Next Article in Journal
Genomics of Wolfram Syndrome 1 (WFS1)
Previous Article in Journal
In Search of Disentanglement in Tandem Mass Spectrometry Datasets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs in the Regulation of RIG-I-like Receptor Signaling Pathway: Possible Strategy for Viral Infection and Cancer

1
Department of Immunology, Zunyi Medical University, Zunyi 563002, China
2
School of Pharmacy, Zunyi Medical University, Zunyi 563002, China
3
Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563002, China
4
Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563002, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2023, 13(9), 1344; https://doi.org/10.3390/biom13091344
Submission received: 6 August 2023 / Revised: 30 August 2023 / Accepted: 30 August 2023 / Published: 4 September 2023

Abstract

:
The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) play a crucial role as pattern-recognition receptors within the innate immune system. These receptors, present in various cell and tissue types, serve as essential sensors for viral infections, enhancing the immune system’s capacity to combat infections through the induction of type I interferons (IFN-I) and inflammatory cytokines. RLRs are involved in a variety of physiological and pathological processes, including viral infections, autoimmune disorders, and cancer. An increasing body of research has examined the possibility of RLRs or microRNAs as therapeutic targets for antiviral infections and malignancies, despite the fact that few studies have focused on the regulatory function of microRNAs on RLR signaling. Consequently, our main emphasis in this review is on elucidating the role of microRNAs in modulating the signaling pathways of RLRs in the context of cancer and viral infections. The aim is to establish a robust knowledge base that can serve as a basis for future comprehensive investigations into the interplay between microRNAs and RIG-I, while also facilitating the advancement of therapeutic drug development.

1. Introduction

1.1. RLR Signaling

1.1.1. The Structural Characterization of RLRs

RLRs are classified within the DExD/H-box RNA helicase family and encompass three distinct members, namely RIG-I, melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2), which are encoded by the genes DDX58, IFIH1, and DHX58, respectively. These three proteins exhibit 30% similarity in their amino acid sequences, particularly in their key domains (RNA helicase, pincer, and CTD domains) [1]. In general, the central helicase domain and the C-terminal domain of these RLRs are responsible for their RNA recognition capability [2]. The N-terminal region of RIG-I and MDA5 contains a tandem caspase recruitment domain (CARD) (Figure 1). This domain is important for the interaction of these proteins with the adaptor molecule, named the mitochondrial antiviral signaling protein (MAVS; also referred to as VISA, Cardiff, and IPS-I). The MAVS protein possesses a solitary CARD domain at its N-terminal region, which interacts with the N-terminal CARD domain of RIG-I. This interaction prompts the assembly of prion-like aggregates of MAVS proteins, subsequently leading to the activation of the transcription factors nuclear factor kappa B (NF-κB) and interferon regulatory factor 3 (IRF3) [3]. LGP2 is mostly similar to RIG-I and MDA5, with the only difference being that it lacks the N-terminal CARD.

1.1.2. The Recognition of RNAs by RLRs

RIG-I primarily detects two main types of RNA: single-stranded RNAs (ssRNAs) with a triphosphate group (PPP) at their 5′ end, and short double-stranded RNAs (dsRNAs) (<300 bp) [4,5,6,7,8]. Conversely, MDA5 predominantly recognizes long dsRNAs (>1000 bp) [9,10]. Unlike RIG-I and MDA5, LGP2 lacks the CARD structural domain, rendering it unable to recognize RNA. However, studies have demonstrated that LGP2 could be involved in coordinating RIG-I and MDA5 [11,12]. Emerging investigations have shed light on the involvement of LGP2 in the recognition of viral RNA [13,14] and its significant role in self-RNA sensing and RNA silencing [14,15]. Moreover, studies have indicated that RNase L-mediated degradation of parainfluenza viral products preferentially activates MDA5 [4]. Notably, the nucleocapsid proteins of paramyxoviruses effectively shield the viral genomic RNA, safeguarding it from RNase digestion and host proteins like MDA5 [16]. Consequently, the lack of dsRNA detection in paramyxovirus infections by dsRNA recognition antibodies is unsurprising [17]. Furthermore, because the nascent genomic RNA is encapsulated, the dsRNA segment within the replication/transcription complex is limited (>2 kb is regarded as an MDA5 trigger). Additionally, coronaviruses have been found to evade detection by MDA5 through 2′-O-methylation of mRNA [18]. However, it is evident that future advancements should take into account higher-order RNA structure, sequence, and modifications, in addition to length. Notably, recent discoveries regarding the biological function of RIG-I in various cellular activities confirm its ability to interact with different endogenous RNAs (e.g., microRNA [19], panhandle RNA [20], circRNA [21], and snRNA [22]). Given that the majority of viral RNA possesses a 5′-PPP group, the RIG-I receptor primarily recognizes and distinguishes viral infections [23].

1.1.3. The RLR Signal Transduction

Under normal circumstances, RIG-I/MDA5 remains in an inactive and inhibited state within the cytoplasm [24,25]. However, during viral infection, RIG-I/MDA5 recognizes viral RNA, resulting in conformational changes within the RIG-I/MDA5. These conformational changes facilitate the interaction between RIG-I/MDA5 and MAVS on the surface of mitochondria [26]. MAVS dimerization then serves as a scaffold to assemble signaling complexes at the outer mitochondrial membrane, attracting molecules such as tumor necrosis factor receptor-associated factor 6 (TRAF6). Upon recruitment to the receptor-associated interleukin-1 receptor-associated kinase 1 (IRAK1)-IRAK4-MyD88 (myeloid differentiation primary response 88) adaptor protein complex, TRAF6 undergoes lysine 63 (K63)-linked polyubiquitination. This event promotes the recruitment of downstream regulators like transforming growth factor-beta-activated kinase 1 (TAK1) and TAK1-binding protein 2/3 (TAB2/3), which in turn activate NF-κB, leading to the production of inflammatory cytokines [27].
In parallel, linker proteins TRAF family-member-associated NF-κB activator (TANK), nucleosome assembly protein 1 (NAP1), and, similar to NAP1, TBK1 adaptor (SINTBAD), are recruited by the TRAF3 bound to MAVS. TANK binding to TANK-binding kinase 1 (TBK1), which is linked to upstream RLR signaling through TANK, leads to the phosphorylation of IRF3. Phosphorylated IRF3 (p-IRF3) dimerizes and translocates to the nucleus to initiate the transcription of the IFN-β gene [28,29]. This process triggers the expression of antiviral genes; increases the production of pro-inflammatory cytokines and IFN-I [30,31]; and ultimately collaborates with IFNs, inflammatory cytokines, and other antiviral products to exert antiviral effects. Conversely, IFN-I can also provide feedback modulation to the RIG-I signaling pathway. For instance, the IFN-induced long noncoding RNA, lnc-Lsm3b, competes with viral RNA for binding to RIG-I monomers and serves as a feedback mechanism to deactivate RIG-I’s innate activity at a later stage of the innate immune response [32].

1.1.4. Regulation of RLR Signaling by LGP2 and E3 Ubiquitin Ligases

While LGP2 lacks the ability to directly recognize RNA and activate downstream pathways, it plays a crucial role in the regulation of RLR signaling. LGP2 exerts its inhibitory effects on the RIG-I-mediated signaling pathway through various mechanisms. Firstly, the C-terminal RD domain of LGP2 forms a heterodimeric complex with the RD domain of RIG-I, hindering RIG-I multimerization and thus impeding its recognition of viral dsRNAs. Also, LGP2 can hinder the interaction between RIG-I and MAVS, attenuating RIG-I signaling. Furthermore, LGP2 can also dampen RIG-I signaling by inhibiting RIG-I’s binding to MAVS [33]. Moreover, LGP2 demonstrates the ability to inhibit the activation of IRF3, a downstream transcription factor of RIG-I, thereby potentially exerting a negative regulatory effect [34].
Earlier studies have identified several E3 ubiquitin ligases, including Riplet, tripartite motif-containing protein 25 (TRIM25), and ring finger protein 123 (RNF123), which modulate RIG-I activity [35,36]. Initially, TRIM25 was believed to be crucial for the ubiquitination of RIG-I [37]. However, recent investigations have established that Riplet, rather than TRIM25, is responsible for RIG-I ubiquitination. To further comprehend the specific recognition of RIG-I by Riplet and determine the E3 ligase responsible for MDA5 recognition, future studies are warranted [38,39]. In addition to the regulation of RLR signaling through LGP2 and E3 ubiquitin ligases, increasing evidence confirms that microRNAs are also involved in the regulation of RLR signaling [19]. In recent years, there has been a notable surge in research interest dedicated to exploring the regulation of the RIG-I signaling system.

