Abstract
Pathogens usurp a variety of host pathways via protein–protein interactions to ensure efficient pathogen replication. Despite the existence of an impressive toolkit of systematic and unbiased approaches, we still lack a comprehensive list of these PPIs and an understanding of their functional implications. Here, we highlight the importance of harnessing genetic diversity of hosts and pathogens for uncovering the biochemical basis of pathogen restriction, virulence, fitness, and pathogenesis. We further suggest that integrating physical interaction data with orthogonal types of data will allow researchers to draw meaningful conclusions both for basic and translational science.
Introduction
During the course of infection, pathogens use their proteins to hijack and re-wire a myriad of host biochemical processes — events that are required for efficient pathogen propagation. Therefore, characterization of host–pathogen protein–protein interactions (PPIs) greatly aids in the understanding of the mechanisms underlying pathogen replication. To date, several approaches have been employed to identify host–pathogen PPIs for viruses, bacteria, and parasites, including yeast two-hybrid (Y2H) and affinity purification coupled with mass spectrometry (AP/MS) [1–8] (Box 1). However, moving from systematic descriptions to functional/clinical relevance requires establishment of a genotype–phenotype relationship through the integration of global and reductionist approaches [9]. Usually, this can be accomplished through targeted characterization of interactions using secondary and even tertiary screens after initial, unbiased proteomic interrogation. In contrast, a comparative approach where PPIs are probed against functionally distinct genetic variants of a pathogen or host protein can yield biochemical insight into the observed phenotype and help to functionally prioritize host proteins. This approach, referred to as ‘comparative proteomics’, leverages functional host and/or pathogen diversity to infer the biochemical basis for genotype–phenotype relationships. Examining PPIs between physiologically relevant genetic variants across the host–pathogen interface will provide a basis for uncovering molecular determinants of disease outcomes.
Box 1. Detection of protein–protein interactions.
Protein–protein interactions (PPIs) can be detected using yeast-two-hybrid (Y2H) or affinity purification/mass spectrometry (AP/MS). Y2H yields insights into pairwise PPIs, and takes advantage of baits and preys that are both linked to yeast transcriptional activators. When a bait successfully interacts with a prey, the transcriptional activators are brought together to drive yeast colony growth or reporter gene expression. Y2H technology has the advantage of scale and speed, as many baits can be screened rapidly once an appropriate prey library of complementary DNA (cDNA) has been created. In AP/MS, bait proteins are affinity purified using a bait-specific antibody, or via over-expression of affinity-tagged proteins. The resulting purified protein complexes are analyzed by MS to determine interacting prey. While more labor intensive, AP/MS is better suited to studying PPIs in the context of stoichiometric protein complexes. Unlike Y2H, preys detected by AP/MS are not subject to cloning-related biases and are present in their endogenous context. While low levels of endogenous expression may favor PPI detection by Y2H, AP/MS is better suited to studying PPIs for membrane proteins that may not effectively translocate to the nucleus for Y2H screening. Recent advances in MS, such as selected reaction monitoring, allow for a highly quantitative assessment of differential PPIs across diverse sets of sample that vary in host, pathogen or time [57]. Finally, when combined with double affinity purification of a viral bait and host prey, AP/MS has the added flexibility to be used to deduce other members of multi-protein complexes [6,20•].
In this review, we highlight the importance of harnessing genetic diversity of both the host and pathogen when designing global proteomics studies. Comparative proteomics can explore the diversity of a population at the level of single nucleotide polymorphisms (SNPs) or as broadly as millions of years of evolutionary history, from the perspective of both the host and/or the pathogen (Figure 1a). Genetic determinants of virulence and/or pathogenesis have been described for several pathogens and comparative PPI mapping has the potential to uncover the underlying biochemical basis of these in vivo outcomes. Furthermore, comparing within or between species can highlight conserved and unique cellular pathways that are hijacked by pathogens (Figure 1b). In conjunction with orthogonal approaches, leveraging host/pathogen diversity with a comparative proteomics framework can greatly advance basic science and clinical goals (Figure 2).
Figure 1.
