System-Based Approaches to Delineate the Antiviral Innate Immune Landscape
Abstract
1. Introduction
1.1. The Intracellular Antiviral Responses
1.2. Experimental Systems to Study ISG Responses
2. Differential Gene Expression Analysis
3. Genetic Screens
3.1. Gain-of-Function Screens
3.2. Loss-of-Function Screens
3.2.1. RNA Interference
3.2.2. Haploid Screens
3.2.3. CRISPR/Cas9
3.3. Validation of Primary Screens
4. Assay Design and Screening Format
4.1. Choosing a Target Gene Library
4.2. The State of the Cell: Requirement of Pre-Stimulation to Allow Appropriate Experimental Conditions
4.3. Pooled Screens
4.4. Arrayed Screens
5. Interaction Screens and Mass-Spectrometry-Based Screens
5.1. Interaction Screens
5.2. Proteomic Approaches
5.3. Virus as Baits
5.4. Secretomics
6. Future Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Appendix A. Box 1 (Methodology)
- RNA Interference (RNAi).
- Haploid Genetic Screens.
- CRISPR/Cas9.
- CRISPRa/i.
References
- Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef] [PubMed]
- Wu, D.; Sanin, D.E.; Everts, B.; Chen, Q.; Qiu, J.; Buck, M.D.; Patterson, A.; Smith, A.M.; Chang, C.-H.; Liu, Z.; et al. Type 1 Interferons Induce Changes in Core Metabolism that Are Critical for Immune Function. Immunity 2016, 44, 1325–1336. [Google Scholar] [CrossRef] [PubMed]
- Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef] [PubMed]
- Schlee, M.; Hartmann, G. Discriminating self from non-self in nucleic acid sensing. Nat. Rev. Immunol. 2016, 16, 566–580. [Google Scholar] [CrossRef] [PubMed]
- Arimoto, K.-I.; Miyauchi, S.; Stoner, S.A.; Fan, J.-B.; Zhang, D.-E. Negative regulation of type I IFN signaling. J. Leukoc. Biol. 2018, 103. [Google Scholar] [CrossRef] [PubMed]
- Blach-Olszewska, Z.; Leszek, J. Mechanisms of over-activated innate immune system regulation in autoimmune and neurodegenerative disorders. Neuropsychiatr. Dis. Treat. 2007, 3, 365–372. [Google Scholar]
- Liu, J.; Cao, X. Cellular and molecular regulation of innate inflammatory responses. Cell. Mol. Immunol. 2016, 13, 711–721. [Google Scholar] [CrossRef]
- Borden, E.C.; Sen, G.C.; Uze, G.; Silverman, R.H.; Ransohoff, R.M.; Foster, G.R.; Stark, G.R. Interferons at age 50: Past, current and future impact on biomedicine. Nat. Rev. Drug Discov. 2007, 6, 975–990. [Google Scholar] [CrossRef]
- Der, S.D.; Zhou, A.; Williams, B.R.; Silverman, R.H. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc. Natl. Acad. Sci. USA 1998, 95, 15623–15628. [Google Scholar] [CrossRef]
- Zhang, X.; Yang, W.; Wang, X.; Zhang, X.; Tian, H.; Deng, H.; Zhang, L.; Gao, G. Identification of new type I interferon-stimulated genes and investigation of their involvement in IFN-β activation. Protein Cell 2018, 9, 799–807. [Google Scholar] [CrossRef]
- Lazear, H.M.; Schoggins, J.W.; Diamond, M.S. Shared and Distinct Functions of Type I and Type III Interferons. Immunity 2019, 50, 907–923. [Google Scholar] [CrossRef] [PubMed]
- Stanifer, M.L.; Pervolaraki, K.; Boulant, S. Differential Regulation of Type I and Type III Interferon Signaling. Int. J. Mol. Sci. 2019, 20, 1445. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Xu, L.; Su, J.; Peppelenbosch, M.P.; Pan, Q. Transcriptional Regulation of Antiviral Interferon-Stimulated Genes. Trends Microbiol. 2017, 25, 573–584. [Google Scholar] [CrossRef] [PubMed]
- Ivashkiv, L.B.; Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49. [Google Scholar] [CrossRef] [PubMed]
- Nishitsuji, H.; Ujino, S.; Yoshio, S.; Sugiyama, M.; Mizokami, M.; Kanto, T.; Shimotohno, K. Long noncoding RNA #32 contributes to antiviral responses by controlling interferon-stimulated gene expression. Proc. Natl. Acad. Sci. USA 2016, 113, 10388–10393. [Google Scholar] [CrossRef] [PubMed]
- Shaw, A.E.; Hughes, J.; Gu, Q.; Behdenna, A.; Singer, J.B.; Dennis, T.; Orton, R.J.; Varela, M.; Gifford, R.J.; Wilson, S.J.; et al. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017, 15, e2004086. [Google Scholar] [CrossRef] [PubMed]
- Subramanian, G.; Kuzmanovic, T.; Zhang, Y.; Peter, C.B.; Veleeparambil, M.; Chakravarti, R.; Sen, G.C.; Chattopadhyay, S. A new mechanism of interferon’s antiviral action: Induction of autophagy, essential for paramyxovirus replication, is inhibited by the interferon stimulated gene, TDRD7. PLoS Pathog. 2018, 14, e1006877. [Google Scholar] [CrossRef]
- Neil, S.J.D.; Zang, T.; Bieniasz, P.D. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 2008, 451, 425–430. [Google Scholar] [CrossRef]
- Neil, S.J.D.; Sandrin, V.; Sundquist, W.I.; Bieniasz, P.D. An Interferon-α-Induced Tethering Mechanism Inhibits HIV-1 and Ebola Virus Particle Release but Is Counteracted by the HIV-1 Vpu Protein. Cell Host Microbe 2007, 2, 193–203. [Google Scholar] [CrossRef]
- Yao, H.; Dittmann, M.; Peisley, A.; Hoffmann, H.-H.; Gilmore, R.H.; Schmidt, T.; Schmidt-Burgk, J.; Hornung, V.; Rice, C.M.; Hur, S. ATP-dependent effector-like functions of RIG-I-like receptors. Mol. Cell 2015, 58, 541–548. [Google Scholar] [CrossRef]
