You are viewing the site in preview mode

Skip to main content

Regulatory role of microRNAs in virus-mediated inflammation

Abstract

Viral infections in humans often cause excessive inflammation. In some viral infections, inflammation can be serious and even fatal, while in other infections it can promote viral clearance. Viruses can escape from the host immune system via regulating inflammatory pathways, thus worsening the illness. MicroRNAs (miRNAs) are tiny non-coding RNA molecules expressed within diverse tissues as well as cells and are engaged in different normal pathological and physiological pathways. Emerging proof suggests that miRNAs can impact innate and adaptive immunity, inflammatory responses, cell invasion, and the progression of viral infections. We discuss some intriguing new findings in the current work, focusing on the impacts of different miRNAs on host inflammatory responses and virus-mediated inflammation. A better understanding of dysregulated miRNAs in viral infections could improve the identification, prevention, and treatment of several serious diseases.

Introduction

Viruses have evolved methods to evade the host’s immune system by manipulating specific biological mechanisms within the host cells [1]. One of the prominent tactics that viruses utilize to impair the host’s innate immune response is through the use of small regulatory RNAs [2]. Small RNAs, particularly microRNAs (miRNAs), play a vital role in regulating host gene expression programs related to antiviral innate immunity genes [3]. MiRNAs have been recognized for their ability to regulate protein-coding genes by means of posttranscriptional repression, either by suppressing translation or promoting the degradation of mRNA [4]. MiRNAs have the ability to regulate the expression of the majority of protein-coding genes, resulting in a significant impact on regulatory networks [2]. The significant role of miRNAs in coordinating innate and adaptive immune respo1nses and inflammatory processes within various cell and tissue types has been well established [5].

Interestingly, viruses have been discovered to disrupt the regulatory role of host miRNAs while also encoding their own miRNAs to evade the host’s immune response. Furthermore, viruses are capable of manipulating the host’s transcriptional machinery for their own benefit, allowing them to produce viral miRNAs [6]. Despite the clearly defined role of viral miRNAs in evading the host immune response, the mechanisms by which virus-derived miRNAs function and elude the immune system’s defenses remain incompletely understood [6]. In this review, we will explore how viruses can control inflammatory responses by altering host miRNAs and using viral miRNAs. Our aim is to provide insights on improving prevention and treatment methods for a range of viral infections.

Role of inflammation in viral pathogenesis

Many innate immune system receptors are expressed either within or on the surface of the host cell. These receptors can recognize invasive viruses and their intermediaries employed in their reproduction. The best-studied types of innate immune receptors linked to viral infections are RIG-I (retinoic acid-inducible gene I), TLRs (Toll-like receptors), as well as NLRs (NOD-like receptors). Viral infections often activate TLRs which are capable of identifying intermediate double-stranded RNA and viral nucleic acids [7, 8]. In fact, the cytoplasmic RIG-I-like receptors are capable of recognizing viral genomic RNA or RNA encoded by genomic DNA, while the NLRs is able to recognize viral DNA genomes [9]. When many of these receptors are stimulated, they produce interferons (IFNs), pro-inflammatory cytokines (PICs), and signals that activates inflammation along with adaptive immunity. The network of intrinsic immune reactions set off by the initial viral entrance into a cell may affect how an infection develops. Many viruses stimulate innate immune cells like natural killer (NK) cells, macrophages, as well as dendritic cells, to generate anti-inflammatory mediators like transforming growth factor β (TGFβ) as well as interleukin-10 (IL-10). For instance, monocytes isolated from humans infected by human immunodeficiency virus (HIV), hepatitis B virus (HBV) or hepatitis C virus HCV were found to produce TGF-β and Il-10. For instance, dendritic cells (DCs), isolated from lymphocytic choriomeningitis virus (LCMV)- infected mice produced high amounts of Il-10 [8, 10]. The interaction between tnfplasmacytoid DCs (pDCs) and the virus present in the lungs was in favor of the anti-inflammatory effect [11]. This conclusion was supported because the elimination of pDCs prior to infection promoted inflammation inside the lungs after the respiratory syncytial virus (RSV) infection. When viruses can interfere with one or more of the host innate defense mechanisms, a more severe viral infection is more likely to occur [8].

Moreover, the adaptive immune system effector cells could also cause damage in the host tissues after their generation in reaction to viral infection. For example, T cells are capable of killing virus-infected cells and releasing harmful cytokines such as tumor necrosis factor alpha (TNF-α). The major cause of liver injury in some noncytopathic viral infections like HCV and HBV, is due to an attempt by host CD8 effector T cells to eradicate infected cells [8]. Different subtypes of CD4 T cells coordinate inflammatory responses to combat persistent viruses inside infected cells, and thereby can cause tissue damage. Infection with HIV, HCV, as well as influenza viruses might invoke TH17 cells to be a part of inflammatory response, although T helper 1 cells (TH1 cells) are more commonly involved [12]. In responses driven by TH17 cells, neutrophils are attracted to the infected area and secrete antimicrobial substances that can also cause tissue damage. However, in extreme cases of pulmonary RSV infection, TH2 cell responses may also occur [13]. Nevertheless, generally TH2 cells rarely participate in inflammatory responses to a viral infection.

An inflammatory reaction is started when an antibody links to one infected cell and activates the complement system. Alternatively, toxicity can also occur when IgG molecules bind to Fc receptors expressed on inflammatory and phagocytic cells, causing the release of inflammatory mediators [8]. If the viruses are still present and IgG continues to be generated, immune complexes can be deposited in the host tissue, leading to chronic conditions like nephritis, polyarteritis, or arthritis. The first diseases in which immune complex lesions were identified were LCMV infection, chronic HCV and HBV infection, and HIV infection-related idiotypic IgA nephropathy. The pulmonary lesions observed in some children with RSV infection may be explained in part by a type I hypersensitivity reaction, because viruses like RSV produce antigens which can cause an IgE reaction [8, 14].

The blood-brain barrier (BBB) is a special semipermeable barrier which allows passage of a limited number of circulating host cells, respiratory gases, and specific metabolites, and physically isolates the central nervous system (CNS) from other body parts. The BBB can protect neuronal cells from systemic inflammation. Microglia resemble macrophage-like immune cells, and together with oligodendrocytes and astrocytes, are examples of glial cells, which protect and repair damaged neural tissue. They provide critical structural support for the neurons. The ability of microglial cells to affect immune responses is employed to control pathological inflammation [15, 16]. A number of viruses, including coronaviruses, picornaviruses, flaviviridae, and togaviridae can attack the central nervous system when it is weakened [16, 17]. Some viruses are confined within the surrounding tissue in the absence of an obvious entry point, before entering peripheral neurons or BBB micro vessels [16, 17]. Several viruses, such as West Nile virus (WNV), Japanese encephalitis virus (JEV), Zika virus (ZIKAV), as well as poliovirus are more likely to cross BBB and damage brain and spinal cord. Both WNV and JEV can lead to BBB disruption within the CNS. It has been suggested that the BBB is disrupted by PICs released via the infected endothelial cells, and not by the viruses themselves [8, 17]. Tissue damage is the result of either an excessive inflammatory reaction driven via PICs or CD8 cytotoxic T lymphocytes (CTLs), or by direct neuronal infections that activate the apoptosis in the neurons. Infected neurons release chemokines in order to attract white blood cells such as monocytes and lymphocytes. However, the CNS is characterized by a very fine line that must be drawn between “good” and “bad” immune responses. Viral eradication and survival are strongly influenced by the migration of CTLs expressing receptors for chemokines, such as CXCL9–11, CXCL2-4, CCL5, and their timing relative to the onset of infection. Through a Fas-Fas ligand (FasL)-mediated or a granzyme-perforin dependent mechanism, CTLs can kill host cells in order to carry out their antiviral function. Moreover, some PICs secreted during a viral infection could also result in the direct death of neuronal cells [16, 17].

Exogenous regulation of these inflammatory responses by disrupting or supplementing key regulators has been proposed as an anti-viral therapeutic measure, since a poorly controlled inflammatory response can have negative effects [16]. The advantages and disadvantages of various inflammation-related strategies are still under investigation. These approaches have been successfully used to treat some viral diseases, because they may protect the affected subjects without any direct effect on the virus life cycle. In addition, if inflammation-related approaches can be shown to be effective, they could reduce the morbidity and mortality caused by some viral infections, which have proven particularly challenging to treat, using conventional antiviral drugs or prophylactic vaccines.

MicroRNAs and inflammation

Biogenesis of microRNAs

Short endogenous non-coding RNAs known as microRNAs (miRNAs) control the translation along with stability of mRNAs. RNA polymerase II converts the miRNA gene into a primary miRNA (pri-miRNA) having a stem-loop structure, which is the initial stage of standard miRNA production. The Drosha microprocessor complex as well as DGCR8 endonuclease subsequently cleave the pri-miRNA for generating hairpin precursor miRNAs (pre-miRNA) of almost 70 nt in length [18]. Pre-miRNAs are transferred to the cytoplasm via exportin-5, where they’re divided into miRNA duplexes by TRBP complexes (two strands) and Dicer [19, 20]. Consequently, the mature miRNA duplex is added to the RNA-induced silencing complex (RISC). This duplex must be further processed by cleavage or thermodynamic instability [21, 22]. The key element Argonaute 2 in the RISC complex guides mature miRNAs to their destination after strand selection, and then shortens the 3’-poly-A tail of each target complementary mRNA, or else cleaves the mRNA to destabilize it. The target mRNA-encoded protein experiences translational inhibition as a result of this whole process [21, 23].

Role of microRNAs in the regulation of inflammatory responses

It was indicated that during inflammatory reactions, a number of miRNAs regulate gene expression at transcriptional level. At late stages of inflammatory transcriptional programs, numerous miRNAs can be activated. MiRNAs are governed similarly to protein-coding genes, according to recent research, but the timing of their synthesis and their type of activity may provide these short RNA molecules with special regulatory properties [24, 25]. Several proteins that are produced during inflammatory responses, are able to control miRNA processing. These include TGF-β-activated SMAD proteins and p68 RNA helicase, which is a part of the Drosha endoribonuclease complex. When ADAR (adenosine deaminase acting on RNA) is triggered via inflammation, double-stranded miRNA precursors can acquire mutations that alter the target selectivity of the miRNAs [26]. Many miRNA transcripts can be processed by tumor-suppressing protein p53 that is increased throughout specific inflammatory responses. Moreover, the expression of Dicer as well as other biogenesis factors is suppressed by interferon-γ [27]. Each miRNA can undergo highly specific posttranscriptional processing. For instance, the processing of miRNA let-7 was controlled by the Lin28 RNA-binding protein [28]. It is unknown whether any other regulatory elements related to a single or to a handful of miRNAs are present throughout inflammation. Moreover, whether miRNA biogenesis regulates immune responses through any other pathways remains to be elucidated. MiRNAs could regulate inflammation mainly via changing expression of particular mRNAs within activated immune cells or bystander cells [29]. There are proofs that inflammatory responses can regulate miRNA biogenesis via changing the transcription, processing, or stability of precursor or mature miRNA transcripts [30]. MiRNAs are involved in both favorable and unfavorable regulatory mechanisms that control the initiation, propagation, and termination of inflammation. A positive feedback loop triggers a series of molecular mechanisms, which successfully repairs tissue damage and prevents the invasion of microbial pathogens. A negative feedback loop, which occurs only when inflammation is at its highest, should prevent potentially harmful results and maintain tissue homeostasis [31].

For example, recent research has shown that miR-21 is crucial for reducing inflammation [32]. It is likely that miR-21 has an anti-inflammatory effect since its overexpression within macrophages decreased secretion of IL-6 whilst raising the IL-10 secretion [24, 31]. In one study, miR-21 has been discovered to have a substantial part in regulating the coordination of Th1 and Th2 responses. Treatment of DCs that were non-responsive to lipopolysaccharide (LPS) with miR-21, increased the production of IL-12. Similarly, the generation of IFN-γ has been increased via ovalbumin, while IL-4 production was decreased. This occurred when miR-21-deficient CD4 T cells were stimulated [33]. Considering the preventing effect of miR-21 on lipid buildup and LPS-induced inflammatory responses in macrophages, miR-21 could serve as a possible therapeutic strategy for the treatment of atherosclerosis [34]. Accordingly, when atherogenesis occurs, macrophages lacking miR-21, could promote endothelial inflammation [31]. In fact, overexpression of miR-21 is found to suppress the classical pro-inflammatory activation (M1) phenotype, while it enhanced the alternative anti-inflammatory activation (M2) phenotype of macrophage [35].

The production of miR-146a and miR-146b may function as a negative feedback loop to stop excessive inflammation, whilst reacting to pro-inflammatory stimuli [36, 37]. Several inflammatory disorders like lupus, psoriasis, rheumatoid arthritis, osteoarthritis, along with viral infection, were connected to low expression of these miRNAs [36, 37]. In fact, miR-146 transcription must be induced via nuclear factor κB (NF-kB), while JNK-1/2 as well as MEK-1/2 are then involved in post-transcriptional processing to produce mature miR-146 [38]. MiR-146a and miR-146b influence TLRs and their downstream effectors to control the inflammatory procedure. MiR-146a negatively controls IFN responses as well as adaptive immunity via selectively targeting adaptor protein (AP)-1 transcription factor, immune cell activation, IL-2 expression, and cytokine production [39, 40]. Moreover, adenosine deaminase 2, which has been reported to control diabetic retinal inflammation, was also suppressed by miR-146b [41].

The MIRHG155 gene, originally known as BIC (B-cell integrating cluster) encodes a pri-miRNA that is converted into miR-155. Several signaling pathways, including NF-kB, SMAD4, ISRE (interferon stimulated response element), IRFs (interferon-regulatory factors) as well as AP -1 are established to have binding sites in BIC gene, and it can regulate miR-155 expression whilst reacting to various cues [42]. Numerous works have indicated that miR-155 expression has been abnormally increased within various activated immune cells, thus demonstrating the critical part that miR-155 plays in immune response [43,44,45]. Several inflammatory stimuli, including TLR ligands, alarmins (e.g. IL-1), damage-associated molecular patterns (DAMPs), interferons, and pathogen-associated molecular patterns (PAMPs) are particularly sensitive to miR-155 in different cells, especially macrophage/monocytes [46]. Indeed, various signaling pathways can control miR-155 expression [46]. The anti-inflammatory protein IRF3 is found to regulate miR-155-3p along with miR-155-5p expression within astrocytes [47]. Moreover, it has been demonstrated that through inhibiting miR-155 expression, resolvin D1 can reduce inflammation during experimental corneal immunopathology [48]. In addition, in mice lacking Ets2, LPS-induced miR-155 expression was shown to be lower. Moreover, IL-10 suppressed the Ets2 gene to control miR-155 expression [49]. It is confirmed that the NF-κB-miR-155 along with NF-κB-miR-146a axis can collaborate in inflammatory responses to control the severity and length of inflammation. Thus, miR-146a-deficient mice were more prone to cancer, autoimmune illnesses, as well as inflammatory disorders. Interestingly, miR-146a-deficient cells expressed more miR-155, which can result in reduced pro-inflammatory phenotype [46]. Furthermore, NF-κB activity is controlled via a two-step system that combines the activity of miR-146a along with miR-155. NF-κB signaling is autoregulated during inflammation. In the initial twelve-hour period of an inflammatory reaction, miR-155 quickly increases NF-κB. In addition, miR-155 targets SHIP1 and, through PI3K/Akt-dependent activation of the IKK signaling complex, creates a positive feedback loop for signal amplification. Furthermore, miR-146a inhibited IRAK1 and TNF receptor associated factor 6 (TRAF6), result in a negative feedback loop that reduced NF- κB activity in later stages of inflammation [50].

Recently, miR-149 was reported to control innate immune defense mechanisms. When miR-149 is increased in macrophages, it can decrease the MyD88 protein expression and the generation of inflammatory mediators, TNF-α, IL-6 as well as NF-kB in reaction to LPS stimulation or actual infection. Additionally, miR-149 inhibited STAT3-mediated signaling, which decreased inflammatory responses in the liver [51, 52]. Moreover, it has been established that tumor necrosis factor (TNF-α) downregulates miR-149 and subsequently promotes the expression of IL-6, Inducible nitric oxide synthase (iNOS), and matrix metalloproteinase − 9 (MMP-9). In that context, it is shown that the transfection with a miR-149 mimic, can block this effect [53]. Several studies have connected osteoarthritis, a joint condition typified by uncontrolled inflammatory responses, to the downregulation of miR-149 [54]. These results are relevant to TNF-α-associated immune disorders, and may provide useful suggestions for possible future therapeutic approaches.

Virus-mediated regulation of inflammatory pathways by host-derived miRNAs

Viral infections can alter host miRNA expression, which then affects host immune responses via transcriptional and translational mechanisms. Changes in miRNA expression can be a result of cell differentiation, cell cycle alteration, apoptosis, host immune signals, and antiviral defenses all of which can be affected by viruses. Severe viral infections often cause widespread inflammation and tissue damage. However, these inflammatory responses are required to rapidly control the initial infection, as well as to start the adaptive immune responses, which will finally stop viral replication. Various mediators or cell types involved in triggering the immune response, can also cause tissue damage and cell death at high concentrations or numbers. In addition, instead of providing protection, some mediators and signaling pathways have been linked to increased death and injuries in tissues. In order to lower the viral load and reduce disease severity, combination therapy with both antiviral and anti-inflammatory drugs is essential. In addition to being effective after the onset of symptoms, the optimized anti-inflammatory drugs can work in tandem with established antiviral drugs, thus decreasing the probability of subsequent bacterial infection, while not interfering with viral clearance mechanisms [55]. Therefore, it is critical to select the most effective therapeutic approaches and/or targets to reduce inflammation during viral infection. Because inflammation is a multifactorial phenomenon, the ability of miRNAs in regulating relevant genes at transcriptional as well as posttranscriptional levels could influence one or more pathways in the growth of viral illnesses. Each miRNA’s ability to act on multiple genes opens the possibility of genetic interference with multiple disease mechanisms [56].

Influenza virus

Influenza virus infections are in charge for excessive mortality and morbidity globally. The yearly influenza epidemics can cause about 500,000 deaths and almost 5 million cases globally in a bad year [57]. The viruses that cause influenza B Victoria, influenza B Yamagata, influenza A H1N1 (swine flu), and influenza A H3N2 (H3N2v), which are mostly limited to the respiratory system, are the most common reasons behind influenza infections in humans [58]. Numerous lines of evidence point to specific miRNAs acting as important regulators in development of influenza infections. For instance, Zhang et al. discovered that H1N1 infection increased miR-29c expression within A549 cells, which raised inflammatory as well as antiviral responses [59]. In addition, Podsiad et al. discovered that miR-155 could inhibit the IL-23/IL-17 signaling pathway, which leads to reduced bacterial clearance and increased mortality due to post-influenza pneumonia [60]. On the other hand, Guo et al. reported that H1N1 infection could cause significant cell damage by increasing cytokine production, decreasing viability, and inducing apoptosis. They found that inoculation with H1N1 virus reduced miR-4485 expression within A549 cells, while miR-4485 overexpression inhibited H1N1-induced cell damage [61]. Traber et al. showed that STAT3 could control the generation of chemokine CXCL5 (C-X-C motif ligand 5) that is necessary for neutrophil and macrophage recruitment to the lungs in pneumonia [62]. Small molecule inhibition of STAT3 reduced LPS-induced infiltration of inflammatory cells and macrophages within bronchoalveolar lavage fluid [63]. Guo et al. demonstrated that STAT3 has been a miR-4485 direct target gene. Downregulation of miR-4485 and cell damage induced by H1N1 virus were reversed by STAT3 silencing. They also found that the knockdown of STAT3 significantly reduced the activity of PI3K/mTOR/AKT pathway, suggesting that miR-4485 regulates the activity of STAT3-mediated pathways. Their study showed that STAT3 has been upregulated whilst miR-4485 has been downregulated, and that PI3K/AKT/mTOR pathway’s activation could increase damage induced by H1N1 infection in A549 cells [61].

With 3.23 million fatalities from chronic obstructive pulmonary disease (COPD) in 2019, it is the third most common reason behind mortality in the world. COPD is defined by emphysema, increasing inflammation in the airways, as well as diminished lung function [64]. IAV infection can worsen the symptoms in COPD patients. Hsu et al. reported that when mice having experimental COPD have been infected with IAV, they showed increased inflammatory cytokines but lower antiviral responses [65]. The expression levels of miR-125a and miR-125b were increased within IAV-infected cells. The group reported that levels of protein A20 were lower in IAV infection as well as in COPD, which led to increased NF-kB-mediated inflammation, because A20 is the inhibitor of NF-kB. Treatment with a selective antagonist of miR-125a/b lowered NF-kB activation, while increasing the release of I/III IFNs as well as reducing infection. MiR-125a and miR-125b both inhibit the induction of type I/III IFNs along with NF-kB, by directly targeting MAVS (mitochondrial antiviral signaling) and A20. They concluded that miR-125-mediated signaling lowered the expressions of MAVS and A20, thereby inducing excessive inflammation, and increasing the susceptibility of COPD patients to IAV infection (Fig. 1) [66]. Therefore, miRNAs could be used in the therapy of IAV infection, by improving antiviral immunity and protecting against excessive inflammatory reactions (Table 1).

Fig. 1
figure 1

Regulation of inflammation by miR-125a/b throughout IAV infection. miR-125a/b increases inflammation and suppresses antiviral responses in COPD by inhibiting A20 and MAVS [66]

Table 1 Virus-mediated regulation of inflammation by host and viral microRNAs

SARS-CoV-2 virus

The SARS-CoV-2 coronavirus infection is to blame for 2019 coronavirus epidemic (COVID-19), and has resulted in over 623 million new cases along with 6.55 million mortalities globally until February 2021. Severe COVID-19 infections have a significant mortality rate and can result in lethal lung hypoxia and a cytokine storm. Apoptosis and necrosis can damage the alveolar epithelial and vascular endothelial cells causing tissue damage due to excessive cytokine production [113, 114].

The SARS-CoV envelope protein (E) is regarded as a virulence factor that supports pulmonary inflammation caused by SARS-CoV through several mechanisms [115, 116]. NF-kB-dependent proinflammatory responses can be triggered by the E protein [117]. The p38 mitogen-activated protein kinase (MAPK) signaling pathway is activated when the PDZ-binding motif of E protein interacts specifically with the PDZ domains of syntenin [118]. In addition, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes and the secretion of IL-1β are triggered by Ca (2+) transport into infected cells. Acute respiratory distress syndrome (ARDS) etiology, includes this mechanism as a critical element, an extreme pulmonary inflammation suffered by SARS and COVID-19 patients who have severe disease [119, 120].

Recently, Morales et al. aimed to elucidate the effect of host’s miRNAs on protein E-mediated virulence. In mice infected via pathogenic SARS-CoV wild type (WT) or even attenuated mutant deficient in the E gene (SARS-CoV-ΔE), miR-223 might function as a regulator of lung inflammation [121]. In virulent SARS-CoV-WT infection, miR-223 levels were significantly higher than in an infection with attenuated SARS-CoV-ΔE. It was found that miR-223 could particularly target inflammasomes and the cystic fibrosis transmembrane conductance regulator (CFTR). Then, in viral lung infections, miRNA-223 might help to regulate excessive inflammation by suppressing pro-inflammatory cytokines as well as the NLRP3 inflammasomes in vivo. The CFTR transporter, which reduces edema, was noticeably increased in the lung of mice infected with virulent SARS-CoV-WT as a result of the suppression of miRNA-223-3p. At the histological level, miR-223-3p inhibition, decreased pulmonary edema, indicating that miRNA-223-3p could regulate SARS-CoV-induced inflammation [121]. Since miRNA-223 can target several host mRNAs involved in lung inflammation during SARS-CoV infection, it could be a viable treatment target in SARS-CoV infection.

There is evidence that the miRNAs Let-7c and Let-7a can inhibit IL-6 expression that is a typical pro-inflammatory cytokine induced via SARS-Cov-2 [122]. It is possible that up-regulation of Let-7 could reduce the SARS-CoV-2-induced cytokine storm by down-regulating inflammatory factors other than IL-6. To test this hypothesis, Xie et al. studied Let-7 and C1632 for their ability to reduce inflammation caused by SARS-CoV-2 [123]. They found that Let-7 which is a universally expressed miRNA within human cells, could target the S and M viral proteins thus preventing SARS-CoV-2 replication. Several inflammatory factors like IL-6, IL-8, IL-1, C-C motif chemokine ligand 2 (CCL2), granulocyte macrophage-colony stimulating factor )GM-CSF(, vascular endothelial growth factor α (VEGFα) and TNF-α, could all be simultaneously suppressed by Let-7. More importantly, Let-7 expression could be increased by the Let-7 stimulating factor C1632, which lowers viral replication as well as inflammatory cytokine secretion [123]. The effectiveness of C1632 in limiting SARS-CoV-2 replication along with lowering virus-induced inflammation via upregulating Let-7 might be therapeutically useful [123].

Respiratory syncytial virus

The RSV is a lipid-enclosed virus which is a member of genus Pneumoviridae and family Paramyxoviridae. The 15.2 kb single-stranded negative RNA genome of RSV has 10 genes that code for 11 different viral proteins. These include small hydrophobic protein (SH), an envelope protein, as well as two major surface proteins, fusion protein (F) along with glycoprotein (G). Serious RSV infections are characterized by substantial airway inflammation, and release and synthesis of multiple NF-kB-controlled chemokines and cytokines [124, 125]. TLRs, along with cytokine receptors like the IL-1 receptor, TNF receptor (TNFR), as well as pattern recognition receptors are examples of receptors that can be stimulated in activating NF-kB [126]. Overexpression of NF-kB-mediated inflammatory genes is most likely caused by persistent NF-kB activity after RSV infection [127, 128]. TLR4 has been indicated to have a significant part in controlling innate immune reaction to RSV infection [129]. RSV F protein can activate pattern recognition receptors CD14, TLR-4, and MD-2, which can activate the transcription factor NF-kB and raise pro-inflammatory cytokines [130]. However, it is still not completely known how TLR4 is regulated after RSV infection. According to a recent publication, miR-140-5p may function via Myeloid differentiation factor 88 (MyD88)/NF-kB pathway to target TLR4 as well as decrease inflammatory cytokines in acute respiratory distress syndrome [131]. Zhang et al. discovered that the amounts of miR-140-5p have been dramatically lower in peripheral blood and nasal mucosa specimens of RSV-infected individuals [74]. Moreover, exogenous expression of miR-140-5p has been discovered to substantially lower the pro-inflammatory cytokines, IL-1, IL-6, and IL-8 along with TNF-α. Moreover, the generation of TNF-α, IL-1, and IL-6, along with IL-8 has been substantially raised when miR-140-5p was inhibited. These outcomes recommend that the immune response to RSV is impaired by lower amounts of miR-140-5p within RSV-infected cells, probably because miR-140-5p targets TLR4 [74].

MiR-146a was reported to be a possible biomarker of sepsis as it was involved in the control of inflammation [132]. Since miR-146a regulates the inflammatory response in both acute lung injury and asthma, it has been considered as a reliable predictor of acute lung damage [133]. However, it is still unclear if miR-146a could contribute to lung damage caused by RSV infection. Stress-induced cytokine secretion has been substantially decreased by both miR-146a expression as well as TRAF-6 inhibition compared to the control group, suggesting that miR-146a has an influence on TRAF-6 in airway inflammation. Huang et al. examined the mechanisms underlying the impact of miR-146a upon RSV transmission [134]. In young rats, as well as in HEp-2 and A549 cells, miR-146a mimics were administered before exposure to RSV. RSV infection substantially decreased miR-146a levels in rat lung tissue, HEp-2, and A549 cells. A more rapid growth, overexpression of TRAF-6 and inflammatory cytokines, and activated JNK signaling pathway were observed in RSV-infected cells. In rats, the lungs exhibited inflammatory infiltrates and worse pathology scores. The group confirmed that overexpression of miR-146a decreased IL-6 and TNF-α production. Furthermore, downregulation of TRAF-6 and suppressed the JNK/ERK/MAPK/NF-B signaling cascade resulted in inhibited lung damage caused by RSV infection. Similar results were obtained with HEp-2 and A549 cells. These results suggested that miR-146a could target TRAF-6 as well as regulating inflammatory pathways [134] (Fig. 2).

Fig. 2
figure 2

MiR-146a decreases inflammation-induced acute lung injury via regulating JNK/ERKMAPK signaling pathway and targeting TRAF-6 [134]

Viral hepatitis

Both HBV and HCV are two primary hepatotropic viruses responsible for chronic liver infections [135, 136]. Upon viral infection, the inflammasomes are activated in the liver cells, thereby chronic viral hepatitis frequently results in excessive liver inflammation. During this process, pro-inflammatory cytokines and type I interferons are processed and released by the inflammasomes, which function as a signaling hub [137]. Various cell types, including microglia, dendritic cells, epithelial cells, monocytes, macrophages, and fibroblasts are able to release CCL2 (referred to as monocyte chemoattractant protein 1) an 11 kDa cytokine of CC chemokine family [138, 139]. Individuals having chronic liver illnesses like alcoholic hepatitis as well as HCV infection, show increased liver inflammation along with elevated levels of CCL2 [140, 141]. It has been demonstrated that reduction of IL-6 and IL-6R complex, along with STAT3 in mice results in downregulation of CCL2 [142]. Cell proliferation and differentiation are tightly regulated via homodimeric bZIP transcription factor CCAAT/CEBPα [143]. A subpopulation of HCC patients shows higher CEBPα levels that can influence the expression of many genes like hepatocyte-specific miR-122, which has been implicated in hepatocarcinogenesis [144]. According to Sarma et al., patients with HCV-induced liver damage show increased miR-107 levels as well as lower miR-449a after infection. These changes affected parts of the IL-6R complex and regulated CCL2 expression [83]. Furthermore, they demonstrated how STAT3, CEBPα, and PU.1 could interact with one another to bind to the promoter and enhance CCL2 expression. Measurements of JAK1 and IL-6R expression in samples from HCV patients indicated that these markers were higher when expressions of miR-107 and miR-449a has been disrupted. This was linked to higher levels of PU.1 and STAT3, but not CEBPα. In vitro research with human hepatocytes, indicated that miR-449a along with miR-107 could target IL-6R and JAK1 expressions respectively, decrease IL-6 signaling, and modify STAT3 activation. Together, their findings unveiled a new gene regulatory system explaining how HCV-induced alterations within miRNAs (miR-449a and miR-107) could control CCL2 expression via triggering IL-6 signaling cascade, thus causing excessive inflammation and fibrosis [83].

Hepatocellular carcinoma as well as liver cirrhosis are often caused by HBV infection [145]. The HBV X (HBx) protein is a multipurpose protein which binds to host regulatory elements, causing inflammatory responses, and can eventually result in liver cancer and cirrhosis [146]. In one study, Wu et al. investigated how microRNAs could contribute to the inflammation caused by HBX [80]. Microarray technology was used to compare miR-203a expression in liver samples from individuals with and without HBV. They used HepG2 cells in vitro to overexpress HBx protein using a plasmid that encodes HBx. The obtained result indicated that the overexpression of HBx promotes miR-203a activity. Forced overexpression of miR-203a within HepG2 cells resulted in increased proliferation and suppressed apoptosis. In addition, it was found that overexpression of miR-203a could shift cells from the G0/G1 stage to the G2/M stage. Upregulation of cytokines, such as IL-8 and IL-6 in HepG2 cells was noticeable following miR-203a overexpression, while TGF-β and IFN levels were downregulated. By using luciferase assays, protein mass spectrometry analysis, and real-time PCR, the target gene of miR-203a has been found to be Rap1a (member of RAS oncogene family 1a). Further investigation revealed that the PI3K/ERK/p38/NF-kB signaling pathway controlled these changes. Such outcomes recommend that HBV infection might increase the miR-203a expression, thus decreasing Rap1a expression and affecting the PI3K/ERK/p38/NF-kB signaling pathway, ultimately triggering inflammation associated with hepatitis [80]. Interestingly, the crucial inflammatory cytokine IL-8 was recently discovered to have a part in the development of HBV-induced chronic hepatitis B. (CHB). Earlier studies had demonstrated that HBV-encoded HBX regulatory protein could transactivate IL-8 promoter, and therefore increase its secretion [147]. It has been demonstrated that IL-8 expression in asthmatic bronchial epithelium is modulated by a variety of miRNAs, including miR-18a, miR-128, miR-27a, and miR-155 [148]. According to Dalbeth et al. miR-146a dramatically decreased IL-8 gene expression induced by MSU (monosodium urate) crystals. In addition, miR-106a is found to considerably suppress IL-8 expression at protein and mRNA levels [149]. Moreover, miR-106a was downregulated in human peripheral blood mononuclear cells (PBMCs) isolated from CHB patients. Considering the essential role of IL-8 in liver inflammation caused by HBV infection, inhibition of IL-8 by miR-106a might have a substantial effect on the progression of CHB [150].

The regulation of inflammatory pathways by miR-21 during HBV-associated HCC is shown in Fig. 3.

Fig. 3
figure 3

The function of HBV HBx protein-induced miR-21 in the regulation of inflammation during HBV infection and hepatocarcinogenesis [151]

HIV infection

The family Retroviridae includes members of the lentivirus genus, such as HIV. At the present time, over 50% of patients infected with HIV throughout the world are taking combination antiretroviral therapy (cART). Moreover, CNS issues, often referred to as NeuroHIV, continue to affect people with HIV infection regardless of the capability of cART to reduce viremia. One of the nine HIV-1-encoded viral proteins which has attracted much interest, is HIV-1 transcriptional transactivator (Tat) that has the ability to damage CNS cells [152]. Also, HIV-1 Tat causes neuroinflammation via increasing viral replication within latently infected cells and by activating glial cells [153]. In the CNS and different tissues, transcription factor NF-kB controls the transcription of some inducible miRNAs like miRNA-34a. Pro-inflammatory cytokines can be secreted in response to elevated NF-kB activity, and this can cause neurodegeneration and cancer [154, 155]. Increased miRNA34a signaling has been correlated with the occurrence of neurological, neuroimmunological, neuroinflammatory, or neurodegenerative diseases in several studies [154, 156]. Periyasami et al. discovered that miR-34a has been raised in a dose and time-dependent manner when mouse microglia have been treated with HIV-1-Tat, while NLRC5-a (NOD-like receptor caspase recruitment domain having 5), a repressor of NF-kB signaling, was concurrently decreased [84]. When primary microglia from mice were transfected with miRNA-34a mimics, NLRC5 expression was markedly reduced, which increased p65 NF-kB expression, whereas, cells transfected with a miR-34a inhibitor produced more NLRC5. This suggests that the downregulation of NLRC5 via miR-34a and activation of NF-kB signaling can occur during HIV1-Tat-mediated microglial activation, while miR-34a inhibition prevented HIV-1 Tat from activating microglia. Therefore, the miR-34a-NLRC5-NF-kB axis could act as an important mechanism of HIV-1 Tat-mediated microglial activation. Overall, HIV-1 Tat upregulates miR-34a expression, that targets NLRC5 and promotes the generation of pro-inflammatory cytokines including IL-1 and IL-6 in microglia (Fig. 4) [84]. There are 2 subtypes of HIV-1 called HIV-1 B and HIV-1 C, which generate slightly various forms of Tat protein called Tat B and Tat C. It has been discovered that human microglial cells treated with HIV-1 Tat C upregulated miR-32 gene expression more than Tat B. It has been found that miR-32 bound to 3’UTR of TRAF3 to inhibit its expression, leading to increased expression of inflammatory genes within microglial cells [85].

Fig. 4
figure 4

HIV Tat-induced miR-34a induces microglial inflammation by increasing level of IL-1β and IL-6 by targeting NLRC5 [84]

Microglial cells secrete a range of chemokines as well as cytokines in response to HIV infection, especially monocyte chemoattractant proteins (MCPs). The primary nuclear receptor for macrophage-tropic HIV-1 is CCR5 (R5), among a range of chemokine receptors [157, 158]. In primary human fetal microglia infected with HIV-1, the expression of miR-146a has been determined, and it has been identified that miR-146a raised as the infection developed [159]. They discovered that overexpression of miR-146a before HIV-1 infection limited the production of MCP-2 within infected cells, and identified CCL8/MCP-2 mRNA to be a direct target for miR-146a. By analyzing the miR-146a expression along with MCP-2 in HIV-1 infected brain tissue, an inverse relationship was found between the level of miR-146a and the expression of its target gene MCP-2 [159]. Because MCP-2 is a powerful CCR5 ligand, the fact that miR-146a can inhibit its expression raises the possibility that the miR-146a/MCP-2 axis could be involved in controlling of viral shedding in brain. Production of the pro-inflammatory chemokine MCP-2 is generally inhibited by HIV-mediated upregulation of miR-146a, thereby maintaining chronic brain inflammation in HIV encephalitis [159].

The inflammatory cytokines overproduced in HIV-infected patients include IFN-γ, TNF-α, IL -18, as well as interferon-inducible protein 10 (IP-10) [160]. IP-10 is a pro-inflammatory chemokine linked to inflammatory disorders, immune system dysfunction, and the formation of tumors [161]. In HIV infection, increased blood levels of IP-10 were linked to both accelerated disease progression as well as persistent immune system activation [160, 162]. Prior research has shown that IP-10 can promote HIV multiplication while impairing T and NK cell activity [163, 164]. This explains how regulating IP-10 expression in HIV infection could prevent inflammation and slow the development of the disease. It was found that HIV-1 infection activated IP-10 synthesis by MDA5/Rig-I or TLR7/9-dependent mechanisms [165]. Although IP-10 is known to be upregulated in HIV infection, the exact mechanism is still unknown. Wu et al. investigated whether miRNAs that targeted IP-10 could decrease IP-10 levels in monocytes [166]. Their findings suggested that miR-21 might prevent monocytes from secreting IP-10, and that monocytes isolated from HIV-positive individuals had lower levels of miR-21. The increased synthesis of ISG15 (interferon-stimulated gene 15) may help to explain why alterations in miR-21 expression had no impact on IP-10 secretion in macrophages. Their data indicated that miR-21 could regulate IP-10 levels, shedding some light on the molecular control of IP-10 [166].

Japanese encephalitis virus

The Japanese encephalitis virus (JEV) is a positive-stranded encapsulated RNA virus, and a member of Flaviviridae family. JEV is a neurotropic virus that attacks the CNS as well as causing severe inflammation within the brain [94, 167]. When JEV infection occurs, pro-inflammatory cytokines are released, the BBB is disrupted, and CNS infiltration by T and B lymphocytes, DCs, along with NK cells occurs, especially at periphery, which worsens the inflammation [94, 167].

The E3 ubiquitin ligase family includes the pellino proteins PELI1, PELI2, and PELI3 [168, 169]. Pellino proteins can cause pro-inflammatory and antiviral responses in response to cell damage such as that found in neuropathic pain [168, 169]. The MAPK/NF-kB signaling cascade has been linked to PELI1 in chronic constriction injury (CCI) [170]. PELI1 controls NF-kB signaling as well as TRAF3 degradation [171]. Rastoghi et al. found that in microglial cells, miR-155 expression was dramatically higher 24 h after JEV infection. JEV replication increased upon miR-155 overexpression, while it was lower after miR-155 suppression. Inhibition of miR-155 reduced JEV infection and IL-6 level, which suggests a role for miR-155 during inflammation. Moreover, these findings demonstrated that JEV boosted the miR-155 expression, which within human microglial cells targeted PELI1 to block non-canonical NF-kB signaling along with pro-inflammatory factors like TNF-α and IL-6 (Fig. 5) [172].

Another study investigated the role of miR-146a in regulation of neuroinflammation caused by JEV infection [91]. The obtained result, demonstrated that the miR-146 and pro-inflammatory cytokines (TNF-α, IFN-β, IL -1b, IFN-α and IL-6) are upregulated in the brains of BALB/c mice and in cultured mouse microglial cells (C8-B4) following the JEV infection. They discovered that miR-146a had negative regulatory impacts on TNF-α, IL-1b, IFN-β, IL-6, and IFN-α in C8-B4 cells. Therefore, TNF-α, IL-1b, IFN-β, IL-6, and IFN-α were upregulated when miR-146a was downregulated. Such outcomes recommend that miR-146a can regulate inflammation during JEV infection, and could be involved in JEV infection-related seizures [91].

Fig. 5
figure 5

Japanese Encephalitis Virus (JEV) suppresses inflammation within microglial cells through the inhibition of the non-canonical NF-κB pathway through regulating miR-155 expression [91]

Investigating the impacts of JEV infection upon the miRNA expression profiles of microglia have shown that miRNAs, such as miR-301a can serve in the modulation of inflammatory responses [173]. MiR-301a has been indicated to regulate the autoimmune demyelination activity of Th cells and Th17 differentiation [174]. In JEV infection, miR-301a was found to be essential for controlling antiviral IFN-b response via inhibiting the production of IFN regulatory factor 1 (IRF1) as well as suppressor of cytokine signaling (SOCS). Hazara et al. confirmed that overexpressed miR-301a in JEV-infected human and mouse microglial cells, suppresses NKRF, the NF-kB inhibitory factor, and prevents NF-kB activation [175, 176]. Furthermore, it has been demonstrated that miR-301a knockdown in JEV-infected microglial cells elevated the NKRF expression, conforming the role of JEV-induced miR-301a in downregulating NKRF [95]. The miR-301a-mediated suppression of NKRF increased inflammation by promoting NF-kB nuclear translocation. However, when NKRF was overexpressed and miR-301a was inhibited, the nuclear accumulation of NF-kB decreased to baseline values. This study established that while JEV infection stimulated the M1 phenotype of microglia with increased proinflammatory cytokines, it reduced the M2 phenotype, which reduced the inflammatory response. Moreover, in vivo inhibition of miR-301a in the mouse brain restored expression of NKRF while decreasing neuronal cell death, inflammation, and microglial activation. Therefore, the NKRF production could be inhibited by JEV-induced miR-301a expression, and this may be employed to mitigate virus-induced neuroinflammation (Fig. 6) [95].

It has been established that JEV downregulating miR-432, results in upregulation of suppressor of cytokine signaling 5 (SOCS5) in CHME3 cells [177]. As a member of the suppressor of cytokine signaling (SOCS) protein family, SOCS5 has a negative impact on the ability of infected cells to respond to viruses [178]. Overexpression of miR-432 results in suppression of SOCS5 which in turn increases the phosphorylation of signal transducer and transcription activator 1 (STAT1) showed increased phosphorylation at Y-701 amino acid. This resulted in both increased ISRE activity along with increased pro-inflammatory cytokines’ expression. Moreover, miR-432 overexpression boosted the production of the inflammatory cytokines TNF-a and IL-6. As a result, miR-432 overexpression promoted the shedding of viral particles into the culture supernatant along with a powerful antiviral milieu within the cells, both of which led to reduced viral replication within the cells. Overall, JEV infection resulted in miR-432 downregulation and SOCS5 upregulation that helped the virus to escape from the cellular immune antiviral response [177]. This microRNA-mediated strategy is used by JEV to evade cellular immunity and promote its pathogenesis.

Coxsackie virus

Coxsackievirus group B type 3 (CVB3) is a member of the Picornaviridae family, and the main cause of viral myocarditis (VMC) [179]. VMC can result in myocardial damage through inducing an overactive host immune response [180]. Despite recent discoveries, there is currently no recognized therapy for VMC, and its pathogenesis is not well understood. The mechanism underlying the immune response and excessive inflammation to VMC infection is yet to be elucidated [181].

Fig. 6
figure 6

Japanese Encephalitis Virus (JEV) promotes inflammatory response via inhibiting NKRF production through upregulating the expression level of miR-301a [95]

NLRX1, a member of the NLR family, is found to be associated with several cancer, inflammatory, and neurodegenerative diseases [182]. However, the involvement of NLRX1 and its role in VMC remain unknown. It is established that during viral infection, NLRX1 suppresses the inflammation and innate immune response [183, 184]. Tong et al. investigated the implication of miR-15–NLRX1 axis in VMC development. In that context, they infected H9c2 cells with CVB3 which resulted in upregulation of miR-15 expression. In fact, knockdown of miR-15 reduced the levels of LDH, CK-MB, and cTn-I in CVB3-infected cells. Moreover, miR-15 knockdown increased cell survival, reduced apoptosis, and decreased the expression level of IL-1, IL-6, and IL-18 [96]. The expression of NLRP3 as well as caspase-1 p20 was also downregulated by miR-15 blockade, which considerably decreased the activation of the NLRP3 inflammasomes. Inhibition of NLRX1 suppressed NLRP3 inflammasomes and reversed the protective effect of miR-15 suppression versus CVB3-induced cardiac cell damage. Overall, the findings showed that CVB3-induced myocardial cell damage and inflammation were abrogated by miR-15 suppression [96].

Various cytokines (IL-1, IFN-γ , TNF-α, TGF-β, and IL-6) are secreted via different cells in reaction to viral infection and are responsible for causing myocardial damage [185]. Studies of cardiac hypertrophy and heart failure have shown that TGF-β is the major cause of fibrosis in both diseases [186]. In a viral myocarditis model in mice, TGF-β expression has been increased in CVB3-infected hearts [187]. Recently, Li et al. found that mice with viral myocarditis had decreased expression of miR-425-3p within their myocardial tissues [97]. In mice with myocarditis, the miR-425-3p overexpression gene alleviated the pathological symptoms, reduced cardiomyocyte death, boosted Bcl-2 expression, and decreased the levels of inflammatory markers and increased cardiac function and survival. They discovered that miR-425-3p can target TGF-β1, and indicated that miR-425-3p overexpression suppressed the expression of TGF-β1, p-smad2/smad2, as well as p-smad3/smad3. In fact, miR-425-3p overexpression could prevent apoptosis in CVB3-HL-1 cells, while the addition of TGF-β1 could counteract this effect according to in vitro studies. Overall, their outcomes indicated that miR-425-3p overexpression decreased cardiac inflammation as well as cardiomyocyte death in mice having viral myocarditis, and increased their survival by blocking the TGF1/smad axis [97].

Enteroviruses

The positive single-stranded RNA virus HEV71, also referred to as human enterovirus 71, is a member of the Picornaviridae family and Enterovirus genus [188]. A chemokine cascade involving overexpression of IL-8, CCL2, and CXCL10 has been reported to trigger the immune responses occurring after HEV71 infection [189]. CCL2 levels have been shown to be linked with the intensity of HEV71 encephalitis [190]. An alternative classification system of chemokines has been provided based on the miRNA-mediated regulation of chemokines, and has e wide range of variety for each chemokine. For instance, in macrophages of atherosclerotic lesions CCL2 is upregulated, while in inflammatory macrophages, CCL2 is expressed by miR-155 [191]. Zhang and colleagues have established that miR-206 is downregulated in severe HEV71 encephalitis. On the other hand, CCL2 is found to be upregulated during the transition between mild and severe HEV71 [101]. They also found that through targeting the 3’-UTR of CCL2, miR-206 can reduce the CCL2 expression in the human brain. Altogether, suggests that the severity of HEV71 encephalitis might be worsened by upregulation of CCL2 and downregulation of miR-206 [101].

IL-6 is one of many cytokines and proinflammatory mediators that can be expressed during EV71 infection [192, 193]. These cytokines and proinflammatory mediators (i.e., IL-6, IL-10, and COX-2) could stimulate STAT3 activity [194]. For example, IL-6 may bind to IL-6R by promoting STAT3 phosphorylation and activating downstream STAT3 signaling. The EV71 virus induced the activation of the MAPK, NF-kB, along with platelet-derived growth factor receptor (PDGFR) signaling pathways within neurons, thus upregulating the COX-2 expression [195]. Besides COX-2, STAT3 can also be activated by upstream signaling molecules including EGFR/PDGFR and NF-kB [194]. These findings collectively imply that STAT3 is involved in EV71 infection. In the traditional Janus kinase (JAK)-STAT3 signaling pathway, the major activators of STAT3 are IL-6, CNTF, IL-11, IL-27, IL-10, IL-22, along with other cytokines like TNF-α, IFN-α, and IFN-λ [196]. The life cycles of several viruses like varicella-zoster virus (VZV), SARS-CoV, HCV, and HBV are all affected by STAT3 [197]. Chang et al. found that after EV71 infection, miR-124 is upregulated while p-STAT3 and STAT3 levels were decreased [100]. MiR-124 was initially found to be a crucial regulator of the biological activity of microglia within the spinal cord and brain [198]. Current studies suggest that miR-124 play a substantial part in controlling immunity and inflammation [199]. Further studies have revealed that miR-124 can promote infection by EV71. Based on the collected data, they concluded that the antiviral activity of STAT3 against EV71 could be inhibited by virus-induced miR-124. It is likely that EV71-induced miR-124 could target the host STAT3 pathway and IL-6R thus mediating immune evasion [100].

The mechanism by which macromolecules, including transcription factors, move from the cytoplasm to nucleus is called nucleocytoplasmic transport, and is conserved across all species. During such procedure, a nuclear localization signal (NLS) must usually be present in a cargo molecule. Through the action of importin proteins that bind to the NLS specifically, cargo proteins can cross the nuclear pore complex, which connects the cytoplasm to the nucleus [200]. Importin 1 is a component of the importin family, which acts as a nucleocytoplasmic transporter, and is also known as karyopherin 2 (KPNA2) [201]. However, there are not many studies discussing the relationship between viral infections and KPNA2. In one example, the ORF6 protein of coronavirus was found to bind to KPNA2 and disassemble STAT1 complex inside nucleus, leading to severe acute respiratory syndrome [202]. Peng et al. discovered that the miR-302 cluster directly targeted KPNA2, thereby preventing the production of proinflammatory cytokines triggered by EV71 infection. KPNA2 regulated JNK1/JNK2, p65, and p38 nuclear translocation that in turn regulated the expression of cytokines stimulated by EV71 infection [99]. In general, the expression of the miR-302 cluster was inhibited via EV71 to increase KPNA2 expression (Fig. 7) [99]. When KPNA2 interacts with such transcription factors, this leads to the translocation of JNK1, JNK2, as well as p38 from the cytoplasm into nucleus, which in turn stimulates the secretion of inflammatory cytokines. Details of some other relevant studies are summarized in Table 1.

Fig. 7
figure 7

Enterovirus 71 (EV71) induces production of inflammatory cytokines by regulating KPNA2 expression by suppression of the miR-302 cluster [99]

Virus-mediated regulation of inflammatory pathway by virus-derived miRNAs

EBV-microRNA-BART11-mediated inflammation

A double-stranded DNA virus called Epstein-Barr virus (EBV) replicates in the human oral epithelium (lytic phase) and spreads via saliva. Sarcomas and cancers, including lung, breast, salivary gland, and gastric cancers, have all been associated with EBV [203,204,205]. How EBV promotes gastric cancer (GC) has been studied in detail [206, 207]. Viral elements that regulate the pathogenesis and development of these EBV-associated malignancies include viral proteins and viral miRNAs (EBV miRNAs) [203].

However, not much is known about the connection between viral miRNAs and GC. According to Song et al. EBV promoted inflammation-induced carcinogenesis by expression of the viral miRNA EBV-miR-BART11, which subsequently downregulated forkhead box protein P1 (FOXP1) [208]. This process promoted the development of tumor-associated macrophages (TAMs) in the host. Cytokine expression and tumor-promoting properties of TAMs were both decreased when FOXP1 was inhibited. The same researchers investigated the impacts of EBV-miR-BART11 upon epithelial-mesenchymal transition (EMT) along with metastasis in GC cells via focusing on FOXP1, an important human tumor suppressor gene [110]. Their findings indicated that increased EBV-miR-BART11 can downregulate FOXP1 in both GC tissues and cell lines. The FOXP1 downregulation could lead to cancer cells to release IL-1, IL-6, and 1 L-10, and in patients with GC this would lead to a worse prognosis. Moreover, TAMs from conditioned media promoted phenotypic changes and expression of EMT-related molecules in GC cells. Additionally, EMT changes were significantly promoted in GC cells cultured in a conditioned medium from TAMs infected with EBV-miR-BART11-containing lentivirus. In contrast, GC cells cultured in a conditioned medium from TAMs infected with FOXP1- encoding lentivirus exhibited little or no EMT change. Taken together, the upregulation of EBV-miR-BART11 level induces gastric cancer cell progression and metastasis mediated by promoting inflammation [110] (Fig. 8).

Fig. 8
figure 8

Regulation of immune responses and inflammation by EBV-encoded miRNAs [209]

HSV encoded-miR-H6-mediated inflammation

The most common herpes simplex virus is HSV-1, a linear double-stranded DNA virus which mostly affects neuronal and epithelial cells. For a successful HSV-1 infection, the following processes must occur: viral DNA replication, virus assembly, and virus shedding. The infected cell 4 polypeptide (ICP4) is an immediate early (IE) protein needed for efficient transcription of the early as well as late HSV-1 viral genes, and for allowing the HSV-1 replication cycle to proceed [210]. HSV-1 reactivation after a period of latency is supported by ICP4. ICP4 appears to be regulated by latency-associated transcripts (LATs) at the posttranscriptional level [211]. Several cell types are prevented from producing viruses when the ICP4 gene is inhibited [212]. The pleiotropic cytokine IL-6 has been linked to the immunological response to HSV-1. IL-6 can exert anti-inflammatory and pro-inflammatory activities depending on context [213]. At least 21 distinct miRNAs, including miR-H1 and miR-H6, are encoded by the HSV-1 genome. These miRNAs are placed upstream of the LAT (latency-associated transcript) promoter [214]. ICP4 interacts with RNA polymerase II-associated transcription factors to downregulate IE-genes in lytic HSV-1 infections, while at the same time upregulating early and late genes and repressing the LAT-promoter to silence LAT production [215, 216]. Duan et al. investigated how miR-H6 could affect HSV-1 replication as well as the generation of IL-6 in herpes simplex keratitis, a disease where HSV-1 infection can result in corneal blindness [111]. They found that in human retinal pigment epithelial cells and human corneal epithelial (HCE) cells, miR-H6 reduced HSV-1 infection and downregulated ICP4 protein. They concluded that the virus could trigger IL-6, and that the miR-H6-mediated reduction of IL-6 generation within HSV-1-infected HCE cells could explain the inhibitory activity of ICP4 in virus infections [217]. MiR-H6 mimics could inhibit the production of IL-6 following the HSV-1 infection within HCE cells. Because miR-H6 can both prevent viral replication and suppress inflammation caused by IL-6, it may be possible to treat HSV by regulating the expression of viral miRNAs [111].

HHV-8 (human herpesvirus 8) encoded miRNAs

The enveloped virus known as HHV-8 (human herpesvirus 8) is a member of Rhadinovirus genus of the Herpesviridae family [218]. HHV-8 can cause KS (Kaposi’s sarcoma), a cancer that affects the skin and mucosal surface. Factors involved in both the lytic and latent stages of the virus life cycle are involved in pathogenesis of KS [218, 219]. According to the available evidence, cytokines like IL-10 and IL-6 could have a substantial part in the pathogenesis of HHV-8-related cancers by suppressing T-cell activation along with promoting tumor cell growth and angiogenesis [220, 221]. Further research linked specific genes encoded via the HHV-8 genome, including the viral glycoprotein vOX2, to IL-6 production by macrophages and monocytes [222]. Although the HHV-8 genome encodes 25 mature miRNAs (KSHV miRNAs) [223], only a few studies have been performed to determine whether HHV-8 miRNAs can regulate cytokine expression. One of these works has been conducted by Qing et al. [112]. They showed that macrophages and myelomonocyte cells preferentially secreted IL-10 and IL-6 after HHV-8 infection. They also found that the HHV-8 miRNAs (miR-K12-1, -3, -7, -9, and − 11) could target the C/EBP-β isoform LIP and induce macrophages to secrete IL-6 and IL-10 [112]. How these HHV-8 miRNAs could be utilized to boost immune responses or block other HHV-8-induced cancer-promoting pathways by preventing HHV-8-macrophage interactions, requires additional research.

Conclusion

Thanks to tremendous advances in miRNA research, a deeper understanding of the processes that underpin the reaction of the innate immune system to viral pathogens could now be expected. Our knowledge of the cell physiology and immunology of viral infections has grown substantially over the past 10 years due to studies on the role of miRNAs in viral pathogenesis. It has been found that a large amount of miRNAs are engaged in the regulation of inflammation in viral infections. These miRNAs are mostly derived from the host cells, but a few of them are encoded by the viral genome itself. Viral infections are extremely complex since a single miRNA could regulate various targets involved in distinct stages of infection. The precise processes behind the control of miRNAs in viral encephalitis, myocarditis, respiratory infections, and neuroinflammation need to be further investigated. To further comprehend these complex networks, future research should examine the potential effects of distinct host and viral miRNAs on diverse targets that may influence the host response to viral infections as a whole.

Data availability

Not applicable

References

  1. Lucas M, Karrer U, Lucas A, Klenerman P. Viral escape mechanisms–escapology taught by viruses. Int J Exp Pathol. 2001;82(5):269–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Skalsky RL, Cullen BR. Viruses, microRNAs, and host interactions. Annu Rev Microbiol. 2010;64:123–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Girardi E, López P, Pfeffer S. On the importance of host microRNAs during viral infection. Front Genet. 2018;9: 439.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Singh RP, Massachi I, Manickavel S, Singh S, Rao NP, Hasan S, et al. The role of miRNA in inflammation and autoimmunity. Autoimmun rev. 2013;12(12):1160–5.

    Article  PubMed  CAS  Google Scholar 

  5. Sonkoly E, Pivarcsi A. microRNAs in inflammation. Int Rev Immunol. 2009;28(6):535–61.

    Article  PubMed  CAS  Google Scholar 

  6. Mishra R, Kumar A, Ingle H, Kumar H. The interplay between viral-derived miRNAs and host immunity during infection. Front Immunol. 2020;10:3079.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Pichlmair A. e Sousa CR. Innate recognit viruses Immunity. 2007;27(3):370–83.

    CAS  Google Scholar 

  8. Rouse BT, Sehrawat SJNRI. Immunity and immunopathology to viruses: what decides the outcome? 2010;10(7):514–26.

    CAS  Google Scholar 

  9. Iwasaki A, Medzhitov RJN. Toll-like receptor control of the adaptive immune responses. 2004;5(10):987–95.

    CAS  Google Scholar 

  10. Brockman MA, Kwon DS, Tighe DP, Pavlik DF, Rosato PC, Sela J, et al. IL-10 is up-regulated in multiple cell types during viremic HIV infection and reversibly inhibits virus-specific. T Cells. 2009;114(2):346–56.

  11. Smit JJ, Rudd BD, Lukacs NWJTJ. Plasmacytoid dendritic cells inhibit pulmonary immunopathology and promote clearance of respiratory syncytial virus. 2006;203(5):1153–9.

    CAS  Google Scholar 

  12. Bermejo-Martin JF, Ortiz de Lejarazu R, Pumarola T, Rello J, Almansa R, Ramírez P et al. Th1 and Th17 hypercytokinemia as early host response signature in severe pandemic influenza. Crit Care. 2009;13(6):R201.

  13. Culley FJ, Pennycook AM, Tregoning JS, Hussell T, Openshaw PJ. Differential chemokine expression following respiratory virus infection reflects Th1-or Th2-biased immunopathology. J Virol. 2006;80(9):4521–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Dakhama A, Lee YM, Ohnishi H, Jing X, Balhorn A, Takeda K et al. Virus-specific IgE enhances airway responsiveness on reinfection with respiratory syncytial virus in newborn mice. J Allergy Clin Immunol. 2009;123(1):138–45.e5.

  15. Koyuncu O, Hogue I, Enquist L. Virus infections in the nervous system. Cell Host Microbe. 2013;13(4):379–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Bhattacharyya S. Inflammation during virus infection: swings and roundabouts: Dynamics of Immune activation in viral diseases. 2019 Nov 5:43–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-981-15-1045-8_3

  17. Dahm T, Rudolph H, Schwerk C, Schroten H, Tenenbaum T. Neuroinvasion and inflammation in viral Central Nervous System infections. Mediat Inflamm. 2016;2016:8562805.

    Article  Google Scholar 

  18. Bartel DPJc. MicroRNAs: genomics, biogenesis, mechanism, and function. 2004;116(2):281–97.

  19. Yi R, Qin Y, Macara IG, Cullen BR. Development. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17(24):3011–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Eslami M, Khazeni S, Khanaghah XM, Asadi MH, Ansari MA, Garjan JH, et al. MiRNA-related metastasis in oral cancer: moving and shaking. Cancer Cell Int. 2023;23(1):182.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Yoshida T, Asano Y, Ui-Tei KJN-cR. Modulation of MicroRNA Processing by Dicer via its Associated dsRNA. Binding Proteins. 2021;7(3):57.

    CAS  Google Scholar 

  22. Sadri Nahand J, Salmaninejad A, Mollazadeh S, Tamehri Zadeh SS, Rezaee M, Sheida AH et al. Virus, exosome, and MicroRNA: new insights into autophagy. Cell biology and translational medicine, volume 17: stem cells in tissue differentiation, regulation and disease: Springer; 2022. pp. 97–162.

  23. Rahimian N, Nahand JS, Hamblin MR, Mirzaei H. Exosomal microRNA profiling. MicroRNA Profiling: Methods Mol Biol. 2023:2595:13–47.

  24. Contreras J, Rao D. MicroRNAs in inflammation and immune responses. Leukemia. 2012;26(3):404–13.

  25. Hussen BM, Ahmadi G, Marzban H, Azar MEF, Sorayyayi S, Karampour R, et al. The role of HPV gene expression and selected cellular MiRNAs in lung cancer development. Microb Pathog. 2021;150:104692.

    Article  PubMed  CAS  Google Scholar 

  26. Yang W, Chendrimada TP, Wang Q, Higuchi M, Seeburg PH, Shiekhattar R, et al. Modulation of microRNA processing and expression through RNA editing by ADAR deaminases. Nat Struct Mol Biol. 2006;13(1):13–21.

    Article  PubMed  CAS  Google Scholar 

  27. Wiesen JL, Tomasi TB. Dicer is regulated by cellular stresses and interferons. Mol Immunol. 2009;46(6):1222–8.

    Article  PubMed  CAS  Google Scholar 

  28. Viswanathan SR, Daley GQ, Gregory RI. Selective blockade of microRNA processing by Lin28. Science. 2008;320(5872):97–100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Liu G, Abraham E. MicroRNAs in immune response and macrophage polarization. Arterioscler Thromb Vasc Biol. 2013;33(2):170–7.

    Article  PubMed  PubMed Central  Google Scholar 

  30. O’Connell RM, Rao DS, Baltimore D. microRNA regulation of inflammatory responses. Annu Rev Immunol. 2012;30:295–312.

    Article  PubMed  Google Scholar 

  31. Tahamtan A, Teymoori-Rad M, Nakstad B, Salimi V. Anti-inflammatory microRNAs and their potential for inflammatory diseases treatment. Front Immunol. 2018;9: 1377.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sheedy FJ, Palsson-McDermott E, Hennessy EJ, Martin C, O’leary JJ, Ruan Q, et al. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol. 2010;11(2):141–7.

    Article  PubMed  CAS  Google Scholar 

  33. Lu TX, Hartner J, Lim E-J, Fabry V, Mingler MK, Cole ET, et al. MicroRNA-21 limits in vivo immune response-mediated activation of the IL-12/IFN-γ pathway, Th1 polarization, and the severity of delayed-type hypersensitivity. J Immunol. 2011;187(6):3362–73.

    Article  PubMed  CAS  Google Scholar 

  34. Feng J, Li A, Deng J, Yang Y, Dang L, Ye Y, et al. miR-21 attenuates lipopolysaccharide-induced lipid accumulation and inflammatory response: potential role in cerebrovascular disease. Lipids Health Dis. 2014;13(1):1–9.

    Article  Google Scholar 

  35. Caescu CI, Guo X, Tesfa L, Bhagat TD, Verma A, Zheng D, et al. Colony stimulating factor-1 receptor signaling networks inhibit mouse macrophage inflammatory responses by induction of microRNA-21 . Blood. J Am Soc Hematol. 2015;125(8):e1–13.

    CAS  Google Scholar 

  36. Xu W-D, Lu M-M, Pan H-F, Ye D-Q. Association of MicroRNA-146a with autoimmune diseases. Inflammation. 2012;35(4):1525–9.

    Article  PubMed  CAS  Google Scholar 

  37. Kutty RK, Nagineni CN, Samuel W, Vijayasarathy C, Jaworski C, Duncan T, et al. Differential regulation of microRNA-146a and microRNA-146b-5p in human retinal pigment epithelial cells by interleukin-1β, tumor necrosis factor-α, and interferon-γ. Mol Vis. 2013;19:737.

    PubMed  PubMed Central  CAS  Google Scholar 

  38. Larner-Svensson HM, Williams AE, Tsitsiou E, Perry MM, Jiang X, Chung KF, et al. Pharmacological studies of the mechanism and function of interleukin-1β-induced miRNA-146a expression in primary human airway smooth muscle. Respir Res. 2010;11(1):1–13.

    Article  Google Scholar 

  39. Jurkin J, Schichl YM, Koeffel R, Bauer T, Richter S, Konradi S, et al. miR-146a is differentially expressed by myeloid dendritic cell subsets and desensitizes cells to TLR2-dependent activation. J Immunol. 2010;184(9):4955–65.

    Article  PubMed  CAS  Google Scholar 

  40. Curtale G, Citarella F, Carissimi C, Goldoni M, Carucci N, Fulci V, et al. An emerging player in the adaptive immune response: microRNA-146a is a modulator of IL-2 expression and activation-induced cell death in T lymphocytes. Blood. J Am Soc Hematol. 2010;115(2):265–73.

    CAS  Google Scholar 

  41. Fulzele S, El-Sherbini A, Ahmad S, Sangani R, Matragoon S, El-Remessy A et al. MicroRNA-146b-3p regulates retinal inflammation by suppressing adenosine deaminase-2 in diabetes. BioMed research international. 2015;2015:846501.

    Article  Google Scholar 

  42. Bruning U, Cerone L, Neufeld Z, Fitzpatrick SF, Cheong A, Scholz CC, et al. MicroRNA-155 promotes resolution of hypoxia-inducible factor 1α activity during prolonged hypoxia. Mol Cell Biol. 2011;31(19):4087–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR, et al. Requirement of bic/microRNA-155 for normal immune function. Science. 2007;316(5824):608–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Calame K. MicroRNA-155 function in B cells. Immunity. 2007;27(6):825–7.

    Article  PubMed  CAS  Google Scholar 

  45. Abbasi-Kolli M, Nahand JS, Kiani SJ, Khanaliha K, Khatami AR, Taghizadieh M, et al. The expression patterns of MALAT-1, NEAT-1, THRIL, and mir-155-5p in the acute to the post-acute phase of COVID-19 disease. Brazilian J Infect Dis. 2022;26:102354.

    Article  CAS  Google Scholar 

  46. Mahesh G, Biswas R. MicroRNA-155: a Master Regulator of inflammation. J Interferon Cytokine Research: Official J Int Soc Interferon Cytokine Res. 2019;39(6):321–30.

    Article  CAS  Google Scholar 

  47. Tarassishin L, Loudig O, Bauman A, Shafit-Zagardo B, Suh HS, Lee SC. Interferon regulatory factor 3 inhibits astrocyte inflammatory gene expression through suppression of the proinflammatory miR‐155 and miR‐155. Glia. 2011;59(12):1911–22.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Rajasagi NK, Bhela S, Varanasi SK, Rouse BT. Frontline Science: aspirin-triggered resolvin D1 controls herpes simplex virus‐induced corneal immunopathology. J Leukoc Biol. 2017;102(5):1159–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Quinn SR, Mangan NE, Caffrey BE, Gantier MP, Williams BR, Hertzog PJ, et al. The role of Ets2 transcription factor in the induction of microRNA-155 (miR-155) by lipopolysaccharide and its targeting by interleukin-10. J Biol Chem. 2014;289(7):4316–25.

    Article  PubMed  CAS  Google Scholar 

  50. Mann M, Mehta A, Zhao J, Lee K, Marinov G, Garcia-Flores Y, et al. An NF-κB-microRNA regulatory network tunes macrophage inflammatory responses. Nat Commun. 2017;8(1):851.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zhang Q, Su J, Wang Z, Qi H, Ge Z, Li Z, et al. MicroRNA-149* suppresses hepatic inflammatory response through antagonizing STAT3 signaling pathway. Oncotarget. 2017;8(39):65397.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Xu G, Zhang Z, Xing Y, Wei J, Ge Z, Liu X, et al. Microrna-149 negatively regulates tlr‐triggered inflammatory response in macrophages by targeting myd88. J Cell Biochem. 2014;115(5):919–27.

    Article  PubMed  CAS  Google Scholar 

  53. Palmieri D, Capponi S, Geroldi A, Mura M, Mandich P, Palombo D. TNFα induces the expression of genes associated with endothelial dysfunction through p38MAPK-mediated down-regulation of miR-149. Biochem Biophys Res Commun. 2014;443(1):246–51.

    Article  PubMed  CAS  Google Scholar 

  54. Chen Q, Wu S, Wu Y, Chen L, Pang Q. miR-149 suppresses the inflammatory response of chondrocytes in osteoarthritis by down-regulating the activation of TAK1/NF-κB. Biomed Pharmacother. 2018;101:763–8.

    Article  PubMed  CAS  Google Scholar 

  55. Yaghoobi R, Kazerouni A. Evidence for clinical use of honey in wound healing as an anti-bacterial, anti-inflammatory anti-oxidant and anti-viral agent: a review. Jundishapur J Nat Pharm Prod. 2013;8(3):100.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Lu Q, Wu R, Zhao M, Garcia-Gomez A, Ballestar E. miRNAs as therapeutic targets in inflammatory disease. Trends Pharmacol Sci. 2019;40(11):853–65.

    Article  PubMed  CAS  Google Scholar 

  57. Berri F, Lê VB, Jandrot-Perrus M, Lina B, Riteau B. Switch from protective to adverse inflammation during influenza: viral determinants and hemostasis are caught as culprits. Cell Mol Life Sci. 2014;71(5):885–98.

    Article  PubMed  CAS  Google Scholar 

  58. Yoon SW, Webby RJ, Webster RG. Evolution and ecology of influenza A viruses. Influenza pathogenesis and control-volume I. 2014:359-75.

  59. Zhang X, Dong C, Sun X, Li Z, Zhang M, Guan Z, et al. Induction of the cellular miR-29c by influenza virus inhibits the innate immune response through protection of A20 mRNA. Biochem Biophys Res Commun. 2014;450(1):755–61.

  60. Podsiad A, Standiford TJ, Ballinger MN, Eakin R, Park P, Kunkel SL, et al. MicroRNA-155 regulates host immune response to postviral bacterial pneumonia via IL-23/IL-17 pathway. Am J Physiology-Lung Cell Mol Physiol. 2016;310(5):L465–75.

    Article  Google Scholar 

  61. Guo L, Wang Q, Zhang D. MicroRNA–4485 ameliorates severe influenza pneumonia via inhibition of the STAT3/PI3K/AKT signaling pathway. Oncol Lett. 2020;20(5):1.

    Google Scholar 

  62. Traber KE, Hilliard KL, Allen E, Wasserman GA, Yamamoto K, Jones MR, et al. Induction of STAT3-dependent CXCL5 expression and neutrophil recruitment by oncostatin-M during pneumonia. Am J Respir Cell Mol Biol. 2015;53(4):479–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Zhao J, Yu H, Liu Y, Gibson SA, Yan Z, Xu X, et al. Protective effect of suppressing STAT3 activity in LPS-induced acute lung injury. Am J Physiology-Lung Cell Mol Physiol. 2016;311(5):L868–880.

    Article  Google Scholar 

  64. Kaur M, Chandel J, Malik J, Naura AS. Particulate matter in COPD pathogenesis: an overview. Inflamm Res. 2022;71(7–8):797–815.

  65. Kopsaftis Z, Wood-Baker R, Poole P. Influenza vaccine for chronic obstructive pulmonary disease (COPD). Cochrane Database Syst Rev. 2018;6:CD002733.

    PubMed  Google Scholar 

  66. Hsu AC, Dua K, Starkey MR, Haw T-J, Nair PM, Nichol K et al. MicroRNA-125a and-b inhibit A20 and MAVS to promote inflammation and impair antiviral response in COPD. JCI insight. 2017;2(7):e90443.

  67. Tu Y-H, Guo Y, Ji S, Shen J-L, Fei G-HJF. The Influenza A Virus H3N2 triggers the hypersusceptibility of Airway Inflammatory Response via activating the lncRNA TUG1/miR-145-5p/NF-κB. Pathw COPD. 2021;12:604590.

    CAS  Google Scholar 

  68. Zhang N, Ma Y, Tian Y, Zhou Y, Tang Y, Hu S. Downregulation of microRNA–221 facilitates H1N1 influenza a virus replication through suppression of type–IFN response by targeting the SOCS1/NF–κB pathway. Mol Med Rep. 2021;24(1):1–11.

    Article  Google Scholar 

  69. Zhou Y, Xia Z, Cheng Z, Xu G, Yang X, Liu S, et al. Inducible microRNA-590-5p inhibits host antiviral response by targeting the soluble interleukin-6 (IL6) receptor. 2018;293(47):18168–79.

    CAS  Google Scholar 

  70. Chen X, Zhou L, Peng N, Yu H, Li M, Cao Z, et al. MicroRNA-302a suppresses influenza a virus–stimulated interferon regulatory factor-5 expression and cytokine storm induction. J Biol Chem. 2017;292(52):21291–303.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Hsu AC, Dua K, Starkey MR, Haw TJ, Nair PM, Nichol K, et al. MicroRNA-125a and -b inhibit A20 and MAVS to promote inflammation and impair antiviral response in COPD. JCI Insight. 2017;2(7):e90443.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Xie C, Chen Y, Luo D, Zhuang Z, Jin H, Zhou H, et al. Therapeutic potential of C1632 by inhibition of SARS-CoV-2 replication and viral-induced inflammation through upregulating let-7. Signal Transduct Target Ther. 2021;6(1):84.

  73. Morales L, Oliveros JC, Enjuanes L, Sola IJM. Contribution of host miRNA-223-3p to SARS-CoV-Induced. Lung Inflamm Pathol. 2022;13(2):e03135–03121.

    Google Scholar 

  74. Zhang Y, Shao L. Decreased microRNA-140-5p contributes to respiratory syncytial virus disease through targeting toll-like receptor 4. Exp Ther Med. 2018;16(2):993–9.

    PubMed  PubMed Central  Google Scholar 

  75. Huang Z, Liu X, Wu X, Chen M, Yu W. MiR-146a alleviates lung injury caused by RSV infection in young rats by targeting TRAF-6 and regulating JNK/ERKMAPK signaling pathways. Sci Rep. 2022;12(1):1–12.

    Google Scholar 

  76. Laanesoo A, Urgard E, Periyasamy K, Laan M, Bochkov YA, Aab A, et al. Dual role of the miR-146 family in rhinovirus-induced airway inflammation and allergic asthma exacerbation. Clin Transl Med. 2021;11(6):e427.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Fu X, Ouyang Y, Mo J, Li R, Fu L, Peng S. Upregulation of microRNA-328-3p by hepatitis B virus contributes to THLE-2 cell injury by downregulating FOXO4. J Transl Med. 2020;18(1):1–15.

    Article  CAS  Google Scholar 

  78. Huang JY, Chou SF, Lee JW, Chen HL, Chen CM, Tao MH, et al. MicroRNA-130a can inhibit hepatitis B virus replication via targeting PGC1α and PPARγ. RNA. 2015;21(3):385–400.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Li JF, Dai XP, Zhang W, Sun SH, Zeng Y, Zhao GY, et al. Upregulation of microRNA-146a by hepatitis B virus X protein contributes to hepatitis development by downregulating complement factor H. mBio. 2015;6(2):e02459–14.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Wu A, Chen H, Xu C, Zhou J, Chen S, Shi Y, et al. miR-203a is involved in HBx-induced inflammation by targeting Rap1a. Exp Cell Res. 2016;349(1):191–7.

    Article  PubMed  CAS  Google Scholar 

  81. Hong Z, Hong H, Liu J, Zheng X, Huang M, Li C, et al. miR-106a is downregulated in peripheral blood mononuclear cells of chronic hepatitis B and associated with enhanced levels of interleukin-8. Mediators Inflamm. 2015;2015:629862.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Yan J, Zhang Y, Su Y, Tian L, Qin P, Xu X, et al. microRNA-125a targets MAVS and TRAF6 to modulate interferon signaling and promote HCV infection. Virus Res. 2021;296:198336.

    Article  PubMed  CAS  Google Scholar 

  83. Sarma NJ, Tiriveedhi V, Crippin JS, Chapman WC, Mohanakumar T. Hepatitis C virus-induced changes in microRNA 107 (miRNA-107) and miRNA-449a modulate CCL2 by targeting the interleukin-6 receptor complex in hepatitis. J Virol. 2014;88(7):3733–43.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Periyasamy P, Thangaraj A, Bendi VS, Buch S. HIV-1 Tat-mediated microglial inflammation involves a novel miRNA-34a-NLRC5-NFκB signaling axis. Brain, behavior, and immunity. 2019;80:227–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Mishra R, Chhatbar C, Singh SK. HIV-1 Tat C-mediated regulation of tumor necrosis factor receptor-associated factor-3 by microRNA 32 in human microglia. J Neuroinflamm. 2012;9(1):1–15.

    Article  Google Scholar 

  86. Huang J, Zhu L, Qiu C, Xu X, Zhang L, Ding X, et al. MicroRNA miR-126-5p enhances the inflammatory responses of monocytes to lipopolysaccharide stimulation by suppressing cylindromatosis in chronic HIV-1 infection. J Virol. 2017;91(10):e02048–16.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Wu X, Zhang LL, Yin LB, Fu YJ, Jiang YJ, Ding HB, et al. Deregulated microRNA-21 expression in monocytes from HIV-infected patients contributes to elevated IP-10 secretion in HIV infection. Front Immunol. 2017;8:1122.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Rom S, Rom I, Passiatore G, Pacifici M, Radhakrishnan S, Del Valle L, et al. CCL8/MCP-2 is a target for mir-146a in HIV-1-infected human microglial cells. FASEB J. 2010;24(7):2292–300.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Rastogi M, Singh SK. Japanese Encephalitis Virus exploits microRNA-155 to suppress the non-canonical NF-κB pathway in human microglial cells. Biochim Biophys Acta Gene Regul Mech. 2020;1863(11):194639.

    Article  PubMed  CAS  Google Scholar 

  90. Jiang H, Bai L, Ji L, Bai Z, Su J, Qin T et al. Degradation of microRNA miR-466d-3p by Japanese encephalitis virus NS3 facilitates viral replication and interleukin-1β expression. J Virol. 2020;94(15):e00294–20.

  91. Deng M, Du G, Zhao J, Du X. miR-146a negatively regulates the induction of proinflammatory cytokines in response to Japanese encephalitis virus infection in microglial cells. Arch Virol. 2017;162(6):1495–505.

    Article  PubMed  CAS  Google Scholar 

  92. Zhu B, Ye J, Nie Y, Ashraf U, Zohaib A, Duan X, et al. MicroRNA-15b modulates Japanese encephalitis virus–mediated inflammation via targeting RNF125. J Immunol. 2015;195(5):2251–62.

    Article  PubMed  CAS  Google Scholar 

  93. Ashraf U, Zhu B, Ye J, Wan S, Nie Y, et al. MicroRNA-19b-3p Modulates Japanese Encephalitis Virus-Mediated Inflammation via Targeting. Virol. 2016;90(9):4780–95.

    Article  CAS  Google Scholar 

  94. Thounaojam MC, Kundu K, Kaushik DK, Swaroop S, Mahadevan A, Shankar SK, et al. MicroRNA 155 regulates Japanese encephalitis virus-induced inflammatory response by targeting src homology 2-containing inositol phosphatase 1. J Virol. 2014;88(9):4798–810.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Hazra B, Chakraborty S, Bhaskar M, Mukherjee S, Mahadevan A, Basu A. miR-301a regulates inflammatory response to Japanese encephalitis virus infection via suppression of NKRF activity. J Immunol. 2019;203(8):2222–38.

    Article  PubMed  CAS  Google Scholar 

  96. Tong R, Jia T, Shi R, Yan F. Inhibition of microRNA-15 protects H9c2 cells against CVB3-induced myocardial injury by targeting NLRX1 to regulate the NLRP3 inflammasome. Cell Mol Biol Lett. 2020;25(1):1–14.

    Article  Google Scholar 

  97. Li J, Tu J, Gao H, Tang L. MicroRNA-425-3p inhibits myocardial inflammation and cardiomyocyte apoptosis in mice with viral myocarditis through targeting TGF-β1. Immun Inflamm Dis. 2021;9(1):288–98.

    Article  PubMed  CAS  Google Scholar 

  98. Corsten MF, Heggermont W, Papageorgiou AP, Deckx S, Tijsma A, Verhesen W, et al. The microRNA-221/-222 cluster balances the antiviral and inflammatory response in viral myocarditis. Eur Heart J. 2015;36(42):2909–19.

    Article  PubMed  CAS  Google Scholar 

  99. Peng N, Yang X, Zhu C, Zhou L, Yu H, Li M, et al. MicroRNA-302 cluster downregulates Enterovirus 71–Induced Innate Immune response by targeting KPNA2. J Immunol. 2018;201(1):145–56.

    Article  PubMed  CAS  Google Scholar 

  100. Chang Z, Wang Y, Bian L, Liu Q, Long J-E. Enterovirus 71 antagonizes the antiviral activity of host STAT3 and IL-6R with partial dependence on virus-induced miR-124. J Gen Virol. 2017;98(12):3008–25.

    Article  PubMed  CAS  Google Scholar 

  101. Zhang G, Wang J, Yao G, Shi B. Downregulation of CCL2 induced by the upregulation of microRNA-206 is associated with the severity of HEV71 encephalitis. Mol Med Rep. 2017;16(4):4620–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Pu J, Wu S, Xie H, Li Y, Yang Z, Wu X, et al. miR-146a Inhibits dengue-virus-induced autophagy by targeting TRAF6. Arch Virol. 2017;162(12):3645–59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Zhang X, Guo Y, Xu X, Tang T, Sun L, Wang H, et al. miR-146a promotes Borna Disease virus 1 replication through IRAK1/TRAF6/NF-κB signaling pathway. Virus Res. 2019;271:197671.

    Article  PubMed  CAS  Google Scholar 

  104. Natekar JP, Rothan HA, Arora K, Strate PG, Kumar M. Cellular microRNA-155 regulates virus-induced inflammatory response and protects against lethal West Nile virus infection. Viruses. 2019;12(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Yan B, Ma H, Jiang S, Shi J, Yang Z, Zhu W, et al. microRNA-221 restricts human cytomegalovirus replication via promoting type I IFN production by targeting SOCS1/NF-κB pathway. Cell Cycle. 2019;18(22):3072–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Choi B, Kim HA, Suh CH, Byun HO, Jung JY, Sohn S. The relevance of miRNA-21 in HSV-induced inflammation in a mouse model. Int J Mol Sci. 2015;16(4):7413–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Selvamani SP, Mishra R, Singh SK. Chikungunya virus exploits miR-146a to regulate NF-κB pathway in human synovial fibroblasts. PloS One. 2014;9(8):e103624.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Shukla A, Rastogi M, Singh SKJIJBM. Zika virus NS1 suppresses the innate immune responses via miR-146a in human microglial cells. 2021;193:2290–6.

    CAS  Google Scholar 

  109. Pandey N, Rastogi M, Singh SKJJBS. Chandipura virus dysregulates the expression of hsa-mir-21-5p to activate NF-κB in human microglial cells. 2021;28(1):1–13.

    Google Scholar 

  110. Song Y, Li Q, Liao S, Zhong K, Jin Y, Zeng T. Epstein-Barr virus-encoded miR-BART11 promotes tumor-associated macrophage-induced epithelial-mesenchymal transition via targeting FOXP1 in gastric cancer. Virology. 2020;548:6–16.

    Article  PubMed  CAS  Google Scholar 

  111. Duan F, Liao J, Huang Q, Nie Y, Wu K. HSV-1 miR-H6 inhibits HSV-1 replication and IL-6 expression in human corneal epithelial cells in vitro. Clin Dev Immunol. 2012;2012:192791.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Qin Z, Kearney P, Plaisance K, Parsons CH. Pivotal advance: Kaposi’s sarcoma-associated herpesvirus (KSHV)-encoded microRNA specifically induce IL-6 and IL-10 secretion by macrophages and monocytes. J Leukoc Biol. 2010;87(1):25–34.

    Article  PubMed  CAS  Google Scholar 

  113. Pum A, Ennemoser M, Adage T, Kungl AJ. Cytokines and chemokines in SARS-CoV-2 infections—therapeutic strategies targeting cytokine storm. Biomolecules. 2021;11(1): 91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Eslami N, Aghbash PS, Shamekh A, Entezari-Maleki T, Nahand JS, Sales AJ, et al. SARS-CoV-2: receptor and co-receptor Tropism Probability. Curr Microbiol. 2022;79(5):133.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe. 2016;19(2):181–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Lamirande EW, DeDiego ML, Roberts A, Jackson JP, Alvarez E, Sheahan T, et al. A live attenuated severe acute respiratory syndrome coronavirus is immunogenic and efficacious in golden Syrian hamsters. J Virol. 2008;82(15):7721–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. DeDiego ML, Nieto-Torres JL, Regla-Nava JA, Jimenez-Guardeño JM, Fernandez-Delgado R, Fett C, et al. Inhibition of NF-κB-mediated inflammation in severe acute respiratory syndrome coronavirus-infected mice increases survival. J Virol. 2014;88(2):913–24.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Schoeman D, Cloete R, Fielding BC. The Flexible, Extended Coil of the PDZ-Binding motif of the three Deadly Human Coronavirus E proteins plays a role in pathogenicity. Viruses. 2022;14(8): 1707.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Cauchois R, Koubi M, Delarbre D, Manet C, Carvelli J, Blasco VB, et al. Early IL-1 receptor blockade in severe inflammatory respiratory failure complicating COVID-19. Proceedings of the National Academy of Sciences. 2020;117(32):18951–3.

    Article  CAS  Google Scholar 

  120. Yoshikawa T, Hill T, Li K, Peters CJ, Tseng C-TK. Severe acute respiratory syndrome (SARS) coronavirus-induced lung epithelial cytokines exacerbate SARS pathogenesis by modulating intrinsic functions of monocyte-derived macrophages and dendritic cells. J Virol. 2009;83(7):3039–48.

    Article  PubMed  CAS  Google Scholar 

  121. Morales L, Oliveros JC, Enjuanes L, Sola I. Contribution of host miRNA-223-3p to SARS-CoV-Induced Lung Inflammatory Pathology. mBio. 2022;13(2):e0313521.

    Article  PubMed  Google Scholar 

  122. Iliopoulos D, Hirsch HA, Struhl K. An epigenetic switch involving NF-κB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell. 2009;139(4):693–706.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Xie C, Chen Y, Luo D, Zhuang Z, Jin H, Zhou H, et al. Therapeutic potential of C1632 by inhibition of SARS-CoV-2 replication and viral-induced inflammation through upregulating let-7. Signal Transduct Target Therapy. 2021;6(1):1–3.

    CAS  Google Scholar 

  124. Garofalo R, Sabry M, Jamaluddin M, Yu RK, Casola A, Ogra PL, et al. Transcriptional activation of the interleukin-8 gene by respiratory syncytial virus infection in alveolar epithelial cells: nuclear translocation of the RelA transcription factor as a mechanism producing airway mucosal inflammation. J Virol. 1996;70(12):8773–81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Tian B, Zhang Y, Luxon BA, Garofalo RP, Casola A, Sinha M, et al. Identification of NF-κB-dependent gene networks in respiratory syncytial virus-infected cells. J Virol. 2002;76(13):6800–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Dev A, Iyer S, Razani B, Cheng G. NF-κB and innate immunity. NF-kB Health Disease. Curr Top Microbiol Immunol. 2011:349:115−43.

  127. Fiedler MA, Wernke-Dollries K. Incomplete regulation of NF-κB by IκBα during respiratory syncytial virus infection in A549 cells. J Virol. 1999;73(5):4502–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Bitko V, Barik S. Persistent activation of RelA by respiratory syncytial virus involves protein kinase C, underphosphorylated IκBβ, and sequestration of protein phosphatase 2A by the viral phosphoprotein. J Virol. 1998;72(7):5610–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Marr N, Turvey SE. Role of human TLR4 in respiratory syncytial virus-induced NF-κB activation, viral entry and replication. Innate Immun. 2012;18(6):856–65.

    Article  PubMed  Google Scholar 

  130. Rallabhandi P, Phillips RL, Boukhvalova MS, Pletneva LM, Shirey KA, Gioannini TL, et al. Respiratory syncytial virus fusion protein-induced toll-like receptor 4 (TLR4) signaling is inhibited by the TLR4 antagonists Rhodobacter sphaeroides lipopolysaccharide and eritoran (E5564) and requires direct interaction with MD-2. mBio. 2012;3(4):e00218–00212.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Yang Y, Liu D, Xi Y, Li J, Liu B, Li J. Upregulation of miRNA–140–5p inhibits inflammatory cytokines in acute lung injury through the MyD88/NF–κB signaling pathway by targeting TLR4. Experimental Therapeutic Med. 2018;16(5):3913–20.

    Google Scholar 

  132. Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences. 2006;103(33):12481–6.

    Article  CAS  Google Scholar 

  133. Shi Z, Sun Y, Wang K, Jia J, Yang J, Li Y. Effects of miR-26a/miR-146a/miR-31 on airway inflammation of asthma mice and asthma children. Eur Rev Med Pharmacol Sci. 2019;23(12):5432–40.

    PubMed  Google Scholar 

  134. Huang Z, Liu X, Wu X, Chen M, Yu W. MiR-146a alleviates lung injury caused by RSV infection in young rats by targeting TRAF-6 and regulating JNK/ERKMAPK signaling pathways. Sci Rep. 2022;12(1):3481.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Lucifora J, Protzer U. Attacking hepatitis B virus cccDNA–The holy grail to hepatitis B cure. J Hepatol. 2016;64(1):S41–8.

    Article  PubMed  CAS  Google Scholar 

  136. Yuen M-F, Chen D-S, Dusheiko GM, Janssen HL, Lau DT, Locarnini SA, et al. Hepatitis B virus infection. Nat Reviews Disease Primers. 2018;4(1):1–20.

    Google Scholar 

  137. Elpek GO. Molecular pathways in viral hepatitis-associated liver carcinogenesis: an update. World J Clin Cases. 2021;9(19): 4890.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Xu LL, Warren MK, Rose WL, Gong W, Wang JM. Human recombinant monocyte chemotactic protein and other C-C chemokines bind and induce directional migration of dendritic cells in vitro. J Leukoc Biol. 1996;60(3):365–71.

    Article  PubMed  CAS  Google Scholar 

  139. Carr MW, Roth SJ, Luther E, Rose SS, Springer TA. Monocyte chemoattractant protein 1 acts as a T-lymphocyte chemoattractant. Proc Natl Acad Sci. 1994;91(9):3652–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Narumi S, Tominaga Y, Tamaru M, Shimai S, Okumura H, Nishioji K, et al. Expression of IFN-inducible protein-10 in chronic hepatitis. J Immunol. 1997;158(11):5536–44.

    Article  PubMed  CAS  Google Scholar 

  141. Degré D, Lemmers A, Gustot T, Ouziel R, Trépo E, Demetter P, et al. Hepatic expression of CCL2 in alcoholic liver disease is associated with disease severity and neutrophil infiltrates. Clin Experimental Immunol. 2012;169(3):302–10.

    Article  Google Scholar 

  142. Zhang C, Li Y, Wu Y, Wang L, Wang X, Du J. Interleukin-6/signal transducer and activator of transcription 3 (STAT3) pathway is essential for macrophage infiltration and myoblast proliferation during muscle regeneration. J Biol Chem. 2013;288(3):1489–99.

    Article  PubMed  CAS  Google Scholar 

  143. Johnson PF. Molecular stop signs: regulation of cell-cycle arrest by C/EBP transcription factors. J Cell Sci. 2005;118(12):2545–55.

    Article  PubMed  CAS  Google Scholar 

  144. Zeng C, Wang R, Li D, Lin XJ, Wei QK, Yuan Y, et al. A novel GSK-3 beta–C/EBP alpha–miR‐122–insulin‐like growth factor 1 receptor regulatory circuitry in human hepatocellular carcinoma. Hepatology. 2010;52(5):1702–12.

    Article  PubMed  CAS  Google Scholar 

  145. Sadri Nahand J, Bokharaei-Salim F, Salmaninejad A, Nesaei A, Mohajeri F, Moshtzan A, et al. microRNAs: Key players in virus‐associated hepatocellular carcinoma. J Cell Physiol. 2019;234(8):12188–225.

    Article  PubMed  CAS  Google Scholar 

  146. Zhang X-D, Wang Y, Ye L-H. Hepatitis B virus X protein accelerates the development of hepatoma. Cancer Biology Med. 2014;11(3):182.

    CAS  Google Scholar 

  147. Mahe Y, Mukaida N, Kuno K, Akiyama M, Ikeda N, Matsushima K, et al. Hepatitis B virus X protein transactivates human interleukin-8 gene through acting on nuclear factor kB and CCAAT/enhancer-binding protein-like cis-elements. J Biol Chem. 1991;266(21):13759–63.

    Article  PubMed  CAS  Google Scholar 

  148. Martinez-Nunez RT, Bondanese VP, Louafi F, Francisco-Garcia AS, Rupani H, Bedke N, et al. A microRNA network dysregulated in asthma controls IL-6 production in bronchial epithelial cells. PLoS ONE. 2014;9(10): e111659.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Dalbeth N, Pool B, Shaw OM, Harper JL, Tan P, Franklin C, et al. Role of miR-146a in regulation of the acute inflammatory response to monosodium urate crystals. Ann Rheum Dis. 2015;74(4):786–90.

    Article  PubMed  CAS  Google Scholar 

  150. Hong Z, Hong H, Liu J, Zheng X, Huang M, Li C, et al. miR-106a is downregulated in Peripheral Blood mononuclear cells of Chronic Hepatitis B and Associated with enhanced levels of Interleukin-8. Mediators of inflammation. 2015;2015: 629862.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Sartorius K, Swadling L, An P, Makarova J, Winkler C, Chuturgoon A, et al. The multiple roles of hepatitis B virus X protein (HBx) dysregulated microRNA in hepatitis B virus-associated hepatocellular carcinoma (HBV-HCC) and immune pathways. Viruses. 2020;12(7): 746.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Bagashev A, Sawaya BE. Roles and functions of HIV-1 Tat protein in the CNS: an overview. Virol J. 2013;10(1):1–20.

    Article  Google Scholar 

  153. Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell. 1988;55(6):1189–93.

    Article  PubMed  CAS  Google Scholar 

  154. Bhattacharjee S, Zhao Y, Dua P, Rogaev EI, Lukiw WJ. microRNA-34a-mediated down-regulation of the microglial-enriched triggering receptor and phagocytosis-sensor TREM2 in age-related macular degeneration. PLoS ONE. 2016;11(3): e0150211.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Lukiw WJ. NF-кB-regulated micro RNAs (miRNAs) in primary human brain cells. Exp Neurol. 2012;235(2):484–90.

    Article  PubMed  CAS  Google Scholar 

  156. Kofman A, Kim J, Park S, Dupart E, Letson C, Bao Y, et al. microRNA-34a promotes DNA damage and mitotic catastrophe. Cell Cycle. 2013;12(22):3500–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, et al. HIV-1 entry into CD4 + cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381(6584):667–73.

    Article  PubMed  CAS  Google Scholar 

  158. Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, et al. CC CKR5: a RANTES, MIP-1α, MIP-1β receptor as a fusion cofactor for macrophage-tropic HIV-1. Science. 1996;272(5270):1955–8.

    Article  PubMed  CAS  Google Scholar 

  159. Rom S, Rom I, Passiatore G, Pacifici M, Radhakrishnan S, Del Valle L, et al. CCL8/MCP-2 is a target for mir-146a in HIV-1-infected human microglial cells. FASEB Journal: Official Publication Federation Am Soc Experimental Biology. 2010;24(7):2292–300.

    Article  CAS  Google Scholar 

  160. Jiao Y, Zhang T, Wang R, Zhang H, Huang X, Yin J, et al. Plasma IP-10 is associated with rapid disease progression in early HIV-1 infection. Viral Immunol. 2012;25(4):333–7.

    Article  PubMed  CAS  Google Scholar 

  161. Ahmadi Z, Arababadi MK, Hassanshahi G. CXCL10 activities, biological structure, and source along with its significant role played in pathophysiology of type I diabetes mellitus. Inflammation. 2013;36(2):364–71.

    Article  PubMed  CAS  Google Scholar 

  162. Noel N, Boufassa F, Lécuroux C, Saez-Cirion A, Bourgeois C, Dunyach-Remy C, et al. Elevated IP10 levels are associated with immune activation and low CD4 + T-cell counts in HIV controller patients. Aids. 2014;28(4):467–76.

    Article  PubMed  CAS  Google Scholar 

  163. Wang Z, Wu T, Ma M, Zhang Z, Fu Y, Liu J, et al. Elevated interferon-γ–induced protein 10 and its receptor CXCR3 impair NK cell function during HIV infection. J Leukoc Biol. 2017;102(1):163–70.

    Article  PubMed  CAS  Google Scholar 

  164. Ramirez L, Arango T, Thompson E, Naji M, Tebas P, Boyer J. High IP-10 levels decrease T cell function in HIV‐1‐infected individuals on ART. J Leukoc Biol. 2014;96(6):1055–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Simmons RP, Scully EP, Groden EE, Benedict KF, Chang JJ, Kim L, et al. HIV-1 infection induces strong production of IP-10 through TLR7/9-dependent pathways. AIDS. 2013;27(16):2505.

    Article  PubMed  CAS  Google Scholar 

  166. Wu X, Zhang LL, Yin LB, Fu YJ, Jiang YJ, Ding HB, et al. Deregulated MicroRNA-21 expression in Monocytes from HIV-Infected patients contributes to elevated IP-10 secretion in HIV infection. Front Immunol. 2017;8: 1122.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Li F, Wang Y, Yu L, Cao S, Wang K, Yuan J, et al. Viral infection of the central nervous system and neuroinflammation precede blood-brain barrier disruption during Japanese encephalitis virus infection. J Virol. 2015;89(10):5602–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Ordureau A, Enesa K, Nanda S, Le Francois B, Peggie M, Prescott A, et al. DEAF1 is a Pellino1-interacting protein required for interferon production by Sendai virus and double-stranded RNA. J Biol Chem. 2013;288(34):24569–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Jiang Z, Johnson HJ, Nie H, Qin J, Bird TA, Li X. Pellino 1 is required for interleukin-1 (IL-1)-mediated signaling through its interaction with the IL-1 receptor-associated kinase 4 (IRAK4)-IRAK-tumor necrosis factor receptor-associated factor 6 (TRAF6) complex. J Biol Chem. 2003;278(13):10952–6.

    Article  PubMed  CAS  Google Scholar 

  170. Marques RE, Del Sarto JL, Rocha RP, Gomes GF, Cramer A, Rachid MA, et al. Development of a model of Saint Louis encephalitis infection and disease in mice. J Neuroinflamm. 2017;14(1):1–14.

    Article  Google Scholar 

  171. Ghoshal A, Das S, Ghosh S, Mishra MK, Sharma V, Koli P, et al. Proinflammatory mediators released by activated microglia induces neuronal death in Japanese encephalitis. Glia. 2007;55(5):483–96.

    Article  PubMed  Google Scholar 

  172. Rastogi M, Singh SK. Japanese Encephalitis Virus exploits microRNA-155 to suppress the non-canonical NF-κB pathway in human microglial cells. Biochim et Biophys Acta (BBA)-Gene Regul Mech. 2020;1863(11):194639.

    Article  CAS  Google Scholar 

  173. Thounaojam MC, Kaushik DK, Kundu K, Basu A. Micro RNA-29b modulates Japanese encephalitis virus-induced microglia activation by targeting tumor necrosis factor alpha-induced protein 3. J Neurochem. 2014;129(1):143–54.

    Article  PubMed  CAS  Google Scholar 

  174. Mycko MP, Cichalewska M, Machlanska A, Cwiklinska H, Mariasiewicz M, Selmaj KW. MicroRNA-301a regulation of a T-helper 17 immune response controls autoimmune demyelination. Proc Natl Acad Sci. 2012;109(20):E1248–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Lu Z, Li Y, Takwi A, Li B, Zhang J, Conklin DJ, et al. miR-301a as an NF‐κB activator in pancreatic cancer cells. EMBO J. 2011;30(1):57–67.

    Article  PubMed  CAS  Google Scholar 

  176. Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, et al. New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci. 2016;113(12):E1738–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Sharma N, Kumawat KL, Rastogi M, Basu A, Singh SK. Japanese Encephalitis Virus exploits the microRNA-432 to regulate the expression of suppressor of Cytokine Signaling (SOCS) 5. Sci Rep. 2016;6:27685.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Trengove MC, Ward AC. SOCS proteins in development and disease. Am J Clin Experimental Immunol. 2013;2(1):1.

    Google Scholar 

  179. Shen Y, Xu W, Chu Y-W, Wang Y, Liu Q-S, Xiong S-D. Coxsackievirus group B type 3 infection upregulates expression of monocyte chemoattractant protein 1 in cardiac myocytes, which leads to enhanced migration of mononuclear cells in viral myocarditis. J Virol. 2004;78(22):12548–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Fairweather D, Rose NR. Coxsackievirus-induced myocarditis in mice: a model of autoimmune disease for studying immunotoxicity. Methods. 2007;41(1):118–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Corsten MF, Schroen B, Heymans S. Inflammation in viral myocarditis: friend or foe? Trends Mol Med. 2012;18(7):426–37.

    Article  PubMed  CAS  Google Scholar 

  182. Nagai-Singer MA, Morrison HA, Allen IC. NLRX1 is a multifaceted and enigmatic regulator of immune system function. Front Immunol. 2019;10:2419.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. O’Neill LA. Innate immunity: squelching anti-viral signalling with NLRX1. Curr Biol. 2008;18(7):R302–4.

    Article  PubMed  Google Scholar 

  184. Allen IC, Moore CB, Schneider M, Lei Y, Davis BK, Scull MA, et al. NLRX1 protein attenuates inflammatory responses to infection by interfering with the RIG-I-MAVS and TRAF6-NF-κB signaling pathways. Immunity. 2011;34(6):854–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  185. Xie Y, Chen R, Zhang X, Yu Y, Yang Y, Zou Y, et al. Blockade of interleukin-17A protects against coxsackievirus B3-induced myocarditis by increasing COX-2/PGE2 production in the heart. FEMS Immunol Med Microbiol. 2012;64(3):343–51.

    Article  PubMed  CAS  Google Scholar 

  186. Gong W, Yan M, Chen J, Chaugai S, Chen C, Wang D. Chronic inhibition of cyclic guanosine monophosphate-specific phosphodiesterase 5 prevented cardiac fibrosis through inhibition of transforming growth factor β-induced smad signaling. Front Med. 2014;8(4):445–55.

    Article  PubMed  Google Scholar 

  187. Glück B, Schmidtke M, Merkle I, Stelzner A, Gemsa D. Persistent expression of Cytokines inÈthe Chronic Stage of CVB3-induced myocarditis in NMRI mice. J Mol Cell Cardiol. 2001;33(9):1615–26.

    Article  PubMed  Google Scholar 

  188. Wang S-M, Liu C-C. Enterovirus 71: epidemiology, pathogenesis and management. Expert Rev anti-infective Therapy. 2009;7(6):735–42.

    Article  PubMed  Google Scholar 

  189. Jin Y, Zhang R, Wu W, Duan G. Antiviral and inflammatory cellular signaling associated with enterovirus 71 infection. Viruses. 2018;10(4): 155.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Li J-a, Chen Z-b, Lv T-g, Han Z-l. Genetic polymorphism of CCL2 – 2518, CXCL10 – 201, IL8 + 781 and susceptibility to severity of Enterovirus-71 infection in a Chinese population. Inflamm Res. 2014;63(7):549–56.

    Article  PubMed  CAS  Google Scholar 

  191. Nazari-Jahantigh M, Wei Y, Noels H, Akhtar S, Zhou Z, Koenen RR, et al. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J Clin Investig. 2012;122(11):4190–202.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Wang S-M, Lei H-Y, Huang M-C, Su L-Y, Lin H-C, Yu C-K, et al. Modulation of cytokine production by intravenous immunoglobulin in patients with enterovirus 71-associated brainstem encephalitis. J Clin Virol. 2006;37(1):47–52.

    Article  PubMed  CAS  Google Scholar 

  193. 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 PGE2 in viral replication. Cell Signal. 2010;22(2):234–46.

    Article  PubMed  CAS  Google Scholar 

  194. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9(11):798–809.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  195. Tung WH, Lee IT, Hsieh HL, Yang CM. EV71 induces COX-2 expression via c‐Src/PDGFR/PI3K/Akt/p42/p44 MAPK/AP‐1 and NF‐κB in rat brain astrocytes. J Cell Physiol. 2010;224(2):376–86.

    Article  PubMed  CAS  Google Scholar 

  196. Walter MR. The molecular basis of IL-10 function: from receptor structure to the onset of signaling. Interleukin-10 Health Dis. 2014;380:191–212.

    Article  CAS  Google Scholar 

  197. Chang Z, Wang Y, Zhou X, Long J-E. STAT3 roles in viral infection: antiviral or proviral? Future Virol. 2018;13(08):557–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Ponomarev ED, Veremeyko T, Barteneva N, Krichevsky AM, Weiner HL. MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-α–PU. 1 pathway. Nat Med. 2011;17(1):64–70.

    Article  PubMed  CAS  Google Scholar 

  199. Qin Z, Wang P-Y, Su D-F, Liu X. miRNA-124 in immune system and immune disorders. Front Immunol. 2016;7:406.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Mehmood R, Jibiki K, Shibazaki N, Yasuhara N. Molecular profiling of nucleocytoplasmic transport factor genes in breast cancer. Heliyon. 2021;7(1): e06039.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Liang P, Zhang H, Wang G, Li S, Cong S, Luo Y, et al. KPNB1, XPO7 and IPO8 mediate the translocation of NF-κB/p65 into the Nucleus. Traffic. 2013;14(11):1132–43.

    Article  PubMed  CAS  Google Scholar 

  202. Frieman M, Yount B, Heise M, Kopecky-Bromberg SA, Palese P, Baric RS. Severe acute respiratory syndrome coronavirus ORF6 antagonizes STAT1 function by sequestering nuclear import factors on the rough endoplasmic reticulum/Golgi membrane. J Virol. 2007;81(18):9812–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. De Re V, Caggiari L, De Zorzi M, Fanotto V, Miolo G, Puglisi F, et al. Epstein-Barr virus BART microRNAs in EBV- associated Hodgkin lymphoma and gastric cancer. Infect Agents cancer. 2020;15:42.

    Article  Google Scholar 

  204. Nahand JS, Rabiei N, Fathazam R, Taghizadieh M, Ebrahimi MS, Mahjoubin-Tehran M, et al. Oncogenic viruses and chemoresistance: what do we know? Pharmacol Res. 2021;170: 105730.

    Article  Google Scholar 

  205. Mostafaei S, Vahidi Manesh P, Sadri Nahand J, Nesaei A, Sorayyayi S, Abasabadi F, et al. The role of Epstein-Barr virus-expressed genes in breast cancer development. Breast J. 2020;26(11):2323–6.

  206. He B, Li W, Wu Y, Wei F, Gong Z, Bo H, et al. Epstein-Barr virus-encoded miR-BART6-3p inhibits cancer cell metastasis and invasion by targeting long non-coding RNA LOC553103. Cell Death Dis. 2016;7(9):e2353–e.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Liang Q, Yao X, Tang S, Zhang J, Yau TO, Li X, et al. Integrative identification of Epstein–Barr virus–associated mutations and epigenetic alterations in gastric cancer. Gastroenterology. 2014;147(6):1350–62 e4.

    Article  PubMed  CAS  Google Scholar 

  208. Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7(24):36783.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Zebardast A, Tehrani SS, Latifi T, Sadeghi F. Critical review of Epstein–Barr virus microRNAs relation with EBV-associated gastric cancer. J Cell Physiol. 2021;236(9):6136–53.

    Article  PubMed  CAS  Google Scholar 

  210. Taylor TJ, Brockman MA, McNamee EE, Knipe DM. Herpes simplex virus. Front Bioscience-Landmark. 2002;7(4):752–64.

    Article  Google Scholar 

  211. Umbach JL, Kramer MF, Jurak I, Karnowski HW, Coen DM, Cullen BR. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature. 2008;454(7205):780–3.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  212. Chaturvedi S, Engel R, Weinberger L. The HSV-1 ICP4 Transcriptional Auto-Repression Circuit Functions as a Transcriptional Accelerator Circuit. Front Cell Infect Microbiol. 2020;10:265.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Heinrich PC, Behrmann I, Müller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J. 1998;334(2):297–314.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  214. Jurak I, Kramer MF, Mellor JC, Van Lint AL, Roth FP, Knipe DM, et al. Numerous conserved and divergent microRNAs expressed by herpes simplex viruses 1 and 2. J Virol. 2010;84(9):4659–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  215. Smith C, Bates P, Rivera-Gonzalez R, Gu B, DeLuca N. ICP4, the major transcriptional regulatory protein of herpes simplex virus type 1, forms a tripartite complex with TATA-binding protein and TFIIB. J Virol. 1993;67(8):4676–87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. DeLuca NA, Schaffer PA. Activation of immediate-early, early, and late promoters by temperature-sensitive and wild-type forms of herpes simplex virus type 1 protein ICP4. Mol Cell Biol. 1985;5(8):1997–2008.

    PubMed  PubMed Central  CAS  Google Scholar 

  217. Paludan SR. Requirements for the induction of interleukin-6 by herpes simplex virus-infected leukocytes. J Virol. 2001;75(17):8008–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. Dittmer DP, Damania B. Kaposi sarcoma–associated herpesvirus: immunobiology, oncogenesis, and therapy. J Clin Investig. 2016;126(9):3165–75.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Giffin L, Damania B. KSHV: pathways to tumorigenesis and persistent infection. Adv Virus Res. 2014;88:111–59.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Cirone M, Lucania G, Aleandri S, Borgia G, Trivedi P, Cuomo L, et al. Suppression of dendritic cell differentiation through cytokines released by primary effusion lymphoma cells. Immunol Lett. 2008;120(1–2):37–41.

    Article  PubMed  CAS  Google Scholar 

  221. Jones KD, Aoki Y, Chang Y, Moore PS, Yarchoan R, Tosato G. Involvement of interleukin-10 (IL-10) and viral IL-6 in the spontaneous growth of Kaposi’s sarcoma herpesvirus-associated infected primary effusion lymphoma cells. Blood J Am Soc Hematol. 1999;94(8):2871–9.

    CAS  Google Scholar 

  222. Chung Y-H, Means RE, Choi J-K, Lee B-S, Jung JU. Kaposi’s sarcoma-associated herpesvirus OX2 glycoprotein activates myeloid-lineage cells to induce inflammatory cytokine production. J Virol. 2002;76(10):4688–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  223. Qin J, Li W, Gao S-J, Lu C. KSHV microRNAs: tricks of the devil. Trends Microbiol. 2017;25(8):648–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The authors declare that no funds, grants, or other supports were received during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

H.M. and J.S.N. contributed to conception, design, and drafting of the manuscript. H.B.B, M.B, P, M, S.M.A.A, M.H.L, M.Y.M, S.P.T, F.Z, M.R.H and SMR.H contributed in data collection and manuscript drafting. All authors approved the final version of the manuscript for submission.

Corresponding authors

Correspondence to Javid Sadri Nahand or Hamed Mirzaei.

Ethics declarations

Ethical approval and consent to participate

This article does not contain any studies with human participants or animals performed by any of the authors.

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bannazadeh Baghi, H., Bayat, M., Mehrasa, P. et al. Regulatory role of microRNAs in virus-mediated inflammation. J Inflamm 21, 43 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12950-024-00417-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12950-024-00417-7

Keywords