1.2. MicroRNAs

The short-length, noncoding RNA molecules referred to as microRNAs (miRNAs) occur in a size range of 19–25 nucleotides and are reported to regulate the post-transcriptional silencing of target genes. A single miRNA can target hundreds of mRNAs simultaneously, thereby affecting the expression of numerous genes that are frequently involved in a functionally interconnected pathway [40]. The majority of miRNAs exist in the form of primary miRNAs (pri-miRNAs), which are transcribed from DNA sequences and typically range in length from approximately 300 to 1000 nucleotides. The processing of pri-miRNAs generates precursor miRNAs (pre-miRNAs), which are approximately 70 to 90 nt in length and are considered the precursor of microRNA. After being cleaved by the Dicer enzyme, the pre-miRNA is re-digested to generate mature miRNA of approximately 19~25 nt in length [41,42,43] (Figure 2).
Two miRNAs named lin-4 and let-7 have been verified in previous studies to be partially complementary to the 3′ region of several messenger RNA (mRNA) targets of interest. The noncoding regions (3′-UTRs) are responsible for the inhibition of protein translation, thereby inhibiting protein synthesis ultimately [44]. MiR-125b, a human counterpart of lin-4, along with let-7, are closely linked to cancer development and hold promise as potential diagnostic and prognostic markers [45,46]. Additionally, microRNAs have been found to play significant roles in the pathogenesis of cardiovascular diseases (CVDs) [47,48], cancer initiation and progression [49,50,51], diabetes [52,53], Alzheimer’s disease [54,55], and inflammation [56,57]. It is worth noting that cellular miRNAs can serve as biomarkers for the diagnosis and monitoring of COVID-19 [58,59,60,61]. Therefore, microRNAs are considered novel means of diagnosis, therapy, and prognosis in the context of the above-stated diseases.