Comparative PPIs: genetic diversity impacts host–pathogen PPIs. (a) Resolution of pathogen or host diversity variables for comparative PPI study design. Each class of experimental variable (pathogen or host) is ordered according to resolution or complexity. Along each scale are either published examples (referenced) or hypothetical comparative PPI experiments. The examples listed are by no means exhaustive. (b) Representation of host–pathogen comparative approach. The interrogation of how pathogen diversity can interact with the host can highlight unique and shared cellular pathways. This can manifest itself as inhibitory (red lines), or as promoting replication (black arrows). Abbreviations: CRL: cullin-RING ubiquitin ligase, TALEN: transcription activator-like effector nuclease, CRISPR: clustered regularly interspaced short palindromic repeat, APC: antigen-presenting cell.
Figure 2.
Using comparative proteomics to move from systems to function. Building and refining systems models of biological processes requires the integration of orthogonal tools and datasets. The feedback between systems and reductionist approaches greatly improves the resolution of models of basic biological processes and strategies for translating these discoveries to the clinic. Using sequence diversity to inform comparative PPI studies can facilitate the translation of biochemical data into therapies. Abbreviations: PPI: protein–protein interaction, NGS: next generation sequencing, 1000 Genomes: http://www.1000genomes.org, HapMap: http://hapmap.ncbi.nlm.nih.gov, CirSeq: circular sequencing, RNAi: RNA interference, TALEN: transcription activator-like effector nuclease, CRISPR: clustered regularly interspaced short palindromic repeat, AP/MS: affinity purification mass spectrometry, Y2H: yeast-2-hybrid, XL-MS: cross-linking mas spectrometry, SRM: selected reaction monitoring, DIA: data-independent acquisition, PTM: post-translation modification, iPSCs: induced pluripotent stem cells.
The pathogen diversity axis
Genetic diversity among pathogens can directly impact host–pathogen PPIs and functional outcomes (Figure 1b), which has been demonstrated in several studies. For example, Greninger and colleagues explored the conservation of host–virus PPIs across several picornavirus 3A proteins [10•]. Picornavirus 3A remodels Golgi membranes into virus replication compartments and the host protein PI4KIIIβ, a regulator of Golgi membranes, is required for the replication of diverse picornaviruses [11]. The authors utilized a comparative AP/MS approach with a diverse panel of picornaviruses and found that in most cases, the interaction of 3A with PI4KIIIβ was mediated by the acyl-CoA binding protein ACBD3. A panel of 3A point mutants was used to establish a positive correlation between the ability of 3A to interact with PI4KIIIβ and the efficiency of virus replication. This study also highlighted that distinct but functionally related proteins may mediate a shared host factor dependence on virus replication. Although PI4KIIIβ kinase activity is required for entero-virus 71 replication [12], its 3A protein was shown to recruit PI4KIIIβ via an interaction with ACAD9, another acyl-CoA binding protein, and not ACBD3 [10•]. Similarly, a systematic comparative PPI study of the lentivirus protein Vif revealed differential biochemical requirements for conserved pathogen protein function. Vif is an accessory factor that is required to counteract the host cytidine deaminase APOBEC3 [13]. In the absence of Vif, APO-BEC3 is packaged into daughter virions and causes the hypermutation of viral genomes upon infection through the deamination of cytidines into uridines [14–17]. Vif binds to and degrades APOBEC3 through the recruitment of a Cullin-RING ubiquitin ligase complex [18]. Importantly, Vif-mediated degradation of APOBEC3 is conserved across the lentivirus family [19]. Through comparative AP/MS of different Vif lineages, Kane and colleagues demonstrated that primate lentivirus Vifs require the non-canonical host transcription factor CBFβ for complex formation, whereas non-primate Vifs required either no host co-factor, or a different non-canonical host co-factor, the cellular peptidyl prolyl isomerase CYPA [20•]. Thus, the Vif protein co-evolved with its hosts to hijack unrelated co-factors in order to counteract a potent cellular innate antiviral response. The authors further suggest that the use of CYPA as a non-canonical co-factor for Vif-APOBEC3 complex formation could potentially serve two purposes by disrupting both APOBEC3 and CYPA function, in a manner similar to the dual-hijacking of CBFβ by HIV-1 Vif [21].
