Next Article in Journal
Overcoming Barriers in Oncolytic Virotherapy with HDAC Inhibitors and Immune Checkpoint Blockade
Next Article in Special Issue
Direct Identification of Enteroviruses in Cerebrospinal Fluid of Patients with Suspected Meningitis by Nested PCR Amplification
Previous Article in Journal
PRMT5 Is Upregulated in HTLV-1-Mediated T-Cell Transformation and Selective Inhibition Alters Viral Gene Expression and Infected Cell Survival
Previous Article in Special Issue
Development of Novel Vaccines against Enterovirus-71
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNA and Pathogenesis of Enterovirus Infection

1
Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, No. 1 Chang-Te Street, Taipei 10048, Taiwan
2
Center of Genomic Medicine, National Taiwan University, Taipei 10048, Taiwan
3
Department of Internal Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan
4
Institute of Biomedical Sciences, Academia Sinica, Taipei 10048, Taiwan
5
Center for Optoelectronic Biomedicine, College of Medicine, National Taiwan University, Taipei 10048, Taiwan
6
Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei 10048, Taiwan
7
Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan
*
Author to whom correspondence should be addressed.
Viruses 2016, 8(1), 11; https://doi.org/10.3390/v8010011
Submission received: 1 October 2015 / Revised: 4 December 2015 / Accepted: 18 December 2015 / Published: 6 January 2016
(This article belongs to the Special Issue Recent Progress in Enterovirus Research)

Abstract

:
There are no currently available specific antiviral therapies for non-polio Enterovirus infections. Although several vaccines have entered clinical trials, the efficacy requires further evaluation, particularly for cross-strain protective activity. Curing patients with viral infections is a public health problem due to antigen alterations and drug resistance caused by the high genomic mutation rate. To conquer these limits in the development of anti-Enterovirus treatments, a comprehensive understanding of the interactions between Enterovirus and host cells is urgently needed. MicroRNA (miRNA) constitutes the biggest family of gene regulators in mammalian cells and regulates almost a half of all human genes. The roles of miRNAs in Enterovirus pathogenesis have recently begun to be noted. In this review, we shed light on recent advances in the understanding of Enterovirus infection-modulated miRNAs. The impacts of altered host miRNAs on cellular processes, including immune escape, apoptosis, signal transduction, shutdown of host protein synthesis and viral replication, are discussed. Finally, miRNA-based medication provides a promising strategy for the development of antiviral therapy.

Graphical Abstract

1. Introduction

Enterovirus (EV) infections have emerged as a major public health problem, and EV outbreaks occur frequently in summer and early fall as epidemics throughout the world [1,2,3,4,5,6,7,8,9,10,11,12,13]. Human EVs are divided into seven species: Enterovirus species A (Coxsackie A viruses 2–8, 10, 12, 14, and 16 and Enteroviruses 71, 76, 89–92, 114, and 119), Enterovirus species B (Coxsackie A virus 9; Coxsackie B viruses 1–6; Echoviruses 1–7, 9, 11–21, 24–27, and 29–33; and Enteroviruses 69, 73–75, and 77–88), Enterovirus species C (Polioviruses 1–3 and Coxsackie A viruses 1, 11, 13, 15, 17–22, and 24), Enterovirus species D (Enteroviruses 68 and 70), Rhinovirus species A (Rhinovirus A viruses 1, 2, 7–13, 15, 16, 18–25, 28–34, and 36), Rhinovirus species B (Rhinovirus B viruses 3–6, 1, 17, 26, and 27) and Rhinovirus species C (Rhinovirus C viruses 1–51). These viruses depend on the host’s physiological features, including age, sex, immune response and nutritional status [14,15]. EVs can infect people of all age groups, but they severely affect younger children in particular and occasionally cause permanent paralysis and neurological complications [16]. Growing evidence indicates that EVs have caused symptomatic infections in North America, Malaysia, Singapore, Australia, and Taiwan [11,17,18,19,20]. Enterovirus 71 (EV71), a member of Enterovirus species A, is a non-enveloped virion with positive-strand RNA. EV71 was first identified in California in 1969 and has caused several outbreaks in the United States, Europe, and Asia over the subsequent 40 years [15]. Recently, EV71 has become an emerging life-threatening pathogen, particularly in the Asia-Pacific region [21,22]. EV71 possesses extensive tissue tropisms, involving the central nervous system and muscle, skeletal muscle, intestine, and immune cells, and can cause such fatal diseases as aseptic meningitis, paralysis, pulmonary edema, encephalomyelitis or even neurologic and psychiatric symptoms [21,22,23,24,25,26].
MicroRNAs (miRNAs) are a newly discovered class of small non-protein-coding RNAs that act via endogenous RNA interference [27,28,29,30,31,32]. To date, more than 1000 cellular miRNAs have been identified that mediate the post-transcriptional regulation of more than 30% of animal genes [28,33]. MiRNA biogenesis is initiated by RNA polymerase II or III, and the newly synthesized primary miRNA transcripts (pri-miRNAs) are cleaved by RNase III and Drosha into precursor miRNAs (pre-miRNAs). Precursor miRNAs are transported into the cytoplasm and further processed by Dicer to form miRNA-miRNA duplexes [34,35,36,37,38]. The miRNA-miRNA duplexes harbor a guide strand that generally functions as the mature miRNA. Mature miRNAs associated with an Ago protein form an RNA-induced silencing complex (RISC) by which miRNAs suppress specific mRNAs by targeting complementary sites of specific mRNAs mainly located in mRNA 3′ untranslated regions (3′ UTRs) [39,40,41]. Occasionally, 5′ UTRs and protein-coding regions can act as the potential binding sites for miRNAs [42,43,44,45]. By post-transcriptional regulation, miRNAs govern a wide range of biological functions, including cell proliferation, differentiation, apoptosis and host-pathogen interactions [46,47,48]. Certain DNA viruses encode viral miRNAs that are able to regulate viral or cellular gene expressions and to contribute to viral pathogenesis [49,50,51]. In contrast, some cellular miRNAs play a role in the virus life cycle, including viral genome replication and virus propagation [52,53].
In general, viruses are predisposed to evolve more genetic diversity to overcome environmental stresses and increase competitive advantages. However, this unique virus tendency generates various antigenic variations and potentiates drug resistance that impedes the development of effective antiviral therapies. Moreover, some viruses produce viral regulatory molecules, such as miRNAs and long non-coding RNAs (lncRNAs), to play a part in viral pathogenesis and life cycle [54,55,56,57]. On the other hand, viruses utilize cellular factors to hijack host bio-energy, evade immune attacks and enforce viral replication [58,59]. In this review, we discuss the emerging roles of cellular and virus-encoded miRNAs in host-pathogen interactions and provide potential strategies for antiviral therapies by manipulating such regulatory molecules.