2. Regulation of RLR Signaling by microRNAs

2.1. The Regulation of RLR Signaling by microRNAs during Viral Infection

2.1.1. MicroRNAs Targeting RIG-I/MDA5 during Viral Infection

RIG-I and MDA5, which are pivotal upstream molecules of the RLR signaling pathway, play crucial roles in response to viral infections and relay signals downstream. Activation of the RLR signaling pathway triggers the production of IFN-I. The antiviral effects of IFNs mainly involve the following major pathways: I. Phosphorylation of IFN-induced proteins and RNA degradation facilitated by the protein kinase R (PKR) kinase and 2′-5′-oligoadenylate-dependent RNase L; II. RNA editing mediated by IFN-induced RNA-specific adenosine deaminase (ADAR1) and Mx protein GTPase; III. Induction of inducible nitric oxide synthase (iNOS2) and major histocompatibility complex class I and II proteins [62]. Furthermore, the activation of RLR signaling leads to IFN-dependent and non-IFN-dependent apoptosis of target cells, which is crucial for inhibiting viral replication [63,64]. MicroRNAs that target RIG-I/MDA5 can modulate RLR signaling and thereby impact viral replication. Therefore, in this section, we provide a comprehensive summary of the role of microRNAs in viral infections through their targeting of RIG-I/MDA5 to regulate RLR signaling.
While several proteins have been reported to modulate the RLR pathway and subsequent IFN signal transduction [65,66], studies on the regulation of this pathway by microRNAs remain limited. The RLR signaling pathway may give rise to certain microRNAs, such as miR-146a and miR-4661 [67,68], which in turn regulate the expression or function of various components of the RLR signaling pathway and IFN-I through a feedback mechanism. Additionally, RIG-I directly regulates the replication of vesicular stomatitis virus (VSV) in conjunction with miR-582-3p [69]. Furthermore, miR-92a has been found to diminish the VSV-induced production of IFN-I and promote viral replication in macrophages by targeting RIG-I [70]. Notably, downregulation of miR-340-5p has been reported in A549 cells infected with influenza A virus (IAV), and it reduces viral replication by targeting RIG-I and 2′-5′-oligoadenylate synthetase 2 (OAS2) [71].
Furthermore, miR-136 serves as a RIG-I immune agonist [72]. In general, miR-136 exhibits two functions in regulating host innate immunity against viruses. Another study reported that miR-4423-3p promoted the replication of the hepatitis C virus (HCV) by inhibiting the activation of the IFN pathway via RIG-I targeting [73]. However, whether miR-4423-3p blocks downstream molecules due to RIG-I inhibition only, or whether other mechanisms are also involved, remains to be investigated. Correspondingly, in response to viral infection, the host produces miR-485, which targets and destroys the RIG-I mRNA, decelerating the antiviral response and accelerating viral multiplication. Consequently, the host suppresses the expression of miR-485, which leads to markedly reduced Newcastle disease virus (NDV) and H5N1 influenza virus multiplication in mammalian cells. Unexpectedly, miR-485 also inhibits the replication of the H5N1 virus by binding to the RNA polymerase encoded by the H5N1 gene polymerase basic protein 1 (PB1) in a sequence-specific manner. In addition, miR-485 demonstrates dual selectivity: during elevated levels of the H5N1 virus, it targets PB1 in the cells, while during low levels of the virus, it targets RIG-I in the cells [74]. Therefore, miR-485 plays a dual role in preventing inappropriate antiviral signaling activation and reducing influenza virus infection.
Studies have reported that miR-146a holds potential as a novel biomarker for distinguishing between the acute and post-acute phases of COVID-19 [75]. However, a study reported that the hepatitis B virus (HBV) worsened the HBV infection by causing miR-146a to target RIG-I and RIG-G (retinoic acid-inducible gene G), thereby facilitating the negative regulation of the RIG-I signaling pathway [76]. Therefore, it is important to explore whether SARS-CoV-2 and other viruses are capable of immune escape via miR-146a.
Nevertheless, a fascinating investigation revealed that the Epstein–Barr virus (EBV)-encoded miR-BART6-3p, which targets RIG-I, promoted viral infection and was linked to the immune surveillance evasion of EBV [77]. Another correlational study identified miR-218 as a novel virus-induced miRNA that interfered with RIG-I expression and impaired the production of interferons to promote viral immune escape [78]. These studies suggest that virus-encoded microRNAs may interact with the host genes to influence the host’s ability to combat viruses, although the precise underlying mechanism remains unknown to date.
On the other hand, the overexpression of miR-34b-5p inhibited the MDA5 signaling pathway but not the RIG-I signaling pathway, thereby promoting the proliferation of the avian leukosis virus subgroup J (ALV-J)-infected DF-1 cells and facilitating the replication of ALV-J [79]. Recent findings indicate that microRNAs may play a role in regulating RLR signaling in the miiuy croaker. For instance, miR-203 was ascertained to target the 3′-UTR of MDA5, which then activated the downstream genes IRF3/7 and ultimately promoted the production of IFN-I and inflammatory cytokines and prevented the viral infection [80].

