Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 2.
Published in final edited form as: Virus Res. 2015 Oct 23;212:1–11. doi: 10.1016/j.virusres.2015.10.002

Long noncoding RNAs in viral infections

Puri Fortes 1,*, Kevin Morris 2,3
PMCID: PMC4744516  NIHMSID: NIHMS732475  PMID: 26454188

Abstract

Viral infections induce strong modifications in the cell transcriptome. Among the RNAs whose expression is altered by infection are long noncoding RNAs (lncRNAs). LncRNAs are transcripts with potential to function as RNA molecules. Infected cells may express viral lncRNAs, cellular lncRNAs and chimeric lncRNAs formed by viral and cellular sequences. Some viruses express viral lncRNAs whose function is essential for viral viability. They are transcribed by polymerase II or III and some of them can be processed by unique maturation steps performed by host cell machineries. Some viral lncRNAs control transcription, stability or translation of cellular and viral genes. Surprisingly, similar functions can be exerted by cellular lncRNAs induced by infection. Expression of cellular lncRNAs may be altered in response to viral replication or viral protein expression. However, many cellular lncRNAs respond to the antiviral pathways induced by infection. In fact, many lncRNAs function as positive or negative regulators of the innate antiviral response. Our current knowledge about the identity and function of lncRNAs in infected cells is very limited. However, research into this field has already helped in the identification of novel cellular pathways and may help in the development of therapeutic tools for the treatment of viral infections, autoimmune diseases, neurological disorders and cancer.

Keywords: lncRNAs, proviral, antiviral, IFN response, lncRNA processing, transcription regulation, RNA interference

1. Long non coding RNAs

The firsts transcriptome analyses, carried out a decade ago, led to the unexpected discovery that while the majority of the genome is transcribed, only ~2% of the genome is transcribed into mRNAs that are then translated into proteins. While most investigation to date has focused on the study protein functionality in health and disease, it is now apparent that the majority of the genome is transcribed as noncoding RNAs (Cawley et al., 2004; ENCODE Project Consortium et al., 2007). Among the noncoding genome, long noncoding RNAs (lncRNAs) are a particularly rich category. Some estimates suggest that the human genome contains more than 90,000 genes and approximately 60,000 of them are lncRNAs, while other estimates suggest that the number of lncRNA genes could reach closer to 200,000 (Iyer et al., 2015). Notably the number of lncRNAs scales with developmental complexity, with primates exhibiting the largest number of lncRNA genes followed by mouse and scaling down accordingly to bacteria (Ulitsky et al., 2011). The function of most of these non-coding transcripts remains largely unknown (Cech and Steitz, 2014). Indeed great efforts are being made to identify the complete collection of lncRNA genes in different species and assign to them functional categories.

LncRNAs are generally classified as transcripts longer than 200 nucleotides with no identifiable protein coding capacity and with potential to function predominantly as RNA molecules (Cech and Steitz, 2014). LncRNAs are generally thought to be transcribed by RNA polymerase II, have one or more introns eliminated by splicing, and some appear to be polyadenylated at their 3′ end (Derrien et al., 2012). In essence lncRNAs are generally similar to mRNAs (Garitano et al., 2013). However, compared to mRNAs, lncRNAs are preferentially nuclear, more tissue specific, poorly expressed and poorly conserved (Djebali et al., 2012). This may be one reason as to why these transcripts, until recently, have gone unnoticed. Once transcribed, some lncRNAs form a secondary/tertiary structure and interact with proteins, DNA or other RNAs to fulfil regulatory functions (Cech and Steitz, 2014). Some lncRNAs act in trans, away from their site of transcription, while other act in cis, controlling the expression of neighbouring genes (Gupta et al., 2010). Several examples exist of lncRNAs that control gene expression by guiding chromatin remodelers, inducing DNA bending, regulating transcription, splicing, translation, or RNA stability (Gupta et al., 2010; Khalil et al., 2009; Tsai et al., 2010; Yao et al., 2010; Feng et al., 2006; Tripathi et al., 2010; Carrieri et al., 2012; Cesana et al., 2011; Yoon et al., 2012). Other lncRNAs have been described to serve as scaffolds of subnuclear structures or to control protein transport or post-translational modifications (Mao et al., 2011; Willingham et al., 2005; Wang et al., 2014). The function of several lncRNAs has been shown to be essential in cell homeostasis, growth and differentiation. Further, several lncRNAs have been described as playing relevant roles in viral infection and in the antiviral response (reviewed in this Special Issue).

2. Protein-modulated control of the antiviral response

The antiviral response is induced in response to viral infection by detection of pathogen-associated molecular patterns (PAMPs) (Schneider et al., 2014). PAMPs can be detected by canonical sensors, such as the retinoic acid-inducible gene I (RIG-I) or toll-like receptors (TLRs) or by non-canonical factors such as the protein kinase R (PKR) (Andrejeva et al., 2004; Kang et al., 2002; Yoneyama et al., 2004; Alexopoulou et al., 2001; Diebold et al., 2004; Arnaud et al., 2010; Arnaud et al., 2011). Most TLRs signal through the myeloid differentiation primary response gene 88 (MyD88) and the tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6) to activate the nuclear factor-κB (NF-κB). The endosome-bound TLR3 activates the innate immune response through the toll-IL-1 receptor domain-containing adaptor inducing interferon (TRIF) (Li et al., 2012) and RIG-I requires the E3 ubiquitin ligase TRIM25 (Gack et al., 2007), which facilitates the interaction of RIG-I with the mitochondrial antiviral-signaling protein (MAVS) (Fig. 1). When MAVS or TRIF are triggered, they lead to the activation of the Interferon (IFN) regulatory factor (IRF) 3 and NF-κB (Gack et al., 2007; Loo and Gale, 2011). Together, IRF3 and NF-κB induce transcription of type I IFN. Further, IRF3 and NF-κB on their own can induce transcription of IFN regulated genes (ISGs) and proinflammatory cytokines, respectively.

Fig. 1. Several lncRNAs regulate the antiviral pathway.

Fig. 1

See the text for details. Proteins involved in the antiviral response are in blue. LncRNAs are in red. Ubiquitin is in yellow. P denotes phosphorylation. Activation is shown with an arrow. Inhibition is depicted with an interrupted line.

Secreted type I IFN (IFNα, IFNβ and others) binds to IFN receptors and activates JAK/STAT (Janus-activated kinase/signal transducer and activator of transcription) signaling (Ivashkiv and Donlin, 2014). STAT1 and 2 coupled to IRF9 activate the transcription of ISGs which function to induce an antiviral state in the cell. STAT1, STAT2, IRF1, 3, 7, 9 or OAS (2′-5′ oligoadenylate synthase) are ISGs that work to increase cell sensitivity to PAMPs and reinforce IFN signaling. Other ISGs block viral infection by affecting several steps of the viral cell cycle, including viral entry and release, and RNA replication, translation and stability. Surprisingly, some ISGs function as negative regulators of the IFN response (Schneider et al., 2014). These regulators are essential to control the response and to help the cell return to homeostasis.

Among the regulators of the IFN pathway, factors involved in protein ubiquitination seem to have special relevance (reviewed in Davis and Gack, 2015). Ubiquitin is a polypeptide that can be covalently bound to a protein substrate (reviewed in Clague et al., 2015). Ubiquitin binding requires an E1 activating enzyme, an E2 conjugating enzyme and an E3 ligase that selects the target. Ubiquitin release from the target requires a deubiquitinating enzyme (DUB). Many polyubiquitinated proteins are targeted to the proteasome for degradation. However, ubiquitination can also induce endosomal and autophagosomal degradation or alter protein functionality. Several sensors and mediators of IFN synthesis and signaling modify their stability or functionality by ubiquitination. These include MyD88, TRIF or nuclear IRF3, degraded after the action of several E3 ligases, whose increase reduces IFN synthesis (Xue et al., 2012; Yang et al., 2013a; Wang et al., 2015). In contrast, USP13 (Ubiquitin Specific Peptidase 13) is a DUB that deubiquitinates STAT1 to avoid degradation and potentiate IFN signaling (Yeh et al., 2013). More complex is the ubiquitin-regulation of RIG-I. RIG-I can be ubiquitinated and degraded by RNF125 (Ring Finger Protein 125) and deubiquitinated and stabilized by USP4 (Wang et al., 2013; Luo et al., 2013). Further, RIG-I can be ubiquitinated and activated by TRIM25, which, in turn, can be ubiquitinated and degraded by LUBAC (linear ubiquitin chain assembly complex) and deubiquitinated and stabilized by USP15 (Davis and Gack, 2015) (Fig. 1). Interestingly, many ubiquitin related proteins are induced by IFN to enhance (TRIM25) or to block (TRIM13 or 21) the IFN response (Zhang et al., 2013a; Narayan et al., 2014). Similarly, many viruses express ubiquitin ligases or DUBs to alter ubiquitin regulation and favor replication (Kwon and Ahn, 2013) or express proteins that block the action of several ubiquitin enzymes that lead to the activation of the IFN pathway (Rajsbaum et al., 2012).

Similar regulation is exerted by ubiquitin-like proteins such as the small ubiquitin-like modifier (SUMO) or ISG15. IFN induces SUMOylation of STAT-1, which leads to decreased STAT1 phosphorylation and signaling (Maarifi et al., 2015). ISG15 and the complete ISGylation machinery are induced by IFN and other viral sensors to play proviral and antiviral roles (Arnaud et al., 2011). As the ISGylation machinery acts cotranscriptionally, ISG15 conjugation occurs preferentially in viral proteins and cellular antiviral factors whose translation is increased after infection (Durfee et al., 2010). ISGylation of viral proteins affects their functionality and therefore, exerts and antiviral function (Zhao et al., 2013). ISGylation of IRF3 and RIG-I affects their ubiquitination, leading to an increased stability of IRF3 and decreased functionality of RIG-I. This leads to decreased IFN production and increased viral replication (Shi et al., 2010; Broering et al., 2010; Kim et al., 2008). In line with this proviral role, free ISG15 also stabilizes the deISGylase USP18, which binds IFNA2R (IFN alpha 2 receptor) and blocks IFN signaling (Malakhova et al., 2003; Malakhova et al., 2006; Zhang et al., 2015).

A tight regulation of functionality and stability by ubiquitin and ubiquitin-like proteins is only one of the constraints that host and viral proteins suffer after infection. Further, protein translation is also blocked, mainly, by PKR activation. PKR is an IFN-induced kinase with several functions (reviewed in Dabo and Meurs, 2012). One of the key functions of PKR after activation is the phosphorylation of eukaryotic translation initiation factor eIF2a, a subunit of the eIF2 factor (Fig. 1). eIF2 together with GTP and Met-tRNA, forms a ternary complex required to position the initiating Met-tRNA codon and start translation. GTP hydrolysis allows recycling of the complex. Phosphorylation of eIF2a prevents the regeneration of GTP in the ternary complex and then blocks cap-dependent translation.

3. Viruses and lncRNAs

Viruses have developed several strategies to fight the antiviral immune system and to increase the efficacy of replication (reviewed in Horner and Gale, 2013). Recent studies show that some of these strategies function through lncRNA molecules. This is not surprising because:

  1. Viruses adopt similar molecular mechanisms than those that function in the host cell. In this way the virus can employ host cell machineries. Host cells express many functional lncRNAs, making likely that viruses also express them.