2. Host Cellular miRNAs in Enterovirus Pathogenesis

2.1. Host miRNAs Participate in Antiviral Responses and Immune Escape in Enterovirus Infections

Generally, certain microbe-unique molecules, such as double-stranded RNA and cytidine-phosphate-guanosine DNA (CpG DNA), are recognized by host pattern-recognition receptors and activate endosomal toll-like receptor (TLR) signaling to produce type I interferons (IFNs), the first-line immune responses against viral infection [60,61,62,63]. The resulting IFNs can establish antiviral machinery in virus-infected subjects by inducing downstream interferon-stimulated genes, promoting T cell proliferation, stimulating IFNγ production and activating dendritic cells or natural killer cells [64,65,66,67]. Intriguingly, EV71 infection fails to elicit type I IFN production efficiently [68,69]. Ho and his colleagues found that miR-146a is induced in EV71 infection and further suppressed two critical components in interferon production, Interleukin-1 receptor-associated kinase 1 (IRAK1) and TNF receptor-associated factor 6 (TRAF6). Knocking out miR-146a or neutralizing virus-induced miR-146a restores IRAK1 and TRAF6 expression, augments IFNβ production, inhibits viral propagation and finally improves the survival of virus-infected neonatal mice [58]. In this case, the authors provided a clue to help develop preventive and therapeutic strategies against Enterovirus infections by manipulating miRNA expressions. Interferon regulatory factors can mediate the upregulation of miR-526a, which further targets Cylindromatosis (turban tumor syndrome), also known as CYLD, a well-known deubiquitinase, to enforce K63-linked RIG-I ubiquitination and then triggers IRF3 and NF-κB signaling. EV71 3C protease cleaves host Interferon regulatory factor 7 (IRF7) to block IRF-mediated miR-526a upregulation and then suppresses RIG-I-dependent IFNs production [70]. In addition to EV71, Coxsackie virus B3 (CVB3) infection also influences the host miRNA expression profile [71]. Zhang and his coworkers identified five differentially expressed miRNAs in CVB3-infected mice hearts using a miRNA microarray. As determined by bioinformatics analysis, these differentially expressed miRNAs might be involved in certain important immune and antiviral pathways, such as the Toll-like receptor signaling pathway, RIG-I-like receptor signaling pathway, NOD-like receptor signaling pathway, cytokine-cytokine receptor interaction, Mitogen-activated protein kinase (MAPK) signaling pathway, Janus kinase-Signal Transducer and Activator of Transcription (JAK-STAT) signaling pathway, and natural killer cell-mediated cytotoxicity. In contrast, the host also manipulates miRNA expression to establish immune attacks or eliminate viral pathogenesis [72,73]. CVB3-infected subjects activate immune responses to attack the virus, while elevated immune responses might result in cardiac myocyte destruction, reparative fibrosis, myocarditis and even heart failure [72,74]. Both miR-155 and miR-148a were upregulated in cardiac biopsies from viral myocarditis patients caused by CVB3 infection. RelA, an important molecule in NF-κB signaling, was demonstrated in vitro as the direct target of these two upregulated miRNAs [72]. Subsequently, the regulatory role of miR-155 was demonstrated in mouse cardiac myocytes and in a mouse infection model due to the sequence conservation of miR-155 between human and mouse. MiR-155 targets RelA and functions as a negative regulator in the host immune system, by which cardiac myoblast cytokine expression is reduced and the infected subjects are protected from an overdriven immune response; thus, the survival is improved in CVB3 infection [72]. In addition, miRNA-548 mimics present an inhibitory effect on IFNλ1 by targeting its 3′ UTR to regulate the IFNλ1-mediated antiviral response [73]. Li et al. [73] highlighted miR-548 as downregulated in both Vesicular stomatitis virus and EV71 infections, whereas hosts might establish an antiviral response. In these cases, hosts and viruses regulate cellular miRNAs expression to establish immune attacks against the virus infections or evolve immune escape machineries to create a beneficial situation for viral replication, respectively.

2.2. Host miRNAs Are Involved in Enterovirus Infection-Induced Apoptosis

Apoptosis is an important cellular defense mechanism, particularly in the early phase of pathogen infection, by which pathogen-infected cells can be eliminated. Although viral spread can be partly restricted, apoptosis may cause tissue damage in the late stages of infection. Several studies have indicated that apoptosis is the major pathogenic feature of Enterovirus infection, resulting in host cell death and tissue damage [75,76,77]. Hence, the roles of miRNAs in Enterovirus infection-induced apoptosis were investigated [55,78,79]. The researchers found that miR-21 expression was suppressed in CVB3-induced myocarditis, while ectopic miR-21 expression significantly alleviated CVB3-induced myocarditis, including myocardial injury recovery, myocarditis score reduction and increased survival rate [78]. Programmed Cell Death 4 (PDCD4) was validated as the other important miR-21 target involved in apoptosis in several cell types [80,81,82]. The authors illustrated the underlying molecular mechanism in which miR-21 serves as an anti-apoptotic molecule in viral myocarditis via targeting PDCD4 [78]. On the other hand, EV71 infection induced let-7b expression in SH-SY5Y cells, which directly targets cyclin D1 (CCND1), a key element in cell cycle and apoptosis processes [79]. In contrast, the inhibition of let-7b by 2′-O-Methyl-RNA restored CCND1 expression, reduced the G2/M phase and restored SH-SY5Y proliferation. This evidence suggests that the proliferation retardation and apoptosis of EV71-infected cells are at least partly attributed to miRNA-mediated mechanisms [79]. More recently, Chang and her colleagues found that miR-146a is upregulated, while miR-370 is downregulated in EV71 infection [55]. Son Of Sevenless Homolog 1 (SOS1) and Growth arrest and DNA-damage-inducible β (GADD45β) are the targets of miR-146a and miR-370, respectively. Further functional studies indicated that the silencing of miR-146a restores SOS1 expression and partially attenuates EV71-induced apoptosis, while the ectopic expression of miR-370 decreases EV71-induced GADD45β expression and diminishes apoptosis. Moreover, the co-expression of antagomiR-146a and miR-370 showed an additive effect on attenuating EV71-induced apoptosis [55]. These studies clearly demonstrate that host miRNAs play a critical role in Enterovirus infection-induced apoptosis by regulating the key elements that are involved in cellular apoptosis and imply that miRNAs might act as potential therapeutic candidates by attenuating Enterovirus infection-induced apoptosis [55,78,79]. The miRNAs that are involved in the regulation of Enterovirus-induced apoptosis are elucidated in Figure 1 and summarized in Table 1.
Figure 1. Cellular miRNAs are involved in Enterovirus infection-induced apoptosis. Hexagons indicate Enterovirus, hairpins indicate pre-miRNAs, red arrows indicate upregulated expression and green arrows indicate downregulated expression.
Figure 1. Cellular miRNAs are involved in Enterovirus infection-induced apoptosis. Hexagons indicate Enterovirus, hairpins indicate pre-miRNAs, red arrows indicate upregulated expression and green arrows indicate downregulated expression.
Viruses 08 00011 g001
Apoptosis is the major pathogenic feature of Enterovirus infections. MiR-21, which targets Programmed Cell Death 4 (PDCD4), is suppressed in CVB3-induced myocarditis and serves as an anti-apoptotic molecule. EV71-induced let-7b directly targets cyclin D1 (CCND1) and contributes to the retardation of host cell proliferation and the trigger of apoptosis. The other two altered miRNAs that were identified in EV71 infection, miR-146a and miR-370, target SOS1 and GADD45β, respectively. The suppression of SOS1 by miR-146a and the relief of GADD45β by miR-370 induce the apoptosis of EV71-infected host cells. These cellular miRNAs play critical roles in Enterovirus infection-induced apoptosis by regulating the key elements in the cellular apoptosis process.