Additionally, pathogen diversity can be used to reveal common themes for host–pathogen PPIs, as was done in a broad study of host–virus PPIs using 70 viral proteins from 30 distinct human viruses. Pichlmair and colleagues found that DNA viruses were specifically enriched for PPIs that link cell cycle to chromosome biology and transcription, whereas PPIs with RNA viruses are enriched for processes that degrade or detect viral RNA, both processes that are known to be important for the respective class of viruses [22]. In another notable study, Rozenblatt-Rosen and colleagues studied 123 tumor-causing virus proteins using AP/MS and demonstrated how different viruses can manipulate the Notch signaling pathway to influence cell proliferation via distinct host–pathogen interactions [23••]. Future proteomic studies of this nature that are more quantitative as well as systematic across larger families of pathogens will provide additional key information about the host–pathogen interface.
Furthermore, comparative approaches utilizing the significant diversity within a given pathogen species in virulence and/or pathogenicity afford the opportunity to uncover PPIs that correlate, and ultimately determine clinical outcomes in the host. In a pair of papers led by White and colleagues, the oncogenic E6 and E7 proteins from several strains of human papilloma virus (HPV) with differing cancer risks were subjected to AP/MS in an effort to identify differential PPIs that could contribute to tumorigenesis [24,25]. These studies are notable for their comprehensive nature and experimental design; however, future work remains to elucidate the biochemical determinants of tumorigenesis for high-risk HPV strains. In summary, by carefully selecting pathogen variants, from highly related strains to evolutionary distinct species, comparative proteomics can identify disease-specific PPIs and general classes of hijacked cellular pathways.
The host diversity axis
Host diversity can impact mechanisms of pathogen restriction, virulence, and fitness, and the resolution to examine this diversity can span from SNPs to phyla (Figure 1b). Diversity between hosts at a single gene can result in profoundly different host–pathogen PPIs and phenotypes, the most well known instance being the disruption of a PPI between CCR5 and the human immunodeficiency virus-1 (HIV-1) envelope protein. A 32 nucleotide deletion in CCR5 renders homozygous carriers resistant to HIV-1 infection [26], and is the basis for an anti-HIV-1 gene therapy currently in clinical trials [27]. At the species level, host-specific PPIs can result in pathogen restriction in one species, and permissivity in the other [28•]. For example, Ashour and colleagues showed that dengue virus (DENV) NS5 binds to and degrades human STAT2 to restrict antiviral signaling. However, this restriction is based on a species-specific host–pathogen PPI, as NS5 does not bind mouse STAT2, which is thus able to restrict DENV replication in mouse cells [29]. Global proteomic studies with pathogen virulence effectors conducted in disparate species are well suited to uncover functionally relevant targets of pathogen antagonists, such as STAT2 and DENV NS5, or alternatively, viral targets of cellular restriction factors such as TRIM5α and HIV-1 Capsid [30]. In contrast, conservation of host–pathogen PPIs across species can lead to insights into virulence. For example, Memisevic and colleagues discovered three previously unknown virulence factors of the pathogenic bacterium Burkholderia mallei by screening secreted B. mallei proteins for interactions with human and mouse proteins using Y2H. The mouse model of B. mallei infection recapitulates much of the pathophysiology observed in human infections, and the authors hypothesized that host–pathogen PPIs that are conserved between the two hosts may mediate virulence [31•]. This comparative approach filtered the number of potential virulence factors from 49 to eight, three of which resulted in reduced virulence in vivo when mutated. Finally, vector-borne pathogens, such as DENV, are ideal candidates for cross-phylum comparisons. In one such Y2H analysis, interactions were determined between DENV and its two hosts (human and mosquito), and 14 PPIs were conserved across host species [32]. While the function of these PPIs remains to be elucidated, these conserved interactions could facilitate replication in two divergent hosts and could serve as ideal drug targets. These studies highlight how host diversity can be harnessed to systematically identify mechanisms of restriction, virulence and potentially fitness.
Coupling orthogonal datasets to advance functional understanding
No single approach is sufficient to fully characterize the host–pathogen interface, and PPI analysis is one of several complementary approaches that, when integrated together, can provide a more comprehensive network view of hijacked cellular processes across hosts and pathogens. Orthogonal technologies include monitoring post-translational modifications [33–36], gene expression profiling [23••,37•], and genome-wide genetic screening [38–42]. Moreover, as more host–pathogen PPIs are uncovered, common themes and differences will be revealed when studies are compared to one another [1]. Non-pathogen related disease datasets could also be used to provide functional insight into host–pathogen PPIs. For example, integration of host genetic data from The Cancer Genome Atlas (http://cancergenome.nih.gov/), which globally describes mutations in different types of cancer, could be used to highlight the importance of specific PPIs in cancer development by identifying common oncogenic pathways that are also targeted by cancer-causing viruses, including HPV, hepatitis C virus and Kaposi’s sarcoma-associated herpesvirus (KSHV) [43]. Finally, visualization of integrated orthogonal datasets can help identify common themes, and software such as Cytoscape (www.cytoscape.org), STRING (http://string.embl.de), and NeXO (www.nexontology.org) are examples of intuitive tools for such a task. The integration of these data into network-centric models will undoubtedly lead to a deeper understanding of how pathogens rewire host cellular processes [44].
Analysis of host and pathogen genetics on evolutionary timescales can also provide insights into the evolutionary interplay between host and pathogen. For example, Jäger and colleagues found a significantly higher level of sequence conservation for human proteins that interact with HIV-1 as compared to the complete human proteome [6]. This observation suggests that HIV-1 targets essential cellular pathways via physical interaction with highly conserved host factors. In a different approach, Davis and colleagues leveraged the 1000 Genomes Project sequence data to identify signatures of recent positive selection in host factors that physically interact with KSHV-specific viral proteins [1]. Positive selection implies that these mutations confer a fitness advantage to the organism, and such signatures of evolution have been used to identify the biochemical basis of host restriction factor activity [45], and their antagonism by pathogens [46]. Thus, enrichment for positive selection in pathogen-interacting host factors suggests that interactions at these nodes may represent instances of host–pathogen antagonism, and such evolutionary analysis could be used to guide functional studies. Similarly, sequence variation among well-established clinical cohorts could be used to identify susceptibility or protective mutations that impact host–pathogen PPIs. Moreover, with the advent of induced pluripotent stem cell (iPSC) technology, disease-related haplotypes could be tested directly for differential host–pathogen PPIs in patient-derived iPSCs. Pathogen sequence databases could also be used to inform how highly conserved (or variable) residues impact host–pathogen PPIs (Table 1). Moreover, new methods such as CirSeq [47] will allow researchers to map dynamic fitness landscapes of pathogens by tracking the genetic composition of RNA virus populations over time, which can be used to inform PPI studies. When coupled with genetic interaction mapping of well-characterized point mutants [48], mapping high and low fitness pathogen mutations to specific host–pathogen PPIs will provide significant functional and structural insight into the pathogen fitness landscape.
Table 1.
Complementary pathogen databases. Several publicly available databases compile a variety of systems-level analysis, including host–pathogen interactions, pathogen sequence diversity and protein structures.
| Tool | Description | Website |
|---|---|---|
| EuPathDB | Database portal for non-viral eukaryotic pathogens with access to genomic-scale data deposited in other databases (e.g. PlasmoDB, ToxoDB, etc.) | http://eupathdb.org/eupathdb/ |
| FLAVIdB | Database of antigenic data of flaviviruses | http://cvc.dfci.harvard.edu/flavi/ |
| HIV systems biology [44] | Collection of web-based tools for exploring basics of HIV lifecycle, gene enrichment analysis, and viral association studies | http://hivsystemsbiology.org/ |
| Influenza Research Database | Collection of influenza sequences, structures, immunological and host factor data | http://www.fludb.org/ |
| Los Alamos National Lab HIV sequence database | Authoritative collection of HIV sequence variants | http://www.hiv.lanl.gov/ |
| PlasmoDB | Comprehensive database of systems data with user friendly data mining tools for various Plasmodium species | http://www.plasmodb.org/ |
| VirusMentha | Resource of virus-virus and virus-host PPIs | http://virusmentha.uniroma2.it/ |
| NCBI Viral Genomes | Database of viral and viroid genome sequences | http://www.ncbi.nlm.nih.gov/genome/viruses/ |
Caveats and potential solutions
To best capture physiologically relevant PPIs with available technologies, several caveats must be considered. While the caveats discussed here are by no means exhaustive, they represent some of the major limitations associated with comparative host–pathogen PPI mapping. PPIs are dynamic by nature and the context by which PPIs are probed can greatly influence results. Traditional PPI experimental approaches (Box 1) are best suited for detecting stable PPIs; however, weak and/or transient PPIs can also be captured using specialized techniques such as cross-linking mass spectrometry [49,50] or BioID, a novel proximity-based biotinylation method [51•,52], in combination with AP/MS. Pathogen replication is itself a dynamic process and comparative host–pathogen PPI mapping across the timescale of pathogen replication or in specific cellular compartments could be used to infer temporal and/or spatial roles of host–pathogen PPIs. Cristea and colleagues recently demonstrated that time-dependent host–pathogen interactions could be captured during Sindbis virus replication by AP/MS [53,54]. Similarly, Moorman and colleagues identified spatiotemporally regulated PPIs during human cytomegalovirus infection involved in virion assembly [55]. As new quantitative mass spectrometry techniques such as selected reaction monitoring or data-independent acquisition are combined with AP/MS [56,57], spatial-dependent and time-dependent host–pathogen PPIs can be quantified during pathogen infection.
One of the greatest limitations for probing host–pathogen PPIs relates to the cellular context in which these PPIs are examined. Some interactions across the host–pathogen interface may only exist in the context of infection or in select cell types. While ideal, genetically inserting an affinity-tag into a pathogen genome and examining PPIs during natural infection is not possible in many cases. However, recent advances in genome engineering technologies such as CRISPR (clustered regularly interspaced short palindromic repeat) now make it possible to affinity-tag host proteins in their natural context [58], allowing for the examination of host–pathogen PPIs during infection. Both of these approaches simultaneously eliminate the possibility of artifacts introduced via over-expression of affinity-tagged proteins and could be used to inform analysis of over-expression PPI datasets. Finally, differential expression levels of bait proteins between cell or tissue types could confound comparative PPI results, but global protein abundance quantification could help correct these biases. Importantly, careful targeted experiments should be used to address whether a differential host–pathogen PPI is the result of a meaningful biophysical difference in the binding interface, or differential gene expression.
Conclusions
Our understanding of genetic diversity and our ability to quantitatively track changes in PPIs is rapidly improving with the development of new technologies, such that comparative mapping of host–pathogen PPIs has the potential to transform how genotype–phenotype relationships are linked to the underlying biochemical interaction. By taking advantage of host and pathogen diversity, researchers can now conduct studies to identify conserved and differential interactions, which will ultimately offer insight into the basic biology of pathogenesis, and lead to more effective therapeutics.
Acknowledgments
We would like to acknowledge Mike Shales, Ruth Hüttenhain, Erik Verschueren, Judd Hultquist and Joshua Kane for critical reading of the manuscript. Funding was provided by NIAID AI112262 to PSS, NIAID AI116410 to JAW and P50 GM082250, P01 AI090935, P50 GM081879, P01 AI091575, DARPA-10-93-Prophecy-PA-008, R01 DA033773, P01 HL089707, U19 AI106754, R01 GM084279 and P01 CA177322-01A1 to NJK.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1.Davis ZH, et al. Global mapping of herpesvirus–host protein complexes reveals a transcription strategy for late genes. Mol Cell. 2015;57:349–360. doi: 10.1016/j.molcel.2014.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ramage HR, et al. A combined proteomics/genomics approach links hepatitis C virus infection with nonsense-mediated mRNA decay. Mol Cell. 2015;57:329–340. doi: 10.1016/j.molcel.2014.12.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mirrashidi KM, et al. Global mapping of the inc-human interactome reveals that retromer restricts chlamydia infection. Cell Host Microbe. 2015;18:109–121. doi: 10.1016/j.chom.2015.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Watanabe T, et al. Influenza virus–host interactome screen as a platform for antiviral drug development. Cell Host Microbe. 2014;16:795–805. doi: 10.1016/j.chom.2014.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mehra A, et al. Mycobacterium tuberculosis type VII secreted effector EsxH targets host ESCRT to impair trafficking. PLoS Pathog. 2013;9:e1003734. doi: 10.1371/journal.ppat.1003734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jäger S, et al. Global landscape of HIV–human protein complexes. Nature. 2012;481:365–370. doi: 10.1038/nature10719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dyer MD, et al. The human–bacterial pathogen protein interaction networks of Bacillus anthracis, Francisella tularensis, and Yersinia pestis. PLoS ONE. 2010;5:e12089. doi: 10.1371/journal.pone.0012089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ahn H-J, Kim S, Kim H-E, Nam H-W. Interactions between secreted GRA proteins and host cell proteins across the paratitophorous vacuolar membrane in the parasitism of Toxoplasma gondii. Korean J Parasitol. 2006;44:303–312. doi: 10.3347/kjp.2006.44.4.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Carter H, Hofree M, Ideker T. Genotype to phenotype via network analysis. Curr Opin Genet Dev. 2013;23:611–621. doi: 10.1016/j.gde.2013.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10•.Greninger AL, Knudsen GM, Betegon M, Burlingame AL, Derisi JL. The 3A protein from multiple picornaviruses utilizes the golgi adaptor protein ACBD3 to recruit PI4KIIIβ. J Virol. 2012;86:3605–3616. doi: 10.1128/JVI.06778-11. A comprehensive comparative PPI study that utilized not only diverse species of picornaviruses and a thorough mutational scan of the Aichi 3A protein, but also functional viral fitness assays. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Jackson WT. Poliovirus-induced changes in cellular membranes throughout infection. Curr Opin Virol. 2014;9:67–73. doi: 10.1016/j.coviro.2014.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Arita M, et al. Phosphatidylinositol 4-kinase III beta is a target of enviroxime-like compounds for antipoliovirus activity. J Virol. 2011;85:2364–2372. doi: 10.1128/JVI.02249-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sheehy AM, Gaddis NC, Choi JD, Malim MH. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature. 2002;418:646–650. doi: 10.1038/nature00939. [DOI] [PubMed] [Google Scholar]
- 14.Lecossier D, Bouchonnet F, Clavel F, Hance AJ. Hypermutation of HIV-1 DNA in the absence of the Vif protein. Science. 2003;300:1112. doi: 10.1126/science.1083338. [DOI] [PubMed] [Google Scholar]
- 15.Zhang H, et al. The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA. Nature. 2003;424:94–98. doi: 10.1038/nature01707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mangeat B, et al. Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature. 2003;424:99–103. doi: 10.1038/nature01709. [DOI] [PubMed] [Google Scholar]
- 17.Harris RS, et al. DNA deamination mediates innate immunity to retroviral infection. Cell. 2003;113:803–809. doi: 10.1016/s0092-8674(03)00423-9. [DOI] [PubMed] [Google Scholar]
- 18.Yu X, et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science. 2003;302:1056–1060. doi: 10.1126/science.1089591. [DOI] [PubMed] [Google Scholar]