- Schneider, W.M.; Chevillotte, M.D.; Rice, C.M. Interferon-stimulated genes: A complex web of host defenses. Annu. Rev. Immunol. 2014, 32, 513–545. [Google Scholar] [CrossRef] [PubMed]
- Bruns, A.M.; Leser, G.P.; Lamb, R.A.; Horvath, C.M. The innate immune sensor LGP2 activates antiviral signaling by regulating MDA5-RNA interaction and filament assembly. Mol. Cell 2014, 55, 771–781. [Google Scholar] [CrossRef] [PubMed]
- Almine, J.F.; O’Hare, C.A.J.; Dunphy, G.; Haga, I.R.; Naik, R.J.; Atrih, A.; Connolly, D.J.; Taylor, J.; Kelsall, I.R.; Bowie, A.G.; et al. IFI16 and cGAS cooperate in the activation of STING during DNA sensing in human keratinocytes. Nat. Commun. 2017, 8, 14392. [Google Scholar] [CrossRef] [PubMed]
- Jønsson, K.L.; Laustsen, A.; Krapp, C.; Skipper, K.A.; Thavachelvam, K.; Hotter, D.; Egedal, J.H.; Kjolby, M.; Mohammadi, P.; Prabakaran, T.; et al. IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP. Nat. Commun. 2017, 8, 14391. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef]
- Liau, N.P.D.; Laktyushin, A.; Lucet, I.S.; Murphy, J.M.; Yao, S.; Whitlock, E.; Callaghan, K.; Nicola, N.A.; Kershaw, N.J.; Babon, J.J. The molecular basis of JAK/STAT inhibition by SOCS1. Nat. Commun. 2018, 9, 1558. [Google Scholar] [CrossRef]
- Blumer, T.; Coto-Llerena, M.; Duong, F.H.T.; Heim, M.H. SOCS1 is an inducible negative regulator of interferon λ (IFN-λ)–induced gene expression in vivo. J. Biol. Chem. 2017, 292, 17928–17938. [Google Scholar] [CrossRef]
- Akhtar, L.N.; Benveniste, E.N. Viral exploitation of host SOCS protein functions. J. Virol. 2011, 85, 1912–1921. [Google Scholar] [CrossRef]
- Liu, S.; Cai, X.; Wu, J.; Cong, Q.; Chen, X.; Li, T.; Du, F.; Ren, J.; Wu, Y.-T.; Grishin, N.V.; et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 2015, 347, aaa2630. [Google Scholar] [CrossRef]
- Ablasser, A.; Hur, S. Regulation of cGAS- and RLR-mediated immunity to nucleic acids. Nature immunology 2020, 21, 17–29. [Google Scholar] [CrossRef]
- Willemsen, J.; Wicht, O.; Wolanski, J.C.; Baur, N.; Bastian, S.; Haas, D.A.; Matula, P.; Knapp, B.; Meyniel-Schicklin, L.; Wang, C.; et al. Phosphorylation-Dependent Feedback Inhibition of RIG-I by DAPK1 Identified by Kinome-wide siRNA Screening. Mol. Cell 2017, 65, 403–415. [Google Scholar] [CrossRef] [PubMed]
- Pervolaraki, K.; Rastgou-Talemi, S.; Albrecht, D.; Bormann, F.; Bamford, C.; Mendoza, J.L.; Garcia, K.C.; McLauchlan, J.; Höfer, T.; Stanifer, M.L.; et al. Differential induction of interferon stimulated genes between type I and type III interferons is independent of interferon receptor abundance. PLoS Pathog. 2018, 14, e1007420. [Google Scholar] [CrossRef]
- Voigt, E.A.; Yin, J. Kinetic Differences and Synergistic Antiviral Effects Between Type I and Type III Interferon Signaling Indicate Pathway Independence. J. Interferon Cytokine Res. 2015, 35, 734–747. [Google Scholar] [CrossRef] [PubMed]
- Pommerenke, C.; Wilk, E.; Srivastava, B.; Schulze, A.; Novoselova, N.; Geffers, R.; Schughart, K. Global transcriptome analysis in influenza-infected mouse lungs reveals the kinetics of innate and adaptive host immune responses. PLoS ONE 2012, 7, e41169. [Google Scholar] [CrossRef] [PubMed]
- Michalska, A.; Blaszczyk, K.; Wesoly, J.; Bluyssen, H.A.R. A Positive Feedback Amplifier Circuit That Regulates Interferon (IFN)-Stimulated Gene Expression and Controls Type I and Type II IFN Responses. Front. Immunol. 2018, 9, 1135. [Google Scholar] [CrossRef]
- Larner, A.C.; Chaudhuri, A.; Darnell, J.E. Transcriptional induction by interferon. New protein(s) determine the extent and length of the induction. J. Biol. Chem. 1986, 261, 453–459. [Google Scholar]
- Lumb, J.H.; Li, Q.; Popov, L.M.; Ding, S.; Keith, M.T.; Merrill, B.D.; Greenberg, H.B.; Li, J.B.; Carette, J.E. DDX6 Represses Aberrant Activation of Interferon-Stimulated Genes. Cell Rep. 2017, 20, 819–831. [Google Scholar] [CrossRef]
- Thompson, S.L.; Compton, D.A. Chromosomes and cancer cells. Chromosome Res. 2011, 19, 433–444. [Google Scholar] [CrossRef]
- McCormick, D.; Lin, Y.-T.; Grey, F. Identification of Host Factors Involved in Human Cytomegalovirus Replication, Assembly, and Egress Using a Two-Step Small Interfering RNA Screen. mBio 2018, 9. [Google Scholar] [CrossRef]
- Konno, H.; Yamauchi, S.; Berglund, A.; Putney, R.M.; Mulé, J.J.; Barber, G.N. Suppression of STING signaling through epigenetic silencing and missense mutation impedes DNA damage mediated cytokine production. Oncogene 2018, 37, 2037–2051. [Google Scholar] [CrossRef]
- Sun, B.; Sundström, K.B.; Chew, J.J.; Bist, P.; Gan, E.S.; Tan, H.C.; Goh, K.C.; Chawla, T.; Tang, C.K.; Ooi, E.E. Dengue virus activates cGAS through the release of mitochondrial DNA. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [PubMed]
- Moriyama, M.; Koshiba, T.; Ichinohe, T. Influenza A virus M2 protein triggers mitochondrial DNA-mediated antiviral immune responses. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