  2. Viruses are under strong selective pressure from the cell. Molecules that constrain viral replication should tend to be selected more easily. It should be easier for RNA viruses, which employ error-prone polymerases, to utilize ncRNAs to elude selective pressures exerted by the cell on the virus. This is because lncRNAs appear to tolerate mutations better than coding genes (Ulitsky et al., 2011; Ponjavic et al., 2007). As such, one would predict that it is more likely that novel viral protective and/or viral repressive cellular functions will develop from lncRNAs.

  3. Effectiveness of molecules that boost replication should be enhanced when they are invisible to the cellular antiviral sensors or the acquired immune system. This is an advantage of lncRNAs compared to protein factors and PAMPs.

  4. As indicated above, the cell responds to infection with upregulation of machineries that affect the cellular proteome. Translation of new proteins is blocked by PKR activation and pre-existing proteins are affected in their stability and functionality by the binding of ubiquitin and ubiquitin-like proteins. LncRNAs should be immune to this protein-hostile environment.

These are reasons why viral lncRNAs are expected to be generated, function to improve viral viability and resist the cellular antiviral atmosphere. Noticeable, lncRNAs employed by the virus to enhance replication could be cellular lncRNAs whose expression is induced by the virus in the infected cell. Similarly, the infected cell has also evolved to express cellular lncRNAs that counteract viral infection.

3. 1. lncRNAs in infected cells

Transcriptome analysis of cells infected with different viruses has led to the identification of viral lncRNAs and cellular lncRNAs whose expression is altered after infection.

3.1.1. Cell lncRNAs altered by infection

When transcriptomes of infected and control cells are contrasted, there are distinct changes in particular lncRNAs. Expression of some of these lncRNAs is altered in response to the expression of viral proteins. For example the expression of X protein (HBx) from Hepatitis B virus (HBV) in cultured cells or transgenic mice, leads to the downregulation of the tumor suppressor lncRNA DREH (down-regulated expression by HBx) (Huang et al., 2013; Moyo et al., 2015). Similarly, expression of Nef at early times post-infection with human immunodeficiency virus (HIV) reduces expression of the lncRNA NRON (noncoding repressor of nuclear factor of T cells (NFAT)) (Imam et al., 2015; Lazar et al., 2015). As NRON is a repressor of NFAT, decreased levels of NRON allow NFAT-mediated transcription of HIV genes at early times post-infection. Interestingly, expression of Vpu protein at late times post-infection with HIV increases NRON expression. This results in decreased NFAT functionality and decreased transcription of HIV genes at late times, prior to viral release and apoptosis. Thus, levels of NRON change during HIV infection to allow transcription regulation. These results also teach us that a proper evaluation of the levels of lncRNAs in infected cells should be performed at different times post-infection.

Expression of cellular lncRNAs can also be altered in response to the antiviral response induced by infection. Activation of canonical or non-canonical PAMP sensors, or treatment with IFN or TNFα (which induces transcription by NF-κB) leads to changes in the expression of many lncRNAs (reviewed in Valadkhan and Gunawardane, 2015; Carpenter, 2015; Aune and Spurlock, 2015). Similar to the induction pattern observed for coding genes, most lncRNAs appear upregulated by the treatment. However, 80% of the lncRNAs whose expression is altered after treatment of THP1 cells with a TLR2 agonist were downregulated (Li et al., 2014). Similarly, treatment of primary liver cells or the liver HuH7 cell line with IFN also resulted in the downregulation of many lncRNAs while most altered coding genes were upregulated (Kambara et al., 2014; Carnero et al., 2014; Barriocanal et al., 2015). While the function of these downregulated lncRNAs is unknown, these results suggest that IFN-regulated lncRNA genes could play different regulatory roles than IFN-induced coding genes in the control of the antiviral response.

Interestingly, the function of several lncRNAs whose expression is altered during the antiviral response has been described. As expected, most seem to regulate inflammation or antiviral gene expression by several mechanisms. Some are inducers, such as NEAT1 (nuclear enriched abundant transcript 1), which increases IL8 (interleukin 8) by sequestering the IL8-inhibitor SFPQ (splicing factor proline/glutamine rich) in paraspeckles (see below) (Imamura et al., 2014) (Fig. 1). Many are negative regulators, such as lincR-COX2 (cyclooxygenase 2), Lethe, IL7R (interleukin 7 receptor), NRAV (negative regulator of antiviral response) or NRIR (negative regulator of IFN response) (see below) (Carpenter et al., 2013; Rapicavoli et al., 2013; Cui et al., 2014; Kambara et al., 2014; Ouyang et al., 2014). Considering the small number of infection-related lncRNAs studied to date and the relatively high proportion that function as negative regulators it is tempting to speculate that lncRNAs may play a key role in the silencing of the inflammatory or the IFN pathway (Fig. 1).

3.1.2. Viral lncRNAs

The existence of viral noncoding RNAs has been known for years (Mathews, 1975; Marx, 1972). In fact, viroids were first described in 1971. Viroids and some virusoids are plant pathogens whose genome is a lncRNA, as it lacks any protein coding capacity (reviewed in Gago-Zachert, 2015; Shimura and Masuta, 2015). Viroid genomes are small 246–401 nucleotide, circular, single-stranded RNA molecules capable of autonomous replication. Viroids have been considered living relics of the hypothetical RNA world (Flores et al., 2014). As they do not encode viral proteins, the genome is transmitted by tools used to process plants for agricultural purposes. Contamination is facilitated by the high stability of the viroid genome. The genome remains infectious for long periods in dry tissue or on dry surfaces as the viroids are resistant to heat and many antiviral chemicals. Inactivation is achieved with hypochlorite and a non-rigorous cleaning of the tools causes plant contaminations and huge economic problems. Some viroids are transmitted by aphids or transferred from plant to plant by leaf contact.

The expression of noncoding RNAs from animal viruses has also been described decades ago. Some of them are very abundant, such as VA (virus-associated) RNAs from adenovirus, EBERs (Epstein-Barr virus (EBV)-encoded small RNA) from EBV or PAN (polyadenylated nuclear) RNA from Kaposi’s sarcoma-associated herpesvirus (KSHV) (reviewed in Vachon and Conn, 2015; Iwakiri, 2015; Conrad, 2016). Viral non-coding RNAs are transcribed from polymerase III (VA RNAs, EBERs) or polymerase II promoters (PAN RNA) and some can be polyadenylated (PAN RNA). Then, some accumulate preferentially in the nucleus (PAN RNA, EBERs) or in the cytoplasm (VA RNAs) of infected cells. Interestingly, some viral lncRNAs are processed by unique maturation steps performed by host cell machineries. This is the case of subgenomic flavivirus RNAs (sfRNAs) or the LAT (latency-associated transcript) from Herpes simplex virus type 1 (HSV-1).

Flavivirus genomic RNA can be degraded by the 5′ to 3′ exonuclease Xrn1. While processing, Xrn1 stalls on secondary structures located towards the 3′ end and creates degradation intermediates named sfRNAs or Xrn1-resistant RNAs (xrRNAs) (reviewed in Charley and Wilusz, 2015). This is very rare, as well-studied RNAs with strong secondary or tertiary structures are not resistant to degradation by Xrn1. Instead, flavivirus RNAs have developed a special structure to stop Xrn1 processing and accumulate to high levels. The resulting sfRNAs from arthropod-borne flaviviruses are noncoding, 300–500 nucleotides long and contain the 3′-end of the genomic RNA. Recently, it has been described that few nucleotides from the 5′ end of hepatitis C virus (HCV) genomic RNA can also be degraded by Xrn1. In this case the enzyme stalls in the IRES sequence and xrRNAs are generated (Moon et al., 2015). Whether these subgenomic HCV RNAs are non-coding transcripts is currently unknown.

During latency, HSV-1 produces only the capped, spliced and polyadenylated LAT (reviewed in Tycowski et al., 2015). Surprisingly, the spliced exon RNA is hard to detect, as it is processed further to generate several miRNAs, while the lariat with intronic sequences is very stable (Kang et al., 2006). Intron stability seems related with a secondary structure that selects a non-consensus branch site sequence for splicing (using guanosine instead of adenosine) (Mukerjee et al., 2004). Other stable intronic sequence RNAs (sisRNAs) have been recently identify in eukaryotes (Hesselberth, 2013).

Finally, it should be highlighted that, similar to coding transcripts, expression of viral lncRNAs may be tightly regulated. Some viral lncRNAs, such as LAT, are only expressed in the latent phase, while others in the lytic phase. This is the case of PAN RNA, whose high levels of expression in the lytic phase are due to increased transcription and, primarily, to several molecular mechanisms that work together or independently to result in PAN RNA stability (reviewed in Conrad, 2015).

3.1.3. Chimeric lncRNAs

The study of HBV-associated hepatocellular carcinomas (HCC) led to the identification of transcripts that result from fusions between human and viral sequences (Lau et al., 2014; reviewed in Moyo et al., 2015). This is not surprising as HBV genome can integrate into the host genome. Integration occurs preferentially within repetitive sequences such as long interspersed nuclear elements (LINEs). Unexpectedly, 23.3 % of HCC express chimeras between HBx protein and host LINE1 sequences. These chimeric transcripts, named HBx-LINE1, have oncogenic effects as they activate Wnt signaling. It is currently unknown whether many viruses can benefit from transcription of chimeric lncRNAs.

3. 2. Function of lncRNAs in infected cells

Given that viruses have such compact genomes and packaging of nucleic acids into virions is at a premium it is reasonable to surmise that every viral lncRNA should have a relevant function. Similar to what has been described for cellular lncRNAs, functionality requires RNA structure and lncRNA binding to other factors such as proteins or other transcripts. The structure of several lncRNAs has been studied in detail. Some viroid RNAs have self-complementarity to form rod-shape structures (Gago-Zachert, 2015). Their dsRNA nature protects the viroid genome but makes it targetable by the cellular RNA interference (RNAi) machinery. Structure of EBERs and VA RNAs is very similar and in fact EBERs can partially substitute VA RNAs in adenoviral infections (Rosa et al., 1981). In some cases the secondary structure is required for protein binding. Many cellular proteins have been described to bind viral ncRNAs. These include:

  1. Proteins required for RNA processing, such as Xrn1 in the case of sfRNAs (see above) or La in the case of EBERs and VA RNAs. La is required for proper folding and stability of polymerase III transcripts (Copela et al., 2006).

  2. Factors from the RNAi machinery: Exportin 5 and Dicer.

  3. Proteins involved in the immune response: Canonical and non-canonical sensors and mediators of IFN synthesis and signaling.

  4. Transcription regulators: transcription factors and chromatin modifying factors.

Binding to ncRNAs alters protein functionality. In some cases the protein is sequestered or mislocalized by interactions with the ncRNA (Imamura et al., 2014). There are also examples of protein destabilization upon binding to ncRNAs (Manokaran et al., 2015). Similarly, some lncRNAs bind cellular RNAs, including miRNAs (Lee et al. 2015; Cazalla et al., 2010). This serves, for example, to localize specific factors close to a target or to alter the stability or function of the miRNAs. All these interactions translate into functional effects in the infected cell.

3.2.1. Viral replication

Viroid genomes are circular, single-stranded RNA molecules capable of autonomous replication (Gago-Zachert, 2015). Replication occurs by a rolling-circle mechanism that involves three steps: generation of polymers containing multiple copies of the genome, cleavage of the polymers to monomers and circularization of the monomers. Nuclear viroids replicate using as host factors the RNA polymerase II and the ligase I. Interestingly, they have managed to redirect the DNA dependent RNA polymerase II and the DNA ligase I to function on viroid RNA sequences. In the case of chloroplastic viroids cleavage is carried out by a hammerhead ribozyme motif contained within the viral genome (Navarro and Flores., 1997).

3.2.2. Regulation of gene expression

Several lncRNAs expressed in the infected cells are used to regulate the expression of viral and host genes.

3.2.2.1. Regulation of gene expression by affecting transcription

In most cases described to date, regulation of viral gene expression by lncRNAs is exerted at the level of transcription and this may change early-to-late infection or latent-to-lytic phase. Several examples exist. Human citomegalovirus (CMV) RNA 4.9 binds the repressive complex PRC2 to inhibit immediate-early transcription during latency (Rossetto et al., 2013a; Noriega et al., 2014). Similarly, PAN RNA interacts with LANA, the KSHV latency associated nuclear antigen. LANA is expressed during latency to repress expression from the promoters of lytic genes. When PAN RNA is expressed, PAN binds to LANA and blocks LANAs DNA binding activity, leading to activation of the lytic cycle (Conrad et al., 2015; Campbell et al., 2014). Further, PAN RNA can also regulate gene expression by binding viral and cellular promoters and factors required for chromatin remodeling (Rossetto et al., 2013b). It has been shown than PAN RNA can bind the promoter of the viral lytic regulator Rta and the demethylases JMJD3 (Jumonji domain containing 3) and UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome) (Rossetto et al., 2013b; Rossetto and Pari, 2012). In PAN knocked-out mutants, JMJD3 and UTX association with the Rta promoter decreases and the repressive H3K27 trimethylation at the Rta promoter is higher (Rossetto and Pari, 2012). Interestingly, PAN RNA can also bind the activating methyltransferase MLL2 (mixed-lineage leukemia protein 2), indicating that PAN could bind promoters of viral genes to eliminate repressive marks and add activating chromatin marks (Rossetto and Pari, 2012). As PAN RNA can also bind to proteins of the repressive PRC2 (polycomb repressive complex 2) such as SUZ12 (suppressor of zeste 12) and EZH2 (enhancer of zeste 2), it has been suggested that it can also work as a negative regulator (Rossetto et al., 2013b). Finally, PAN RNA can also bind the transcription factor IRF4 and block transcription of cellular targets (Rossetto and Pari, 2011). PAN RNA expression results in decreased expression of several immune responsive genes and in increased cell proliferation (Rossetto et al., 2013b).

Other viral lncRNAs have also been shown to bind chromatin or transcription factors. Antisense HIV transcripts repress HIV expression (Kobayashi-Ishihara et al., 2012; Saayman et al., 2014) by binding silencing factors such as DNA methyltransferase 3A (DNMT3a), EZH2 and HDAC1 (histone deacetylase 1) (Lazar et al., 2015). EBER2 can also localize to specific sites of DNA to guide a transcription factor (Lee et al. 2015; Tycowski et al., 2015). EBER2 binding occurs at the terminal repeats of the latent EBV genome, where the master regulator of B-lymphocytes PAX5 (paired box protein 5) also binds. Thus, EBER2 and PAX5 function to decrease the expression of genes located close to the terminal repeats and increase lytic replication. In fact, PAX5 is recruited by EBER2 to the EBV DNA. Interestingly, EBER2 binding to DNA requires that EBER2 base-pairs with nascent transcripts derived from the terminal repeats. It would be interesting to identify whether cellular lncRNAs can guide transcription factors to specific position in the genome by base-pairing to nascent transcripts.

HIV viruses use several mechanisms to control expression of host and viral genes, including by affecting functionality of cellular transcription factors. As described above, HIV viral proteins regulate the expression of NRON, which, in turn, regulates NFAT activity. NRON is a cytoplasmic lncRNA that forms a ribonucleoprotein complex together with NFAT kinases and NFAT (Willingham et al., 2005; Sharma et al., 2011). The complex keeps NFAT phosphorylated and retained in the cytoplasm. Upon NRON depletion, NFAT can be dephosphorylated by calcineurin and translocated to the nucleus to activate expression of several host and HIV virus genes (Lazar et al., 2015; Aune and Spurlock, 2015).

3.2.2.2. Regulation of gene expression by affecting RNA stability

Regulation of host gene expression is exerted mainly by transcription regulation (see above) but also by affecting RNA stability. This is achieved by the control of exoribonucleases such as Xrn1 or by using the RNAi pathway. As explained above, sfRNAs are produced by stalled Xrn1 (Charley and Wilusz, 2015). As a result, the Xrn1 enzyme is repressed and flavivirus infected cells accumulate Xrn1 substrates such as decapped mRNAs (Moon et al., 2012; Moon et al., 2015; Jinek et al., 2011). Surprisingly, in cells infected by some flaviviruses, it seems that the entire 5′ to 3′ decay pathway is repressed, leading to the accumulation and translation of otherwise unstable transcripts and a dramatic dysregulation of cellular gene expression (Moon et al., 2012; Moon et al., 2015). As expected, deregulated genes include normally short-lived cytokines, growth factors, cell cycle regulators and oncogenes (Rigby and Rehwinkel, 2015). Therefore, Xrn1 inactivation could be the underline mechanism that results in the cytokine storm observed during some flavivirus infections and in the oncogenic potential of HCV-infected cells (Pijlman et al., 2008; Carnero and Fortes, 2015).

Besides Xrn1 inhibition, sfRNAs can repress the RNAi machinery in both insect and mammalian cells (Schnettler et al., 2012; Schnettler et al., 2014). This could be achieved by sequestration of Dicer due to the large amount of secondary structure in the sfRNA. This is similar to what has been described for VA RNAs. The structure and the high abundancy of VA RNAs allow the saturation of several key members of the RNAi pathway such as Exportin 5 and Dicer (Andersson et al., 2005). Interestingly, at the same time, VA RNAs are processed by the RNAi machinery to produce viral micro RNAs that are not essential for lytic replication (Andersson et al., 2005; Aparicio et al., 2006; Aparicio et al., 2010; Kamel et al., 2013). However, these VA RNA-derived miRNAs can alter the expression of host cell genes. Finally, cells infected with viroids contain dsRNAs present in the highly structured regions of the genome or generated by genome replication. These dsRNA viroid molecules are processed by the cellular RNAi machinery to produce antiviral small interference RNAs (siRNAs) (Molnar et al., 2005; Mlotshwa et al., 2008; Gago-Zachert, 2015). Some viroid-derived siRNAs can also target cellular genes and result in several of the symptoms associated with viroid infection (Eamens et al., 2014).

The interplay between the RNAi machinery and the viral lncRNAs is also observed in cells infected with Herpesvirus saimiri (HVS). Latent infection with HVS produces high amounts of several HSURs (HVS U-rich RNAs) (Lee at al., 1988). HSUR1 and 2 are required for efficient viral replication and their expression results in upregulation of several host genes involved in T-cell activation (Murthy et al., 1989; Cook et al., 2005). This is mediated by HSUR1 binding and degradation of miR27 (Cazalla et al., 2010). After degradation of miR27, miR27 targets are increased and lead to T-cell activation and, probably, to proliferation of HVS-infected T cells (Tycowski et al., 2015). Several cellular lncRNAs have been described to interact with miRNAs. In most cases, they behave as sponges that sequester miRNAs and lead to the upregulation of miR-target genes. This is the case of HULC (highly upregulated in liver cancer), a lncRNA whose expression is particularly increased in the liver and serum of patients with HCC and an underlying HBV infection and serves as a marker for poor prognosis (Xie et al., 2013; Panzitt et al., 2007). A single nucleotide polymorphism in HULC shows decreased susceptibility to developing HCC in chronic HBV patients (Liu et al., 2012). HBx from HBV seems in charge of HULC upregulation and HULC silencing blocks HBx protein-mediated enhanced cell proliferation (Du et al., 2012; Wang et al., 2010). Interestingly, HULC sequesters miR-372, involved in regulation of cell cycle, apoptosis, invasion, and proliferation in many types of human cancers (Wu et al., 2015).

3.2.3. Regulation of the antiviral response

Within the regulation of host genes, control of those required to build an antiviral response deserves a special chapter. It is expected that viral lncRNAs should decrease the antiviral response, while cellular lncRNAs induced by infection could either induce the pathway or decrease IFN synthesis and signaling to control the duration and the strength of the antiviral response.

3.2.3.1. Control of the antiviral response by viral ncRNAs

Viral ncRNAs can be detected by several canonical and non-canonical PAMP sensors, such as TLR3, RIG-I, PKR and OAS, and induce the antiviral pathway. RIG-I is activated by dsRNAs with a triphosphate group at their 5′ end. This is a characteristic of EBERs and VA RNAs. Therefore, both EBERs and VA RNAs should activate RIG-I and led to IFN production (Iwakiri, 2015; Vachon and Conn, 2015). Further, VA RNAs can also activate OAS, which is another sensor for dsRNA. OAS synthesizes 2′-5′-linked oligoadenylate second messengers that activate RNase L, an endoribonuclease able to degrade single stranded RNAs. These RNA fragments can enhance RIG-I activation, leading to a further amplification of the IFN response (Malathi et al., 2007). Finally, EBERs are secreted by infected cells in complex with La protein (Iwakiri et al., 2009; Ahmed et al., 2014). Secreted EBERs activate TLR3 signaling in immune cells leading to production of inflammatory cytokines which could account for the immunopathologic diseases caused by EBV infection (Iwakiri et al., 2009; Iwakiri, 2015).

Interestingly, some ncRNAs have been described to block the activity of PKR or regulators, such as the RIG-I E3 ligase TRIM25. Both VA RNAs and EBERs are sensed by PKR, which requires a short dsRNA sequence. Interestingly, binding to PKR by these ncRNAs does not allow PKR activation. Instead, it avoids PKR dimerization and auto-phosphorylation. Therefore, signaling through PKR to eIF2a does not occur and translation of viral genes is properly initiated (Iwakiri, 2015; Vachon and Conn, 2015) (Fig. 1). In this case, therefore, the ncRNA behaves as a negative regulator of the antiviral pathway. Another negative regulator of the IFN synthesis pathway is sfRNA. Surprisingly, a particular sfRNA, PR-2B, has been described to bind TRIM25, an E3 ligase with unknown RNA binding capacity (Fig. 1). Binding of TRIM25 by PR-2B sfRNA prevents its deubiquitinylation by ubiquitin-specific peptidase 15 (USP15). Ubiquitinated TRIM25 is not active to polyubiquitinate and stabilize RIG-I, resulting in a dramatic decrease in the interferon response to infection by the cell (Manokaran et al., 2015).

3.2.3.2. Control of the antiviral response by cellular lncRNAs induced by infection
Positive regulators of the INF pathway

Among the lncRNAs induced by infection that function to activate the antiviral response are NEAT1 and BISPR (BST2 (Bone Marrow Stromal Antigen 2) IFN-stimulated positive regulator) (Fig. 1). IFN induces the expression of lncRNA BST2 or BISPR in several cell lines (Kambara et al., 2015; Barriocanal et al., 2015). BISPR is located head-to-head with BST2/tetherin, a well-characterized antiviral factor that attaches viruses to the cell surface and impedes viral release (Dafa-Berger et al., 2012; Neil et al., 2008). Both BISPR and BST2 are corregulated and it has been shown that BISPR acts in trans to induce transcription of BST2 gene, probably, by counteracting the repressive action of PRC2 (Kambara et al., 2015; Barriocanal et al., 2015).

HIV, influenza virus and other infections increase the levels of NEAT1, required for formation of paraspeckles (Zhang et al., 2013b; Clemson et al., 2009). Paraspeckles retain hyperedited and non-spliced RNAs such as many HIV RNAs. Further, paraspeckles retain the regulator SFPQ (Imamura et al., 2014). SFPQ is required for proper expression of several innate immune related genes, including IL8, RIG-I and MDA5. Increase of NEAT1 by HIV and other viruses, increases paraspeckles, decreases available SFPQ and augments IL8 and other antiviral factors (Landeras-Bueno and Ortín, 2015; Lazar et al., 2015) (Fig. 1).

Negative regulators of the IFN pathway

As mentioned above, many lncRNAs have been described to function as negative regulators of IFN synthesis or signaling and they could be required to help the cell to return to homeostasis after infection. Interestingly, some of these lncRNAs could be induced by certain viruses to counteract the cellular antiviral response. Many lncRNAs that negatively control the IFN pathway are regulated by activation of TLR receptors, such as lincRNA-COX2, IL7R or THRIL (Carpenter, 2015; Aune and Spurlock, 2015). Linc-RNA-COX2, induced after TLR activation, represses the expression of ISGs by binding hnRNP A/B proteins (heterogeneous nuclear ribonucleoprotein A/B) and, in part, by regulating the recruitment of RNA polymerase II to the promoter of target genes (Carpenter et al., 2013). Lnc-IL7R gene, induced after LPS and TLR2 stimulation, overlaps with the 3′UTR of the IL7R gene and functions as a negative regulator of the LPS-induced inflammatory response by regulating levels of H3K27 trimethylation at the promoters of target genes (Cui et al., 2014). THRIL (TNF-α and heterogeneous nuclear ribonucleoprotein L (hnRNP L) related immunoregulatory LincRNA), repressed by TLR and TNF activation, is required for induction of gene expression by binding hnRNP L and the promoter/enhancers of TNF targets (Li et al., 2014). While THRIL is indeed an inducer of TNF transcription, the fact that is downregulated by TLR and TNF activation results in decreased expression of target genes during the antiviral response. TNF also increases the expression of Lethe, a pseudogene of the ribosomal protein S15a (Rps15a) gene. Lethe inhibits NF-κB by titrating the RelA or p65 component of the active NF-κB p60/p65 heterodimer away from the NF-κB DNA response elements (Rapicavoli et al., 2013) (Fig. 1).

Similarly, IFN treatment induces the expression of NRIR gene (negative regulator of IFNα response), which is located in the genome close to two key ISGs: CMPK2 and viperin (Kambara et al., 2014). Surprisingly, NRIR expression inhibits transcription of CMPK2 and viperin, but also of other ISGs located far away in the genome, such as ISG15, CXCL10, IFIT3 or IFITM1 (Fig. 1). However, other ISGs such as Mx1, IFIT1 or IFNβ seemed resistant to NRIR. The molecular mechanism of transcription regulation is still unknown, but it could involve regulation of chromatin remodelling factors. This is the case of NRAV, a lncRNA downregulated by influenza virus expression (Ouyang et al., 2014). Downregulation of NRAV resulted in an increase in the H3K4me3 active mark and a decrease in the H3K27me3 repressive mark at the promoter of MxA and IFITM3 genes. Therefore, decreased levels of NRAV caused the upregulation of these ISGs.

3.2.4. Induction of mitochondrial energy

The most abundant lncRNA produced during the lytic infection with HCMV is expressed from the viral Beta 2.7 gene (Poole et al., 2015; Gatherer et al., 2011). Although with some coding potential, surprisingly, the RNA interacts with GRIM19 (genes associated with retinoid/IFN-induced mortality 19), a subunit of mitochondrial Complex I essential for complex assembly and function (Reeves et al., 2007). Binding of Beta 2.7 RNA to GRIM19 stabilizes mitochondrial membrane potential and results in continued ATP production, which is critical for the successful completion of the virus’ life cycle. Interestingly, interaction of the Beta 2.7 RNA with Complex I inhibits rotenone stress-induced apoptosis in neuronal cells.

3.2.5. Induction of pathogenesis

Some viral lncRNAs induce part of the pathogenicity exerted by viruses. It has been already described how viroid-derived siRNAs can target host genes and result in several plant symptoms, how inhibition of Xrn1 by sfRNA stabilizes short-lived cytokine mRNAs and may generate a cytokine storm associated to the infection or how secreted EBERs are sensed by TLR3 and lead to immunopathological diseases.

Similarly, it has been already described how HBV infection regulates the expression of several lncRNAs (DREH, HBx-LINE or HULC) which may mediate the increased risk of HCC development observed in HBV infected patients. Actually, chronic infection with either HBV or HCV leads to a chronic damage and liver inflammation that helps the development of HCC. In line with this, infection with HCV has been also shown to induce the expression of several lncRNAs with oncogenic potential (Carnero et al., submitted; Carnero and Fortes, 2015). Only in few cases the molecular mechanisms of oncogenicity have been studied in detail. DREH, inhibited by HBV infection, decreases proliferation by binding to vimentin (Huang et al., 2013); HBx-LINE induces the Wnt pathway by increasing the nuclear localization of b-catenin (Lau et al., 2014); HULC binds miR372 and avoids activation of CREB, cell proliferation and differentiation (Wang et al., 2010); HEIH (High Expression In HCC), increased in HBV-derived HCC, inhibits several cyclin dependent kinases by binding the repressive factor EZH2 (Yang et al., 2011); LET (Low Expression in Tumor), decreased in HBV-derived HCC, binds to and destabilizes NF90, a well-known regulator of factors related to tumor growth and metastasis (Yang et al., 2013b); MVIH (microvascular invasion in HCC), increased in HBV-derived HCCs, avoids secretion of PGK-1, which is an inhibitor of angiogenesis (Yuan et al., 2012); PVT-1 (plasmocytoma variant translocation 1), induced by HCV infection, is a positive regulator of c-myc (Tseng et al., 2014) and UCA1 (urothelial carcinoma associated 1), also increased by HCV infection, sequesters PTB and decreased PTB-mediated translation of p27, causing a decrease of this tumor suppressor (Huang et al., 2014).

Final considerations

Investigation into the role and function of lncRNAs in viral infections is still largely in its infancy. Future studies are required to understand in depth how the lncRNA transcriptome is altered in the infected cell and how these alterations impact viral fitness and cell survival. These studies may help to identify novel cellular pathways. In fact, studies with viruses have been transformative with regards to our collective knowledge of several key steps of gene expression such as transcription, splicing, polyadenylation and transport. More recently, the study of the stability of PAN RNA has allowed the discovery of a novel nuclear decay pathway and the identification of a triple helix structure that serves to stabilize viral and cellular transcripts (Wilusz et al., 2012; Brown et al., 2012; Brown et al., 2014). In line with this, it would be very interesting to determine whether Xrn1 serves to process cellular RNAs similarly to how it processes sfRNAs.

Furthermore, it is expected that the study of lncRNAs in infected cells leads to the development of RNAs with therapeutic relevance. In fact, this has been already proven to be the case for Beta 2.7 RNA, involved in the induction of mitochondrial energy (Poole et al., 2015). A domain of Beta 2.7 RNA has been identified that retains the functionality of the full-length transcript (Kuan et al., 2012). When bound to a RVG peptide with 9 arginine residues (RVG-9R), which crosses the blood-brain barrier, it can be delivered to neurons (Kuan et al., 2012; Kumar et al., 2007). There, the Beta 2.7 domain can protect mitochondrial complex I from stress-induced apoptosis and avoid neuron death. This treatment protects dopaminergic neurons in a rat model of Parkinson disease and could also be applied for the treatment of other mitochondrial and neurological diseases.

Finally, some viral lncRNAs may be used as therapeutic targets. This may be especially relevant for those lncRNAs that are expressed in latent infections, with the aim of eliminating latency-infected populations (Lazar et al., 2015). Similarly, virus-related lncRNAs secreted into the serum may serve as prognostic markers for disease progression. Interestingly, the fact that several lncRNAs behave as inducers or inhibitors of the IFN response, suggests that by modulating the cellular levels of these lncRNAs, the IFN response can be enhanced or repressed. This modulation may have therapeutic relevance for the treatment of several autoimmune diseases, viral infections and cancer.

Highlights.

  1. Infected cells change their transcriptome to express proviral and antiviral lncRNAs.

  2. Many viruses express viral noncoding transcripts essential for viral viability.

  3. Many lncRNAs expressed in the infected cell function as positive or negative regulators of the innate antiviral response.

Acknowledgments

We want to apologize to all the scientists whose work has not been mentioned due to space limitations. This study was supported by grants from the Ministry of Science and Innovation (BIO2009/09295, SAF2012-40003, BFU2013-50517-EXP), Fondo de Investigación Sanitaria (PI13/01989), financed by the Instituto de Salud Carlos III, Fundació La Marató de TV3 (20132130-31-32), Caja Navarra Foundation (70020), FEDER funding, funds from the “UTE project CIMA” and by the project RNAREG [CSD2009-00080], funded by the Ministry of Science and Innovation under the program CONSOLIDER INGENIO 2010. Support for KVM is acknowledged from NIAID PO1 AI099783-01, R01 AI111139-01, R01 DK104681-01 and Australian Research Council FT1300100572.

Footnotes

The authors declare no conflict of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Ahmed W, Phillip PS, Taniq S, Khan G. Epstein-Barr virus-encoded small RNAs (EBERs) are present in fractions related to exosomes released by EBV-transformed cells. PLoS One. 2014;9:e99163. doi: 10.1371/journal.pone.0099163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 2001;413:732–738. doi: 10.1038/35099560. [DOI] [PubMed] [Google Scholar]
  3. Andersson MG, Haasnoot PC, Xu N, Berenjian S, Berkhout B, Akusjarvi G. Suppression of RNA interference by adenovirus virus-associated RNA. J Virol. 2005;79:9556–9565. doi: 10.1128/JVI.79.15.9556-9565.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Andrejeva J, Childs KS, Young DF, Carlos TS, Stock N, Goodbourn S, Randall RE. 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 U S A. 2004;101:17264–17269. doi: 10.1073/pnas.0407639101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Aparicio O, Razquin N, Zaratiegui M, Narvaiza I, Fortes P. Adenovirus virus-associated RNA is processed to functional interfering RNAs involved in virus production. J Virol. 2006;80:1376–1384. doi: 10.1128/JVI.80.3.1376-1384.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Aparicio O, Carnero E, Abad X, Razquin N, Guruceaga E, Segura V, Fortes P. Adenovirus VA RNA-derived miRNAs target cellular genes involved in cell growth, gene expression and DNA repair. Nucleic Acids Res. 2010;38:750–763. doi: 10.1093/nar/gkp1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Arnaud N, Dabo S, Akazawa D, Fukasawa M, Shinkai-Ouchi F, Hugon J, Wakita T, Meurs EF. Hepatitis C virus reveals a novel early control in acute immune response. PLoS Pathog. 2011;7:e1002289. doi: 10.1371/journal.ppat.1002289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Arnaud N, Dabo S, Maillard P, Budkowska A, Kalliampakou KI, Mavromara P, Garcin D, Hugon J, Gatignol A, Akazawa D, Wakita T, Meurs EF. Hepatitis C virus controls interferon production through PKR activation. PLoS One. 2010;5:e10575. doi: 10.1371/journal.pone.0010575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Aune TM, Spurlock CF. Long non-coding RNAs in innate and adaptive immunity. Virus Res. 2015 doi: 10.1016/j.virusres.2015.07.003. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Barriocanal M, Carnero E, Segura V, Fortes P. Long Non-Coding RNA BST2/BISPR is Induced by IFN and Regulates the Expression of the Antiviral Factor Tetherin. Front Immunol. 2015;5:655. doi: 10.3389/fimmu.2014.00655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Broering R, Zhang X, Kottilil S, Trippler M, Jiang M, Lu M, Gerken G, Schlaak JF. The interferon stimulated gene 15 functions as a proviral factor for the hepatitis C virus and as a regulator of the IFN response. Gut. 2010;59:1111–1119. doi: 10.1136/gut.2009.195545. [DOI] [PubMed] [Google Scholar]
  12. Brown JA, Bulkley D, Wang J, Valenstein ML, Yario TA, Steitz TA, Steitz JA. Structural insights into the stabilization of MALAT1 noncoding RNA by a bipartite triple helix. Nat Struct Mol Biol. 2014;21(7):633–40. doi: 10.1038/nsmb.2844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brown JA, Valenstein ML, Yario TA, Tycowski KT, Steitz JA. Formation of triple-helical structures by the 3′-end sequences of MALAT1 and MENβ noncoding RNAs. Proc Natl Acad Sci U S A. 2012;109(47):19202–7. doi: 10.1073/pnas.1217338109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Campbell M, Kim KY, Chang PC, Huerta S, Shevchenko B, Wang DH, Izumiya C, Kung HJ, Izumiya Y. A lytic viral long noncoding RNA modulates the function of a latent protein. J Virol. 2014;88:1843–1848. doi: 10.1128/JVI.03251-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Carnero E, Barriocanal M, Segura V, Guruceaga E, Prior C, Borner K, Grimm D, Fortes P. Type I Interferon Regulates the Expression of Long Non-Coding RNAs. Front Immunol. 2014;5:548. doi: 10.3389/fimmu.2014.00548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Carnero E, Fortes P. HCV infection, IFN response and the coding and non-coding host cell genome. Virus Res. 2015 doi: 10.1016/j.virusres.2015.10.001. In press. [DOI] [PubMed] [Google Scholar]
  17. Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, Byron M, Monks B, Henry-Bezy M, Lawrence JB, O’Neill LAJ, Moore MJ, Caffrey DR, Fitzgerald KA. A Long Noncoding RNA Mediates Both Activation and Repression of Immune Response Genes. Science. 2013;341:789–792. doi: 10.1126/science.1240925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Carpenter S. Long noncoding RNA: novel links between gene expression and innate immunity. Virus Res. 2015 doi: 10.1016/j.virusres.2015.08.019. In press. [DOI] [PubMed] [Google Scholar]
  19. Carrieri C, Cimatti L, Biagioli M, Beugnet A, Zucchelli S, Fedele S, Pesce E, Ferrer I, Collavin L, Santoro C, Forrest AR, Carninci P, Biffo S, Stupka E, Gustincich S. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature. 2012;491:454–457. doi: 10.1038/nature11508. [DOI] [PubMed] [Google Scholar]
  20. Cawley S, Bekiranov S, Ng HH, Kapranov P, Sekinger EA, Kampa D, Piccolboni A, Sementchenko V, Cheng J, Williams AJ, Wheeler R, Wong B, Drenkow J, Yamanaka M, Patel S, Brubaker S, Tammana H, Helt G, Struhl K, Gingeras TR. Unbiased mapping of transcription factor binding sites along human chromosomes 21 and 22 points to widespread regulation of noncoding RNAs. Cell. 2004;116:499–509. doi: 10.1016/s0092-8674(04)00127-8. [DOI] [PubMed] [Google Scholar]
  21. Cazalla D, Yario T, Steitz JA. Down-regulation of a host microRNA by a Herpesvirus saimiri noncoding RNA. Science. 2010;328:1563–1566. doi: 10.1126/science.1187197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell. 2014;157:77–94. doi: 10.1016/j.cell.2014.03.008. [DOI] [PubMed] [Google Scholar]
  23. Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A, Bozzoni I. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 2011;147:358–369. doi: 10.1016/j.cell.2011.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Charley PA, Wilusz J. Standing your Ground to Exoribonucleases: Function of Flavivirus Long Non-coding RNAs. Virus Res. 2015 doi: 10.1016/j.virusres.2015.09.009. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Clague MJ, Heride C, Urbé S. The demographics of the ubiquitin system. Trends Cell Biol. 2015;25:417–426. doi: 10.1016/j.tcb.2015.03.002. [DOI] [PubMed] [Google Scholar]
  26. Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A, Lawrence JB. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Molecular cell. 2009;33(6):717–726. doi: 10.1016/j.molcel.2009.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Conrad NK. New insights into the expression and functions of the Kaposi’s sarcoma-associated herpesvirus long noncoding PAN RNA. Virus Res. 2015 doi: 10.1016/j.virusres.2015.06.012. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cook HL, Lytle JR, Mischo HE, Li MJ, Rossi JJ, Silva DP, Desrosiers RC, Steitz JA. Small nuclear RNAs encoded by Herpesvirus saimiri upregulate the expression of genes linked to T cell activation in virally transformed T cells. Curr Biol. 2005;15:974–979. doi: 10.1016/j.cub.2005.04.034. [DOI] [PubMed] [Google Scholar]
  29. Copela LA, Chakshusmathi G, Sherrer RL, Wolin SL. The La protein functions redundantly with tRNA modification enzymes to ensure tRNA structural stability. RNA. 2006;12:644–54. doi: 10.1261/rna.2307206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Cui H, Xie N, Tan Z, Banerjee S, Thannickal VJ, Abraham E, Liu G. The human long noncoding RNA lnc-IL7R regulates the inflammatory response. Eur J Immunol. 2014;44:2085–95. doi: 10.1002/eji.201344126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dabo S, Meurs EF. dsRNA-dependent protein kinase PKR and its role in stress, signaling and HCV infection. Viruses. 2012;4:2598–635. doi: 10.3390/v4112598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dafa-Berger A, Kuzmina A, Fassler M, Yitzhak-Asraf H, Shemer-Avni Y, Taube R. Modulation of hepatitis C virus release by the interferon-induced protein BST-2/tetherin. Virology. 2012;428:98–111. doi: 10.1016/j.virol.2012.03.011. [DOI] [PubMed] [Google Scholar]
  33. Davis ME, Gack MU. Ubiquitination in the antiviral immune response. Virology. 2015;479–480:52–65. doi: 10.1016/j.virol.2015.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, Lagarde J, Veeravalli L, Ruan X, Ruan Y, Lassmann T, Carninci P, Brown JB, Lipovich L, Gonzalez JM, Thomas M, Davis CA, Shiekhattar R, Gingeras TR, Hubbard TJ, Notredame C, Harrow J, Guigó R. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 2012;22:1775–1789. doi: 10.1101/gr.132159.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303:1529–1531. doi: 10.1126/science.1093616. [DOI] [PubMed] [Google Scholar]
  36. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigo R, Gingeras TR. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Du Y, Kong G, You X, Zhang S, Zhang T, Gao Y, Ye L, Zhang X. Elevation of highly up regulated in liver cancer (HULC) by hepatitis B virus X protein promotes hepatoma cell proliferation via down-regulating p18. J Biol Chem. 2012;287:26302–26311. doi: 10.1074/jbc.M112.342113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Durfee LA, Lyon N, Seo K, Huibregtse JM. The ISG15 conjugation system broadly targets newly synthesized proteins: implications for the antiviral function of ISG15. Mol Cell. 2010;38:722–732. doi: 10.1016/j.molcel.2010.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Eamens AL, Smith NA, Dennis ES, Wassenegger M, Wang MB. In Nicotiana species, an artificial microRNA corresponding to the virulence modulating region of Potato spindle tuber viroid directs RNA silencing of a soluble inorganic pyrophosphatase gene and the development of abnormal phenotypes. Virology. 2014;450–451:266–277. doi: 10.1016/j.virol.2013.12.019. [DOI] [PubMed] [Google Scholar]
  40. Birney E, Stamatoyannopoulos JA, Dutta A, Guigo R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, Thurman RE, Kuehn MS, Taylor CM, Neph S, Koch CM, Asthana S, Malhotra A, Adzhubei I, Greenbaum JA, Andrews RM, Flicek P, Boyle PJ, Cao H, Carter NP, Clelland GK, Davis S, Day N, Dhami P, Dillon SC, Dorschner MO, Fiegler H, Giresi PG, Goldy J, Hawrylycz M, Haydock A, Humbert R, James KD, Johnson BE, Johnson EM, Frum TT, Rosenzweig ER, Karnani N, Lee K, Lefebvre GC, Navas PA, Neri F, Parker SC, Sabo PJ, Sandstrom R, Shafer A, Vetrie D, Weaver M, Wilcox S, Yu M, Collins FS, Dekker J, Lieb JD, Tullius TD, Crawford GE, Sunyaev S, Noble WS, Dunham I, Denoeud F, Reymond A, Kapranov P, Rozowsky J, Zheng D, Castelo R, Frankish A, Harrow J, Ghosh S, Sandelin A, Hofacker IL, Baertsch R, Keefe D, Dike S, Cheng J, Hirsch HA, Sekinger EA, Lagarde J, Abril JF, Shahab A, Flamm C, Fried C, Hackermuller J, Hertel J, Lindemeyer M, Missal K, Tanzer A, Washietl S, Korbel J, Emanuelsson O, Pedersen JS, Holroyd N, Taylor R, Swarbreck D, Matthews N, Dickson MC, Thomas DJ, Weirauch MT, Gilbert J, Drenkow J, Bell I, Zhao X, Srinivasan KG, Sung WK, Ooi HS, Chiu KP, Foissac S, Alioto T, Brent M, Pachter L, Tress ML, Valencia A, Choo SW, Choo CY, Ucla C, Manzano C, Wyss C, Cheung E, Clark TG, Brown JB, Ganesh M, Patel S, Tammana H, Chrast J, Henrichsen CN, Kai C, Kawai J, Nagalakshmi U, Wu J, Lian Z, Lian J, Newburger P, Zhang X, Bickel P, Mattick JS, Carninci P, Hayashizaki Y, Weissman S, Hubbard T, Myers RM, Rogers J, Stadler PF, Lowe TM, Wei CL, Ruan Y, Struhl K, Gerstein M, Antonarakis SE, Fu Y, Green ED, Karaoz U, Siepel A, Taylor J, Liefer LA, Wetterstrand KA, Good PJ, Feingold EA, Guyer MS, Cooper GM, Asimenos G, Dewey CN, Hou M, Nikolaev S, Montoya-Burgos JI, Loytynoja A, Whelan S, Pardi F, Massingham T, Huang H, Zhang NR, Holmes I, Mullikin JC, Ureta-Vidal A, Paten B, Seringhaus M, Church D, Rosenbloom K, Kent WJ, Stone EA, Batzoglou S, Goldman N, Hardison RC, Haussler D, Miller W, Sidow A, Trinklein ND, Zhang ZD, Barrera L, Stuart R, King DC, Ameur A, Enroth S, Bieda MC, Kim J, Bhinge AA, Jiang N, Liu J, Yao F, Vega VB, Lee CW, Ng P, Shahab A, Yang A, Moqtaderi Z, Zhu Z, Xu X, Squazzo S, Oberley MJ, Inman D, Singer MA, Richmond TA, Munn KJ, Rada-Iglesias A, Wallerman O, Komorowski J, Fowler JC, Couttet P, Bruce AW, Dovey OM, Ellis PD, Langford CF, Nix DA, Euskirchen G, Hartman S, Urban AE, Kraus P, Van Calcar S, Heintzman N, Kim TH, Wang K, Qu C, Hon G, Luna R, Glass CK, Rosenfeld MG, Aldred SF, Cooper SJ, Halees A, Lin JM, Shulha HP, Zhang X, Xu M, Haidar JN, Yu Y, Ruan Y, Iyer VR, Green RD, Wadelius C, Farnham PJ, Ren B, Harte RA, Hinrichs AS, Trumbower H, Clawson H, Hillman-Jackson J, Zweig AS, Smith K, Thakkapallayil A, Barber G, Kuhn RM, Karolchik D, Armengol L, Bird CP, de Bakker PI, Kern AD, Lopez-Bigas N, Martin JD, Stranger BE, Woodroffe A, Davydov E, Dimas A, Eyras E, Hallgrimsdottir IB, Huppert J, Zody MC, Abecasis GR, Estivill X, Bouffard GG, Guan X, Hansen NF, Idol JR, Maduro VV, Maskeri B, McDowell JC, Park M, Thomas PJ, Young AC, Blakesley RW, Muzny DM, Sodergren E, Wheeler DA, Worley KC, Jiang H, Weinstock GM, Gibbs RA, Graves T, Fulton R, Mardis ER, Wilson RK, Clamp M, Cuff J, Gnerre S, Jaffe DB, Chang JL, Lindblad-Toh K, Lander ES, Koriabine M, Nefedov M, Osoegawa K, Yoshinaga Y, Zhu B, de Jong PJ ENCODE Project Consortium NISC Comparative Sequencing Program Baylor College of Medicine Human Genome Sequencing Center Washington University Genome Sequencing Center Broad Institute Children's Hospital Oakland Research Institute. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816. doi: 10.1038/nature05874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Feng J, Bi C, Clark BS, Mady R, Shah P, Kohtz JD. The Evf-2 noncoding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as a Dlx-2 transcriptional coactivator. Genes Dev. 2006;20:1470–1484. doi: 10.1101/gad.1416106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Flores R, Gago-Zachert S, Serra P, Sanjuán R, Elena SF. Viroids: survivors from the RNA world? Annu Rev Microbiol. 2014;68:395–414. doi: 10.1146/annurev-micro-091313-103416. [DOI] [PubMed] [Google Scholar]
  43. Gack MU, Shin YC, Joo CH, Urano T, Liang C, Sun L, Takeuchi O, Akira S, Chen Z, Inoue S, Jung JU. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature. 2007;446:916–920. doi: 10.1038/nature05732. [DOI] [PubMed] [Google Scholar]
  44. Gago-Zachert S. Viroids, infectious long non-coding RNAs with autonomous replication. Virus Res. 2015 doi: 10.1016/j.virusres.2015.08.018. In press. [DOI] [PubMed] [Google Scholar]
  45. Garitano-Trojaola A, Agirre X, Prosper F, Fortes P. Long non-coding RNAs in haematological malignancies. Int J Mol Sci. 2013;14:15386–15422. doi: 10.3390/ijms140815386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gatherer D, Seirafian S, Cunningham C, Holton M, Dargan DJ, Baluchova K, Hector RD, Galbraith J, Herzyk P, Wilkinson GW, Davison AJ. High-resolution human cytomegalovirus transcriptome. Proc Natl Acad Sci U S A. 2011;108(49):19755–19760. doi: 10.1073/pnas.1115861108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Hesselberth JR. Lives that introns lead after splicing. Wiley Interdiscip Rev RNA. 2013;4:677–691. doi: 10.1002/wrna.1187. [DOI] [PubMed] [Google Scholar]
  49. Horner SM, Gale M., Jr Regulation of hepatic innate immunity by hepatitis C virus. Nat Med. 2013;19:879–88. doi: 10.1038/nm.3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Huang J, Zhou N, Watabe K, Lu Z, Wu F, Xu M, Mo YY. Long non-coding RNA UCA1 promotes breast tumor growth by suppression of p27 (Kip1) Cell Death Dis. 2014;5:e1008. doi: 10.1038/cddis.2013.541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Huang JF, Guo YJ, Zhao CX, Yuan SX, Wang Y, Tang GN, Zhou WP, Sun SH. Hepatitis B virus X protein (HBx)-related long noncoding RNA (lncRNA) down-regulated expression by HBx (Dreh) inhibits hepatocellular carcinoma metastasis by targeting the intermediate filament protein vimentin. Hepatology. 2013;57:1882–1892. doi: 10.1002/hep.26195. [DOI] [PubMed] [Google Scholar]
  52. Imam H, Bano AS, Patel P, Holla P, Jameel S. The lncRNA NRON modulates HIV-1 replication in a NFAT-dependent manner and is differentially regulated by early and late viral proteins. Scientific reports. 2015;5:8639. doi: 10.1038/srep08639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Imamura K, Imamachi N, Akizuki G, Kumakura M, Kawaguchi A, Nagata K, Kato A, Kawaguchi Y, Sato H, Yoneda M, Kai C, Yada T, Suzuki Y, Yamada T, Ozawa T, Kaneki K, Inoue T, Kobayashi M, Kodama T, Wada Y, Sekimizu K, Akimitsu N. Long noncoding RNA NEAT1-dependent SFPQ relocation from promoter region to paraspeckle mediates IL8 expression upon immune stimuli. Molecular Cell. 2014;53:393–406. doi: 10.1016/j.molcel.2014.01.009. [DOI] [PubMed] [Google Scholar]
  54. Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14:36–49. doi: 10.1038/nri3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, Poliakov A, Cao X, Dhanasekaran SM, Wu YM, Robinson DR, Beer DG, Feng FY, Iyer HK, Chinnaiyan AM. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 2015;47:199–208. doi: 10.1038/ng.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Iwakiri D, Zhou L, Samanta M, Matsumoto M, Ebihara T, Seya T, Imai S, Fujieda M, Kawa K, Takada K. Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from Toll-like receptor 3. J Exp Med. 2009;206:2091–2099. doi: 10.1084/jem.20081761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Iwakiri D. Multifunctional non-coding Epstein-Barr virus encoded RNAs (EBERs) contribute to viral pathogenesis. Virus Res. 2015 doi: 10.1016/j.virusres.2015.08.007. In press. [DOI] [PubMed] [Google Scholar]
  58. Jinek M, Coyle SM, Doudna JA. Coupled 5′ nucleotide recognition and processivity in Xrn1-mediated mRNA decay. Mol Cell. 2011;41:600–8. doi: 10.1016/j.molcel.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kambara H, Gunawardane L, Zebrowski E, Kostadinova L, Jobava R, Krokowski D, Hatzoglou M, Anthony DD, Valadkhan S. Regulation of Interferon-Stimulated Gene BST2 by a lncRNA Transcribed from a Shared Bidirectional Promoter. Front Immunol. 2015;5:676. doi: 10.3389/fimmu.2014.00676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kambara H, Niazi F, Kostadinova L, Moonka DK, Siegel CT, Post AB, Carnero E, Barriocanal M, Fortes P, Anthony DD, Valadkhan S. Negative regulation of the interferon response by an interferon-induced long non-coding RNA. Nucleic Acids Res. 2014;42:10668–10680. doi: 10.1093/nar/gku713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kamel W, Segerman B, Oberg D, Punga T, Akusjarvi G. The adenovirus VARNA-derived miRNAs are not essential for lytic virus growth in tissue culture cells. Nucleic Acids Res. 2013;41:4802–4812. doi: 10.1093/nar/gkt172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kang W, Mukerjee R, Gartner JJ, Hatzigeorgiou AG, Sandri-Goldin RM, Fraser NW. Characterization of a spliced exon product of herpes simplex type-1 latency-associated transcript in productively infected cells. Virology. 2006;356:106–114. doi: 10.1016/j.virol.2006.07.033. [DOI] [PubMed] [Google Scholar]
  63. Kang DC, Gopalkrishnan RV, Wu Q, Jankowsky E, Pyle AM, Fisher PB. Mda-5: An interferon-inducible putative RNA helicase with double-stranded RNA-dependent ATPase activity and melanoma growth-suppressive properties. Proc Natl Acad Sci U S A. 2002;99:637–642. doi: 10.1073/pnas.022637199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, Regev A, Lander ES, Rinn JL. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A. 2009;106:11667–11672. doi: 10.1073/pnas.0904715106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kim MJ, Hwang SY, Imaizumi T, Yoo JY. Negative feedback regulation of RIG-I-mediated antiviral signaling by interferon-induced ISG15 conjugation. J Virol. 2008;82:1474–1483. doi: 10.1128/JVI.01650-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kobayashi-Ishihara M, Yamagishi M, Hara T, Matsuda Y, Takahashi R, Miyake A, Nakano K, Yamochi T, Ishida T, Watanabe T. HIV-1-encoded antisense RNA suppresses viral replication for a prolonged period. Retrovirology. 2012;9:38. doi: 10.1186/1742-4690-9-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kuan WL, Poole E, Fletcher M, Karniely S, Tyers P, Wills M, Barker RA, Sinclair JH. A novel neuroprotective therapy for Parkinson’s disease using a viral noncoding RNA that protects mitochondrial complex I activity. J Exp Med. 2012;209(1):1–10. doi: 10.1084/jem.20111126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kumar P, Wu H, McBride JL, Jung KE, Kim MH, Davidson BL, Lee SK, Shankar P, Manjunath N. Transvascular delivery of small interfering RNA to the central nervous system. Nature. 2007;448(7149):39–43. doi: 10.1038/nature05901. [DOI] [PubMed] [Google Scholar]
  69. Kwon KM, Ahn J-H. Herpesvirus-encoded Deubiquitinating Proteases and Their Roles in Regulating Immune Signaling Pathways. J Bacteriol Virol. 2013;43:244–252. [Google Scholar]
  70. Landeras-Bueno S, Ortín J. Regulation of influenza virus infection by long non-coding RNAs. Virus Res. 2015 doi: 10.1016/j.virusres.2015.08.008. In press. [DOI] [PubMed] [Google Scholar]
  71. Lau CC, Sun T, Ching AK, He M, Li JW, Wong AM, Co NN, Chan AW, Li PS, Lung RW, Tong JH, Lai PB, Chan HL, To KF, Chan TF, Wong N. Viral-human chimeric transcript predisposes risk to liver cancer development and progression. Cancer Cell. 2014;25:335–49. doi: 10.1016/j.ccr.2014.01.030. [DOI] [PubMed] [Google Scholar]
  72. Lazar DC, Morris KV, Saayman SM. The emerging role of long non-coding RNAs in HIV infection. Virus Res. 2015 doi: 10.1016/j.virusres.2015.07.023. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lee N, Moss WN, Yario TA, Steitz JA. EBV noncoding RNA binds nascent RNA to drive host PAX5 to viral DNA. Cell. 2015;160:607–18. doi: 10.1016/j.cell.2015.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lee SI, Murthy SC, Trimble JJ, Desrosiers RC, Steitz JA. Four novel U RNAs are encoded by a herpesvirus. Cell. 1988;54:599–607. doi: 10.1016/s0092-8674(88)80004-7. [DOI] [PubMed] [Google Scholar]
  75. Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, Head SR, Burns JC, Rana TM. The long noncoding RNA THRIL regulates TNFalpha expression through its interaction with hnRNPL. Proc Natl Acad Sci U S A. 2014;111:1002–7. doi: 10.1073/pnas.1313768111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Li K, Li NL, Wei D, Pfeffer SR, Fan M, Pfeffer LM. Activation of chemokine and inflammatory cytokine response in hepatitis C virus-infected hepatocytes depends on Toll-like receptor 3 sensing of hepatitis C virus double-stranded RNA intermediates. Hepatology. 2012;55:666–675. doi: 10.1002/hep.24763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Liu Y, Pan S, Liu L, Zhai X, Liu J, Wen J, Zhang Y, Chen J, Shen H, Hu Z. A genetic variant in long non-coding RNA HULC contributes to risk of HBV-related hepatocellular carcinoma in a Chinese population. PLoS One. 2012;7:e35145. doi: 10.1371/journal.pone.0035145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Loo YM, Gale M., Jr Immune signaling by RIG-I-like receptors. Immunity. 2011;34:680–692. doi: 10.1016/j.immuni.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Luo H, Zhang Z, Zheng Z, Ke X, Zhang X, Li Q, Liu Y, Bai B, Mao P, Hu Q, Wang H. Human bocavirus VP2 upregulates IFN-β pathway by inhibiting ring finger protein 125-mediated ubiquitination of retinoic acid-inducible gene-I. J Immunol. 2013;191:660–669. doi: 10.4049/jimmunol.1202933. [DOI] [PubMed] [Google Scholar]
  80. Maarifi G, Maroui MA, Dutrieux J, Dianoux L, Nisole S, Chelbi-Alix MK. Small Ubiquitin-like Modifier Alters IFN Response. J Immunol. 2015;195:2312–2324. doi: 10.4049/jimmunol.1500035. [DOI] [PubMed] [Google Scholar]
  81. Malakhova OA, Kim KI, Luo JK, Zou W, Kumar KG, Fuchs SY, Shuai K, Zhang DE. UBP43 is a novel regulator of interferon signaling independent of its ISG15 isopeptidase activity. EMBO J. 2006;25:2358–2367. doi: 10.1038/sj.emboj.7601149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Malakhova OA, Yan M, Malakhov MP, Yuan Y, Ritchie KJ, Kim KI, Peterson LF, Shuai K, Zhang DE. Protein ISGylation modulates the JAK-STAT signaling pathway. Genes Dev. 2003;17:455–460. doi: 10.1101/gad.1056303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Malathi K, Dong B, Gale M, Jr, Silverman RH. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature. 2007;448(7155):816–819. doi: 10.1038/nature06042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Manokaran G, Finol E, Wang C, Gunaratne J, Bahl J, Ong EZ, Tan HC, Sessions OM, Ward AM, Gubler DJ, Harris E, Garcia-Blanco MA, Ooi EE. Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness. Science. 2015 doi: 10.1126/science.aab3369. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Mao YS, Sunwoo H, Zhang B, Spector DL. Direct visualization of the co-transcriptional assembly of a nuclear body by noncoding RNAs. Nat Cell Biol. 2011;13:95–101. doi: 10.1038/ncb2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Marx JL. “Viroids”: a new kind of pathogen? Science. 1972;178:734. doi: 10.1126/science.178.4062.734. [DOI] [PubMed] [Google Scholar]
  87. Mathews MB. Genes for VA-RNA in adenovirus 2. Cell. 1975;6:223–9. doi: 10.1016/0092-8674(75)90013-6. [DOI] [PubMed] [Google Scholar]
  88. Mlotshwa S, Pruss GJ, Vance V. Small RNAs in viral infection and host defense. Trends Plant Sci. 2008;13:375–382. doi: 10.1016/j.tplants.2008.04.009. [DOI] [PubMed] [Google Scholar]
  89. Molnar A, Csorba T, Lakatos L, Varallyay E, Lacomme C, Burgyan J. Plant virus-derived small interfering RNAs originate predominantly from highly structured single-stranded viral RNAs. J Virol. 2005;79:7812–7818. doi: 10.1128/JVI.79.12.7812-7818.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Moon SL, Blackinton JG, Anderson JR, Dozier MK, Dodd BJ, Keene JD, Wilusz CJ, Bradrick SS, Wilusz J. XRN1 stalling in the 5′ UTR of Hepatitis C virus and Bovine Viral Diarrhea virus is associated with dysregulated host mRNA stability. PLoS Pathog. 2015;11:e1004708. doi: 10.1371/journal.ppat.1004708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Moon SL, Anderson JR, Kumagai Y, Wilusz CJ, Akira S, Khromykh AA, Wilusz J. A noncoding RNA produced by arthropod-borne flaviviruses inhibits the cellular exoribonuclease XRN1 and alters host mRNA stability. RNA. 2012;18:2029–40. doi: 10.1261/rna.034330.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Moyo B, Nicholson SA, Arbuthnot PB. The role of long non-coding RNAs in hepatitis B virus-related hepatocellular carcinoma. Virus Res. 2015 doi: 10.1016/j.virusres.2015.07.025. In press. [DOI] [PubMed] [Google Scholar]
  93. Mukerjee R, Kang W, Suri V, Fraser NW. A non-consensus branch point plays an important role in determining the stability of the 2-kb LAT intron during acute and latent infections of herpes simplex virus type-1. Virology. 2004;324:340–349. doi: 10.1016/j.virol.2004.03.043. [DOI] [PubMed] [Google Scholar]
  94. Murthy SC, Trimble JJ, Desrosiers RC. Deletion mutants of Herpesvirus saimiri define an open reading frame necessary for transformation. J Virol. 1989;63:3307–3314. doi: 10.1128/jvi.63.8.3307-3314.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Narayan K, Waggoner L, Pham ST, Hendricks GL, Waggoner SN, Conlon J, Wang JP, Fitzgerald KA, Kang J. TRIM13 is a negative regulator of MDA5-mediated type I interferon production. J Virol. 2014;88:10748–10757. doi: 10.1128/JVI.02593-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Navarro B, Flores R. Chrysanthemum chlorotic mottle viroid: unusual structural properties of a subgroup of self-cleaving viroids with hammerhead ribozymes. Proc Natl Acad Sci USA. 1997;94:11262–11267. doi: 10.1073/pnas.94.21.11262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Neil SJ, Zang T, Bieniasz PD. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature. 2008;451:425–430. doi: 10.1038/nature06553. [DOI] [PubMed] [Google Scholar]
  98. Noriega VM, Haye KK, Kraus TA, Kowalsky SR, Ge Y, Moran TM, Tortorella D. Human cytomegalovirus modulates monocyte-mediated innate immune responses during short-term experimental latency in vitro. J Virol. 2014;88:9391–9405. doi: 10.1128/JVI.00934-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Ouyang J, Zhu X, Chen Y, Wei H, Chen Q, Chi X, Qi B, Zhang L, Zhao Y, Gao GF, Wang G, Chen JL. NRAV, a long noncoding RNA, modulates antiviral responses through suppression of interferon-stimulated gene transcription. Cell host & microbe. 2014;16(5):616–626. doi: 10.1016/j.chom.2014.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Panzitt K, Tschernatsch MM, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M, Zatloukal K. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330–342. doi: 10.1053/j.gastro.2006.08.026. [DOI] [PubMed] [Google Scholar]
  101. Pijlman GP, Funk A, Kondratieva N, Leung J, Torres S, van der Aa L, Liu WJ, Palmenberg AC, Shi PY, Hall RA, Khromykh AA. A highly structured, nuclease-resistant, noncoding RNA produced by flaviviruses is required for pathogenicity. Cell Host Microbe. 2008;4:579–91. doi: 10.1016/j.chom.2008.10.007. [DOI] [PubMed] [Google Scholar]
  102. Ponjavic J, Ponting CP, Lunter G. Functionality or transcriptional noise? Evidence for selection within long noncoding RNAs. Genome Res. 2007;17(5):556–65. doi: 10.1101/gr.6036807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Poole E, Kuan WL, Barker R, Sinclair J. The human cytomegalovirus non-coding Beta2.7 RNA as a novel therapeutic for Parkinson’s disease - Translational research with no translation. Virus Res. 2015 doi: 10.1016/j.virusres.2015.05.007. In press. [DOI] [PubMed] [Google Scholar]
  104. Rajsbaum R, Albrecht RA, Wang MK, Maharaj NP, Versteeg GA, Nistal-Villán E, García-Sastre A, Gack MU. Species-specific inhibition of RIG-I ubiquitination and IFN induction by the influenza A virus NS1 protein. PLoS Pathog. 2012;8:e1003059. doi: 10.1371/journal.ppat.1003059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM, Chang HY. A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. eLife. 2013;2:e00762. doi: 10.7554/eLife.00762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Reeves MB, Davies AA, McSharry BP, Wilkinson GW, Sinclair JH. Complex I binding by a virally encoded RNA regulates mitochondria-induced cell death. Science. 2007;316(5829):1345–1348. doi: 10.1126/science.1142984. [DOI] [PubMed] [Google Scholar]
  107. Rigby RE, Rehwinkel J. RNA degradation in antiviral immunity and autoimmunity. Trends Immunol. 2015;36:179–88. doi: 10.1016/j.it.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Rosa MD, Gottlieb E, Lerner MR, Steitz JA. Striking similarities are exhibited by two small Epstein-Barr virus-encoded ribonucleic acids and the adenovirus-associated ribonucleic acids VAI and VAII. Mol Cell Biol. 1981;9:785–796. doi: 10.1128/mcb.1.9.785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Rossetto CC, Tarrant-Elorza M, Pari GS. Cis and trans acting factors involved in human cytomegalovirus experimental and natural latent infection of CD14 (+) monocytes and CD34 (+) cells. PLoS Pathog. 2013a;9(5):e1003366. doi: 10.1371/journal.ppat.1003366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Rossetto CC, Pari G. KSHV PAN RNA associates with demethylases UTX and JMJD3 to activate lytic replication through a physical interaction with the virus genome. PLoS Pathog. 2012;8:e1002680. doi: 10.1371/journal.ppat.1002680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Rossetto CC, Pari GS. Kaposi’s sarcoma-associated herpesvirus noncoding polyadenylated nuclear RNA interacts with virus- and host cell-encoded proteins and suppresses expression of genes involved in immune modulation. J Virol. 2011;85:13290–13297. doi: 10.1128/JVI.05886-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Rossetto CC, Tarrant-Elorza M, Verma S, Purushothaman P, Pari GS. Regulation of viral and cellular gene expression by Kaposi’s sarcoma-associated herpesvirus polyadenylated nuclear RNA. J Virol. 2013b;87:5540–5553. doi: 10.1128/JVI.03111-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Saayman S, Ackley A, Turner AM, Famiglietti M, Bosque A, Clemson M, Planelles V, Morris KV. An HIV-encoded antisense long noncoding RNA epigenetically regulates viral transcription. Molecular Therapy. 2014;22(6):1164–1175. doi: 10.1038/mt.2014.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Schneider WM, Chevillotte MD, Rice CM. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol. 2014;32:513–545. doi: 10.1146/annurev-immunol-032713-120231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Schnettler E, Sterken MG, Leung JY, Metz SW, Geertsema C, Goldbach RW, Vlak JM, Kohl A, Khromykh AA, Pijlman GP. Noncoding flavivirus RNA displays RNA interference suppressor activity in insect and Mammalian cells. J Virol. 2012;86:13486–500. doi: 10.1128/JVI.01104-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Schnettler E, Tykalová H, Watson M, Sharma M, Sterken MG, Obbard DJ, Lewis SH, McFarlane M, Bell-Sakyi L, Barry G, Weisheit S, Best SM, Kuhn RJ, Pijlman GP, Chase-Topping ME, Gould EA, Grubhoffer L, Fazakerley JK, Kohl A. Induction and suppression of tick cell antiviral RNAi responses by tick-borne flaviviruses. Nucleic Acids Res. 2014;42:9436–46. doi: 10.1093/nar/gku657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Sharma S, Findlay GM, Bandukwala HS, Oberdoerffer S, Baust B, Li Z, Schmidt V, Hogan PG, Sacks DB, Rao A. Dephosphorylation of the nuclear factor of activated T cells (NFAT) transcription factor is regulated by an RNA-protein scaffold complex. Proc Natl Acad Sci U S A. 2011;108:11381–6. doi: 10.1073/pnas.1019711108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Shi HX, Yang K, Liu X, Liu XY, Wei B, Shan YF, Zhu LH, Wang C. Positive regulation of interferon regulatory factor 3 activation by Herc5 via ISG15 modification. Mol Cell Biol. 2010;30:2424–36. doi: 10.1128/MCB.01466-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Shimura H, Masuta C. Plant subviral RNAs as a long noncoding RNA (lncRNA): Analogy with animal lncRNAs in host-virus interactions. Virus Res. 2015 doi: 10.1016/j.virusres.2015.06.016. In press. [DOI] [PubMed] [Google Scholar]
  120. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, Blencowe BJ, Prasanth SG, Prasanth KV. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell. 2010;39:925–938. doi: 10.1016/j.molcel.2010.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, Chang HY. Long noncoding RNA as modular scaffold of histone modification complexes. Science. 2010;329:689–693. doi: 10.1126/science.1192002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, Ronning P, Reuland B, Guenther K, Beadnell TC, Essig J, Otto GM, O’Sullivan MG, Largaespada DA, Schwertfeger KL, Marahrens Y, Kawakami Y, Bagchi A. PVT1 dependence in cancer with MYC copy-number increase. Nature. 2014;512:82–86. doi: 10.1038/nature13311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Tycowski KT, Guo YE, Lee N, Moss WN, Vallery TK, Xie M, Steitz JA. Viral noncoding RNAs: more surprises. Genes Dev. 2015;29:567–84. doi: 10.1101/gad.259077.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Ulitsky I, Shkumatava A, Jan CH, Sive H, Bartel DP. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 2011;147:1537–1550. doi: 10.1016/j.cell.2011.11.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Vachon VK, Conn GL. Adenovirus VA RNA: An essential pro-viral non-coding RNA. Virus Res. 2015 doi: 10.1016/j.virusres.2015.06.018. In press. [DOI] [PubMed] [Google Scholar]
  126. Valadkhan S, Gunawardane LS. lncRNA-mediated regulation of the interferon response. Virus Res. 2015 doi: 10.1016/j.virusres.2015.09.023. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Wang J, Liu X, Wu H, Ni P, Gu Z, Qiao Y, Chen N, Sun F, Fan Q. CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer. Nucleic Acids Res. 2010;38:5366–5383. doi: 10.1093/nar/gkq285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Wang L, Zhao W, Zhang M, Wang P, Zhao K, Zhao X, Yang S, Gao C. USP4 positively regulates RIG-I-mediated antiviral response through deubiquitination and stabilization of RIG-I. J Virol. 2013;87:4507–4515. doi: 10.1128/JVI.00031-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Wang P, Zhao W, Zhao K, Zhang L, Gao C. TRIM26 negatively regulates interferon-β production and antiviral response through polyubiquitination and degradation of nuclear IRF3. PLoS Pathog. 2015;11:e1004726. doi: 10.1371/journal.ppat.1004726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Wang P, Xue Y, Han Y, Lin L, Wu C, Xu S, Jiang Z, Xu J, Liu Q, Cao X. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science. 2014;344:310–313. doi: 10.1126/science.1251456. [DOI] [PubMed] [Google Scholar]
  131. Willingham AT, Orth AP, Batalov S, Peters EC, Wen BG, Aza-Blanc P, Hogenesch JB, Schultz PG. A strategy for probing the function of noncoding RNAs finds a repressor of NFAT. Science. 2005;309:1570–1573. doi: 10.1126/science.1115901. [DOI] [PubMed] [Google Scholar]
  132. Wilusz JE, JnBaptiste CK, Lu LY, Kuhn CD, Joshua-Tor L, Sharp PA. A triple helix stabilizes the 3′ ends of long noncoding RNAs that lack poly(A) tails. Genes Dev. 2012;26(21):2392–407. doi: 10.1101/gad.204438.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Wu G, Wang Y, Lu X, He H, Liu H, Meng X, Xia S, Zheng K, Liu B. Low mir-372 expression correlates with poor prognosis and tumor metastasis in hepatocellular carcinoma. BMC Cancer. 2015;15:182. doi: 10.1186/s12885-015-1214-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Xie H, Ma H, Zhou D. Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int. 2013:136106. doi: 10.1155/2013/136106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Xue Q, Zhou Z, Lei X, Liu X, He B, Wang J, Hung T. TRIM38 negatively regulates TLR3-mediated IFN-β signaling by targeting TRIF for degradation. PLoS One. 2012;7:e46825. doi: 10.1371/journal.pone.0046825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Yang F, Zhang L, Huo XS, Yuan JH, Xu D, Yuan SX, Zhu N, Zhou WP, Yang GS, Wang YZ, Shang JL, Gao CF, Zhang FR, Wang F, Sun SH. Long Noncoding RNA High Expression in HCC Facilitates Tumor Growth Through Enhancer of Zeste Homolog 2 in Humans. Hepatology. 2011;54:1679–89. doi: 10.1002/hep.24563. [DOI] [PubMed] [Google Scholar]
  137. Yang Y, Liao B, Wang S, Yan B, Jin Y, Shu HB, Wang YY. E3 ligase WWP2 negatively regulates TLR3-mediated innate immune response by targeting TRIF for ubiquitination and degradation. Proc Natl Acad Sci U S A. 2013a;110:5115–5120. doi: 10.1073/pnas.1220271110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Yang F, Huo XS, Yuan SX, Zhang L, Zhou WP, Wang F, Sun SH. Repression of the long noncoding RNA-LET by histone deacetylase 3 contributes to hypoxia-mediated metastasis. Mol Cell. 2013b;49(6):1083–1096. doi: 10.1016/j.molcel.2013.01.010. [DOI] [PubMed] [Google Scholar]
  139. Yao H, Brick K, Evrard Y, Xiao T, Camerini-Otero RD, Felsenfeld G. Mediation of CTCF transcriptional insulation by DEAD-box RNA-binding protein p68 and steroid receptor RNA activator SRA. Genes Dev. 2010;24:2543–2555. doi: 10.1101/gad.1967810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Yeh HM, Yu CY, Yang HC, Ko SH, Liao CL, Lin YL. Ubiquitin-specific protease 13 regulates IFN signaling by stabilizing STAT1. J Immunol. 2013;191:3328–3336. doi: 10.4049/jimmunol.1300225. [DOI] [PubMed] [Google Scholar]
  141. 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. doi: 10.1038/ni1087. [DOI] [PubMed] [Google Scholar]
  142. Yoon JH, Abdelmohsen K, Srikantan S, Yang X, Martindale JL, De S, Huarte M, Zhan M, Becker KG, Gorospe M. LincRNA-p21 suppresses target mRNA translation. Mol Cell. 2012;47:648–655. doi: 10.1016/j.molcel.2012.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Yuan SX, Yang F, Yang Y, Tao QF, Zhang J, Huang G, Yang Y, Wang RY, Yang S, Huo XS, Zhang L, Wang F, Sun SH, Zhou WP. Long noncoding RNA associated with microvascular invasion in hepatocellular carcinoma promotes angiogenesis and serves as a predictor for hepatocellular carcinoma patients’ poor recurrence-free survival after hepatectomy. Hepatology. 2012;56(6):2231–41. doi: 10.1002/hep.25895. [DOI] [PubMed] [Google Scholar]
  144. Zhang Q, Chen CY, Yedavalli VS, Jeang KT. NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio. 2013b;4(1):e00596–00512. doi: 10.1128/mBio.00596-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Zhang Z, Bao M, Lu N, Weng L, Yuan B, Liu YJ. The E3 ubiquitin ligase TRIM21 negatively regulates the innate immune response to intracellular double-stranded DNA. Nat Immunol. 2013a;14:172–178. doi: 10.1038/ni.2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Zhang X, Bogunovic D, Payelle-Brogard B, Francois-Newton V, Speer SD, Yuan C, Volpi S, Li Z, Sanal O, Mansouri D, Tezcan I, Rice GI, Chen C, Mansouri N, Mahdaviani SA, Itan Y, Boisson B, Okada S, Zeng L, Wang X, Jiang H, Liu W, Han T, Liu D, Ma T, Wang B, Liu M, Liu JY, Wang QK, Yalnizoglu D, Radoshevich L, Uze G, Gros P, Rozenberg F, Zhang SY, Jouanguy E, Bustamante J, Garcia-Sastre A, Abel L, Lebon P, Notarangelo LD, Crow YJ, Boisson-Dupuis S, Casanova JL, Pellegrini S. Human intracellular ISG15 prevents interferon-alpha/beta over-amplification and auto-inflammation. Nature. 2015;517:89–93. doi: 10.1038/nature13801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Zhao C, Collins MN, Hsiang TY, Krug RM. Interferon-induced ISG15 pathway: an ongoing virus-host battle. Trends Microbiol. 2013;21:181–186. doi: 10.1016/j.tim.2013.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES