Elsevier

Virus Research

Volume 283, 2 July 2020, 197962
Virus Research

Review
Role of SUMOylation in Human Oncogenic Herpesvirus Infection

https://doi.org/10.1016/j.virusres.2020.197962Get rights and content

Highlights

  • Regulation of SUMO signaling in the EBV/KSHV latent infection.

  • Role of SUMOylation in reactivation of EBV/KSHV lytic replication.

  • Cross-talk between SUMO and Ubiquitin controls EBV/KSHV latent and lytic replication.

Abstract

Post-translational modification of target proteins by the Small Ubiquitin-like Modifier (SUMO) plays a critical role in regulation of many cellular processes including transcription, RNA processing, protein trafficking, DNA repair, and chromosome segregation, and is also often hijacked by viral infections. Epstein-Barr Virus (EBV) and Kaposi’s sarcoma-associated Herpesvirus (KSHV), two human oncogenic herpesviruses with a typical life cycle of latent and lytic replication, have been shown to be associated with many human cancers. In the past decade, intensive studies have investigated the interplay between tumor virus infection and SUMO-modification. In this review, we summarize the current knowledge as to how SUMOylation can regulate latent and lytic replication of EBV and KSHV, and the strategies by which these oncogenic herpesviruses usurp the SUMO pathways to establish a favorable microenvironment to promote host cell survival and proliferation in latency, and reactivate virion production during lytic replication, which are critical contributors to the development of EBV/KSHV-associated human malignancies.

Introduction

The Small Ubiquitin-like Modifiers (SUMOs) are small peptides that consist of more than 100 amino acid residues, and have a molecular weight of approximately 11 KDa (Bayer et al., 1998; Hay, 2001; Isogai and Shirakawa, 2007)(Mossessova and Lima, 2000). The three-dimension structure of the SUMO molecule is quite similar to ubiquitin protein, while they exert different functions. It has been demonstrated that there are at least four isoforms of SUMO proteins, namely SUMO1, SUMO2, SUMO3, and SUMO4, encoded by human genome (Guo et al., 2015; Melchior, 2000). Among them, SUMO1 has 50% sequence homology with SUMO2, while SUMO2 and SUMO3 are highly similar, with only 3 amino acid differences (a homology of 95%), which is often referred as SUMO2/3 (Matic et al., 2008). Although both SUMO1 and SUMO2/3 play a major role in many cellular processes and are expressed in various tissues (Vertegaal et al., 2006), they differ greatly in function, and the substrate proteins bound to them in vivo are also different (Rosas-Acosta et al., 2005). SUMO1 mainly modifies protein molecules under physiological conditions, whereas SUMO2/3 modifies protein molecules that are in response to stress, and can even compensate for loss of SUMO1 in knockout mice (Saitoh and Chemistry, 2000). SUMO4 is encoded by a sequence within the intron of the TAB2 gene and is expressed mainly in the kidney tissue (Zhong et al., 2009). Even though the function of SUMO4 remains unclear, a few studies have linked it to the susceptibility of type 1 diabetes (Song et al., 2012; Zhong et al., 2009). Interestingly, recent studies have also identified that another isoform SUMO5, as a novel and controversial small ubiquitin-related modifier, is considered to be a nucleosome regulator with restricted tissue expression. However, it needs to be further studies are required to confirm whether SUMO5 is an endogenous protein required for translation (Liang et al., 2016).

Increasing studies have shown that SUMO modifications can act as adapters to regulate the interaction between target proteins and other proteins through a specific conserved cis-acting DNA sequence referred to as the SUMO-interaction motif (SIM)) or SUMO-binding motif (SBM) which is non-covalently bound to SUMO. Once the target protein is modified by SUMO, it can promote the interaction between the target protein and the protein containing the SIM/SBM (Geiss-Friedlander and Melchior, 2007; Kerscher, 2007). Similar to ubiquitylation, SUMOylation is also involved in multiple cellular processes, such as protein trafficking, protein stability, the ability of proteins to interact with substrates including proteins or DNA, and proteins function (Kerscher, 2007; Kerscher et al., 2006). Likewise, SUMOylation also requires a cascade of enzymatic steps catalyzed by the E1-activating enzyme, E2-conjugating enzyme, E3-ligating enzyme (i.e. the protein inhibitor of activated STAT [PIAS] family, RanBP2, MMS21, Pc2, and Topors) (Wimmer et al., 2012), and is removed by SUMO proteases (i.e., the Ulp and SUMO/sentrin-specific peptidase [SENP] family, DESI1, DESI2, and USPL1) (Hickey et al., 2012; Müller et al., 2001). However, in contrast to ubiquitylation, each SUMO isoform is initially synthesized as an immature precursor protein with a C-terminal tail which contains a short peptide of about 2 ∼ 11 residues (Huang et al., 2004). Therefore, it must undergo maturation when a SUMO-specific protease (Ulp) removes the C-terminal tail of the short pro-peptide and exposes the iconic C-terminal Gly-Gly module sequence (Wimmer et al., 2012), so that SUMO can then be transferred to the mature functional proteins to play its reversible role through modification (Geiss-Friedlander and Melchior, 2007).

In the process of SUMOylation, the first step is the activation of a mature protein by the SUMO activating enzyme (E1) (a heterodimer composed of Uba2 and Aos1), mediated through an ATP-dependent SUMO-adenylate intermediate, and is activated by a thioester bond to the cysteinyl residues of Uba2 (Geiss-Friedlander and Melchior, 2007). Next, the activated SUMO is transferred to the cystine residue of SUMO specific binding enzyme (E2) Ubc9 by a transester reaction, which forms a thioester linkage between the catalytic Cys residue of Ubc9 and the C-terminal carboxyl group of SUMO (Johnson and Blobel, 1997; Lee et al., 2000). Finally, Ubc9 transfers SUMO to the substrates by forming an isopeptide bond between the C-terminal Gly residue of SUMO and a Lys side chain of the target proteins (Müller et al., 2001; Saitoh et al., 1998). This process can be promoted by SUMO E3 ligase, which catalyzes the transfer of SUMO from Ubc9 to a substrate (Geiss-Friedlander and Melchior, 2007). Although experiments in vitro have shown that E1 and E2 are sufficient to regulate SUMOylation of various substrates, it has been demonstrated that the process of most SUMO conjugation to the target molecule still requires participation of E3-ligating enzyme in vivo.

Herpesviruses is a large class of DNA viruses that are morphologically distinct from other viruses, and have a linear, double-stranded DNA genome (encoding 100-200 genes) contained within a 16-icosahedral capsid (Davison et al., 2009; M. et al., 2013). According to their physicochemical properties, they are categorized into three subfamilies of α, β, and γ (ME and EM, 2017; Roizmann et al., 1992​). In comparison to other members of the herpesvirus family, γ-herpesvirus is the only lymphotropic viruses that can induce tumors from natural or laboratory host cells (Geng and Wang, 2015; Roizmann et al., 1992). Among these, Epstein-Barr virus (EBV, also referred to as human herpesvirus-4, HHV-4), and Kaposi’s sarcoma-associated herpesvirus (KSHV, also named human herpesvirus-8, HHV-8), are members of the human γ-herpesvirus subfamily, which have been shown to infect different cell types including B cells, endothelial cells, and epithelial cells, and are closely associated with many human malignancies (Table 1). EBV, the first human tumor virus isolated from Burkitt’s Lymphoma Cells, was identified for the first time by Epstein, Achong and Barr in 1964 (Martel et al., 2012), and causes more than 200,000 cases of cancer every year (Ko and Medicine, 2015). This virus could primarily cause various lymphomas, including Burkitt lymphoma (BL), Hodgkins lymphoma (HL), NK/T-cell lymphoma and immunodeficiency-associated lymphoma. In addition, it can also induce some carcinomas, such as nasopharyngeal carcinoma (NPC) and EBV-associated gastric carcinoma (EBV-GC) (Young et al., 2016). KSHV was discovered from Kaposi's sarcoma (KS) patients infected with human immunodeficiency virus by Chang and Moore in 1994 (Chang et al., 1994), and it is also associated with various lymphomas, including primary effusion lymphoma (PEL), Multicentric Castleman’s disease (MCD) and KSHV-associated germinotropic lymphoproliferative disorder (Reid, 2011). Because these cancers often appears in the condition of immunosuppression, it has made KSHV-associated malignancies an increasing global health concern with the persistence of the AIDS epidemic (Giffin and Damania, 2014).

Like other viruses in the herpesviruses family, EBV and KSHV are capable of establishing latent infection in host cells and reactivating for lytic replication under the certain condition (Longnecker and Neipel, 2016). It has been assumed that the transforming capacities of EBV and KSHV are predominantly mediated by viral latent proteins, along with low level of reactivation of infectious virus production from latently infected cells (Okano, 1998). During latency, EBV expresses all eight latency-associated proteins, namely nuclear antigens EBNA1, 2, 3A–3C, LP, and the three latent membrane proteins LMP1 and LMP2A and 2B (Murray et al., 1992; Styles et al., 2017)(Jha et al., 2015). According to the expression patterns of the EBV latent proteins in different tumors, the EBV latency programs are classified into four main types of 0, I, II and III. The latency III program is detected predominantly in EBV-transformed lymphoblastoid cell lines and lymphoproliferative diseases in immunocompromised individuals with all the EBNAs and LMPs expressed (Rea et al., 1994). Latency II is found in the most Hodgkin’s lymphoma and nasopharyngeal carcinoma with only expression of EBNA1, LMP1, LMP2-A and LMP2-B (Thorley-Lawson, 2001). In latency I, there are two EBER genes expressed as in proliferating memory B cells and EBV-associated BL, the BART transcripts, and EBNA1 (EBV nuclear antigen 1) (Dirmeier et al., 2003). In latency 0, the healthy individuals carrying EBV with expression of non-translated viral RNAs (EBERs and miRNAs) but no viral protein (Gregory et al., 1998; Hochberg et al., 2004). In contrast to EBV, KSHV expresses a limited number of proteins including LANA, vFLIP, vCyclin during latent infection (Dittmer et al., 1998; Jennera and Boshoffb, 2002), which play critical roles in cell proliferation, apoptosis, and escape of the host immune surveillance. The majority of the KSHV genome remains silent during latency. However, expression of the lytic master regulator RTA, encoded by ORF50 (Gradoville et al., 2000; Lukac et al., 1999), is necessary and sufficient to switch from latent to lytic replication (Martin et al., 2000). Similarly, the expression of EBV lytic genes Zta and Rta, also play essential roles in switching from latent to lytic replication.

Emerging evidence has shown that many viral proteins can also be a SUMO-binding substrate, and alternation of SUMO signaling can promote virus infection in host cells [The progression about global effect of SUMOylation on virus could refer to recent reviews (Isaacson and Ploegh, 2009)(Zhang et al., 2009)(Peter and Viruses, 2015)(Everett et al., 2013)]. The interaction between the human oncogenic herpesviruses and SUMO modifying system in host cells has also been comprehensively investigated in the past decade. In this review, we mainly summarize the role of SUMOylation in EBV/KSHV infection, particularly as to how the SUMO system alters latent and lytic replication of the EBV/KSHV life cycles, and how these viruses interfere with cell cycle progression of the host by hijacking the SUMO modification pathway.

Section snippets

SUMOylation plays a pivotal role in human oncogenic gammaherpesvirus infection

SUMOylation plays critical roles in a variety of cellular process, including gene transcriptional regulation, cell cycle regulation, signal transduction, the DNA damage response (DDR), and the regulation of protein-protein interactions. Therefore it is not surprising that EBV and KSHV hijack the SUMOylation machinery by expressing viral proteins during latent and lytic replication in the infected host cell. Emerging evidence has suggested that these viruses, EBV and KSHV, have adopted a

Cross-talk between SUMO and Ubiquitin controls latent and lytic replication

Increasing and compelling evidence have elucidated that there is ongoing cross-talk between the SUMO and Ubiquitin systems through an enzyme referred to as SUMO-targeted Ubiquitin ligase (STUbLs), which recognizes SUMO (or poly-SUMO) conjugated proteins and targets them for ubiquitin-mediated degradation. For example, KSHV encodes at least encoded two proteins (including K-Rta and LANA2), which function as STUbLs (Perry et al., 2007). K-Rta contains SIMs and a RING finger domain and can inhibit

Conclusion and Future perspective

In summary, SUMOylation is a highly dynamic and reversible process that covalently attaches SUMO to the lysine residue of its target proteins. SUMO signaling can also regulate protein function by altering their intracellular location, turnover, ability to interact with other proteins, or their ability to interact with DNA (Kerscher, 2007; Kerscher et al., 2006; Kroetz and Medicine, 2005). Therefore, it can play a critical role in a vast array of cellular processes. Given the importance of

Acknowledgments

This work was supported by the National Natural Science Foundation of China (81672015, 81971930), the National Key Research and Development Program of China (2019ZX09721001), and the Research Program on Biosafety Guarantee Technology of High-level Biosafety Laboratory and Important Pathogen Laboratory (2018ZX10734401-004). Public Health Service support from the National Institutes of Health, National Cancer Institute provides ongoing support for ESR (R01-CA171979, P01-CA174439 and R01-CA177423).

References (93)

  • L.K. Chang et al.

    Post-translational Modification of Rta of Epstein-Barr Virus by SUMO-1

    (2004)
  • P.C. Chang et al.

    Kaposi\"s Sarcoma-associated Herpesvirus (KSHV) Encodes a SUMO E3 ligase That Is SIM-dependent and SUMO-2/3-specific.

    (2010)
  • L. Giffin et al.

    KSHV: Pathways to Tumorigenesis and Persistent Infection

    Advances in Virus Research

    (2014)
  • R.T. Hay

    Protein modification by SUMO

    (2001)
  • E.S. Johnson et al.

    Ubc9p Is the Conjugating Enzyme for the Ubiquitin-like Protein Smt3p.

    (1997)
  • I. Matic et al.

    In Vivo Identification of Human Small Ubiquitin-like Modifier Polymerization Sites by High Accuracy Mass Spectrometry and an in Vitro to in Vivo Strategy

    Molecular & cellular proteomics: MCP

    (2008)
  • E. Mossessova et al.

    Ulp1-SUMO Crystal Structure and Genetic Analysis Reveal Conserved Interactions and a Regulatory Element Essential for Cell Growth in Yeast.

    (2000)
  • R.J. Murray et al.

    Identification of target antigens for the human cytotoxic T cell response to Epstein-Barr virus (EBV): implications for the immune control of EBV-positive malignancies

    (1992)
  • H. Saitoh et al.

    Ubc9p and the conjugation of SUMO-1 to RanGAP1 and RanBP2

    (1998)
  • A.C.O. Vertegaal et al.

    Distinct and Overlapping Sets of SUMO-1 and SUMO-2 Target Proteins Revealed by Quantitative Proteomics

    (2006)
  • Y.C. Yang et al.

    Role of RNF4 in the Ubiquitination of Rta of Epstein-Barr Virus

    (2013)
  • L. Zhang et al.

    Interplay between poxviruses and the cellular ubiquitin/ubiquitin-like pathways

    (2009)
  • Bayer, P., Arndt, A., Metzger, S., Mahajan, R., Melchior, F., Jaenicke, R., Becker, J.r., 1998. Structure determination...
  • G.L. Bentz et al.

    LMP1-Induced Sumoylation Influences the Maintenance of Epstein-Barr Virus Latency through KAP1

    (2015)
  • G.L. Bentz et al.

    Epstein-Barr Virus Latent Membrane Protein 1 Regulates the Function of Interferon Regulatory Factor 7 by Inducing Its Sumoylation

    (2012)
  • G.L. Bentz et al.

    Epstein-Barr Virus Latent Membrane Protein 1 (LMP1) C-Terminal-Activating Region 3 Contributes to LMP1-Mediated Cellular Migration via Its Interaction with Ubc9

    (2011)
  • Q. Cai et al.

    A Unique SUMO-2-Interacting Motif within LANA Is Essential for KSHV

    Latency.

    (2013)
  • M.A. Calderwood et al.

    The Epstein-Barr Virus LF2 Protein Inhibits

    Viral Replication.

    (2008)
  • L.K. Chang et al.

    Enhancement of Transactivation Activity of Rta of Epstein-Barr Virus by RanBPM.

    (2008)
  • P.-C. Chang et al.

    SUMO and KSHV Replication.

    (2014)
  • P.C. Chang et al.

    The chromatin modification by SUMO-2/3 but not SUMO-1 prevents the epigenetic activation of key immune-related genes during Kaposi’s sarcoma associated herpesvirus reactivation.

    (2013)
  • P.C. Chang et al.

    Kruppel-Associated Box Domain-Associated Protein-1 as a Latency Regulator for Kaposi’s Sarcoma-Associated Herpesvirus and Its Modulation by the Viral Protein Kinase.

    (2009)
  • Y. Chang et al.

    Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma.

    (1994)
  • A.J. Davison et al.

    The order <Emphasis Type="Italic">Herpesvirales

    (2009)
  • C.F. De La Cruz-Herrera et al.

    A genome-wide screen of Epstein-Barr virus proteins that modulate host SUMOylation identifies a SUMO E3 ligase conserved in herpesviruses

    PLOS Pathogens

    (2018)
  • L. Ding et al.

    Identification of viral SIM-SUMO2-interaction inhibitors for treating primary effusion lymphoma

    PLOS Pathogens

    (2019)
  • U. Dirmeier et al.

    Latent membrane protein 1 is critical for efficient growth transformation of human B cells by Epstein-Barr virus

    (2003)
  • D. Dittmer et al.

    A Cluster of Latently Expressed Genes in Kaposi’s Sarcoma-Associated Herpesvirus

    Journal of Virology

    (1998)
  • R.D. Everett et al.

    Interplay between viruses and host sumoylation pathways

    (2013)
  • R. Geiss-Friedlander et al.

    Concepts in sumoylation: a decade on

    (2007)
  • L. Geng et al.

    Epstein-Barr Virus-associated lymphoproliferative disorders: Experimental and clinical developments

    International Journal of Clinical & Experimental Medicine

    (2015)
  • L. Gradoville et al.

    Kaposi\"s Sarcoma-Associated Herpesvirus Open Reading Frame 50/Rta Protein Activates the Entire Viral Lytic Cycle in the HH-B2 Primary Effusion Lymphoma Cell Line

    (2000)
  • Babcock Gregory et al.

    EBV Persistence in Memory B Cells In Vivo

    (1998)
  • D. Guo et al.

    A functional variant of SUMO4, a new IκBα modifier, is associated with type 1 diabetes

    (2015)
  • S.R. Hagemeier et al.

    Sumoylation of the Epstein-Barr Virus BZLF1 Protein Inhibits Its Transcriptional Activity and Is Regulated by the Virus-Encoded Protein Kinase

    (2010)
  • C.M. Hickey et al.

    Function and regulation of SUMO proteases.

    (2012)
  • D. Hochberg et al.

    Demonstration of the Burkitt\"s lymphoma Epstein-Barr virus phenotype in dividing latently infected memory cells in vivo

    (2004)
  • W.C. Huang et al.

    Crystal structures of the human SUMO-2 protein at 1.6 A and 1.2 A resolution: Implication on the functional differences of SUMO proteins

    (2004)
  • M.K. Isaacson et al.

    Ubiquitination, Ubiquitin-like Modifiers, and Deubiquitination in Viral Infection

    (2009)
  • S. Isogai et al.

    Protein modification by SUMO

    (2007)
  • Y. Izumiya et al.

    Kaposi\"s Sarcoma-Associated Herpesvirus K-bZIP Represses Gene Transcription via SUMO Modification.

    (2005)
  • Y. Izumiya et al.

    Kaposi\"s Sarcoma-Associated Herpesvirus-Encoded Protein Kinase and Its Interaction with K-bZIP.

    (2007)
  • Izumiya, Y., Kobayashi, K., Kim, K.Y., Pochampalli, M., Izumiya, C., Shevchenko, B., Wang, D.-H., Huerta, S.B.,...
  • Izumiya, Y., Kobayashi, K., Kim, K.Y., Pochampalli, M., Izumiya, C., Shevchenko, B., Wang, D.-H., Huerta, S.B.,...
  • R.G. Jennera et al.

    The molecular pathology of Kaposi’s sarcoma-associated herpesvirus

    Biochim Biophys Acta

    (2002)
  • Oliver Kerscher

    SUMO junction—what\"s your function? New insights through SUMO-interacting motifs

    (2007)
  • Cited by (7)

    • Control of animal virus replication by RNA adenosine methylation

      2022, Advances in Virus Research
      Citation Excerpt :

      Relatively little is known about the regulation of the protein factors that function within the m6A and m6Am pathways but as has been shown repeatedly for other regulatory processes such as protein phosphorylation, sumoylation and ubiquitylation, viruses can often prove especially useful in uncovering post-translational modifications that control cellular pathways (Song et al., 2021). For instance, METTL3 can be conjugated to SUMO1 at four or more positions, impacting its catalytic activity (Du et al., 2018) and a number of viruses have developed strategies to interfere SUMO-signaling (Zhang et al., 2020). Global m6A levels are reduced during reactive oxygen species (ROS)-induced stress resulting in increased expression of genes involved in of genome integrity and DNA repair (Yu et al., 2021).

    • Interplays between inflammasomes and viruses, bacteria (pathogenic and probiotic), yeasts and parasites

      2020, Immunology Letters
      Citation Excerpt :

      Human gammaherpesvirus 4 (Epstein−Barr virus (EBV) is a DNA virus of the Herpesvirus (HHV) family. EBV can replicate in B-lymphocytes, epithelial cells, or dendric cells and is associated with a variety of tumors, including Burkitt lymphoma, immunocompromise-associated lymphomas, and nasopharyngeal cell carcinoma [29]. EBV-positive endemic BL cell line HH514-16 and peripheral blood mononuclear cells (PBMC) isolated from the blood of patients with NOMID syndrome had heterozygous germline mutations in their NLRP3 gene which showed assembly of the TXNIP−NLRP3 inflammasome and activation of procaspase-1 and active caspase-1 during lytic growth.

    View all citing articles on Scopus
    View full text