- West, A.P.; Khoury-Hanold, W.; Staron, M.; Tal, M.C.; Pineda, C.M.; Lang, S.M.; Bestwick, M.; Duguay, B.A.; Raimundo, N.; MacDuff, D.A.; et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 2015, 520, 553–557. [Google Scholar] [CrossRef]
- Rivas, C.; Aaronson, S.A.; Munoz-Fontela, C. Dual Role of p53 in Innate Antiviral Immunity. Viruses 2010, 2, 298–313. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Fontela, C.; Pazos, M.; Delgado, I.; Murk, W.; Mungamuri, S.K.; Lee, S.W.; García-Sastre, A.; Moran, T.M.; Aaronson, S.A. p53 serves as a host antiviral factor that enhances innate and adaptive immune responses to influenza A virus. J. Immunol. 2011, 187, 6428–6436. [Google Scholar] [CrossRef]
- Enache, O.M.; Rendo, V.; Abdusamad, M.; Lam, D.; Davison, D.; Pal, S.; Currimjee, N.; Hess, J.; Pantel, S.; Nag, A.; et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat. Genet. 2020, 52, 662–668. [Google Scholar] [CrossRef] [PubMed]
- Thi-Nhu-Thao, T.; Labroussaa, F.; Ebert, N.; V’kovski, P.; Stalder, H.; Portmann, J.; Kelly, J.; Steiner, S.; Holwerda, M.; Kratzel, A.; et al. Rapid reconstruction of SARS-CoV-2 using a synthetic genomics platform. Nature 2020, 582, 561–565. [Google Scholar] [CrossRef]
- Kuroda, M.; Halfmann, P.J.; Hill-Batorski, L.; Ozawa, M.; Lopes, T.J.S.; Neumann, G.; Schoggins, J.W.; Rice, C.M.; Kawaoka, Y. Identification of interferon-stimulated genes that attenuate Ebola virus infection. Nat. Commun. 2020, 11, 2953. [Google Scholar] [CrossRef]
- Halfmann, P.; Kim, J.H.; Ebihara, H.; Noda, T.; Neumann, G.; Feldmann, H.; Kawaoka, Y. Generation of biologically contained Ebola viruses. Proc. Natl. Acad. Sci. USA 2008, 105, 1129–1133. [Google Scholar] [CrossRef]
- Schoggins, J.W. Interferon-Stimulated Genes: What Do They All Do? Annu. Rev. Virol. 2019, 6, 567–584. [Google Scholar] [CrossRef]
- Moll, H.P.; Maier, T.; Zommer, A.; Lavoie, T.; Brostjan, C. The differential activity of interferon-α subtypes is consistent among distinct target genes and cell types. Cytokine 2011, 53, 52–59. [Google Scholar] [CrossRef] [PubMed]
- Thomas, C.; Moraga, I.; Levin, D.; Krutzik, P.O.; Podoplelova, Y.; Trejo, A.; Lee, C.; Yarden, G.; Vleck, S.E.; Glenn, J.S.; et al. Structural linkage between ligand discrimination and receptor activation by type I interferons. Cell 2011, 146, 621–632. [Google Scholar] [CrossRef] [PubMed]
- Bolen, C.R.; Ding, S.; Robek, M.D.; Kleinstein, S.H. Dynamic expression profiling of type I and type III interferon-stimulated hepatocytes reveals a stable hierarchy of gene expression. Hepatology 2014, 59, 1262–1272. [Google Scholar] [CrossRef]
- Steuerman, Y.; Cohen, M.; Peshes-Yaloz, N.; Valadarsky, L.; Cohn, O.; David, E.; Frishberg, A.; Mayo, L.; Bacharach, E.; Amit, I.; et al. Dissection of Influenza Infection In Vivo by Single-Cell RNA Sequencing. Cell Syst. 2018, 6, 679–691. [Google Scholar] [CrossRef] [PubMed]
- Zanini, F.; Robinson, M.L.; Croote, D.; Sahoo, M.K.; Sanz, A.M.; Ortiz-Lasso, E.; Albornoz, L.L.; Rosso, F.; Montoya, J.G.; Goo, L.; et al. Virus-inclusive single-cell RNA sequencing reveals the molecular signature of progression to severe dengue. Proc. Natl. Acad. Sci. USA 2018, 115, E12363–E12369. [Google Scholar] [CrossRef]
- Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef] [PubMed]
- Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed]
- Schoggins, J.W.; MacDuff, D.A.; Imanaka, N.; Gainey, M.D.; Shrestha, B.; Eitson, J.L.; Mar, K.B.; Richardson, R.B.; Ratushny, A.V.; Litvak, V.; et al. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature 2014, 505, 691–695. [Google Scholar] [CrossRef]
- Riley, J.S.; Tait, S.W. Mitochondrial DNA in inflammation and immunity. EMBO Rep. 2020, 21, e49799. [Google Scholar] [CrossRef]
- Seth, R.B.; Sun, L.; Ea, C.-K.; Chen, Z.J. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 2005, 122, 669–682. [Google Scholar] [CrossRef]
- Biacchesi, S.; Mérour, E.; Lamoureux, A.; Bernard, J.; Brémont, M. Both STING and MAVS fish orthologs contribute to the induction of interferon mediated by RIG-I. PLoS ONE 2012, 7, e47737. [Google Scholar] [CrossRef]
- Stremlau, M.; Owens, C.M.; Perron, M.J.; Kiessling, M.; Autissier, P.; Sodroski, J. The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature 2004, 427, 848–853. [Google Scholar] [CrossRef] [PubMed]
- Kane, M.; Zang, T.M.; Rihn, S.J.; Zhang, F.; Kueck, T.; Alim, M.; Schoggins, J.; Rice, C.M.; Wilson, S.J.; Bieniasz, P.D. Identification of Interferon-Stimulated Genes with Antiretroviral Activity. Cell Host Microbe 2016, 20, 392–405. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Wickenhagen, A.; Turnbull, M.L.; Rezelj, V.V.; Kreher, F.; Tilston-Lunel, N.L.; Slack, G.S.; Brennan, B.; Koudriakova, E.; Shaw, A.E.; et al. Interferon-Stimulated Gene (ISG)-Expression Screening Reveals the Specific Antibunyaviral Activity of ISG20. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [PubMed]
