generated a collection of transgenic RNAi lines for those genes that are highly homologous to human disease genes.
Collapse
|
10
|
New tools for old drugs: Functional genetic screens to optimize current chemotherapy. Drug Resist Updat 2018; 36:30-46. [PMID: 29499836 PMCID: PMC5844649 DOI: 10.1016/j.drup.2018.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/29/2017] [Accepted: 01/06/2018] [Indexed: 12/26/2022]
Abstract
Despite substantial advances in the treatment of various cancers, many patients still receive anti-cancer therapies that hardly eradicate tumor cells but inflict considerable side effects. To provide the best treatment regimen for an individual patient, a major goal in molecular oncology is to identify predictive markers for a personalized therapeutic strategy. Regarding novel targeted anti-cancer therapies, there are usually good markers available. Unfortunately, however, targeted therapies alone often result in rather short remissions and little cytotoxic effect on the cancer cells. Therefore, classical chemotherapy with frequent long remissions, cures, and a clear effect on cancer cell eradication remains a corner stone in current anti-cancer therapy. Reliable biomarkers which predict the response of tumors to classical chemotherapy are rare, in contrast to the situation for targeted therapy. For the bulk of cytotoxic therapeutic agents, including DNA-damaging drugs, drugs targeting microtubules or antimetabolites, there are still no reliable biomarkers used in the clinic to predict tumor response. To make progress in this direction, meticulous studies of classical chemotherapeutic drug action and resistance mechanisms are required. For this purpose, novel functional screening technologies have emerged as successful technologies to study chemotherapeutic drug response in a variety of models. They allow a systematic analysis of genetic contributions to a drug-responsive or −sensitive phenotype and facilitate a better understanding of the mode of action of these drugs. These functional genomic approaches are not only useful for the development of novel targeted anti-cancer drugs but may also guide the use of classical chemotherapeutic drugs by deciphering novel mechanisms influencing a tumor’s drug response. Moreover, due to the advances of 3D organoid cultures from patient tumors and in vivo screens in mice, these genetic screens can be applied using conditions that are more representative of the clinical setting. Patient-derived 3D organoid lines furthermore allow the characterization of the “essentialome”, the specific set of genes required for survival of these cells, of an individual tumor, which could be monitored over the course of treatment and help understanding how drug resistance evolves in clinical tumors. Thus, we expect that these functional screens will enable the discovery of novel cancer-specific vulnerabilities, and through clinical validation, move the field of predictive biomarkers forward. This review focuses on novel advanced techniques to decipher the interplay between genetic alterations and drug response.
Collapse
|
11
|
Multiplex shRNA Screening of Germ Cell Development by in Vivo Transfection of Mouse Testis. G3-GENES GENOMES GENETICS 2017; 7:247-255. [PMID: 27856695 PMCID: PMC5217113 DOI: 10.1534/g3.116.036087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Spermatozoa are one of the few mammalian cell types that cannot be fully derived in vitro, severely limiting the application of modern genomic techniques to study germ cell biology. The current gold standard approach of characterizing single-gene knockout mice is slow as generation of each mutant line can take 6–9 months. Here, we describe an in vivo approach to rapid functional screening of germline genes based on a new nonsurgical, nonviral in vivo transfection method to deliver nucleic acids into testicular germ cells. By coupling multiplex transfection of short hairpin RNA (shRNA) constructs with pooled amplicon sequencing as a readout, we were able to screen many genes for spermatogenesis function in a quick and inexpensive experiment. We transfected nine mouse testes with a pilot pool of RNA interference (RNAi) against well-characterized genes to show that this system is highly reproducible and accurate. With a false negative rate of 18% and a false positive rate of 12%, this method has similar performance as other RNAi screens in the well-described Drosophila model system. In a separate experiment, we screened 26 uncharacterized genes computationally predicted to be essential for spermatogenesis and found numerous candidates for follow-up studies. Finally, as a control experiment, we performed a long-term selection screen in neuronal N2a cells, sampling shRNA frequencies at five sequential time points. By characterizing the effect of both libraries on N2a cells, we show that our screening results from testis are tissue-specific. Our calculations indicate that the current implementation of this approach could be used to screen thousands of protein-coding genes simultaneously in a single mouse testis. The experimental protocols and analysis scripts provided will enable other groups to use this procedure to study diverse aspects of germ cell biology ranging from epigenetics to cell physiology. This approach also has great promise as an applied tool for validating diagnoses made from medical genome sequencing, or designing synthetic biological sequences that can act as potent and highly specific male contraceptives.
Collapse
|
12
|
Park HR, Oh R, Wagner P, Panganiban R, Lu Q. New Insights Into Cellular Stress Responses to Environmental Metal Toxicants. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 331:55-82. [PMID: 28325215 DOI: 10.1016/bs.ircmb.2016.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exposures to metal toxicants in the environment disrupt normal physiological functions and have been linked to the development of a myriad of human diseases. While the molecular and cellular mechanisms underlying metal toxicities remain to be fully understood, it is well appreciated that metal toxicants induce cellular stresses and that how cells respond to the stresses plays an important role in metal toxicity. In this review, we focus on how metal exposures induce stresses in the endoplasmic reticulum (ER) to elicit the unfolded protein response (UPR). We document the emerging evidence that induction of ER stress and UPR in the development of human diseases is associated with metal exposures. We also discuss the role of the interplay between ER stress and oxidative stress in metal toxicity. Finally, we review recent advances in functional genomics approaches and discuss how applications of these new tools could help elucidate the molecular mechanisms underlying cellular stresses induced by environmental metal toxicants.
Collapse
Affiliation(s)
- H-R Park
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - R Oh
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - P Wagner
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - R Panganiban
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Q Lu
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
13
|
Howard TP, Vazquez F, Tsherniak A, Hong AL, Rinne M, Aguirre AJ, Boehm JS, Hahn WC. Functional Genomic Characterization of Cancer Genomes. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2016; 81:237-246. [PMID: 27815544 DOI: 10.1101/sqb.2016.81.031070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
International efforts to sequence cancer genomes now provide an overview of the major genetic alterations that occur in most human cancers. These studies have identified many highly recurrent alterations in specific cancer subtypes but have also identified mutations that occur at lower frequency and unstudied variants of known cancer-associated genes. To elucidate the function of such cancer alleles, we have developed several approaches to systematically interrogate genomic changes found in human tumors. In general, we have taken two complementary approaches. In the first approach, we focus on perturbing genes identified as mutated, amplified, or deleted by cancer genome annotation efforts, whereas in the second, we have taken an unbiased approach to identify genes that are essential for cancer cell proliferation or survival in cell lines that are extensively annotated to identify context-specific essential genes. These studies begin to allow us to define a cancer dependencies map.
Collapse
Affiliation(s)
- Thomas P Howard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - Francisca Vazquez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - Aviad Tsherniak
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - Andrew L Hong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142.,Boston Children's Hospital, Boston, Massachusetts 02115
| | - Mik Rinne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - Jesse S Boehm
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| |
Collapse
|
14
|
Cloud-Richardson KM, Smith BR, Macdonald SJ. Genetic dissection of intraspecific variation in a male-specific sexual trait in Drosophila melanogaster. Heredity (Edinb) 2016; 117:417-426. [PMID: 27530909 PMCID: PMC5117841 DOI: 10.1038/hdy.2016.63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 06/07/2016] [Accepted: 06/22/2016] [Indexed: 01/06/2023] Open
Abstract
An open question in evolutionary biology is the relationship between standing variation for a trait and the variation that leads to interspecific divergence. By identifying loci underlying phenotypic variation in intra- and interspecific crosses we can determine the extent to which polymorphism and divergence are controlled by the same genomic regions. Sexual traits provide abundant examples of morphological and behavioral diversity within and among species, and here we leverage variation in the Drosophila sex comb to address this question. The sex comb is an array of modified bristles or ‘teeth' present on the male forelegs of several Drosophilid species. Males use the comb to grasp females during copulation, and ablation experiments have shown that males lacking comb teeth typically fail to mate. We measured tooth number in >700 genotypes derived from a multiparental advanced-intercross population, mapping three moderate-effect loci contributing to trait heritability. Two quantitative trait loci (QTLs) coincide with previously identified intra- and interspecific sex comb QTL, but such overlap can be explained by chance alone, in part because of the broad swathes of the genome implicated by earlier, low-resolution QTL scans. Our mapped QTL regions encompass 70–124 genes, but do not include those genes known to be involved in developmental specification of the comb. Nonetheless, we identified plausible candidates within all QTL intervals, and used RNA interference to validate effects at four loci. Notably, TweedleS expression knockdown substantially reduces tooth number. The genes we highlight are strong candidates to harbor segregating, functional variants contributing to sex comb tooth number.
Collapse
Affiliation(s)
| | - B R Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - S J Macdonald
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA.,Center for Computational Biology, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
15
|
Functional Genomic Strategies for Elucidating Human-Virus Interactions: Will CRISPR Knockout RNAi and Haploid Cells? Adv Virus Res 2016; 94:1-51. [PMID: 26997589 PMCID: PMC7112329 DOI: 10.1016/bs.aivir.2015.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Over the last several years a wealth of transformative human–virus interaction discoveries have been produced using loss-of-function functional genomics. These insights have greatly expanded our understanding of how human pathogenic viruses exploit our cells to replicate. Two technologies have been at the forefront of this genetic revolution, RNA interference (RNAi) and random retroviral insertional mutagenesis using haploid cell lines (haploid cell screening), with the former technology largely predominating. Now the cutting edge gene editing of the CRISPR/Cas9 system has also been harnessed for large-scale functional genomics and is poised to possibly displace these earlier methods. Here we compare and contrast these three screening approaches for elucidating host–virus interactions, outline their key strengths and weaknesses including a comparison of an arrayed multiple orthologous RNAi reagent screen to a pooled CRISPR/Cas9 human rhinovirus 14–human cell interaction screen, and recount some notable insights made possible by each. We conclude with a brief perspective on what might lie ahead for the fast evolving field of human–virus functional genomics.
Collapse
|
16
|
Abstract
The orchestrated action of genes controls complex biological phenotypes, yet the systematic discovery of gene and drug combinations that modulate these phenotypes in human cells is labor intensive and challenging to scale. Here, we created a platform for the massively parallel screening of barcoded combinatorial gene perturbations in human cells and translated these hits into effective drug combinations. This technology leverages the simplicity of the CRISPR-Cas9 system for multiplexed targeting of specific genomic loci and the versatility of combinatorial genetics en masse (CombiGEM) to rapidly assemble barcoded combinatorial genetic libraries that can be tracked with high-throughput sequencing. We applied CombiGEM-CRISPR to create a library of 23,409 barcoded dual guide-RNA (gRNA) combinations and then perform a high-throughput pooled screen to identify gene pairs that inhibited ovarian cancer cell growth when they were targeted. We validated the growth-inhibiting effects of specific gene sets, including epigenetic regulators KDM4C/BRD4 and KDM6B/BRD4, via individual assays with CRISPR-Cas-based knockouts and RNA-interference-based knockdowns. We also tested small-molecule drug pairs directed against our pairwise hits and showed that they exerted synergistic antiproliferative effects against ovarian cancer cells. We envision that the CombiGEM-CRISPR platform will be applicable to a broad range of biological settings and will accelerate the systematic identification of genetic combinations and their translation into novel drug combinations that modulate complex human disease phenotypes.
Collapse
|
17
|
Reagent and Data Resources for Investigation of RNA Binding Protein Functions in Drosophila melanogaster Cultured Cells. G3-GENES GENOMES GENETICS 2015. [PMID: 26199285 PMCID: PMC4555228 DOI: 10.1534/g3.115.019364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RNA binding proteins (RBPs) are involved in many cellular functions. To facilitate functional characterization of RBPs, we generated an RNA interference (RNAi) library for Drosophila cell-based screens comprising reagents targeting known or putative RBPs. To test the quality of the library and provide a baseline analysis of the effects of the RNAi reagents on viability, we screened the library using a total ATP assay and high-throughput imaging in Drosophila S2R+ cultured cells. The results are consistent with production of a high-quality library that will be useful for functional genomics studies using other assays. Altogether, we provide resources in the form of an initial curated list of Drosophila RBPs; an RNAi screening library we expect to be used with additional assays that address more specific biological questions; and total ATP and image data useful for comparison of those additional assay results with fundamental information such as effects of a given reagent in the library on cell viability. Importantly, we make the baseline data, including more than 200,000 images, easily accessible online.
Collapse
|
18
|
Humphrey SE, Kasinski AL. RNA-guided CRISPR-Cas technologies for genome-scale investigation of disease processes. J Hematol Oncol 2015; 8:31. [PMID: 25888285 PMCID: PMC4389696 DOI: 10.1186/s13045-015-0127-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/14/2015] [Indexed: 12/26/2022] Open
Abstract
From its discovery as an adaptive bacterial and archaea immune system, the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system has quickly been developed into a powerful and groundbreaking programmable nuclease technology for the global and precise editing of the genome in cells. This system allows for comprehensive unbiased functional studies and is already advancing the field by revealing genes that have previously unknown roles in disease processes. In this review, we examine and compare recently developed CRISPR-Cas platforms for global genome editing and examine the advancements these platforms have made in guide RNA design, guide RNA/Cas9 interaction, on-target specificity, and target sequence selection. We also explore some of the exciting therapeutic potentials of the CRISPR-Cas technology as well as some of the innovative new uses of this technology beyond genome editing.
Collapse
Affiliation(s)
- Sean E Humphrey
- Department of Biological Sciences, Purdue University, 1203 West State Street, West Lafayette, IN, 47907, USA.
| | - Andrea L Kasinski
- Department of Biological Sciences, Purdue University, 1203 West State Street, West Lafayette, IN, 47907, USA.
| |
Collapse
|
19
|
Shen H, McHale CM, Smith MT, Zhang L. Functional genomic screening approaches in mechanistic toxicology and potential future applications of CRISPR-Cas9. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 764:31-42. [PMID: 26041264 DOI: 10.1016/j.mrrev.2015.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 01/25/2023]
Abstract
Characterizing variability in the extent and nature of responses to environmental exposures is a critical aspect of human health risk assessment. Chemical toxicants act by many different mechanisms, however, and the genes involved in adverse outcome pathways (AOPs) and AOP networks are not yet characterized. Functional genomic approaches can reveal both toxicity pathways and susceptibility genes, through knockdown or knockout of all non-essential genes in a cell of interest, and identification of genes associated with a toxicity phenotype following toxicant exposure. Screening approaches in yeast and human near-haploid leukemic KBM7 cells have identified roles for genes and pathways involved in response to many toxicants but are limited by partial homology among yeast and human genes and limited relevance to normal diploid cells. RNA interference (RNAi) suppresses mRNA expression level but is limited by off-target effects (OTEs) and incomplete knockdown. The recently developed gene editing approach called clustered regularly interspaced short palindrome repeats-associated nuclease (CRISPR)-Cas9, can precisely knock-out most regions of the genome at the DNA level with fewer OTEs than RNAi, in multiple human cell types, thus overcoming the limitations of the other approaches. It has been used to identify genes involved in the response to chemical and microbial toxicants in several human cell types and could readily be extended to the systematic screening of large numbers of environmental chemicals. CRISPR-Cas9 can also repress and activate gene expression, including that of non-coding RNA, with near-saturation, thus offering the potential to more fully characterize AOPs and AOP networks. Finally, CRISPR-Cas9 can generate complex animal models in which to conduct preclinical toxicity testing at the level of individual genotypes or haplotypes. Therefore, CRISPR-Cas9 is a powerful and flexible functional genomic screening approach that can be harnessed to provide unprecedented mechanistic insight in the field of modern toxicology.
Collapse
Affiliation(s)
- Hua Shen
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Cliona M McHale
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Martyn T Smith
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Luoping Zhang
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
20
|
Abstract
The power of Drosophila melanogaster as a model organism lies in its ability to be used for large-scale genetic screens with the capacity to uncover the genetic basis of biological processes. In particular, genetic screens for circadian behavior, which have been performed since 1971, allowed researchers to make groundbreaking discoveries on multiple levels: they discovered that there is a genetic basis for circadian behavior, they identified the so-called core clock genes that govern this process, and they started to paint a detailed picture of the molecular functions of these clock genes and their encoded proteins. Since the discovery that fruit flies sleep in 2000, researchers have successfully been using genetic screening to elucidate the many questions surrounding this basic animal behavior. In this chapter, we briefly recall the history of circadian rhythm and sleep screens and then move on to describe techniques currently employed for mutagenesis and genetic screening in the field. The emphasis lies on comparing the newer approaches of transgenic RNA interference (RNAi) to classical forms of mutagenesis, in particular in their application to circadian behavior and sleep. We discuss the different screening approaches in light of the literature and published and unpublished sleep and rhythm screens utilizing ethyl methanesulfonate mutagenesis and transgenic RNAi from our lab.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, USA
| | - Lino Saez
- Laboratory of Genetics, The Rockefeller University, New York, USA
| | - Michael W Young
- Laboratory of Genetics, The Rockefeller University, New York, USA.
| |
Collapse
|
21
|
Malty RH, Jessulat M, Jin K, Musso G, Vlasblom J, Phanse S, Zhang Z, Babu M. Mitochondrial targets for pharmacological intervention in human disease. J Proteome Res 2014; 14:5-21. [PMID: 25367773 PMCID: PMC4286170 DOI: 10.1021/pr500813f] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Over the past several years, mitochondrial
dysfunction has been
linked to an increasing number of human illnesses, making mitochondrial
proteins (MPs) an ever more appealing target for therapeutic intervention.
With 20% of the mitochondrial proteome (312 of an estimated 1500 MPs)
having known interactions with small molecules, MPs appear to be highly
targetable. Yet, despite these targeted proteins functioning in a
range of biological processes (including induction of apoptosis, calcium
homeostasis, and metabolism), very few of the compounds targeting
MPs find clinical use. Recent work has greatly expanded the number
of proteins known to localize to the mitochondria and has generated
a considerable increase in MP 3D structures available in public databases,
allowing experimental screening and in silico prediction of mitochondrial
drug targets on an unprecedented scale. Here, we summarize the current
literature on clinically active drugs that target MPs, with a focus
on how existing drug targets are distributed across biochemical pathways
and organelle substructures. Also, we examine current strategies for
mitochondrial drug discovery, focusing on genetic, proteomic, and
chemogenomic assays, and relevant model systems. As cell models and
screening techniques improve, MPs appear poised to emerge as relevant
targets for a wide range of complex human diseases, an eventuality
that can be expedited through systematic analysis of MP function.
Collapse
Affiliation(s)
- Ramy H Malty
- Department of Biochemistry, Research and Innovation Centre, University of Regina , Regina, Saskatchewan S4S 0A2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hériché JK, Lees JG, Morilla I, Walter T, Petrova B, Roberti MJ, Hossain MJ, Adler P, Fernández JM, Krallinger M, Haering CH, Vilo J, Valencia A, Ranea JA, Orengo C, Ellenberg J. Integration of biological data by kernels on graph nodes allows prediction of new genes involved in mitotic chromosome condensation. Mol Biol Cell 2014; 25:2522-36. [PMID: 24943848 PMCID: PMC4142622 DOI: 10.1091/mbc.e13-04-0221] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The advent of genome-wide RNA interference (RNAi)-based screens puts us in the position to identify genes for all functions human cells carry out. However, for many functions, assay complexity and cost make genome-scale knockdown experiments impossible. Methods to predict genes required for cell functions are therefore needed to focus RNAi screens from the whole genome on the most likely candidates. Although different bioinformatics tools for gene function prediction exist, they lack experimental validation and are therefore rarely used by experimentalists. To address this, we developed an effective computational gene selection strategy that represents public data about genes as graphs and then analyzes these graphs using kernels on graph nodes to predict functional relationships. To demonstrate its performance, we predicted human genes required for a poorly understood cellular function-mitotic chromosome condensation-and experimentally validated the top 100 candidates with a focused RNAi screen by automated microscopy. Quantitative analysis of the images demonstrated that the candidates were indeed strongly enriched in condensation genes, including the discovery of several new factors. By combining bioinformatics prediction with experimental validation, our study shows that kernels on graph nodes are powerful tools to integrate public biological data and predict genes involved in cellular functions of interest.
Collapse
Affiliation(s)
- Jean-Karim Hériché
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Jon G Lees
- Research Department of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
| | - Ian Morilla
- Department of Molecular Biology and Biochemistry-CIBER de Enfermedades Raras, University of Malaga, Malaga 29071, Spain
| | - Thomas Walter
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Boryana Petrova
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - M Julia Roberti
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - M Julius Hossain
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Priit Adler
- Institute of Molecular and Cell Biology, University of Tartu, 51010 Tartu, Estonia
| | - José M Fernández
- Structural Bioinformatics Group, Spanish National Cancer Research Centre and Spanish National Bioinformatics Institute, 28029 Madrid, Spain
| | - Martin Krallinger
- Structural Bioinformatics Group, Spanish National Cancer Research Centre and Spanish National Bioinformatics Institute, 28029 Madrid, Spain
| | - Christian H Haering
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, 50409 Tartu, Estonia
| | - Alfonso Valencia
- Structural Bioinformatics Group, Spanish National Cancer Research Centre and Spanish National Bioinformatics Institute, 28029 Madrid, Spain
| | - Juan A Ranea
- Department of Molecular Biology and Biochemistry-CIBER de Enfermedades Raras, University of Malaga, Malaga 29071, Spain
| | - Christine Orengo
- Research Department of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
| | - Jan Ellenberg
- Cell Biology/Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| |
Collapse
|
23
|
Mohr SE. RNAi screening in Drosophila cells and in vivo. Methods 2014; 68:82-8. [PMID: 24576618 DOI: 10.1016/j.ymeth.2014.02.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 12/31/2022] Open
Abstract
Here, I discuss how RNAi screening can be used effectively to uncover gene function. Specifically, I discuss the types of high-throughput assays that can be done in Drosophila cells and in vivo, RNAi reagent design and available reagent collections, automated screen pipelines, analysis of screen results, and approaches to RNAi results verification.
Collapse
Affiliation(s)
- Stephanie E Mohr
- Drosophila RNAi Screening Center, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States.
| |
Collapse
|
24
|
Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science 2013; 343:80-4. [PMID: 24336569 DOI: 10.1126/science.1246981] [Citation(s) in RCA: 2040] [Impact Index Per Article: 170.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The bacterial clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system for genome editing has greatly expanded the toolbox for mammalian genetics, enabling the rapid generation of isogenic cell lines and mice with modified alleles. Here, we describe a pooled, loss-of-function genetic screening approach suitable for both positive and negative selection that uses a genome-scale lentiviral single-guide RNA (sgRNA) library. sgRNA expression cassettes were stably integrated into the genome, which enabled a complex mutant pool to be tracked by massively parallel sequencing. We used a library containing 73,000 sgRNAs to generate knockout collections and performed screens in two human cell lines. A screen for resistance to the nucleotide analog 6-thioguanine identified all expected members of the DNA mismatch repair pathway, whereas another for the DNA topoisomerase II (TOP2A) poison etoposide identified TOP2A, as expected, and also cyclin-dependent kinase 6, CDK6. A negative selection screen for essential genes identified numerous gene sets corresponding to fundamental processes. Last, we show that sgRNA efficiency is associated with specific sequence motifs, enabling the prediction of more effective sgRNAs. Collectively, these results establish Cas9/sgRNA screens as a powerful tool for systematic genetic analysis in mammalian cells.
Collapse
Affiliation(s)
- Tim Wang
- Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
25
|
In Vivo RNAi-Based Screens: Studies in Model Organisms. Genes (Basel) 2013; 4:646-65. [PMID: 24705267 PMCID: PMC3927573 DOI: 10.3390/genes4040646] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 10/29/2013] [Accepted: 11/14/2013] [Indexed: 11/23/2022] Open
Abstract
RNA interference (RNAi) is a technique widely used for gene silencing in organisms and cultured cells, and depends on sequence homology between double-stranded RNA (dsRNA) and target mRNA molecules. Numerous cell-based genome-wide screens have successfully identified novel genes involved in various biological processes, including signal transduction, cell viability/death, and cell morphology. However, cell-based screens cannot address cellular processes such as development, behavior, and immunity. Drosophila and Caenorhabditis elegans are two model organisms whose whole bodies and individual body parts have been subjected to RNAi-based genome-wide screening. Moreover, Drosophila RNAi allows the manipulation of gene function in a spatiotemporal manner when it is implemented using the Gal4/UAS system. Using this inducible RNAi technique, various large-scale screens have been performed in Drosophila, demonstrating that the method is straightforward and valuable. However, accumulated results reveal that the results of RNAi-based screens have relatively high levels of error, such as false positives and negatives. Here, we review in vivo RNAi screens in Drosophila and the methods that could be used to remove ambiguity from screening results.
Collapse
|
26
|
Binder M, Sulaimanov N, Clausznitzer D, Schulze M, Hüber CM, Lenz SM, Schlöder JP, Trippler M, Bartenschlager R, Lohmann V, Kaderali L. Replication vesicles are load- and choke-points in the hepatitis C virus lifecycle. PLoS Pathog 2013; 9:e1003561. [PMID: 23990783 PMCID: PMC3749965 DOI: 10.1371/journal.ppat.1003561] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/02/2013] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) infection develops into chronicity in 80% of all patients, characterized by persistent low-level replication. To understand how the virus establishes its tightly controlled intracellular RNA replication cycle, we developed the first detailed mathematical model of the initial dynamic phase of the intracellular HCV RNA replication. We therefore quantitatively measured viral RNA and protein translation upon synchronous delivery of viral genomes to host cells, and thoroughly validated the model using additional, independent experiments. Model analysis was used to predict the efficacy of different classes of inhibitors and identified sensitive substeps of replication that could be targeted by current and future therapeutics. A protective replication compartment proved to be essential for sustained RNA replication, balancing translation versus replication and thus effectively limiting RNA amplification. The model predicts that host factors involved in the formation of this compartment determine cellular permissiveness to HCV replication. In gene expression profiling, we identified several key processes potentially determining cellular HCV replication efficiency. Hepatitis C is a severe disease and a prime cause for liver transplantation. Up to 3% of the world's population are chronically infected with its causative agent, the Hepatitis C virus (HCV). This capacity to establish long (decades) lasting persistent infection sets HCV apart from other plus-strand RNA viruses typically causing acute, self-limiting infections. A prerequisite for its capacity to persist is HCV's complex and tightly regulated intracellular replication strategy. In this study, we therefore wanted to develop a comprehensive understanding of the molecular processes governing HCV RNA replication in order to pinpoint the most vulnerable substeps in the viral life cycle. For that purpose, we used a combination of biological experiments and mathematical modeling. Using the model to study HCV's replication strategy, we recognized diverse but crucial roles for the membraneous replication compartment of HCV in regulating RNA amplification. We further predict the existence of an essential limiting host factor (or function) required for establishing active RNA replication and thereby determining cellular permissiveness for HCV. Our model also proved valuable to understand and predict the effects of pharmacological inhibitors of HCV and might be a solid basis for the development of similar models for other plus-strand RNA viruses.
Collapse
Affiliation(s)
- Marco Binder
- Heidelberg University, Medical Faculty, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
| | - Nurgazy Sulaimanov
- Technische Universität Dresden, Institute for Medical Informatics and Biometry, Dresden, Germany
- Heidelberg University, ViroQuant Research Group Modeling, BioQuant BQ26, Heidelberg, Germany
| | - Diana Clausznitzer
- Technische Universität Dresden, Institute for Medical Informatics and Biometry, Dresden, Germany
| | - Manuel Schulze
- Technische Universität Dresden, Institute for Medical Informatics and Biometry, Dresden, Germany
| | - Christian M. Hüber
- Heidelberg University, Medical Faculty, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
| | - Simon M. Lenz
- Heidelberg University, Interdisciplinary Center for Scientific Computing (IWR), Simulation and Optimization Group, Heidelberg, Germany
| | - Johannes P. Schlöder
- Heidelberg University, Interdisciplinary Center for Scientific Computing (IWR), Simulation and Optimization Group, Heidelberg, Germany
| | - Martin Trippler
- University Hospital of Essen, Department of Gastroenterology and Hepatology, Essen, Germany
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
| | - Volker Lohmann
- Heidelberg University, Medical Faculty, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
| | - Lars Kaderali
- Technische Universität Dresden, Institute for Medical Informatics and Biometry, Dresden, Germany
- Heidelberg University, ViroQuant Research Group Modeling, BioQuant BQ26, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
27
|
Iyer EPR, Iyer SC, Sullivan L, Wang D, Meduri R, Graybeal LL, Cox DN. Functional genomic analyses of two morphologically distinct classes of Drosophila sensory neurons: post-mitotic roles of transcription factors in dendritic patterning. PLoS One 2013; 8:e72434. [PMID: 23977298 PMCID: PMC3744488 DOI: 10.1371/journal.pone.0072434] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/10/2013] [Indexed: 11/19/2022] Open
Abstract
Background Neurons are one of the most structurally and functionally diverse cell types found in nature, owing in large part to their unique class specific dendritic architectures. Dendrites, being highly specialized in receiving and processing neuronal signals, play a key role in the formation of functional neural circuits. Hence, in order to understand the emergence and assembly of a complex nervous system, it is critical to understand the molecular mechanisms that direct class specific dendritogenesis. Methodology/Principal Findings We have used the Drosophila dendritic arborization (da) neurons to gain systems-level insight into dendritogenesis by a comparative study of the morphologically distinct Class-I (C-I) and Class-IV (C-IV) da neurons. We have used a combination of cell-type specific transcriptional expression profiling coupled to a targeted and systematic in vivo RNAi functional validation screen. Our comparative transcriptomic analyses have revealed a large number of differentially enriched/depleted gene-sets between C-I and C-IV neurons, including a broad range of molecular factors and biological processes such as proteolytic and metabolic pathways. Further, using this data, we have identified and validated the role of 37 transcription factors in regulating class specific dendrite development using in vivo class-specific RNAi knockdowns followed by rigorous and quantitative neurometric analysis. Conclusions/Significance This study reports the first global gene-expression profiles from purified Drosophila C-I and C-IV da neurons. We also report the first large-scale semi-automated reconstruction of over 4,900 da neurons, which were used to quantitatively validate the RNAi screen phenotypes. Overall, these analyses shed global and unbiased novel insights into the molecular differences that underlie the morphological diversity of distinct neuronal cell-types. Furthermore, our class-specific gene expression datasets should prove a valuable community resource in guiding further investigations designed to explore the molecular mechanisms underlying class specific neuronal patterning.
Collapse
Affiliation(s)
- Eswar Prasad R. Iyer
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Srividya Chandramouli Iyer
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Luis Sullivan
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Dennis Wang
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Ramakrishna Meduri
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Lacey L. Graybeal
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Daniel N. Cox
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
28
|
Fisher KH, Brown S, Zeidler MP. Designing RNAi screens to identify JAK/STAT pathway components. Methods Mol Biol 2013; 967:81-97. [PMID: 23296723 DOI: 10.1007/978-1-62703-242-1_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The JAK/STAT signaling pathway has essential roles in multiple developmental processes, including stem cell maintenance, immune responses, and cellular proliferation. As a result, it has been extensively studied in both vertebrate systems and lower complexity models, such as Drosophila. Given its connection with such a wide range of biological functions, it is no surprise that pathway misregulation is frequently associated with multiple human diseases including cancer. While the core components of the pathway, and a number of negative regulators, are well known and conserved in many organisms, more subtle levels of regulation and inter-pathway crosstalk are less well understood. With the emergence of RNA interference (RNAi) as a tool to knock down gene expression and so evaluate protein function, high-throughput screens have been developed to identify pathway regulators on a genome-wide scale. Here we discuss the approaches and methods employed thus far for identification of pathway regulators using RNAi in Drosophila. Furthermore, we discuss possible approaches for future screens and the significant potential for applying RNAi technology in vertebrate models.
Collapse
Affiliation(s)
- Katherine H Fisher
- MRC Centre of Developmental Biology and Genetics, Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, UK
| | | | | |
Collapse
|
29
|
St Johnston D. Using mutants, knockdowns, and transgenesis to investigate gene function in Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:587-613. [PMID: 24014449 DOI: 10.1002/wdev.101] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The sophisticated genetic techniques available in Drosophila are largely responsible for its success as a model organism. One of the most important of these is the ability to disrupt gene function in vivo and observe the resulting phenotypes. This review considers the ever-increasing repertoire of approaches for perturbing the functions of specific genes in flies, ranging from classical and transposon-mediated mutageneses to newer techniques, such as homologous recombination and RNA interference. Since most genes are used over and over again in different contexts during development, many important advances have depended on being able to interfere with gene function at specific times or places in the developing animal, and a variety of approaches are now available to do this. Most of these techniques rely on being able to create genetically modified strains of Drosophila and the different methods for generating lines carrying single copy transgenic constructs will be described, along with the advantages and disadvantages of each approach.
Collapse
Affiliation(s)
- Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge CB2 1QN, UK.
| |
Collapse
|
30
|
On the role of PDZ domain-encoding genes in Drosophila border cell migration. G3-GENES GENOMES GENETICS 2012; 2:1379-91. [PMID: 23173089 PMCID: PMC3484668 DOI: 10.1534/g3.112.004093] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 09/10/2012] [Indexed: 12/31/2022]
Abstract
Cells often move as collective groups during normal embryonic development and wound healing, although the mechanisms governing this type of migration are poorly understood. The Drosophila melanogaster border cells migrate as a cluster during late oogenesis and serve as a powerful in vivo genetic model for collective cell migration. To discover new genes that participate in border cell migration, 64 out of 66 genes that encode PDZ domain-containing proteins were systematically targeted by in vivo RNAi knockdown. The PDZ domain is one of the largest families of protein-protein interaction domains found in eukaryotes. Proteins that contain PDZ domains participate in a variety of biological processes, including signal transduction and establishment of epithelial apical-basal polarity. Targeting PDZ proteins effectively assesses a larger number of genes via the protein complexes and pathways through which these proteins function. par-6, a known regulator of border cell migration, was a positive hit and thus validated the approach. Knockdown of 14 PDZ domain genes disrupted migration with multiple RNAi lines. The candidate genes have diverse predicted cellular functions and are anticipated to provide new insights into the mechanisms that control border cell movement. As a test of this concept, two genes that disrupted migration were characterized in more detail: big bang and the Dlg5 homolog CG6509. We present evidence that Big bang regulates JAK/STAT signaling, whereas Dlg5/CG6509 maintains cluster cohesion. Moreover, these results demonstrate that targeting a selected class of genes by RNAi can uncover novel regulators of collective cell migration.
Collapse
|
31
|
Fisher KH, Wright VM, Taylor A, Zeidler MP, Brown S. Advances in genome-wide RNAi cellular screens: a case study using the Drosophila JAK/STAT pathway. BMC Genomics 2012; 13:506. [PMID: 23006893 PMCID: PMC3526451 DOI: 10.1186/1471-2164-13-506] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 09/12/2012] [Indexed: 12/01/2022] Open
Abstract
Background Genome-scale RNA-interference (RNAi) screens are becoming ever more common gene discovery tools. However, whilst every screen identifies interacting genes, less attention has been given to how factors such as library design and post-screening bioinformatics may be effecting the data generated. Results Here we present a new genome-wide RNAi screen of the Drosophila JAK/STAT signalling pathway undertaken in the Sheffield RNAi Screening Facility (SRSF). This screen was carried out using a second-generation, computationally optimised dsRNA library and analysed using current methods and bioinformatic tools. To examine advances in RNAi screening technology, we compare this screen to a biologically very similar screen undertaken in 2005 with a first-generation library. Both screens used the same cell line, reporters and experimental design, with the SRSF screen identifying 42 putative regulators of JAK/STAT signalling, 22 of which verified in a secondary screen and 16 verified with an independent probe design. Following reanalysis of the original screen data, comparisons of the two gene lists allows us to make estimates of false discovery rates in the SRSF data and to conduct an assessment of off-target effects (OTEs) associated with both libraries. We discuss the differences and similarities between the resulting data sets and examine the relative improvements in gene discovery protocols. Conclusions Our work represents one of the first direct comparisons between first- and second-generation libraries and shows that modern library designs together with methodological advances have had a significant influence on genome-scale RNAi screens.
Collapse
Affiliation(s)
- Katherine H Fisher
- The MRC Centre for Developmental and Biomedical Genetics and The Department of Biomedical Science, University of Sheffield, Firth Court,Western Bank, Sheffield S10 2TN, UK
| | | | | | | | | |
Collapse
|
32
|
Humphreys D, Liu T, Davidson AC, Hume PJ, Koronakis V. The Drosophila Arf1 homologue Arf79F is essential for lamellipodium formation. J Cell Sci 2012; 125:5630-5. [PMID: 22992458 DOI: 10.1242/jcs.108092] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The WAVE regulatory complex (WRC) drives the polymerisation of actin filaments located beneath the plasma membrane to generate lamellipodia that are pivotal to cell architecture and movement. By reconstituting WRC-dependent actin assembly at the membrane, we recently discovered that several classes of Arf family GTPases directly recruit and activate WRC in cell extracts, and that Arf cooperates with Rac1 to trigger actin polymerisation. Here, we demonstrate that the Class 1 Arf1 homologue Arf79F colocalises with the WRC at dynamic lamellipodia. We report that Arf79F is required for lamellipodium formation in Drosophila S2R+ cells, which only express one Arf isoform for each class. Impeding Arf function either by dominant-negative Arf expression or by Arf double-stranded RNA interference (dsRNAi)-mediated knockdown uncovered that Arf-dependent lamellipodium formation was specific to Arf79F, establishing that Class 1 Arfs, but not Class 2 or Class 3 Arfs, are crucial for lamellipodia. Lamellipodium formation in Arf79F-silenced cells was restored by expressing mammalian Arf1, but not by constitutively active Rac1, showing that Arf79F does not act via Rac1. Abolition of lamellipodium formation in Arf79F-silenced cells was not due to Golgi disruption. Blocking Arf79F activation with guanine nucleotide exchange factor inhibitors impaired WRC localisation to the plasma membrane and concomitant generation of lamellipodia. Our data indicate that the Class I Arf GTPase is a central component in WRC-driven lamellipodium formation.
Collapse
Affiliation(s)
- Daniel Humphreys
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | | | | | | |
Collapse
|
33
|
Wang X, Castro MA, Mulder KW, Markowetz F. Posterior association networks and functional modules inferred from rich phenotypes of gene perturbations. PLoS Comput Biol 2012; 8:e1002566. [PMID: 22761558 PMCID: PMC3386165 DOI: 10.1371/journal.pcbi.1002566] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/03/2012] [Indexed: 11/19/2022] Open
Abstract
Combinatorial gene perturbations provide rich information for a systematic exploration of genetic interactions. Despite successful applications to bacteria and yeast, the scalability of this approach remains a major challenge for higher organisms such as humans. Here, we report a novel experimental and computational framework to efficiently address this challenge by limiting the 'search space' for important genetic interactions. We propose to integrate rich phenotypes of multiple single gene perturbations to robustly predict functional modules, which can subsequently be subjected to further experimental investigations such as combinatorial gene silencing. We present posterior association networks (PANs) to predict functional interactions between genes estimated using a Bayesian mixture modelling approach. The major advantage of this approach over conventional hypothesis tests is that prior knowledge can be incorporated to enhance predictive power. We demonstrate in a simulation study and on biological data, that integrating complementary information greatly improves prediction accuracy. To search for significant modules, we perform hierarchical clustering with multiscale bootstrap resampling. We demonstrate the power of the proposed methodologies in applications to Ewing's sarcoma and human adult stem cells using publicly available and custom generated data, respectively. In the former application, we identify a gene module including many confirmed and highly promising therapeutic targets. Genes in the module are also significantly overrepresented in signalling pathways that are known to be critical for proliferation of Ewing's sarcoma cells. In the latter application, we predict a functional network of chromatin factors controlling epidermal stem cell fate. Further examinations using ChIP-seq, ChIP-qPCR and RT-qPCR reveal that the basis of their genetic interactions may arise from transcriptional cross regulation. A Bioconductor package implementing PAN is freely available online at http://bioconductor.org/packages/release/bioc/html/PANR.html.
Collapse
Affiliation(s)
- Xin Wang
- Cancer Research UK Cambridge Research Institute, Cambridge, Cambridgeshire, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, Cambridgeshire, United Kingdom
| | - Mauro A. Castro
- Cancer Research UK Cambridge Research Institute, Cambridge, Cambridgeshire, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, Cambridgeshire, United Kingdom
| | - Klaas W. Mulder
- Cancer Research UK Cambridge Research Institute, Cambridge, Cambridgeshire, United Kingdom
| | - Florian Markowetz
- Cancer Research UK Cambridge Research Institute, Cambridge, Cambridgeshire, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, Cambridgeshire, United Kingdom
- * E-mail:
| |
Collapse
|
34
|
Stringent analysis of gene function and protein-protein interactions using fluorescently tagged genes. Genetics 2011; 190:931-40. [PMID: 22174071 DOI: 10.1534/genetics.111.136465] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In Drosophila collections of green fluorescent protein (GFP) trap lines have been used to probe the endogenous expression patterns of trapped genes or the subcellular localization of their protein products. Here, we describe a method, based on nonoverlapping, highly specific, shRNA transgenes directed against GFP, that extends the utility of these collections to loss-of-function studies. Furthermore, we used a MiMIC transposon to generate GFP traps in Drosophila cell lines with distinct subcellular localization patterns, which will permit high-throughput screens using fluorescently tagged proteins. Finally, we show that fluorescent traps, paired with recombinant nanobodies and mass spectrometry, allow the study of endogenous protein complexes in Drosophila.
Collapse
|
35
|
Flockhart IT, Booker M, Hu Y, McElvany B, Gilly Q, Mathey-Prevot B, Perrimon N, Mohr SE. FlyRNAi.org--the database of the Drosophila RNAi screening center: 2012 update. Nucleic Acids Res 2011; 40:D715-9. [PMID: 22067456 PMCID: PMC3245182 DOI: 10.1093/nar/gkr953] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
FlyRNAi (http://www.flyrnai.org), the database and website of the Drosophila RNAi Screening Center (DRSC) at Harvard Medical School, serves a dual role, tracking both production of reagents for RNA interference (RNAi) screening in Drosophila cells and RNAi screen results. The database and website is used as a platform for community availability of protocols, tools, and other resources useful to researchers planning, conducting, analyzing or interpreting the results of Drosophila RNAi screens. Based on our own experience and user feedback, we have made several changes. Specifically, we have restructured the database to accommodate new types of reagents; added information about new RNAi libraries and other reagents; updated the user interface and website; and added new tools of use to the Drosophila community and others. Overall, the result is a more useful, flexible and comprehensive website and database.
Collapse
Affiliation(s)
- Ian T Flockhart
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mohr SE, Perrimon N. RNAi screening: new approaches, understandings, and organisms. WILEY INTERDISCIPLINARY REVIEWS-RNA 2011; 3:145-58. [PMID: 21953743 DOI: 10.1002/wrna.110] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RNA interference (RNAi) leads to sequence-specific knockdown of gene function. The approach can be used in large-scale screens to interrogate function in various model organisms and an increasing number of other species. Genome-scale RNAi screens are routinely performed in cultured or primary cells or in vivo in organisms such as C. elegans. High-throughput RNAi screening is benefitting from the development of sophisticated new instrumentation and software tools for collecting and analyzing data, including high-content image data. The results of large-scale RNAi screens have already proved useful, leading to new understandings of gene function relevant to topics such as infection, cancer, obesity, and aging. Nevertheless, important caveats apply and should be taken into consideration when developing or interpreting RNAi screens. Some level of false discovery is inherent to high-throughput approaches and specific to RNAi screens, false discovery due to off-target effects (OTEs) of RNAi reagents remains a problem. The need to improve our ability to use RNAi to elucidate gene function at large scale and in additional systems continues to be addressed through improved RNAi library design, development of innovative computational and analysis tools and other approaches.
Collapse
Affiliation(s)
- Stephanie E Mohr
- Drosophila RNAi Screening Center, Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|