2.3. Enterovirus Regulates Host Signaling Modulators Resulting in Pathogenesis

Many studies have clearly indicated that Enteroviral proteases cleave host factors and then lead to cellular or tissue malfunctions, such as apoptosis, translation shutdown and cytokine suppression, and finally cause pathogenesis [83,84,85,86,87,88,89,90]. For example, EV68 3C protease cleaved host cell TIR-domain-containing adapter-inducing interferon-β (TRIF), a key molecule downstream of TLR3, to abolish NF-κB signaling and IFN-β production and enabled viruses to escape the host innate immune response [83]. In addition to viral proteases, miRNAs have also been characterized as a critical players in host signaling transduction [54,55,58,78,79,91,92,93,94,95,96,97]. In the investigation of Coxsackie virus-induced cardiovascular pathogenesis, the destruction of cell-cell interactions is one of its key mechanisms. Several components of membrane structure are the targets of Coxsackie virus-regulated miRNAs. CBV3-induced miR-21 enhances desmin degradation by targeting a deubiquitinating enzyme, YOD1 [54], and Gap junction protein alpha 1 (GPJ1) is the target of CBV3-induced miR-1 [93]. MiR-21 also activated mitogen-activated protein kinase (MAPK) signaling cascade by targeting sprouty homolog 1 (SPRY1), a native inhibitor of fibroblast growth factor (FGF) pathways [94]. Moreover, CBV3 infection triggered ERK1/2 activation, leading to miR-126 upregulation. The miR-126 played as a pivot to link the ERK1/2 and WNT/β-catenin signaling pathways and promoted CBV3 propagation [96].
Table 1. MicroRNAs(miRNAs) regulations in Enteroviruses infections.
Table 1. MicroRNAs(miRNAs) regulations in Enteroviruses infections.
miRNATargetEnterovirusExpressionProcessModelReference
miR-146aIRAK1EV71UpregulationImmune responseIn vitro & In vivo[58]
miR-146aTRAF6EV71UpregulationImmune responseIn vitro & In vivo[58]
miR-526aCYLDEV71DownregulationImmune responseIn vitro[70]
miR-155RelACVB3 & VMC aUpregulationImmune responseIn vitro & In vivo[72]
miR-148aRelACVB3 & VMCUpregulationImmune responseIn vitro[72]
miR-548IFNλ1EV71 & HBV-infected subjects bDownregulationImmune responseIn vitro[73]
miR-21PDCD4CVB3DownregulationApoptosisIn vitro & In vivo[78]
let-7bCCND1EV71UpregulationCell cycle and ProliferationIn vitro[79]
miR-146aSOS1EV71UpregulationApoptosisIn vitro[55]
miR-370GADD45bEV71DownregulationApoptosisIn vitro[55]
miR-21YOD1CVB3UpregulationCell-cell interactionIn vitro[54]
miR-1GPJ1CVB3UpregulationCell-cell interactionIn vitro[93]
miR-21SPRY1VMC & DCM cEctopicMAPK signalingIn vitro[94]
miR-126LRPCVB3UpregulationWnt/β-catenin signalingIn vitro[96]
miR-126WRCH1CVB3UpregulationWnt/β-catenin signalingIn vitro[96]
miR-1246DLG3EV71UpregulationCell death signalingIn vitro[95]
miR-27aEGFREV71DownregulationEGFR signalingIn vitro[97]
miR-141eIF4EEV71UpregulationProtein synthesisIn vitro[59]
miR-296-5pEV71 VP1EV71UpregulationViral replicationIn vitro[56]
miR-296-5pEV71 VP3EV71UpregulationViral replicationIn vitro[56]
miR-23bEV71 VP1EV71DownregulationViral replicationIn vitro[98]
miR-342-5pCVB3 2CNA dEctopicViral replicationIn vitro[99]
miR-373EV71 5′ UTRNAEctopicViral replicationIn vitro[100]
miR-542-5pEV71 5′ UTRNAEctopicViral replicationIn vitro[100]
miR-10a*CVB3 3DNAEctopicViral replicationIn vitro[57]
a determined in CVB3 infection and viral myocarditis subjects (VMC); b determined in EV71 infection and HBV-infected subjects; c determined in viral myocarditis subjects (VMC) and dilated cardiomyopathy subjects (DCM); d not determined in virus infection.
Next, we focused on the role of miRNAs in EV71-induced neurological pathogenesis due to the severe neuronal complications in EV71-infected patients [21,24]. The EV71-induced miR-1246 directly repressed the expression of disc-large homolog 3 (DLG3), which is a member of the membrane-associated guanylate kinase protein family and is associated with mental disorders [95]. MiR-1246 might contribute to EV71-associated neurological pathogenesis by targeting DLG3. In addition to the direct destruction of neuronal components, the inflammatory reaction is one of main causes contributing to tissue damage, such as meningitis and encephalitis [101,102]. The increased Cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) induced by Enterovirus infection accelerated EV71 replication and reactive oxygen species (ROS) generation, and the upstream regulation mechanisms have been intensively elucidated [103,104,105]. Among these regulatory mediators, EGFR signaling plays a crucial role in EV71 replication in the human neuroblastoma cell line, SK-N-SH cells [103,104]. Zhang and his colleagues found that the expression of miR-27a is downregulated during EV71 infection and that miR-27a could target EGFR [97]. Further studies have shown that the ectopic expression of miR-27a suppresses EGFR expression and reduces Akt and ERK phosphorylation. Finally, the blockage of the EGFR signaling cascade attenuated EV71 replication [97]. The miRNAs that are involved in the regulation of host signaling pathways are summarized in Table 1.

2.4. Host miRNAs Are Involved in Enterovirus Infection-Induced Protein Synthesis Shutdown

It has long been believed that the viral protease-mediated cleavage of host factors that are involved in the host cap-dependent translation process might almost or even thoroughly account for the shutdown of host protein synthesis caused by Picornavirus infection [85,86,90,106,107]. Enterovirus 2A and 3C proteases digested the host eukaryotic initiation factor 4G (eIF4G) and Poly(A)-binding protein (PABP), and these cleavages led to the shutdown of host cell protein synthesis and further promoted apoptosis, along with nuclear condensation and DNA fragmentation [85,86,90]. In addition to viral proteases, miRNAs have also been characterized as critical players in host protein synthesis shutdown and signaling regulation [54,55,59,78,79,91,92,93,94,95,96,97,108]. The role of miRNAs in the virus-induced translational switch has recently begun to be investigated. Ho and his colleagues found that cellular miR-141 is induced by EV71 infection, which targets eIF4E, the cap-dependent translation initiation factor, and results in the shutdown of host protein synthesis, while the silencing of virus infection-induced miR-141 almost recovers host protein synthesis and blocks viral propagation up to 1000-fold [59]. Hence, virus infection-induced miR-141 expedites the translational switch from cap-dependent translation to cap-independent translation. This study largely increased the understanding of how miRNA facilitates Enterovirus-causing host protein synthesis in addition to the classical concept [59,109].

3. Host miRNAs Are Involved in the Enterovirus Life Cycle

3.1. Cellular miRNAs Target Vial Genome to Suppress Viral Replication

The host and virus influence cellular miRNA expressions during virus infection, and the altered miRNAs target cellular molecules to enforce viral pathogenesis or establish host defense machineries. Recently, several studies have suggested that host miRNAs could target viral genomes and result in the suppression of virus replication [56,57,98,99,100]. The induction of miRNA-296-5p was found in EV71-infected human rhabdomyosarcoma (RD) and SK-N-SH cells, and the silicon analysis results predicted that miRNA-296-5p might target both the VP1 and VP3 regions of the viral genome; this speculation was confirmed by molecular biological assays (Figure 2) [56]. Meanwhile, the suppression of endogenous miR-296-5p promoted EV71 replication, and the introduction of mutations into binding sites on the viral genome led EV71 to escape the inhibitory effects of miR-296-5p [56]. Wang et al. [99] and Wen et al. [98] and also found similar phenomena in CVB3 and EV71 infections, respectively [98,99]. The expression of miR-23b was downregulated in EV71 infection, while the restoration of miR-23b by specific mimic inhibited EV71 production (Figure 2). A further study has evidenced that miR-23b suppressed EV71 replication by targeting the EV71 VPl protein [98]. Wang and the coworkers discovered that miR-342-5p could significantly inhibit CVB3 replication by directly targeting the 2C-coding region of the viral genome (Figure 2) [99]. Both miR-373 and miR-542-5p target the 5′ untranslated region (5′ UTR) of EV71 vRNA and thus attenuate viral propagation as assayed in RD cells (Figure 2) [100].
Figure 2. Host miRNAs are involved in the Enterovirus life cycle by targeting to viral genomes. Hexagons indicate Enterovirus, solid lines indicate viral RNAs, dotted lines indicate inhibited viral RNAs, red arrows indicate upregulated expression and green arrows indicate downregulated expression.
Figure 2. Host miRNAs are involved in the Enterovirus life cycle by targeting to viral genomes. Hexagons indicate Enterovirus, solid lines indicate viral RNAs, dotted lines indicate inhibited viral RNAs, red arrows indicate upregulated expression and green arrows indicate downregulated expression.
Viruses 08 00011 g002
Cellular miRNAs play a regulatory role in the viral life cycle to suppress or promote virus replication by targeting the viral genome. MiR-296-5p is upregulated, while miR-23b is downregulated in EV71 infection. EV71 infection-induced miR-296-5p could target both viral VP1 and VP3 regions and result in the inhibition of virus replication. The downregulation of miR-23b in EV71 infection relieves EV71 viral RNAs (vRNAs) from miRNA-mediated inhibition. In addition to endogenous miRNAs, the ectopic introduction of miRNAs also affects virus replication. The ectopic expression of miR-342-5p, miR-373 or miR-542-5p suppresses Enterovirus replication, but miR-10* facilitates CVB3 replication. MiR-342-5p targets the CVB3 2C-coding region and thus inhibits CVB3 replication. In contrast, miR-10a* acts as a functional RNA molecule to facilitate CVB3 replication by targeting the viral 3D-coding region. Similarly, miR-373 and miR-542-5p attenuate EV71 propagation by targeting the 5′ UTR of EV71 vRNAs.

3.2. Cellular miRNAs Target the Viral Genome to Promote Viral Propagation

Intriguingly, miR-10a*, the passenger strand of the miRNA duplex, is a functional RNA molecule in CVB3 infection, in which miR-10a* directly targets the nt6818-nt6941 sequence of the CVB3 3D-coding region, facilitating CVB3 replication (Figure 2) [57]. Furthermore, miR-10a* was abundant in the cardiac tissues of suckling Balb/c mice, indicating that miR-10a* might be involved in CVB3-induced myocarditis [57]. Despite a large number of reports evidently showing that miRNAs suppress target gene expression via the RISC complex, a few cases have shown that miRNAs are capable of positively regulating target gene expressions [53,110,111]. As mentioned in 2005, a host miRNA, miR-122, targeted Hepatitis C virus (HCV) 5′ noncoding region, facilitating viral RNA replication by promoting RNA folding or sequestration in the replication complexes [53]. This evidence implied that miRNAs might serve as potential therapeutic agents for Enterovirus infection treatment. The miRNAs that are involved in the manipulation of viral genomes are provided in Figure 2 and summarized in Table 1.

4. Conclusions and Perspective

A complete molecular understanding of virus pathogenesis is necessary to develop more efficient strategies against virus infection. Since the discovery of miRNAs in 1993, miRNA is becoming the largest family of gene regulators, accounting for the regulation of one-third of human genes. In the case of Enterovirus infection, virus-induced cellular miRNAs modulate the cellular and infection processes and contribute to pathogenesis by targeting either host mRNAs or virus RNAs. This growing evidence demonstrates the important role of miRNAs in Enterovirus infection-related pathogenesis and accelerates the progress of anti-virus strategies. There are no effective antiviral treatments, approved medications or vaccines for Enterovirus infections except for Poliovirus [16,112,113]. Routine treatment is supportive for reducing clinical symptoms. The efficacy of passive immunoglobulin treatments is not often effective or satisfying for infected subjects. Unfortunately, quarantine might be the best solution to prevent Enterovirus outbreaks. Although anti-EV vaccines are undergoing clinical trials and may soon be approved, the cross-protection of these vaccines requires further evaluation. Herein, miRNAs provide an opportunity to develop alternative miRNA-based therapeutic and preventive strategies for non-polio Enterovirus infections. In fact, the phase I clinical trial of MRX34, a miR-34 mimic, in solid tumors and hematological malignancies is expected to be completed by the end of 2015 [114]. A phase IIa clinical trial was conducted to assess the safety and efficacy of Miravirsen, a 15-nucleotide locked nucleic acid-modified antisense oligonucleotide against mature miR-122 in HCV carriers, and the preliminary results showed exciting therapeutic potential with acceptable adverse drug effects (ADE) [115]. Furthermore, the phase II clinical trial of RG101, another anti-miR-122 antagonist, combined with an anti-HCV agent in HCV patients has been underway since August 2015. For EV infection control, miR-146a-based medication provides new insight into the potential anti-EV71 therapy by showing that the neutralization of EV71-induced miR-146a prevents death, as demonstrated in an EV71 infection mouse model via the restoration of type I interferon production [58]. The potential long-term cytotoxicity and genotoxicity might not be a critical consideration in EV infections compared to chronic infections or cancers due to the short therapeutic period. Moreover, targeting host components is a new strategy to overcome the drug resistance and antigenic variation of viruses, particularly RNA viruses. Since the mystery of miRNAs in Enterovirus infection is generally understood, the preventive and therapeutic activities of miRNAs might be conspicuous in the foreseablefuture.

Acknowledgments

We thank the supports from NRPGM (MOST103-2319-B-002-002), NRPB (MOST103-2325-B-002-026 and MOST 103-2325-B-002-035), and EID (MOST 104-2321-B-002-047). We apologized to the authors whose work we either misinterpreted or failed to cite.

Author Contributions

Bing-Ching Ho, Pan-Chyr Yang and Sung-Liang Yu wrote the review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Badran, S.A.; Midgley, S.; Andersen, P.; Bottiger, B. Clinical and virological features of enterovirus 71 infections in Denmark, 2005 to 2008. Scand. J. Infect. Dis. 2011, 43, 642–648. [Google Scholar] [CrossRef] [PubMed]
  2. Bian, L.; Wang, Y.; Yao, X.; Mao, Q.; Xu, M.; Liang, Z. Coxsackievirus A6: A new emerging pathogen causing hand, foot and mouth disease outbreaks worldwide. Expert Rev. Anti-Infect. Ther. 2015, 13, 1061–1071. [Google Scholar] [CrossRef] [PubMed]
  3. Chua, K.B.; Kasri, A.R. Hand foot and mouth disease due to enterovirus 71 in Malaysia. Virol. Sin. 2011, 26, 221–228. [Google Scholar] [CrossRef] [PubMed]
  4. Gaunt, E.; Harvala, H.; Osterback, R.; Sreenu, V.B.; Thomson, E.; Waris, M.; Simmonds, P. Genetic characterization of human coxsackievirus A6 variants associated with atypical hand, foot and mouth disease: A potential role of recombination in emergence and pathogenicity. J. Gen. Virol. 2015, 96, 1067–1079. [Google Scholar] [CrossRef] [PubMed]
  5. Guan, H.; Wang, J.; Wang, C.; Yang, M.; Liu, L.; Yang, G.; Ma, X. Etiology of multiple Non-EV71 and non-CVA16 enteroviruses associated with hand, foot and mouth disease in Jinan, China, 2009—June 2013. PLoS ONE 2015, 10, e0142733. [Google Scholar] [CrossRef] [PubMed]
  6. Honkanen, H.; Oikarinen, S.; Pakkanen, O.; Ruokoranta, T.; Pulkki, M.M.; Laitinen, O.H.; Tauriainen, S.; Korpela, S.; Lappalainen, M.; Vuorinen, T.; et al. Human enterovirus 71 strains in the background population and in hospital patients in Finland. J. Clin. Virol. 2013, 56, 348–353. [Google Scholar] [CrossRef] [PubMed]
  7. Huang, X.; Wei, H.; Wu, S.; Du, Y.; Liu, L.; Su, J.; Xu, Y.; Wang, H.; Li, X.; Wang, Y.; et al. Epidemiological and etiological characteristics of hand, foot, and mouth disease in Henan, China, 2008–2013. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, H.; Kang, B.; Hwang, S.; Lee, S.W.; Cheon, D.S.; Kim, K.; Jeong, Y.S.; Hyeon, J.Y. Clinical and enterovirus findings associated with acute flaccid paralysis in the Republic of Korea during the recent decade. J. Med. Virol. 2014, 86, 1584–1589. [Google Scholar] [CrossRef] [PubMed]
  9. Lee, T.C.; Guo, H.R.; Su, H.J.; Yang, Y.C.; Chang, H.L.; Chen, K.T. Diseases caused by enterovirus 71 infection. Pediatr. Infect. Dis. J. 2009, 28, 904–910. [Google Scholar] [CrossRef] [PubMed]
  10. Linsuwanon, P.; Puenpa, J.; Huang, S.W.; Wang, Y.F.; Mauleekoonphairoj, J.; Wang, J.R.; Poovorawan, Y. Epidemiology and seroepidemiology of human enterovirus 71 among Thai populations. J. Biomed. Sci. 2014, 21, 1110–1186. [Google Scholar] [CrossRef] [PubMed]
  11. Singh, S.; Poh, C.L.; Chow, V.T. Complete sequence analyses of enterovirus 71 strains from fatal and non-fatal cases of the hand, foot and mouth disease outbreak in Singapore (2000). Microbiol. Immunol. 2002, 46, 801–808. [Google Scholar] [CrossRef] [PubMed]
  12. Wu, J.S.; Zhao, N.; Pan, H.; Wang, C.M.; Wu, B.; Zhang, H.M.; He, H.X.; Liu, D.; Amer, S.; Liu, S.L. Patterns of polymorphism and divergence in the VP1 gene of enterovirus 71 circulating in the Asia-Pacific region between 1994 and 2013. J. Virol. Methods 2013, 193, 713–728. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, W.H.; Kuo, T.C.; Lin, Y.T.; Huang, S.W.; Liu, H.F.; Wang, J.; Chen, Y.M. Molecular epidemiology of enterovirus 71 infection in the central region of Taiwan from 2002 to 2012. PLoS ONE 2013, 8, e83711. [Google Scholar] [CrossRef] [PubMed]
  14. Fields, B.N.; Knipe, D.M.; Howley, P.M. Fields' Virology; Wolters Kluwer Health/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2007. [Google Scholar]
  15. Bible, J.M.; Pantelidis, P.; Chan, P.K.; Tong, C.Y. Genetic evolution of enterovirus 71: Epidemiological and pathological implications. Rev. Med. Virol. 2007, 17, 371–379. [Google Scholar] [CrossRef] [PubMed]
  16. Solomon, T.; Lewthwaite, P.; Perera, D.; Cardosa, M.J.; McMinn, P.; Ooi, M.H. Virology, epidemiology, pathogenesis, and control of enterovirus 71. Lancet Infect. Dis. 2010, 10, 778–790. [Google Scholar] [CrossRef]
  17. Shimizu, H.; Utama, A.; Yoshii, K.; Yoshida, H.; Yoneyama, T.; Sinniah, M.; Yusof, M.A.; Okuno, Y.; Okabe, N.; Shih, S.R.; et al. Enterovirus 71 from fatal and nonfatal cases of hand, foot and mouth disease epidemics in Malaysia, Japan and Taiwan in 1997–1998. Jpn J. Infect. Dis. 1999, 52, 12–15. [Google Scholar] [PubMed]
  18. McMinn, P.; Stratov, I.; Nagarajan, L.; Davis, S. Neurological manifestations of enterovirus 71 infection in children during an outbreak of hand, foot, and mouth disease in Western Australia. Clin. Infect. Dis. 2001, 32, 236–242. [Google Scholar] [CrossRef] [PubMed]
  19. Ryu, W.S.; Kang, B.; Hong, J.; Hwang, S.; Kim, A.; Kim, J.; Cheon, D.S. Enterovirus 71 infection with central nervous system involvement, South Korea. Emerg. Infect. Dis. 2010, 16, 1764–1766. [Google Scholar] [CrossRef] [PubMed]
  20. Pons-Salort, M.; Parker, E.P.; Grassly, N.C. The epidemiology of non-polio enteroviruses: Recent advances and outstanding questions. Curr. Opin. Infect. Dis. 2015, 28, 479–487. [Google Scholar] [CrossRef] [PubMed]
  21. Chang, L.Y.; Huang, L.M.; Gau, S.S.; Wu, Y.Y.; Hsia, S.H.; Fan, T.Y.; Lin, K.L.; Huang, Y.C.; Lu, C.Y.; Lin, T.Y. Neurodevelopment and cognition in children after enterovirus 71 infection. N. Engl. J. Med. 2007, 356, 1226–1234. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, Y.F.; Chou, C.T.; Lei, H.Y.; Liu, C.C.; Wang, S.M.; Yan, J.J.; Su, I.J.; Wang, J.R.; Yeh, T.M.; Chen, S.H.; et al. A mouse-adapted enterovirus 71 strain causes neurological disease in mice after oral infection. J. Virol. 2004, 78, 7916–7924. [Google Scholar] [CrossRef] [PubMed]
  23. Chang, L.Y.; Huang, Y.C.; Lin, T.Y. Fulminant neurogenic pulmonary oedema with hand, foot, and mouth disease. Lancet 1998, 352, 36736–36738. [Google Scholar]
  24. Huang, C.C.; Liu, C.C.; Chang, Y.C.; Chen, C.Y.; Wang, S.T.; Yeh, T.F. Neurologic complications in children with enterovirus 71 infection. N. Engl. J. Med. 1999, 341, 936–942. [Google Scholar] [CrossRef] [PubMed]
  25. Lin, Y.W.; Wang, S.W.; Tung, Y.Y.; Chen, S.H. Enterovirus 71 infection of human dendritic cells. Exp. Biol. Med. 2009, 234, 1166–1173. [Google Scholar] [CrossRef] [PubMed]
  26. Whitton, J.L.; Cornell, C.T.; Feuer, R. Host and virus determinants of picornavirus pathogenesis and tropism. Nat. Rev. Microbiol. 2005, 3, 765–776. [Google Scholar] [CrossRef] [PubMed]
  27. Hammond, S.M. MicroRNAs as oncogenes. Curr. Opin. Genet. Dev. 2006, 16, 4–9. [Google Scholar] [CrossRef] [PubMed]
  28. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [PubMed]
  29. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  30. Park, J.H.; Shin, C. MicroRNA-directed cleavage of targets: Mechanism and experimental approaches. BMB Rep. 2014, 47, 417–423. [Google Scholar] [CrossRef] [PubMed]
  31. Hammond, S.M. An overview of microRNAs. Adv. Drug Deliv. Rev. 2015, 87, 3–14. [Google Scholar] [CrossRef] [PubMed]
  32. Lin, S.; Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 2015, 15, 321–333. [Google Scholar] [CrossRef] [PubMed]
  33. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef] [PubMed]
  34. Cullen, B.R. Transcription and processing of human microRNA precursors. Mol. Cell 2004, 16, 861–865. [Google Scholar] [CrossRef] [PubMed]
  35. Borchert, G.M.; Lanier, W.; Davidson, B.L. RNA polymerase III transcribes human microRNAs. Nat. Struct. Mol. Biol. 2006, 13, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
  36. Hutvagner, G.; McLachlan, J.; Pasquinelli, A.E.; Balint, E.; Tuschl, T.; Zamore, P.D. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 2001, 293, 834–838. [Google Scholar] [CrossRef] [PubMed]
  37. Bernstein, E.; Caudy, A.A.; Hammond, S.M.; Hannon, G.J. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001, 409, 363–366. [Google Scholar] [CrossRef] [PubMed]
  38. Grishok, A.; Pasquinelli, A.E.; Conte, D.; Li, N.; Parrish, S.; Ha, I.; Baillie, D.L.; Fire, A.; Ruvkun, G.; Mello, C.C. Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell 2001, 106, 23–34. [Google Scholar] [CrossRef]
  39. Hammond, S.M.; Bernstein, E.; Beach, D.; Hannon, G.J. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature 2000, 404, 293–296. [Google Scholar] [PubMed]
  40. Wu, L.; Fan, J.; Belasco, J.G. MicroRNAs direct rapid deadenylation of mRNA. Proc. Natl. Acad. Sci. USA 2006, 103, 4034–4039. [Google Scholar] [CrossRef] [PubMed]
  41. Standart, N.; Jackson, R.J. MicroRNAs repress translation of m7Gppp-capped target mRNAs in vitro by inhibiting initiation and promoting deadenylation. Genes Dev. 2007, 21, 1975–1982. [Google Scholar] [CrossRef] [PubMed]
  42. Duursma, A.M.; Kedde, M.; Schrier, M.; le Sage, C.; Agami, R. miR-148 targets human DNMT3b protein coding region. RNA 2008, 14, 872–877. [Google Scholar] [CrossRef] [PubMed]
  43. Rigoutsos, I. New tricks for animal microRNAS: Targeting of amino acid coding regions at conserved and nonconserved sites. Cancer Res. 2009, 69, 3245–3248. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, I.; Ajay, S.S.; Yook, J.I.; Kim, H.S.; Hong, S.H.; Kim, N.H.; Dhanasekaran, S.M.; Chinnaiyan, A.M.; Athey, B.D. New class of microRNA targets containing simultaneous 5′-UTR and 3′-UTR interaction sites. Genom. Res. 2009, 19, 1175–1183. [Google Scholar] [CrossRef] [PubMed]
  45. Grey, F.; Tirabassi, R.; Meyers, H.; Wu, G.; McWeeney, S.; Hook, L.; Nelson, J.A. A viral microRNA down-regulates multiple cell cycle genes through mRNA 5′AUTRs. PLoS Pathog. 2010, 6, e1000967. [Google Scholar] [CrossRef] [PubMed]
  46. Croce, C.M. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009, 10, 704–714. [Google Scholar] [CrossRef] [PubMed]
  47. Umbach, J.L.; Cullen, B.R. The role of RNAi and microRNAs in animal virus replication and antiviral immunity. Genes Dev. 2009, 23, 1151–1164. [Google Scholar] [CrossRef] [PubMed]
  48. Winter, J.; Jung, S.; Keller, S.; Gregory, R.I.; Diederichs, S. Many roads to maturity: MicroRNA biogenesis pathways and their regulation. Nat. Cell Biol. 2009, 11, 228–234. [Google Scholar] [CrossRef] [PubMed]
  49. Umbach, J.L.; Kramer, M.F.; Jurak, I.; Karnowski, H.W.; Coen, D.M.; Cullen, B.R. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature 2008, 454, 780–783. [Google Scholar] [CrossRef] [PubMed]
  50. Pfeffer, S.; Zavolan, M.; Grasser, F.A.; Chien, M.; Russo, J.J.; Ju, J.; John, B.; Enright, A.J.; Marks, D.; Sander, C.; et al. Identification of virus-encoded microRNAs. Science 2004, 304, 734–736. [Google Scholar] [CrossRef] [PubMed]
  51. Gottwein, E.; Mukherjee, N.; Sachse, C.; Frenzel, C.; Majoros, W.H.; Chi, J.T.; Braich, R.; Manoharan, M.; Soutschek, J.; Ohler, U.; et al. A viral microRNA functions as an orthologue of cellular miR-155. Nature 2007, 450, 1096–1099. [Google Scholar] [CrossRef] [PubMed]
  52. Triboulet, R.; Mari, B.; Lin, Y.L.; Chable-Bessia, C.; Bennasser, Y.; Lebrigand, K.; Cardinaud, B.; Maurin, T.; Barbry, P.; Baillat, V.; et al. Suppression of microRNA-silencing pathway by HIV-1 during virus replication. Science 2007, 315, 1579–1582. [Google Scholar] [CrossRef] [PubMed]
  53. Jopling, C.L.; Yi, M.; Lancaster, A.M.; Lemon, S.M.; Sarnow, P. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science 2005, 309, 1577–1581. [Google Scholar] [CrossRef] [PubMed]
  54. Ye, X.; Zhang, H.M.; Qiu, Y.; Hanson, P.J.; Hemida, M.G.; Wei, W.; Hoodless, P.A.; Chu, F.; Yang, D. Coxsackievirus-induced miR-21 disrupts cardiomyocyte interactions via the downregulation of intercalated disk components. PLoS Pathog. 2014, 10, e1004070. [Google Scholar] [CrossRef] [PubMed]
  55. Chang, Y.L.; Ho, B.C.; Sher, S.; Yu, S.L.; Yang, P.C. miR-146a and miR-370 coordinate enterovirus 71-induced cell apoptosis through targeting SOS1 and GADD45β. Cell Microbiol. 2015, 17, 802–818. [Google Scholar] [CrossRef] [PubMed]
  56. Zheng, Z.; Ke, X.; Wang, M.; He, S.; Li, Q.; Zheng, C.; Zhang, Z.; Liu, Y.; Wang, H. Human microRNA hsa-miR-296-5p suppresses enterovirus 71 replication by targeting the viral genome. J. Virol. 2013, 87, 5645–5656. [Google Scholar] [CrossRef] [PubMed]
  57. Tong, L.; Lin, L.; Wu, S.; Guo, Z.; Wang, T.; Qin, Y.; Wang, R.; Zhong, X.; Wu, X.; Wang, Y.; et al. MiR-10a* up-regulates coxsackievirus B3 biosynthesis by targeting the 3D-coding sequence. Nucleic Acids Res. 2013, 41, 3760–3771. [Google Scholar] [CrossRef] [PubMed]
  58. Ho, B.C.; Yu, I.S.; Lu, L.F.; Rudensky, A.; Chen, H.Y.; Tsai, C.W.; Chang, Y.L.; Wu, C.T.; Chang, L.Y.; Shih, S.R.; et al. Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon. Nat. Commun. 2014, 5. [Google Scholar] [CrossRef] [PubMed]
  59. Ho, B.C.; Yu, S.L.; Chen, J.J.; Chang, S.Y.; Yan, B.S.; Hong, Q.S.; Singh, S.; Kao, C.L.; Chen, H.Y.; Su, K.Y.; et al. Enterovirus-induced miR-141 contributes to shutoff of host protein translation by targeting the translation initiation factor eIF4E. Cell Host Microbe 2011, 9, 58–69. [Google Scholar] [CrossRef] [PubMed]
  60. Klotman, M.E.; Chang, T.L. Defensins in innate antiviral immunity. Nat. Rev. Immunol. 2006, 6, 447–456. [Google Scholar] [CrossRef] [PubMed]
  61. Wilson, S.S.; Wiens, M.E.; Smith, J.G. Antiviral mechanisms of human defensins. J. Mol. Biol. 2013, 425, 4965–4980. [Google Scholar] [CrossRef] [PubMed]
  62. Kawai, T.; Akira, S. Innate immune recognition of viral infection. Nat. Immunol. 2006, 7, 131–137. [Google Scholar] [CrossRef] [PubMed]
  63. Thompson, M.R.; Kaminski, J.J.; Kurt-Jones, E.A.; Fitzgerald, K.A. Pattern recognition receptors and the innate immune response to viral infection. Viruses 2011, 3, 920–940. [Google Scholar] [CrossRef] [PubMed]
  64. Biron, C.A.; Nguyen, K.B.; Pien, G.C.; Cousens, L.P.; Salazar-Mather, T.P. Natural killer cells in antiviral defense: Function and regulation by innate cytokines. Annu. Rev. Immunol. 1999, 17, 189–220. [Google Scholar] [CrossRef] [PubMed]
  65. Le Bon, A.; Schiavoni, G.; D’Agostino, G.; Gresser, I.; Belardelli, F.; Tough, D.F. Type I interferons potently enhance humoral immunity and can promote isotype switching by stimulating dendritic cells in vivo. Immunity 2001, 14, 461–470. [Google Scholar] [CrossRef]
  66. Nguyen, K.B.; Watford, W.T.; Salomon, R.; Hofmann, S.R.; Pien, G.C.; Morinobu, A.; Gadina, M.; O’Shea, J.J.; Biron, C.A. Critical role for STAT4 activation by type 1 interferons in the interferon-γ response to viral infection. Science 2002, 297, 2063–2066. [Google Scholar] [CrossRef] [PubMed]
  67. Tough, D.F.; Sun, S.; Zhang, X.; Sprent, J. Stimulation of naive and memory T cells by cytokines. Immunol. Rev. 1999, 170, 39–47. [Google Scholar] [CrossRef] [PubMed]
  68. Ichimura, H.; Shimase, K.; Tamura, I.; Kaneto, E.; Kurimura, O.; Aramitsu, Y.; Kurimura, T. Neutralizing antibody and interferon-α in cerebrospinal fluids and sera of acute aseptic meningitis. J. Med. Virol. 1985, 15, 231–237. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, M.L.; Lee, Y.P.; Wang, Y.F.; Lei, H.Y.; Liu, C.C.; Wang, S.M.; Su, I.J.; Wang, J.R.; Yeh, T.M.; Chen, S.H.; et al. Type I interferons protect mice against enterovirus 71 infection. J. Gen. Virol. 2005, 86Pt 12, 3263–3269. [Google Scholar] [CrossRef] [PubMed]
  70. Xu, C.; He, X.; Zheng, Z.; Zhang, Z.; Wei, C.; Guan, K.; Hou, L.; Zhang, B.; Zhu, L.; Cao, Y.; et al. Downregulation of microRNA miR-526a by enterovirus inhibits RIG-I-dependent innate immune response. J. Virol. 2014, 88, 11356–11368. [Google Scholar] [CrossRef] [PubMed]
  71. Zhang, Q.; Xiao, Z.; He, F.; Zou, J.; Wu, S.; Liu, Z. MicroRNAs regulate the pathogenesis of CVB3-induced viral myocarditis. Intervirology 2013, 56, 104–113. [Google Scholar] [CrossRef] [PubMed]
  72. Bao, J.L.; Lin, L. MiR-155 and miR-148a reduce cardiac injury by inhibiting NF-κB pathway during acute viral myocarditis. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 2349–2356. [Google Scholar] [PubMed]
  73. Li, Y.; Xie, J.; Xu, X.; Wang, J.; Ao, F.; Wan, Y.; Zhu, Y. MicroRNA-548 down-regulates host antiviral response via direct targeting of IFN-lambda1. Protein Cell 2013, 4, 130–141. [Google Scholar] [CrossRef] [PubMed]
  74. Shauer, A.; Gotsman, I.; Keren, A.; Zwas, D.R.; Hellman, Y.; Durst, R.; Admon, D. Acute viral myocarditis: Current concepts in diagnosis and treatment. Isr. Med. Assoc. J. IMAJ 2013, 15, 180–185. [Google Scholar] [PubMed]
  75. Khong, W.X.; Foo, D.G.; Trasti, S.L.; Tan, E.L.; Alonso, S. Sustained high levels of interleukin-6 contribute to the pathogenesis of enterovirus 71 in a neonate mouse model. J. Virol. 2011, 85, 3067–3076. [Google Scholar] [CrossRef] [PubMed]
  76. Ch’ng, W.C.; Stanbridge, E.J.; Ong, K.C.; Wong, K.T.; Yusoff, K.; Shafee, N. Partial protection against enterovirus 71 (EV71) infection in a mouse model immunized with recombinant Newcastle disease virus capsids displaying the EV71 VP1 fragment. J. Med. Virol. 2011, 83, 1783–1791. [Google Scholar] [CrossRef] [PubMed]
  77. Aubert, M.; Jerome, K.R. Apoptosis prevention as a mechanism of immune evasion. Int. Rev. Immunol. 2003, 22, 361–371. [Google Scholar] [CrossRef] [PubMed]
  78. He, J.; Yue, Y.; Dong, C.; Xiong, S. MiR-21 confers resistance against CVB3-induced myocarditis by inhibiting PDCD4-mediated apoptosis. Clin. Investig. Med. Med. Clin. Exp. 2013, 36, E103–E111. [Google Scholar]
  79. Du, X.; Wang, H.; Xu, F.; Huang, Y.; Liu, Z.; Liu, T. Enterovirus 71 induces apoptosis of SHSY5Y human neuroblastoma cells through stimulation of endogenous microRNA let-7b expression. Mol. Med. Rep. 2015, 12, 953–959. [Google Scholar] [PubMed]
  80. Cheng, Y.; Liu, X.; Zhang, S.; Lin, Y.; Yang, J.; Zhang, C. MicroRNA-21 protects against the H2O2-induced injury on cardiac myocytes via its target gene PDCD4. J. Mol. Cell Cardiol. 2009, 47, 5–14. [Google Scholar] [CrossRef] [PubMed]
  81. Lu, P.; Sun, H.; Zhang, L.; Hou, H.; Zhang, L.; Zhao, F.; Ge, C.; Yao, M.; Wang, T.; Li, J. Isocorydine targets the drug-resistant cellular side population through PDCD4-related apoptosis in hepatocellular carcinoma. Mol. Med. 2012, 18, 1136–1146. [Google Scholar] [CrossRef] [PubMed]
  82. Frankel, L.B.; Christoffersen, N.R.; Jacobsen, A.; Lindow, M.; Krogh, A.; Lund, A.H. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 2008, 283, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  83. Xiang, Z.; Li, L.; Lei, X.; Zhou, H.; Zhou, Z.; He, B.; Wang, J. Enterovirus 68 3C protease cleaves TRIF to attenuate antiviral responses mediated by Toll-like receptor 3. J. Virol. 2014, 88, 6650–6659. [Google Scholar] [CrossRef] [PubMed]
  84. Lei, X.; Han, N.; Xiao, X.; Jin, Q.; He, B.; Wang, J. Enterovirus 71 3C inhibits cytokine expression through cleavage of the TAK1/TAB1/TAB2/TAB3 complex. J. Virol. 2014, 88, 9830–9841. [Google Scholar] [CrossRef] [PubMed]
  85. Kuyumcu-Martinez, N.M.; van Eden, M.E.; Younan, P.; Lloyd, R.E. Cleavage of poly(A)-binding protein by poliovirus 3C protease inhibits host cell translation: A novel mechanism for host translation shutoff. Mol. Cell Biol. 2004, 24, 1779–1790. [Google Scholar] [CrossRef] [PubMed]
  86. Kuo, R.L.; Kung, S.H.; Hsu, Y.Y.; Liu, W.T. Infection with enterovirus 71 or expression of its 2A protease induces apoptotic cell death. J. Gen. Virol. 2002, 83, 1367–1376. [Google Scholar] [CrossRef] [PubMed]
  87. Graham, K.L.; Gustin, K.E.; Rivera, C.; Kuyumcu-Martinez, N.M.; Choe, S.S.; Lloyd, R.E.; Sarnow, P.; Utz, P.J. Proteolytic cleavage of the catalytic subunit of DNA-dependent protein kinase during poliovirus infection. J. Virol. 2004, 78, 6313–6321. [Google Scholar] [CrossRef] [PubMed]
  88. Feng, Q.; Langereis, M.A.; Lork, M.; Nguyen, M.; Hato, S.V.; Lanke, K.; Emdad, L.; Bhoopathi, P.; Fisher, P.B.; Lloyd, R.E.; et al. Enterovirus 2Apro targets MDA5 and MAVS in infected cells. J. Virol. 2014, 88, 3369–3378. [Google Scholar] [CrossRef] [PubMed]
  89. Barnabei, M.S.; Sjaastad, F.V.; Townsend, D.; Bedada, F.B.; Metzger, J.M. Severe dystrophic cardiomyopathy caused by the enteroviral protease 2A-mediated C-terminal dystrophin cleavage fragment. Sci. Transl. Med. 2015, 7. [Google Scholar] [CrossRef] [PubMed]
  90. Goldstaub, D.; Gradi, A.; Bercovitch, Z.; Grosmann, Z.; Nophar, Y.; Luria, S.; Sonenberg, N.; Kahana, C. Poliovirus 2A protease induces apoptotic cell death. Mol. Cell Biol. 2000, 20, 1271–1277. [Google Scholar] [CrossRef] [PubMed]
  91. Corsten, M.F.; Papageorgiou, A.; Verhesen, W.; Carai, P.; Lindow, M.; Obad, S.; Summer, G.; Coort, S.L.; Hazebroek, M.; van Leeuwen, R.; et al. MicroRNA profiling identifies microRNA-155 as an adverse mediator of cardiac injury and dysfunction during acute viral myocarditis. Circ. Res. 2012, 111, 415–425. [Google Scholar] [CrossRef] [PubMed]
  92. Lam, W.Y.; Cheung, A.C.; Tung, C.K.; Yeung, A.C.; Ngai, K.L.; Lui, V.W.; Chan, P.K.; Tsui, S.K. miR-466 is putative negative regulator of Coxsackie virus and Adenovirus Receptor. FEBS Lett. 2015, 589, 246–254. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, H.F.; Ding, Y.J.; Shen, Y.W.; Xue, A.M.; Xu, H.M.; Luo, C.L.; Li, B.X.; Liu, Y.L.; Zhao, Z.Q. MicroRNA-1 represses Cx43 expression in viral myocarditis. Mol. Cell Biochem. 2012, 362, 141–148. [Google Scholar] [CrossRef] [PubMed]
  94. Xu, H.F.; Ding, Y.J.; Zhang, Z.X.; Wang, Z.F.; Luo, C.L.; Li, B.X.; Shen, Y.W.; Tao, L.Y.; Zhao, Z.Q. MicroRNA21 regulation of the progression of viral myocarditis to dilated cardiomyopathy. Mol. Med. Rep. 2014, 10, 161–168. [Google Scholar] [PubMed]
  95. Xu, L.J.; Jiang, T.; Zhao, W.; Han, J.F.; Liu, J.; Deng, Y.Q.; Zhu, S.Y.; Li, Y.X.; Nian, Q.G.; Zhang, Y.; et al. Parallel mRNA and microRNA profiling of HEV71-infected human neuroblastoma cells reveal the up-regulation of miR-1246 in association with DLG3 repression. PLoS ONE 2014, 9, e95272. [Google Scholar] [CrossRef] [PubMed]
  96. Ye, X.; Hemida, M.G.; Qiu, Y.; Hanson, P.J.; Zhang, H.M.; Yang, D. MiR-126 promotes coxsackievirus replication by mediating cross-talk of ERK1/2 and Wnt/β-catenin signal pathways. Cell Mol. Life Sci. CMLS 2013, 70, 4631–444. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, L.; Chen, X.; Shi, Y.; Zhou, B.; Du, C.; Liu, Y.; Han, S.; Yin, J.; Peng, B.; He, X.; et al. miR-27a suppresses EV71 replication by directly targeting EGFR. Virus Genes 2014, 49, 373–382. [Google Scholar] [CrossRef] [PubMed]
  98. Wen, B.P.; Dai, H.J.; Yang, Y.H.; Zhuang, Y.; Sheng, R. MicroRNA-23b inhibits enterovirus 71 replication through downregulation of EV71 VPl protein. Intervirology 2013, 56, 195–200. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, L.; Qin, Y.; Tong, L.; Wu, S.; Wang, Q.; Jiao, Q.; Guo, Z.; Lin, L.; Wang, R.; Zhao, W.; et al. MiR-342-5p suppresses coxsackievirus B3 biosynthesis by targeting the 2C-coding region. Antivir. Res. 2012, 93, 270–279. [Google Scholar] [CrossRef] [PubMed]
  100. Yang, Z.; Tien, P. MiR373 and miR542-5p regulate the replication of enterovirus 71 in rhabdomyosarcoma cells. Chin. J. Biotechnol. 2014, 30, 943–953. (in Chinese). [Google Scholar]
  101. Piralla, A.; Mariani, B.; Stronati, M.; Marone, P.; Baldanti, F. Human enterovirus and parechovirus infections in newborns with sepsis-like illness and neurological disorders. Early Hum. Dev. 2014, 90 (Suppl. 1), S75–S77. [Google Scholar] [CrossRef]
  102. Britton, P.N.; Khandaker, G.; Booy, R.; Jones, C.A. The causes and consequences of childhood encephalitis in Asia. Infect. Disord. Drug Targets 2014, 14. [Google Scholar] [CrossRef]
  103. Tung, W.H.; Hsieh, H.L.; Lee, I.T.; Yang, C.M. Enterovirus 71 induces integrin β1/EGFR-Rac1-dependent oxidative stress in SK-N-SH cells: Role of HO-1/CO in viral replication. J. Cell Physiol. 2011, 226, 3316–3329. [Google Scholar] [CrossRef] [PubMed]
  104. Tung, W.H.; Hsieh, H.L.; Lee, I.T.; Yang, C.M. Enterovirus 71 modulates a COX-2/PGE2/cAMP-dependent viral replication in human neuroblastoma cells: Role of the c-Src/EGFR/p42/p44 MAPK/CREB signaling pathway. J. Cell Biochem. 2011, 112, 559–570. [Google Scholar] [CrossRef] [PubMed]
  105. Tung, W.H.; Hsieh, H.L.; Yang, C.M. Enterovirus 71 induces COX-2 expression via MAPKs, NF-κB, and AP-1 in SK-N-SH cells: Role of PGE(2) in viral replication. Cell Signal. 2010, 22, 234–246. [Google Scholar] [CrossRef] [PubMed]
  106. Belsham, G.J.; Sonenberg, N. Picornavirus RNA translation: Roles for cellular proteins. Trends Microbiol. 2000, 8, 330–335. [Google Scholar] [CrossRef]
  107. Schneider, R.J.; Mohr, I. Translation initiation and viral tricks. Trends Biochem. Sci. 2003, 28, 130–136. [Google Scholar] [CrossRef]
  108. Hemida, M.G.; Ye, X.; Zhang, H.M.; Hanson, P.J.; Liu, Z.; McManus, B.M.; Yang, D. MicroRNA-203 enhances coxsackievirus B3 replication through targeting zinc finger protein-148. Cell Mol. Life Sci. CMLS 2013, 70, 277–291. [Google Scholar] [CrossRef] [PubMed]
  109. Abraham, T.M.; Sarnow, P. RNA virus harnesses microRNAs to seize host translation control. Cell Host Microbe 2011, 9, 5–7. [Google Scholar] [CrossRef] [PubMed]
  110. Orom, U.A.; Nielsen, F.C.; Lund, A.H. MicroRNA-10a binds the 5′MUTR of ribosomal protein mRNAs and enhances their translation. Mol. Cell 2008, 30, 460–471. [Google Scholar] [CrossRef] [PubMed]
  111. Roberts, A.P.; Lewis, A.P.; Jopling, C.L. miR-122 activates hepatitis C virus translation by a specialized mechanism requiring particular RNA components. Nucleic Acids Res. 2011, 39, 7716–7729. [Google Scholar] [CrossRef] [PubMed]
  112. Li, R.; Liu, L.; Mo, Z.; Wang, X.; Xia, J.; Liang, Z.; Zhang, Y.; Li, Y.; Mao, Q.; Wang, J.; et al. An inactivated enterovirus 71 vaccine in healthy children. N. Engl. J. Med. 2014, 370, 829–837. [Google Scholar] [CrossRef] [PubMed]
  113. Zhu, F.; Xu, W.; Xia, J.; Liang, Z.; Liu, Y.; Zhang, X.; Tan, X.; Wang, L.; Mao, Q.; Wu, J.; et al. Efficacy, safety, and immunogenicity of an enterovirus 71 vaccine in China. N. Engl. J. Med. 2014, 370, 818–828. [Google Scholar] [CrossRef] [PubMed]
  114. Farooqi, A.A.; Fayyaz, S.; Shatynska-Mytsyk, I.; Javed, Z.; Jabeen, S.; Yaylim, I.; Gasparri, M.L.; Panici, P.B. Is miR-34a a well equipped swordsman to conquer temple of molecular oncology? Chem. Biol. Drug Des. 2015, 86. [Google Scholar] [CrossRef] [PubMed]
  115. Janssen, H.L.; Reesink, H.W.; Lawitz, E.J.; Zeuzem, S.; Rodriguez-Torres, M.; Patel, K.; van der Meer, A.J.; Patick, A.K.; Chen, A.; Zhou, Y.; et al. Treatment of HCV infection by targeting microRNA. N. Engl. J. Med. 2013, 368, 1685–1694. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Ho, B.-C.; Yang, P.-C.; Yu, S.-L. MicroRNA and Pathogenesis of Enterovirus Infection. Viruses 2016, 8, 11. https://doi.org/10.3390/v8010011

AMA Style

Ho B-C, Yang P-C, Yu S-L. MicroRNA and Pathogenesis of Enterovirus Infection. Viruses. 2016; 8(1):11. https://doi.org/10.3390/v8010011

Chicago/Turabian Style

Ho, Bing-Ching, Pan-Chyr Yang, and Sung-Liang Yu. 2016. "MicroRNA and Pathogenesis of Enterovirus Infection" Viruses 8, no. 1: 11. https://doi.org/10.3390/v8010011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop