Trends in Microbiology
Volume 27, Issue 3, March 2019, Pages 268-281
Journal home page for Trends in Microbiology

Review
Advances in Analyzing Virus-Induced Alterations of Host Cell Splicing

https://doi.org/10.1016/j.tim.2018.11.004Get rights and content

Highlights

Numerous viruses manipulate the splicing machinery in the host cell.

snRNP components and SR proteins are common viral targets.

Viral infections induce transcriptome-wide alterations in alternative splicing events.

Recent progress in RNAseq and bioinformatics is opening the way to an improved appreciation of the extent and impact of these alterations on pathogenesis.

Alteration of host cell splicing is a common feature of many viral infections which is underappreciated because of the complexity and technical difficulty of studying alternative splicing (AS) regulation. Recent advances in RNA sequencing technologies revealed that up to several hundreds of host genes can show altered mRNA splicing upon viral infection. The observed changes in AS events can be either a direct consequence of viral manipulation of the host splicing machinery or result indirectly from the virus-induced innate immune response or cellular damage. Analysis at a higher resolution with single-cell RNAseq, and at a higher scale with the integration of multiple omics data sets in a systems biology perspective, will be needed to further comprehend this complex facet of virus–host interactions.

Section snippets

Alteration of Cellular Splicing: A Complex Facet of Virus–Host Interactions

In higher eukaryotic cells most genes are transcribed as precursor messenger RNAs (pre-mRNAs) that undergo splicing, a maturation process through which RNA sequences (introns) are removed and the remaining sequences (exons) are ligated together. Splicing occurs in the nucleus and is catalyzed by the spliceosome, a large and highly dynamic ribonucleoprotein complex 1, 2. Most mammalian pre-mRNAs are subject to alternative splicing (AS), and human genes contain on average 8.8 exons and 7.8

Viral Manipulation of the Host Splicing Machinery

Viruses that replicate in the nucleus of infected cells and gain access to the splicing machinery (e.g., adenoviruses, herpesviruses, and influenza viruses) have evolved an expansion of their coding capacity by producing spliced viral mRNAs. However, manipulation of the host splicing machinery is not exclusive to nuclear viruses and has also been observed with viruses that replicate in the cytoplasm such as picornaviruses or flaviviruses (Table 1). This can be accounted for by the

Peering into Virus-Induced Alterations of Host AS Events

To date there are only a few studies that performed a transcriptome-wide microarray or RNAseq analysis of cellular AS events in virus-infected cells (Table 2). They were carried out with herpesviruses 26, 27, 28, reoviruses 24, 29, dengue viruses (DENV) 12, 30, Zika virus [31], and influenza viruses [32], and reveal several hundreds of host genes that show altered mRNA splicing upon infection. When examined, no correlation was found between changes in AS events and changes in mRNA expression

Toward a More Accurate View: Methodological Challenges

RNAseq transcriptomic analysis of AS is a challenging issue, and even more so when performed on a virus–host system. Accurate quantification of isoform abundance requires a high read number (about 50 million paired-end reads of at least 75 bp are recommended for the human transcriptome), which to date can only be provided by the Illumina technology. However, a serious limitation of Illumina RNAseq is that it relies on short reads, so that the resolution of exon connectivity and full-length

Integration of Isoform Data Sets in a Systems Biology Perspective

The rapid advancement of high-throughput technologies has led to the development of the systems biology field, which aims at modeling the properties of complex biological systems, and predicting their response to biological or chemical perturbations. The commonly used ‘top down’ systems biology approach turns RNAseq measurement of expression levels into a variable which can be included in a mathematical model, such as a generalized linear model or a multivariate analysis (principal component

Concluding Remarks

The importance of characterizing the transcriptome landscape of virus-infected cells at the splicing level is highlighted by recent studies which reveal significant AS alterations in response to viral infection. The observed changes in AS events, which are regulated through a very complex protein–RNA interaction network, can be either a direct consequence of viral manipulation of the host splicing machinery or result indirectly from virus-induced immune responses or cellular damages.

Acknowledgments

U.A. is part of the Pasteur–Paris University (PPU) International PhD Program, which has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 665807, and from the Institut Carnot Pasteur Microbes & Santé. C.B.P. has received funding from the Fondation pour la Recherche Médicale. This study was supported by the ANR ASTER (Grant No. ANR-16-CE23-0001) and the LabEx IBEID (Grant No. 10-LABX-0062).

References (124)

  • S. Ramani

    Human organoid cultures: transformative new tools for human virus studies

    Curr. Opin. Virol.

    (2018)
  • X. Liu

    Transcriptomic signatures differentiate survival from fatal outcomes in humans infected with Ebola virus

    Genome Biol.

    (2017)
  • J. Tisoncik-Go

    Integrated omics analysis of pathogenic host responses during pandemic H1N1 influenza virus infection: the crucial role of lipid metabolism

    Cell Host Microbe

    (2016)
  • A. Eisfeld

    Multi-platform ‘omics analysis of human Ebola virus disease pathogenesis

    Cell Host Microbe

    (2017)
  • Y. Liu

    Impact of alternative splicing on the human proteome

    Cell Rep.

    (2017)
  • M.L. Tress

    Alternative splicing may not be the key to proteome complexity

    Trends Biochem. Sci.

    (2017)
  • H.D. Li

    The emerging era of genomic data integration for analyzing splice isoform function

    Trends Genet.

    (2014)
  • W. Li

    Pushing the annotation of cellular activities to a higher resolution: predicting functions at the isoform level

    Methods

    (2016)
  • X. Yang

    Widespread expansion of protein interaction capabilities by alternative splicing

    Cell

    (2016)
  • I. Marazzi et al.

    Interference of viral effector proteins with chromatin, transcription, and the epigenome

    Curr. Opin. Microbiol.

    (2015)
  • S.R. Gonzales-van Horn et al.

    Making the mark: the role of adenosine modifications in the life cycle of RNA viruses

    Cell Host Microbe

    (2017)
  • E. Park

    The expanding landscape of alternative splicing variation in human populations

    Am. J. Hum. Genet.

    (2018)
  • B. Ibrahim

    A new era of virus bioinformatics

    Virus Res.

    (2018)
  • W. Deng

    Negative regulation of virus-triggered IFN-beta signaling pathway by alternative splicing of TBK1

    J. Biol. Chem.

    (2008)
  • O. Lakhdari

    TLR3 signaling is downregulated by a MAVS isoform in epithelial cells

    Cell Immunol.

    (2016)
  • S.M. Fica et al.

    Cryo-electron microscopy snapshots of the spliceosome: structural insights into a dynamic ribonucleoprotein machine

    Nat. Struct. Mol. Biol.

    (2017)
  • P. Papasaikas et al.

    The spliceosome: the ultimate RNA chaperone and sculptor

    Trends Biochem. Sci.

    (2015)
  • Y. Lee et al.

    Mechanisms and regulation of alternative pre-mRNA splicing

    Annu. Rev. Biochem.

    (2015)
  • M.K. Sakharkar

    Distributions of exons and introns in the human genome

    In Silico Biol.

    (2004)
  • M. Wang

    Circular RNAs: a novel type of non-coding RNA and their potential implications in antiviral immunity

    Int. J. Biol. Sci.

    (2017)
  • A.R. Kornblihtt

    Alternative splicing: a pivotal step between eukaryotic transcription and translation

    Nat. Rev. Mol. Cell Biol.

    (2013)
  • A.L. Richards

    Environmental perturbations lead to extensive directional shifts in RNA processing

    PLoS Genet.

    (2017)
  • A. Harwig

    The battle of RNA synthesis: virus versus host

    Viruses

    (2017)
  • K.M. Herbert et al.

    A tale of two RNAs during viral infection: how viruses antagonize mRNAs and small non-coding RNAs in the host cell

    Viruses

    (2016)
  • H.G. Rivas

    Shutoff of host gene expression in influenza A virus and herpesviruses: similar mechanisms and common themes

    Viruses

    (2016)
  • F.A. De Maio

    The dengue virus NS5 protein intrudes in the cellular spliceosome and modulates splicing

    PLoS Pathog.

    (2016)
  • L. Twyffels

    Shuttling SR proteins: more than splicing factors

    FEBS J.

    (2011)
  • E. Avota

    Measles virus induces expression of SIP110, a constitutively membrane clustered lipid phosphatase, which inhibits T cell proliferation

    Cell. Microbiol.

    (2006)
  • C. Zhu

    EFTUD2 is a novel innate immune regulator restricting hepatitis C virus infection through the RIG-I/MDA5 pathway

    J. Virol.

    (2015)
  • J.M. Howard et al.

    The RNAissance family: SR proteins as multifaceted regulators of gene expression

    Wiley Interdiscip. Rev. RNA

    (2015)
  • C. Estmer Nilsson

    The adenovirus E4-ORF4 splicing enhancer protein interacts with a subset of phosphorylated SR proteins

    EMBO J.

    (2001)
  • E.L. Prescott

    Human papillomavirus type 1 E1^E4 protein is a potent inhibitor of the serine-arginine (SR) protein kinase SRPK1 and inhibits phosphorylation of host SR proteins and of the viral transcription and replication regulator E2

    J. Virol.

    (2014)
  • K.S. Sciabica

    ICP27 interacts with SRPK1 to mediate HSV splicing inhibition by altering SR protein phosphorylation

    EMBO J.

    (2003)
  • T. Guirimand

    VirHostNet 2.0: surfing on the web of virus/host molecular interactions data

    Nucleic Acids Res.

    (2015)
  • E.E. Rivera-Serrano

    A cytoplasmic RNA virus alters the function of the cell splicing protein SRSF2

    J. Virol.

    (2017)
  • R. Batra

    RNA-binding protein CPEB1 remodels host and viral RNA landscapes

    Nat. Struct. Mol. Biol.

    (2016)
  • B. Hu

    Cellular responses to HSV-1 infection are linked to specific types of alterations in the host transcriptome

    Sci. Rep.

    (2016)
  • A.J. Rutkowski

    Widespread disruption of host transcription termination in HSV-1 infection

    Nat. Commun.

    (2015)
  • S. Boudreault

    Global profiling of the cellular alternative RNA splicing landscape during virus–host interactions

    PLoS One

    (2016)
  • O.M. Sessions

    Host cell transcriptome profile during wild-type and attenuated dengue virus infection

    PLoS Negl. Trop. Dis.

    (2013)
  • Cited by (43)

    • DNA virus replication

      2023, Molecular Medical Microbiology, Third Edition
    • Transcriptome-wide changes in gene expression, splicing, and lncRNAs in response to a live attenuated dengue virus vaccine

      2022, Cell Reports
      Citation Excerpt :

      We cannot exclude other mechanisms, such as alternative gene activation states in innate immune cells or many environmental and genetic factors that may affect functional diversity in gene expression patterns. The substantial overlap between lncRNAs and DS signals highlights pre-mRNA splicing as an essential mechanism for post-transcriptional regulation of gene expression, allowing immune cell functions to proceed more rapidly (Ashraf et al., 2019; Carpenter et al., 2014). Application of a modular repertoire framework to the analysis of transcriptional data reduces the bias of using predefined functional pathways and increases the correlation analysis information, assigning biological significance of the co-expressed genes to the observed trends (Gaucher et al., 2008; Querec et al., 2009).

    View all citing articles on Scopus
    View full text