- Pichlmair, A.; Lassnig, C.; Eberle, C.-A.; Górna, M.W.; Baumann, C.L.; Burkard, T.R.; Bürckstümmer, T.; Stefanovic, A.; Krieger, S.; Bennett, K.L.; et al. IFIT1 is an antiviral protein that recognizes 5′-triphosphate RNA. Nat. Immunol. 2011, 12, 624–630. [Google Scholar] [CrossRef]
- Johnson, B.; VanBlargan, L.A.; Xu, W.; White, J.P.; Shan, C.; Shi, P.-Y.; Zhang, R.; Adhikari, J.; Gross, M.L.; Leung, D.W.; et al. Human IFIT3 Modulates IFIT1 RNA Binding Specificity and Protein Stability. Immunity 2018, 48, 487–499. [Google Scholar] [CrossRef]
- Fellmann, C.; Lowe, S.W. Stable RNA interference rules for silencing. Nat. Cell Biol. 2014, 16, 10–18. [Google Scholar] [CrossRef]
- Schuster, A.; Erasimus, H.; Fritah, S.; Nazarov, P.V.; van Dyck, E.; Niclou, S.P.; Golebiewska, A. RNAi/CRISPR Screens: From a Pool to a Valid Hit. Trends Biotechnol. 2019, 37, 38–55. [Google Scholar] [CrossRef]
- Boettcher, M.; McManus, M.T. Choosing the Right Tool for the Job: RNAi, TALEN, or CRISPR. Mol. Cell 2015, 58, 575–585. [Google Scholar] [CrossRef]
- Carthew, R.W.; Sontheimer, E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell 2009, 136, 642–655. [Google Scholar] [CrossRef]
- Agarwal, V.; Bell, G.W.; Nam, J.-W.; Bartel, D.P. Predicting effective microRNA target sites in mammalian mRNAs. Elife 2015, 4. [Google Scholar] [CrossRef] [PubMed]
- Riba, A.; Emmenlauer, M.; Chen, A.; Sigoillot, F.; Cong, F.; Dehio, C.; Jenkins, J.; Zavolan, M. Explicit Modeling of siRNA-Dependent On- and Off-Target Repression Improves the Interpretation of Screening Results. Cell Syst. 2017, 4, 182–193. [Google Scholar] [CrossRef] [PubMed]
- Pillay, S.; Meyer, N.L.; Puschnik, A.S.; Davulcu, O.; Diep, J.; Ishikawa, Y.; Jae, L.T.; Wosen, J.E.; Nagamine, C.M.; Chapman, M.S.; et al. An essential receptor for adeno-associated virus infection. Nature 2016, 530, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Carette, J.E.; Raaben, M.; Wong, A.C.; Herbert, A.S.; Obernosterer, G.; Mulherkar, N.; Kuehne, A.I.; Kranzusch, P.J.; Griffin, A.M.; Ruthel, G.; et al. Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature 2011, 477, 340–343. [Google Scholar] [CrossRef]
- Jae, L.T.; Raaben, M.; Herbert, A.S.; Kuehne, A.I.; Wirchnianski, A.S.; Soh, T.K.; Stubbs, S.H.; Janssen, H.; Damme, M.; Saftig, P.; et al. Virus entry. Lassa virus entry requires a trigger-induced receptor switch. Science 2014, 344, 1506–1510. [Google Scholar] [CrossRef] [PubMed]
- Staring, J.; von Castelmur, E.; Blomen, V.A.; van den Hengel, L.G.; Brockmann, M.; Baggen, J.; Thibaut, H.J.; Nieuwenhuis, J.; Janssen, H.; van Kuppeveld, F.J.M.; et al. PLA2G16 represents a switch between entry and clearance of Picornaviridae. Nature 2017, 541, 412–416. [Google Scholar] [CrossRef] [PubMed]
- Carette, J.E.; Guimaraes, C.P.; Varadarajan, M.; Park, A.S.; Wuethrich, I.; Godarova, A.; Kotecki, M.; Cochran, B.H.; Spooner, E.; Ploegh, H.L.; et al. Haploid genetic screens in human cells identify host factors used by pathogens. Science 2009, 326, 1231–1235. [Google Scholar] [CrossRef]
- Riblett, A.M.; Blomen, V.A.; Jae, L.T.; Altamura, L.A.; Doms, R.W.; Brummelkamp, T.R.; Wojcechowskyj, J.A. A Haploid Genetic Screen Identifies Heparan Sulfate Proteoglycans Supporting Rift Valley Fever Virus Infection. J. Virol. 2016, 90, 1414–1423. [Google Scholar] [CrossRef]
- Luteijn, R.D.; van Diemen, F.; Blomen, V.A.; Boer, I.G.J.; Manikam-Sadasivam, S.; van Kuppevelt, T.H.; Drexler, I.; Brummelkamp, T.R.; Lebbink, R.J.; Wiertz, E.J. A Genome-Wide Haploid Genetic Screen Identifies Heparan Sulfate-Associated Genes and the Macropinocytosis Modulator TMED10 as Factors Supporting Vaccinia Virus Infection. J. Virol. 2019, 93. [Google Scholar] [CrossRef]
- Doench, J.G.; Fusi, N.; Sullender, M.; Hegde, M.; Vaimberg, E.W.; Donovan, K.F.; Smith, I.; Tothova, Z.; Wilen, C.; Orchard, R.; et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 2016, 34, 184–191. [Google Scholar] [CrossRef]
- Munoz, D.M.; Cassiani, P.J.; Li, L.; Billy, E.; Korn, J.M.; Jones, M.D.; Golji, J.; Ruddy, D.A.; Yu, K.; McAllister, G.; et al. CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions. Cancer Discov. 2016, 6, 900–913. [Google Scholar] [CrossRef] [PubMed]
- Duan, J.; Lu, G.; Xie, Z.; Lou, M.; Luo, J.; Guo, L.; Zhang, Y. Genome-wide identification of CRISPR/Cas9 off-targets in human genome. Cell Res. 2014, 24, 1009–1012. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Scott, D.A.; Kriz, A.J.; Chiu, A.C.; Hsu, P.D.; Dadon, D.B.; Cheng, A.W.; Trevino, A.E.; Konermann, S.; Chen, S.; et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 2014, 32, 670–676. [Google Scholar] [CrossRef] [PubMed]
- Kuscu, C.; Arslan, S.; Singh, R.; Thorpe, J.; Adli, M. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat. Biotechnol. 2014, 32, 677–683. [Google Scholar] [CrossRef] [PubMed]
- Tsai, S.Q.; Zheng, Z.; Nguyen, N.T.; Liebers, M.; Topkar, V.V.; Thapar, V.; Wyvekens, N.; Khayter, C.; Iafrate, A.J.; Le, L.P.; et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 2015, 33, 187–197. [Google Scholar] [CrossRef]
- Joung, J.; Konermann, S.; Gootenberg, J.S.; Abudayyeh, O.O.; Platt, R.J.; Brigham, M.D.; Sanjana, N.E.; Zhang, F. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat. Protoc. 2017, 12, 828–863. [Google Scholar] [CrossRef]
- Lee, C.-H.; Griffiths, S.; Digard, P.; Pham, N.; Auer, M.; Haas, J.; Grey, F. Asparagine Deprivation Causes a Reversible Inhibition of Human Cytomegalovirus Acute Virus Replication. mBio 2019, 10. [Google Scholar] [CrossRef]
- van Asten, S.D.; Raaben, M.; Nota, B.; Spaapen, R.M. Secretome Screening Reveals Fibroblast Growth Factors as Novel Inhibitors of Viral Replication. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
- Puschnik, A.S.; Majzoub, K.; Ooi, Y.S.; Carette, J.E. A CRISPR toolbox to study virus-host interactions. Nat. Rev. Microbiol. 2017, 15, 351–364. [Google Scholar] [CrossRef]
- Chou, Y.-C.; Lai, M.M.; Wu, Y.-C.; Hsu, N.-C.; Jeng, K.-S.; Su, W.-C. Variations in genome-wide RNAi screens: Lessons from influenza research. J. Clin. Bioinforma. 2015, 5, 2. [Google Scholar] [CrossRef]
- Hao, L.; He, Q.; Wang, Z.; Craven, M.; Newton, M.A.; Ahlquist, P. Limited agreement of independent RNAi screens for virus-required host genes owes more to false-negative than false-positive factors. PLoS Comput. Biol. 2013, 9, e1003235. [Google Scholar] [CrossRef] [PubMed]
- Sessions, O.M.; Barrows, N.J.; Souza-Neto, J.A.; Robinson, T.J.; Hershey, C.L.; Rodgers, M.A.; Ramirez, J.L.; Dimopoulos, G.; Yang, P.L.; Pearson, J.L.; et al. Discovery of insect and human dengue virus host factors. Nature 2009, 458, 1047–1050. [Google Scholar] [CrossRef]
- Savidis, G.; McDougall, W.M.; Meraner, P.; Perreira, J.M.; Portmann, J.M.; Trincucci, G.; John, S.P.; Aker, A.M.; Renzette, N.; Robbins, D.R.; et al. Identification of Zika Virus and Dengue Virus Dependency Factors using Functional Genomics. Cell Rep. 2016, 16, 232–246. [Google Scholar] [CrossRef]
- Filone, C.M.; Dower, K.; Cowley, G.S.; Hensley, L.E.; Connor, J.H. Probing the virus host interaction in high containment: An approach using pooled short hairpin RNA. Assay Drug Dev. Technol. 2015, 13, 34–43. [Google Scholar] [CrossRef]
- Martin, S.; Chiramel, A.I.; Schmidt, M.L.; Chen, Y.-C.; Whitt, N.; Watt, A.; Dunham, E.C.; Shifflett, K.; Traeger, S.; Leske, A.; et al. A genome-wide siRNA screen identifies a druggable host pathway essential for the Ebola virus life cycle. Genome Med. 2018, 10, 58. [Google Scholar] [CrossRef] [PubMed]
- Tai, A.W.; Benita, Y.; Peng, L.F.; Kim, S.-S.; Sakamoto, N.; Xavier, R.J.; Chung, R.T. A functional genomic screen identifies cellular cofactors of hepatitis C virus replication. Cell Host Microbe 2009, 5, 298–307. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Brass, A.L.; Ng, A.; Hu, Z.; Xavier, R.J.; Liang, T.J.; Elledge, S.J. A genome-wide genetic screen for host factors required for hepatitis C virus propagation. Proc. Natl. Acad. Sci. USA 2009, 106, 16410–16415. [Google Scholar] [CrossRef] [PubMed]
- Ali, H.; Mano, M.; Braga, L.; Naseem, A.; Marini, B.; Vu, D.M.; Collesi, C.; Meroni, G.; Lusic, M.; Giacca, M. Cellular TRIM33 restrains HIV-1 infection by targeting viral integrase for proteasomal degradation. Nat. Commun. 2019, 10, 926. [Google Scholar] [CrossRef]
- Brass, A.L.; Dykxhoorn, D.M.; Benita, Y.; Yan, N.; Engelman, A.; Xavier, R.J.; Lieberman, J.; Elledge, S.J. Identification of host proteins required for HIV infection through a functional genomic screen. Science 2008, 319, 921–926. [Google Scholar] [CrossRef]
- König, R.; Zhou, Y.; Elleder, D.; Diamond, T.L.; Bonamy, G.M.C.; Irelan, J.T.; Chiang, C.-Y.; Tu, B.P.; de Jesus, P.D.; Lilley, C.E.; et al. Global analysis of host-pathogen interactions that regulate early-stage HIV-1 replication. Cell 2008, 135, 49–60. [Google Scholar] [CrossRef]
- Zhou, H.; Xu, M.; Huang, Q.; Gates, A.T.; Zhang, X.D.; Castle, J.C.; Stec, E.; Ferrer, M.; Strulovici, B.; Hazuda, D.J.; et al. Genome-scale RNAi screen for host factors required for HIV replication. Cell Host Microbe 2008, 4, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.-M.; Zhang, X.-Y.; Zhang, Y.-Z.; Liu, L.; Lu, H.-Z. A high throughput RNAi screen reveals determinants of HIV-1 activity in host kinases. Int. J. Clin. Exp. Pathol. 2014, 7, 2229–2237. [Google Scholar] [PubMed]
- König, R.; Stertz, S.; Zhou, Y.; Inoue, A.; Hoffmann, H.-H.; Bhattacharyya, S.; Alamares, J.G.; Tscherne, D.M.; Ortigoza, M.B.; Liang, Y.; et al. Human host factors required for influenza virus replication. Nature 2010, 463, 813–817. [Google Scholar] [CrossRef] [PubMed]
- Su, W.-C.; Chen, Y.-C.; Tseng, C.-H.; Hsu, P.W.-C.; Tung, K.-F.; Jeng, K.-S.; Lai, M.M.C. Pooled RNAi screen identifies ubiquitin ligase Itch as crucial for influenza A virus release from the endosome during virus entry. Proc. Natl. Acad. Sci. USA 2013, 110, 17516–17521. [Google Scholar] [CrossRef]
- Hao, L.; Sakurai, A.; Watanabe, T.; Sorensen, E.; Nidom, C.A.; Newton, M.A.; Ahlquist, P.; Kawaoka, Y. Drosophila RNAi screen identifies host genes important for influenza virus replication. Nature 2008, 454, 890–893. [Google Scholar] [CrossRef]
- Karlas, A.; Machuy, N.; Shin, Y.; Pleissner, K.-P.; Artarini, A.; Heuer, D.; Becker, D.; Khalil, H.; Ogilvie, L.A.; Hess, S.; et al. Genome-wide RNAi screen identifies human host factors crucial for influenza virus replication. Nature 2010, 463, 818–822. [Google Scholar] [CrossRef]
- Cheng, H.; Koning, K.; O’Hearn, A.; Wang, M.; Rumschlag-Booms, E.; Varhegyi, E.; Rong, L. A parallel genome-wide RNAi screening strategy to identify host proteins important for entry of Marburg virus and H5N1 influenza virus. Virol. J. 2015, 12, 194. [Google Scholar] [CrossRef]
- Panda, D.; Rose, P.P.; Hanna, S.L.; Gold, B.; Hopkins, K.C.; Lyde, R.B.; Marks, M.S.; Cherry, S. Genome-wide RNAi screen identifies SEC61A and VCP as conserved regulators of Sindbis virus entry. Cell Rep. 2013, 5, 1737–1748. [Google Scholar] [CrossRef]
- Rose, P.P.; Hanna, S.L.; Spiridigliozzi, A.; Wannissorn, N.; Beiting, D.P.; Ross, S.R.; Hardy, R.W.; Bambina, S.A.; Heise, M.T.; Cherry, S. Natural resistance-associated macrophage protein is a cellular receptor for sindbis virus in both insect and mammalian hosts. Cell Host Microbe 2011, 10, 97–104. [Google Scholar] [CrossRef]
- Krishnan, M.N.; Ng, A.; Sukumaran, B.; Gilfoy, F.D.; Uchil, P.D.; Sultana, H.; Brass, A.L.; Adametz, R.; Tsui, M.; Qian, F.; et al. RNA interference screen for human genes associated with West Nile virus infection. Nature 2008, 455, 242–245. [Google Scholar] [CrossRef]
- Ma, H.; Dang, Y.; Wu, Y.; Jia, G.; Anaya, E.; Zhang, J.; Abraham, S.; Choi, J.-G.; Shi, G.; Qi, L.; et al. A CRISPR-Based Screen Identifies Genes Essential for West-Nile-Virus-Induced Cell Death. Cell Rep. 2015, 12, 673–683. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Clohisey, S.M.; Chia, B.S.; Wang, B.; Cui, A.; Eisenhaure, T.; Schweitzer, L.D.; Hoover, P.; Parkinson, N.J.; Nachshon, A.; et al. Genome-wide CRISPR screen identifies host dependency factors for influenza A virus infection. Nat. Commun. 2020, 11, 164. [Google Scholar] [CrossRef] [PubMed]
- Han, J.; Perez, J.T.; Chen, C.; Li, Y.; Benitez, A.; Kandasamy, M.; Lee, Y.; Andrade, J.; tenOever, B.; Manicassamy, B. Genome-wide CRISPR/Cas9 Screen Identifies Host Factors Essential for Influenza Virus Replication. Cell Rep. 2018, 23, 596–607. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Muffat, J.; Omer-Javed, A.; Keys, H.R.; Lungjangwa, T.; Bosch, I.; Khan, M.; Virgilio, M.C.; Gehrke, L.; Sabatini, D.M.; et al. Genome-wide CRISPR screen for Zika virus resistance in human neural cells. Proc. Natl. Acad. Sci. USA 2019, 116, 9527–9532. [Google Scholar] [CrossRef]
- Wei, J.; Alfajaro, M.M.; Hanna, R.E.; DeWeirdt, P.C.; Strine, M.S.; Lu-Culligan, W.J.; Zhang, S.-M.; Graziano, V.R.; Schmitz, C.O.; Chen, J.S.; et al. Genome-wide CRISPR screen reveals host genes that regulate SARS-CoV-2 infection. bioRxiv 2020. [Google Scholar] [CrossRef]
- Thamamongood, T.; Aebischer, A.; Wagner, V.; Chang, M.W.; Elling, R.; Benner, C.; García-Sastre, A.; Kochs, G.; Beer, M.; Schwemmle, M. A Genome-Wide CRISPR-Cas9 Screen Reveals the Requirement of Host Cell Sulfation for Schmallenberg Virus Infection. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
- Roesch, F.; OhAinle, M.; Emerman, M. A CRISPR screen for factors regulating SAMHD1 degradation identifies IFITMs as potent inhibitors of lentiviral particle delivery. Retrovirology 2018, 15, 26. [Google Scholar] [CrossRef]
- Richardson, R.B.; Ohlson, M.B.; Eitson, J.L.; Kumar, A.; McDougal, M.B.; Boys, I.N.; Mar, K.B.; de la Cruz-Rivera, P.C.; Douglas, C.; Konopka, G.; et al. A CRISPR screen identifies IFI6 as an ER-resident interferon effector that blocks flavivirus replication. Nat. Microbiol. 2018, 3, 1214–1223. [Google Scholar] [CrossRef]
- Varble, A.; Benitez, A.A.; Schmid, S.; Sachs, D.; Shim, J.V.; Rodriguez-Barrueco, R.; Panis, M.; Crumiller, M.; Silva, J.M.; Sachidanandam, R.; et al. An in vivo RNAi screening approach to identify host determinants of virus replication. Cell Host Microbe 2013, 14, 346–356. [Google Scholar] [CrossRef]
- Benaoudia, S.; Martin, A.; Puig-Gamez, M.; Gay, G.; Lagrange, B.; Cornut, M.; Krasnykov, K.; Claude, J.-B.; Bourgeois, C.F.; Hughes, S.; et al. A genome-wide screen identifies IRF2 as a key regulator of caspase-4 in human cells. EMBO Rep. 2019, 20, e48235. [Google Scholar] [CrossRef]
- Orvedahl, A.; McAllaster, M.R.; Sansone, A.; Dunlap, B.F.; Desai, C.; Wang, Y.-T.; Balce, D.R.; Luke, C.J.; Lee, S.; Orchard, R.C.; et al. Autophagy genes in myeloid cells counteract IFNγ-induced TNF-mediated cell death and fatal TNF-induced shock. Proc. Natl. Acad. Sci. USA 2019, 116, 16497–16506. [Google Scholar] [CrossRef] [PubMed]
- Ding, S.; Diep, J.; Feng, N.; Ren, L.; Li, B.; Ooi, Y.S.; Wang, X.; Brulois, K.F.; Yasukawa, L.L.; Li, X.; et al. STAG2 deficiency induces interferon responses via cGAS-STING pathway and restricts virus infection. Nat. Commun. 2018, 9, 1485. [Google Scholar] [CrossRef]
- OhAinle, M.; Helms, L.; Vermeire, J.; Roesch, F.; Humes, D.; Basom, R.; Delrow, J.J.; Overbaugh, J.; Emerman, M. A virus-packageable CRISPR screen identifies host factors mediating interferon inhibition of HIV. Elife 2018, 7. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.S.; Lee, K.; Bae, S.; Park, J.; Lee, C.-K.; Kim, M.; Kim, E.; Kim, M.; Kim, S.; Kim, C.; et al. CRISPR/Cas9-mediated gene knockout screens and target identification via whole-genome sequencing uncover host genes required for picornavirus infection. J. Biol. Chem. 2017, 292, 10664–10671. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Lu, T.; Emanuel, G.; Babcock, H.P.; Zhuang, X. Imaging-based pooled CRISPR screening reveals regulators of lncRNA localization. Proc. Natl. Acad. Sci. USA 2019, 116, 10842–10851. [Google Scholar] [CrossRef] [PubMed]
- Datlinger, P.; Rendeiro, A.F.; Schmidl, C.; Krausgruber, T.; Traxler, P.; Klughammer, J.; Schuster, L.C.; Kuchler, A.; Alpar, D.; Bock, C. Pooled CRISPR screening with single-cell transcriptome readout. Nat Methods 2017, 14, 297–301. [Google Scholar] [CrossRef] [PubMed]
- Metzakopian, E.; Strong, A.; Iyer, V.; Hodgkins, A.; Tzelepis, K.; Antunes, L.; Friedrich, M.J.; Kang, Q.; Davidson, T.; Lamberth, J.; et al. Enhancing the genome editing toolbox: genome wide CRISPR arrayed libraries. Sci. Rep. 2017, 7, 2244. [Google Scholar] [CrossRef]
- Kim, H.S.; Lee, K.; Kim, S.-J.; Cho, S.; Shin, H.J.; Kim, C.; Kim, J.-S. Arrayed CRISPR screen with image-based assay reliably uncovers host genes required for coxsackievirus infection. Genome Res. 2018, 28, 859–868. [Google Scholar] [CrossRef]
- Fields, S.; Song, O. A novel genetic system to detect protein-protein interactions. Nature 1989, 340, 245–246. [Google Scholar] [CrossRef]
- McCraith, S.; Holtzman, T.; Moss, B.; Fields, S. Genome-wide analysis of vaccinia virus protein-protein interactions. Proc. Natl. Acad. Sci. USA 2000, 97, 4879–4884. [Google Scholar] [CrossRef]
- von Brunn, A.; Teepe, C.; Simpson, J.C.; Pepperkok, R.; Friedel, C.C.; Zimmer, R.; Roberts, R.; Baric, R.; Haas, J. Analysis of intraviral protein-protein interactions of the SARS coronavirus ORFeome. PLoS ONE 2007, 2, e459. [Google Scholar] [CrossRef]
- Flajolet, M.; Rotondo, G.; Daviet, L.; Bergametti, F.; Inchauspé, G.; Tiollais, P.; Transy, C.; Legrain, P. A genomic approach of the hepatitis C virus generates a protein interaction map. Gene 2000, 242, 369–379. [Google Scholar] [CrossRef]
- Fossum, E.; Friedel, C.C.; Rajagopala, S.V.; Titz, B.; Baiker, A.; Schmidt, T.; Kraus, T.; Stellberger, T.; Rutenberg, C.; Suthram, S.; et al. Evolutionarily conserved herpesviral protein interaction networks. PLoS Pathog. 2009, 5, e1000570. [Google Scholar] [CrossRef] [PubMed]
- Uetz, P.; Dong, Y.-A.; Zeretzke, C.; Atzler, C.; Baiker, A.; Berger, B.; Rajagopala, S.V.; Roupelieva, M.; Rose, D.; Fossum, E.; et al. Herpesviral protein networks and their interaction with the human proteome. Science 2006, 311, 239–242. [Google Scholar] [CrossRef] [PubMed]
- Calderwood, M.A.; Venkatesan, K.; Xing, L.; Chase, M.R.; Vazquez, A.; Holthaus, A.M.; Ewence, A.E.; Li, N.; Hirozane-Kishikawa, T.; Hill, D.E.; et al. Epstein-Barr virus and virus human protein interaction maps. Proc. Natl. Acad. Sci. USA 2007, 104, 7606–7611. [Google Scholar] [CrossRef] [PubMed]
- de Chassey, B.; Navratil, V.; Tafforeau, L.; Hiet, M.S.; Aublin-Gex, A.; Agaugué, S.; Meiffren, G.; Pradezynski, F.; Faria, B.F.; Chantier, T.; et al. Hepatitis C virus infection protein network. Mol. Syst. Biol. 2008, 4, 230. [Google Scholar] [CrossRef] [PubMed]
- Shapira, S.D.; Gat-Viks, I.; Shum, B.O.V.; Dricot, A.; de Grace, M.M.; Wu, L.; Gupta, P.B.; Hao, T.; Silver, S.J.; Root, D.E.; et al. A physical and regulatory map of host-influenza interactions reveals pathways in H1N1 infection. Cell 2009, 139, 1255–1267. [Google Scholar] [CrossRef]
- Chen, J.-Y.; Chen, W.-N.; Poon, K.-M.V.; Zheng, B.-J.; Lin, X.; Wang, Y.-X.; Wen, Y.-M. Interaction between SARS-CoV helicase and a multifunctional cellular protein (Ddx5) revealed by yeast and mammalian cell two-hybrid systems. Arch. Virol. 2009, 154, 507–512. [Google Scholar] [CrossRef]
- Zhang, L.; Villa, N.Y.; Rahman, M.M.; Smallwood, S.; Shattuck, D.; Neff, C.; Dufford, M.; Lanchbury, J.S.; Labaer, J.; McFadden, G. Analysis of vaccinia virus-host protein-protein interactions: Validations of yeast two-hybrid screenings. J. Proteome Res. 2009, 8, 4311–4318. [Google Scholar] [CrossRef]
- Moerdyk-Schauwecker, M.; Destephanis, D.; Hastie, E.; Grdzelishvili, V.Z. Detecting protein-protein interactions in vesicular stomatitis virus using a cytoplasmic yeast two hybrid system. J. Virol. Methods 2011, 173, 203–212. [Google Scholar] [CrossRef]
- Hubel, P.; Urban, C.; Bergant, V.; Schneider, W.M.; Knauer, B.; Stukalov, A.; Scaturro, P.; Mann, A.; Brunotte, L.; Hoffmann, H.H.; et al. A protein-interaction network of interferon-stimulated genes extends the innate immune system landscape. Nat. Immunol. 2019, 20, 493–502. [Google Scholar] [CrossRef] [PubMed]
- Lum, K.K.; Cristea, I.M. Proteomic approaches to uncovering virus-host protein interactions during the progression of viral infection. Expert Rev. Proteom. 2016, 13, 325–340. [Google Scholar] [CrossRef]
- Greco, T.M.; Diner, B.A.; Cristea, I.M. The Impact of Mass Spectrometry-Based Proteomics on Fundamental Discoveries in Virology. Annu. Rev. Virol. 2014, 1, 581–604. [Google Scholar] [CrossRef] [PubMed]
- Aebersold, R.; Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature 2016, 537, 347–355. [Google Scholar] [CrossRef] [PubMed]
- Eckhardt, M.; Hultquist, J.F.; Kaake, R.M.; Hüttenhain, R.; Krogan, N.J. A systems approach to infectious disease. Nat. Rev. Genet. 2020, 21, 339–354. [Google Scholar] [CrossRef]
- Andrejeva, J.; Childs, K.S.; Young, D.F.; Carlos, T.S.; Stock, N.; Goodbourn, S.; Randall, R.E. The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta promoter. Proc. Natl. Acad. Sci. USA 2004, 101, 17264–17269. [Google Scholar] [CrossRef] [PubMed]
- Pichlmair, A.; Kandasamy, K.; Alvisi, G.; Mulhern, O.; Sacco, R.; Habjan, M.; Binder, M.; Stefanovic, A.; Eberle, C.-A.; Goncalves, A.; et al. Viral immune modulators perturb the human molecular network by common and unique strategies. Nature 2012, 487, 486–490. [Google Scholar] [CrossRef] [PubMed]
- Scaturro, P.; Stukalov, A.; Haas, D.A.; Cortese, M.; Draganova, K.; Płaszczyca, A.; Bartenschlager, R.; Götz, M.; Pichlmair, A. An orthogonal proteomic survey uncovers novel Zika virus host factors. Nature 2018, 561, 253–257. [Google Scholar] [CrossRef] [PubMed]
- Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; White, K.M.; O’Meara, M.J.; Rezelj, V.V.; Guo, J.Z.; Swaney, D.L.; et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020, 583, 459–468. [Google Scholar] [CrossRef]
- Stukalov, A.; Girault, V.; Grass, V.; Bergant, V.; Karayel, O.; Urban, C.; Haas, D.A.; Huang, Y.; Oubraham, L.; Wang, A.; et al. Multi-level proteomics reveals host-perturbation strategies of SARS-CoV-2 and SARS-CoV. bioRxiv 2020. [Google Scholar] [CrossRef]
- Jäger, S.; Cimermancic, P.; Gulbahce, N.; Johnson, J.R.; McGovern, K.E.; Clarke, S.C.; Shales, M.; Mercenne, G.; Pache, L.; Li, K.; et al. Global landscape of HIV-human protein complexes. Nature 2011, 481, 365–370. [Google Scholar] [CrossRef] [PubMed]
- Unterholzner, L.; Keating, S.E.; Baran, M.; Horan, K.A.; Jensen, S.B.; Sharma, S.; Sirois, C.M.; Jin, T.; Latz, E.; Xiao, T.S.; et al. IFI16 is an innate immune sensor for intracellular DNA. Nat. Immunol. 2010, 11, 997–1004. [Google Scholar] [CrossRef] [PubMed]
- Koppenol-Raab, M.; Sjoelund, V.; Manes, N.P.; Gottschalk, R.A.; Dutta, B.; Benet, Z.L.; Fraser, I.D.C.; Nita-Lazar, A. Proteome and Secretome Analysis Reveals Differential Post-transcriptional Regulation of Toll-like Receptor Responses. Mol. Cell. Proteom. 2017, 16, S172–S186. [Google Scholar] [CrossRef] [PubMed]
- Meissner, F.; Scheltema, R.A.; Mollenkopf, H.-J.; Mann, M. Direct proteomic quantification of the secretome of activated immune cells. Science 2013, 340, 475–478. [Google Scholar] [CrossRef] [PubMed]
- Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef] [PubMed]
- Wilson, R.C.; Doudna, J.A. Molecular mechanisms of RNA interference. Annu. Rev. Biophys. 2013, 42, 217–239. [Google Scholar] [CrossRef]
- Essletzbichler, P.; Konopka, T.; Santoro, F.; Chen, D.; Gapp, B.V.; Kralovics, R.; Brummelkamp, T.R.; Nijman, S.M.B.; Bürckstümmer, T. Megabase-scale deletion using CRISPR/Cas9 to generate a fully haploid human cell line. Genome Res. 2014, 24, 2059–2065. [Google Scholar] [CrossRef]
- Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef]
- Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
- Brinkman, E.K.; Chen, T.; de Haas, M.; Holland, H.A.; Akhtar, W.; van Steensel, B. Kinetics and Fidelity of the Repair of Cas9-Induced Double-Strand DNA Breaks. Mol. Cell 2018, 70, 801–813. [Google Scholar] [CrossRef]
- Kurosaki, T.; Maquat, L.E. Nonsense-mediated mRNA decay in humans at a glance. J. Cell Sci. 2016, 129, 461–467. [Google Scholar] [CrossRef] [PubMed]
- Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Krey, K.; Babnis, A.W.; Pichlmair, A. System-Based Approaches to Delineate the Antiviral Innate Immune Landscape. Viruses 2020, 12, 1196. https://doi.org/10.3390/v12101196
Krey K, Babnis AW, Pichlmair A. System-Based Approaches to Delineate the Antiviral Innate Immune Landscape. Viruses. 2020; 12(10):1196. https://doi.org/10.3390/v12101196
Chicago/Turabian StyleKrey, Karsten, Aleksandra W. Babnis, and Andreas Pichlmair. 2020. "System-Based Approaches to Delineate the Antiviral Innate Immune Landscape" Viruses 12, no. 10: 1196. https://doi.org/10.3390/v12101196
APA StyleKrey, K., Babnis, A. W., & Pichlmair, A. (2020). System-Based Approaches to Delineate the Antiviral Innate Immune Landscape. Viruses, 12(10), 1196. https://doi.org/10.3390/v12101196