- 19.Larue RS, Lengyel J, Jónsson SR, Andrésdóttir V, Harris RS. Lentiviral Vif degrades the APOBEC3Z3/APOBEC3H protein of its mammalian host and is capable of cross-species activity. J Virol. 2010;84:8193–8201. doi: 10.1128/JVI.00685-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20•.Kane JR, et al. Lineage-specific viral hijacking of non-canonical E3 ubiquitin ligase cofactors in the evolution of vif anti-APOBEC3 activity. Cell Rep. 2015;11:1236–1250. doi: 10.1016/j.celrep.2015.04.038. This study is an excellent example of how leveraging species diversity in the host can uncover unexpected mechanisms to explain shared cellular pathways targeted by a pathogen. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kim DY, et al. CBFβ stabilizes HIV Vif to counteract APOBEC3 at the expense of RUNX1 target gene expression. Mol Cell. 2013;49:632–644. doi: 10.1016/j.molcel.2012.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pichlmair A, et al. Viral immune modulators perturb the human molecular network by common and unique strategies. Nature. 2012;487:486–490. doi: 10.1038/nature11289. [DOI] [PubMed] [Google Scholar]
- 23••.Rozenblatt-Rosen O, et al. Interpreting cancer genomes using systematic host network perturbations by tumour virus proteins. Nature. 2012;487:491–495. doi: 10.1038/nature11288. This study illustrates the power of not only comparing several disparate species of pathogen, but also how integrating orthogonal datasets can clarify PPI data. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.White EA, et al. Comprehensive analysis of host cellular interactions with human papillomavirus E6 proteins identifies new E6 binding partners and reflects viral diversity. J Virol. 2012;86:13174–13186. doi: 10.1128/JVI.02172-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.White EA, et al. PNAS Plus: systematic identification of interactions between host cell proteins and E7 oncoproteins from diverse human papillomaviruses. Proc Natl Acad Sci U S A. 2012;109:E260–E267. doi: 10.1073/pnas.1116776109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wilen CB, Tilton JC, Doms RW. HIV: cell binding and entry. Cold Spring Harb Perspect Med. 2012;2:a006866. doi: 10.1101/cshperspect.a006866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tebas P, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370:901–910. doi: 10.1056/NEJMoa1300662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28•.Daugherty MD, Malik HS. Rules of engagement: molecular insights from host–virus arms races. Annu Rev Genet. 2012;46:677–700. doi: 10.1146/annurev-genet-110711-155522. An excellent review on the growing focus of co-evolving virulence factors and their host restriction factor targets. [DOI] [PubMed] [Google Scholar]
- 29.Ashour J, et al. Mouse STAT2 restricts early dengue virus replication. Cell Host Microbe. 2010;8:410–421. doi: 10.1016/j.chom.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Puvvada MP, Patel SS. Role of trim5α in the suppression of cross-species transmission and its defence against human immunodeficiency virus. Curr HIV Res. 2013;11:601–609. doi: 10.2174/1570162x12666140306124543. [DOI] [PubMed] [Google Scholar]
- 31•.Memisević V, et al. Novel Burkholderia mallei virulence factors linked to specific host–pathogen protein interactions. Mol Cell Proteomics. 2013;12:3036–3051. doi: 10.1074/mcp.M113.029041. A creative comparative PPI approach that leveraged host species conservation to uncover novel bacterial virulence factors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mairiang D, et al. Identification of new protein interactions between dengue fever virus and its hosts, human and mosquito. PLOS ONE. 2013;8:e53535. doi: 10.1371/journal.pone.0053535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wojcechowskyj JA, et al. Quantitative phosphoproteomics reveals extensive cellular reprogramming during HIV-1 entry. Cell Host Microbe. 2013;13:613–623. doi: 10.1016/j.chom.2013.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Stahl JA, et al. Phosphoproteomic analyses reveal signaling pathways that facilitate lytic gammaherpesvirus replication. PLoS Pathog. 2013;9:e1003583. doi: 10.1371/journal.ppat.1003583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kim W, et al. Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol Cell. 2011;44:325–340. doi: 10.1016/j.molcel.2011.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xu G, Paige JS, Jaffrey SR. Global analysis of lysine ubiquitination by ubiquitin remnant immunoaffinity profiling. Nat Biotechnol. 2010;28:868–873. doi: 10.1038/nbt.1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37•.Shapira SD, et al. A physical and regulatory map of host-influenza interactions reveals pathways in H1N1 infection. Cell. 2009;139:1255–1267. doi: 10.1016/j.cell.2009.12.018. A pioneering ‘systems biology’ study of how a virus, in this case Influenza, rewires and utilizes host cellular pathways for replication. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.König R, et al. Human host factors required for influenza virus replication. Nature. 2010;463:813–817. doi: 10.1038/nature08699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bushman FD, et al. Host cell factors in HIV replication: meta-analysis of genome-wide studies. PLoS Pathog. 2009;5 :e1000437. doi: 10.1371/journal.ppat.1000437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li Q, et al. A genome-wide genetic screen for host factors required for hepatitis C virus propagation. Proc Natl Acad Sci U S A. 2009;106:16410–16415. doi: 10.1073/pnas.0907439106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Derré I, Pypaert M, Dautry-Varsat A, Agaisse H. RNAi screen in Drosophila cells reveals the involvement of the Tom complex in Chlamydia infection. PLoS Pathog. 2007;3:1446–1458. doi: 10.1371/journal.ppat.0030155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Philips JA, Rubin EJ, Perrimon N. Drosophila RNAi screen reveals CD36 family member required for mycobacterial infection. Science. 2005;309:1251–1253. doi: 10.1126/science.1116006. [DOI] [PubMed] [Google Scholar]
- 43.Krogan NJ, Lippman S, Agard DA, Ashworth A, Ideker T. The cancer cell map initiative: defining the hallmark networks of cancer. Mol Cell. 2015;58:690–698. doi: 10.1016/j.molcel.2015.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ideker T, Krogan NJ. Differential network biology. Mol Syst Biol. 2012;8:565. doi: 10.1038/msb.2011.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mitchell PS, et al. Evolution-guided identification of antiviral specificity determinants in the broadly acting interferon-induced innate immunity factor MxA. Cell Host Microbe. 2012;12 :598–604. doi: 10.1016/j.chom.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lim ES, et al. The ability of primate lentiviruses to degrade the monocyte restriction factor SAMHD1 preceded the birth of the viral accessory protein Vpx. Cell Host Microbe. 2012;11 :194–204. doi: 10.1016/j.chom.2012.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Acevedo A, Brodsky L, Andino R. Mutational and fitness landscapes of an RNA virus revealed through population sequencing. Nature. 2014;505:686–690. doi: 10.1038/nature12861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Braberg H, et al. From structure to systems: high-resolution, quantitative genetic analysis of RNA polymerase II. Cell. 2013;154:775–788. doi: 10.1016/j.cell.2013.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kaake RM, et al. A new in vivo cross-linking mass spectrometry platform to define protein–protein interactions in living cells. Mol Cell Proteomics. 2014;13:3533–3543. doi: 10.1074/mcp.M114.042630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sinz A. Investigation of protein–protein interactions in living cells by chemical crosslinking and mass spectrometry. Anal Bioanal Chem. 2010;397:3433–3440. doi: 10.1007/s00216-009-3405-5. [DOI] [PubMed] [Google Scholar]
- 51•.Lambert J-P, Tucholska M, Go C, Knight JDR, Gingras A-C. Proximity biotinylation and affinity purification are complementary approaches for the interactome mapping of chromatin-associated protein complexes. J Proteomics. 2015;118:81–94. doi: 10.1016/j.jprot.2014.09.011. An elegant implementation of the BioID approach to map the interactome of chromatin associated proteins yielding complementary interaction data to traditional AP/MS, by allowing for detection of low-abundance interactors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Roux KJ, Kim DI, Burke B. BioID: a screen for protein–protein interactions. Curr Protoc Protein Sci. 2013;74(Unit 19.23) doi: 10.1002/0471140864.ps1923s74. [DOI] [PubMed] [Google Scholar]
- 53.Cristea IM, et al. Host factors associated with the Sindbis virus RNA-dependent RNA polymerase: role for G3BP1 and G3BP2 in virus replication. J Virol. 2010;84:6720–6732. doi: 10.1128/JVI.01983-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Cristea IM, et al. Tracking and elucidating alphavirus–host protein interactions. J Biol Chem. 2006;281:30269–30278. doi: 10.1074/jbc.M603980200. [DOI] [PubMed] [Google Scholar]
- 55.Moorman NJ, Sharon-Friling R, Shenk T, Cristea IM. A targeted spatial–temporal proteomics approach implicates multiple cellular trafficking pathways in human cytomegalovirus virion maturation. Mol Cell Proteomics. 2010;9:851–860. doi: 10.1074/mcp.M900485-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zheng Y, et al. Temporal regulation of EGF signalling networks by the scaffold protein Shc1. Nature. 2013;499:166–171. doi: 10.1038/nature12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bisson N, et al. Selected reaction monitoring mass spectrometry reveals the dynamics of signaling through the GRB2 adaptor. Nat Biotechnol. 2011;29:653–658. doi: 10.1038/nbt.1905. [DOI] [PubMed] [Google Scholar]
- 58.Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346:1258096. doi: 10.1126/science.1258096. [DOI] [PubMed] [Google Scholar]