2.1.2. Targeting Other Genes of RLR Signaling

Apart from targeting RIG-I/MDA5 to modulate RLR signaling during viral infections, microRNAs also exhibit similar effects by targeting other molecules within the RLR signaling pathway. For example, extensive research has highlighted the functional involvement of miR-33 in cellular metabolism, particularly in the maintenance of cholesterol homeostasis [81,82]. However, a study confirmed that overexpression of miR-33 impairs the RIG-I signaling pathway, strengthens the VSV viral load and lethality, and decreases the production of IFN-I in vitro and in vivo. This is achieved by targeting AMP-activated protein kinase (AMPK) [83]. Interestingly, cyprinid herpesvirus 3 (CyHV-3) infection reportedly induced another microRNA named miR-155, which targets AMPK. AMPK plays a crucial role in impeding the formation of activation aggregates by MAVS, consequently inhibiting the autophagy-mediated elimination of damaged mitochondria, and disrupting mitochondrial homeostasis. This disruption hinders the effective activation of MAVS, leading to a reduction in interferon expression. The research demonstrated that miR-155 exhibited anti-CyHV-3 activity via modulating the AMPK-MAVS-IFN axis and could, therefore, be utilized in studies focused on designing novel anti-CyHV-3 drugs [84].
Similar to miR-146a, let-7b, a microRNA, has been implicated in various conditions, including inflammation [85,86], cancers [87,88], and IgA nephropathy [89,90]. However, a recent study has revealed that let-7b can interfere with HCV infection by targeting the IκB kinase complex 1 (IKK1 or IKKα), leading to enhanced phosphorylation and nuclear translocation of IRF3 mediated by RIG-I. This ultimately results in the expression of IFN-β [91]. These findings suggest that let-7b plays a significant role in multiple disorders and could serve as a promising therapeutic target for the treatment of these diseases. In the context of IAV-infected A549 cells, the host protein contactin-1 (CNTN1) promotes viral replication by directly interacting with miR-200c, thereby inhibiting RIG-I-MAVS-mediated interferon signaling [92]. Rhabdovirus infection substantially upregulated the expression of host miR-3570 in Miichthys miiuy macrophages, and the induced miR-3570 promoted viral replication by targeting and downregulating MAVS [93]. Moreover, RIG-I/TBK1 signaling activation was induced in macrophages against HBV by the hepatic exosomes with decreased levels of miR-27b-3p. The expression level of miR-27-3p in serum exosomes shows promise as a potential biomarker for patients with chronic hepatitis B (CHB) [94]. Moreover, enterovirus infection leads to the downregulation of miR-526a, which targets cylindromatosis (CYLD) and suppresses the RIG-I-dependent innate immune response [95].
A separate investigation demonstrated that the upregulation of miRNA-146a in macrophages infected with VSV was dependent on both the RIG-I/NF-kB pathways. This miR-146a encourages VSV replication by targeting IRAK1, IRAK2, and TRAF6 and by preventing the release of IFN-I that is activated by the virus [67]. On the other hand, the hepatitis A virus (HAV) partly interferes with RIG-I/MDA5-mediated IFN-I signaling by targeting the vital adaptor molecule TRAF6 through miR-146a-5p [96]. The afore-stated study demonstrated that viruses might induce the production of miR-146a to evade host immune responses. Li et al. reported that miR-9-5p inhibits the replication of enterovirus 71 (EV71) by targeting the NF-κB-mediated innate immune response [97].
Notably, miR-30a inhibits TRIM25 expression and TRIM25-mediated RIG-I ubiquitination to enhance the replication of coxsackievirus B3 (CVB3) [98]. In addition, miR-202-5p was also reported to target the TRIM25-mediated ubiquitination of RIG-I and promote red spotted grouper nervous necrosis virus (RGNNV) infection in zebrafish [99]. Furthermore, TRIM25 is involved in the regulation of MDA5 expression. MiR-30a and miR-202-5p target TRIM25 and can activate both RIG-I and MDA5, thereby augmenting the antiviral response.
The miR-302/367 cluster is one of the most studied microRNAs and consists of five members: miR-367, miR-302a, miR-302b, miR-302c, and miR-302d. The miR-302/367 cluster is widely involved in cellular differentiation and development, tumor progression, immune regulation, and other biological processes [100,101]. MiR-302b, as a member of this family, has been shown to control MAVS-mediated antiviral innate immunity by targeting mutations in the solute carrier family 25, member 12 (SLC25A12) transporter protein to regulate the mitochondrial metabolism [102]. MiR-302c mediates IAV-induced IFN-β expression by targeting NF-κB [103]. In addition, miR-302a, another member of the miR-302/367 cluster, inhibits IRF5 expression by directly targeting the IRF5 3′-UTR, leading to the overexpression of inflammatory cytokines and chemokines [104]. Thus, the miR-302/367 cluster could serve as a potential regulator of virus-induced cytokine storms and provide a candidate target for the treatment of viral infections.
The above-stated findings provide fresh insights into how microRNAs are involved in the regulation of RLR signaling. Therefore, this field of research is of great importance in the development of novel antiviral therapies in the future.

2.1.3. The Role of microRNAs and RLRs in COVID-19 Sensing

The infection of COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), involves the activation of the host immune response through the recognition of the viral RNA by RIG-I/MDA5. Consequently, our research focuses on investigating the involvement of RLR signaling and microRNAs in the infection process. The two cytoplasmic viral RNA sensors, RIG-I and MDA5, are vital for the immune escape of SARS-CoV-2. Several coronavirus-encoded proteins have been confirmed to inhibit the IFN response, although the precise mechanism remains to be elucidated [105,106]. Nonetheless, the SARS-CoV-2 membrane (M) protein might bind RIG-I/MDA5 and limit the production of IFN-I and IFN-III, which reveals a role for this protein in antagonizing IFN-mediated innate viral immunity and viral replication [107]. The major protease (Mpro) of SARS-CoV-2, known as NSP5, has garnered attention as a potential therapeutic target for COVID-19 treatment. NSP5 exhibits unique cleavage specificity, as it not only cleaves lengthy viral polypeptides but also targets nucleotide-binding oligomerization-like receptor protein 12 (NLRP12) and TAB1. This distinct cleavage profile differentiates NSP5 from known human proteases [108]. During SARS-CoV-2 infection, the nucleoprotein (N protein) is the most abundant viral protein. It consists of an N-terminal structural domain (NTD) and a C-terminal structural domain (CTD). These domains are primarily responsible for RNA binding and are crucial for assembling the viral genome into viral particles [109]. Attractively, the SARS-CoV-2 NSP5 and N protein reduce the formation of antiviral stress granules (avSGs). NSP5 also disrupts RIG-I signaling by destroying the RIG-I-MAVS complex, thereby reducing the antiviral response [110]. The above results indicate that RLR signaling (particularly RIG-I and MDA5) is the main pathway through which SARS-CoV-2 escapes the innate immune response of the host.
Previous studies have extensively documented the importance of microRNAs in viral infections. Two key aspects have been elucidated: Firstly, host microRNAs enhance the immune response, thereby impacting viral replication. Secondly, viral microRNAs have been demonstrated to target host immune cells, impeding viral clearance [111,112]. Therefore, the identification of microRNAs that are strongly correlated with SARS-CoV-2 may provide fresh insights into the diagnosis and prognosis of COVID-19, and could be used as a feasible strategy to develop novel targeted therapies. The upregulation of miR-146a and miR-155 in the oral cavities of patients with diabetes and periodontitis, as reported by Jelena R. Roganovic, has been suggested to potentially enhance the expression of angiotensin-converting enzyme 2 (ACE2) and alter the host’s antiviral response to the SARS-CoV-2 virus [113]. Nevertheless, further verification is required to confirm this hypothesis.
In a recent study, a correlation was observed between the upregulation of hsa_circ_0000479, RIG-I, and IL-6 and the downregulation of has-miR-149-5p, suggesting that the has-circ-0000479/hsa-miR-149-5p/RIG-I/IL-6 axis may be involved in regulating the immune response to SARS-CoV-2 [114]. As a whole, the findings imply that overexpression of hsa_circ_0000479 may affect the expression of IL-6 and RIG-I via sponging hsa-miR-149-5p in COVID-19. Sponging refers to the phenomenon where circular RNAs (circRNAs), which possess multiple miRNA binding sites, act as competitive endogenous RNAs (ceRNAs) by specifically sequestering miRNAs. This process can be likened to a sponge absorbing water, as circRNAs “isolate” miRNAs from their target mRNAs, thus impacting the negative regulation of target mRNAs by miRNAs [115,116,117]. The regulation of the expression of IL-6 and RIG-I, which are two genes essential for the immunological response to COVID-19 infection, could impact the severity of COVID-19 symptoms (Figure 3). Therefore, the hsa-circ-0000479/hsa-miR-149-5p/IL-6 RIG-I axis could serve as a promising therapeutic target in the treatment of COVID-19.
Furthermore, several other microRNAs have been implicated in relation to COVID-19, including miR-1207-5p [118], miR-320 [119], miR-200c-3p [120], miR-146a [121], miR-421-5p [122], and let-7b [123], among others. Importantly, microRNAs encoded by SARS-CoV-2 have the potential to target host cell mRNAs involved in viral replication, thereby facilitating viral replication and regulating host gene expression [124,125,126]. The elucidation of the detailed mechanism underlying this effect may facilitate the development of microRNA attenuated vaccines against COVID-19.

2.2. Regulation of RLR Signaling by MicroRNAs in Cancer

MicroRNAs in RLR signaling are also involved in regulating cancer development. Once activated, the RIG-I pathway will induce target cell death through a variety of pathways, including endogenous apoptosis, exogenous apoptosis, pyroptosis, and autophagy [127,128,129]. MicroRNAs have been established to modulate tumor progression through the regulation of RLR signaling. Notably, microRNA-545 has been demonstrated to exert significant effects in various cancers, including breast cancer [130], bladder cancer [131], ovarian cancer [132], cervical cancer [133], and other malignancies [134,135,136]. However, further investigation demonstrated that the overexpression or knockdown of miR-545 could promote or inhibit cancer cell proliferation by targeting RIG-I [137,138,139]. Moreover, the inhibition of PTEN (phosphatase and tensin homolog), a tumor suppressor gene frequently mutated, deleted, and functionally inactivated in human cancers, along with RIG-I, has been shown to enhance the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway. This regulation involves the intronic microRNAs miR-374b and miR-545, which are modulated by the intronic miR-421 [140]. Therefore, it is inferred that the various loci that regulate the functional components of tumor progression are relatable. However, little research has been conducted in the area, and the underlying mechanisms also remain unknown so far; therefore, it is necessary to explore novel investigations in this aspect of cancer treatment and prognosis research.

2.3. Others

Except for the above two aspects, studies have also reported that certain microRNAs stimulate the RIG-I signaling pathway to affect IFN-I production. For instance, miR-139 promotes IFN secretion in prostate cancer cells by acting as an immunological agonist of RIG-I [141]. Moreover, miR-1248 was reported to activate IFN-β through a direct association with RIG-I and AGO2 (argonaute 2) [142]. Remarkably, miR-145 was transfected into human mesenchymal stem cells (hBM-MSCs) and human articular chondrocytes (hSCs), where it could induce immune responses by targeting RIG-I [143].
Similarly, studies have revealed that miR-145-5p, miR-122, and miR-489 can target MDA5, DAK, and TRAF6, respectively, in the miiuy croaker to modulate the RLR signaling pathway upon poly(I:C) stimulation [144,145,146]. These findings offer valuable insights into the regulatory role of microRNAs in the immune response of fish. A comprehensive summary of the microRNAs involved in the regulation of the RIG-I signaling pathway, as discussed in the preceding sections, can be found in Figure 4 and Table 1.

3. Conclusions and Prospects

The present review summarizes several roles that microRNAs play in the regulation of various physiological processes, such as cell division, apoptosis, etc. During recent years, numerous microRNAs have been discovered through advancements in high-throughput sequencing technology. Moreover, the regulatory functions of microRNAs have been receiving greater attention all over the world. Innate antiviral immune responses are mainly mediated by RLRs, particularly RIG-I/MDA5. Modulating RLR signaling through noncoding RNAs presents a promising approach for optimizing the immune response against viral infections and inhibiting tumor progression. The regulatory role of miRNAs in RLR signaling primarily involves binding to the 3′ UTR of the mRNAs associated with RLR signaling, thereby modulating the mRNA levels of target genes and subsequently influencing protein expression. This, in turn, enhances or suppresses RLR signaling, leading to the secretion of IFNs and inflammatory cytokines that impact viral replication and cancer progression. While previous studies have demonstrated the regulatory impact of microRNAs on RLR signaling, further investigation is required to elucidate the intricacies of their interactions.
The “energy factories” within the cell, mitochondria are essential organelles in the metabolic processes of the cell. Studies have demonstrated that the mitochondrial route may be used by microRNAs to regulate cellular processes. For instance, in adipose tissue, miR-494-3p directly reduces PGC1 expression and the ensuing mitochondrial biogenesis [147]. MiR-125 promotes the demise of cancer cells via regulating mitochondrial dynamics and metabolism [148]. Furthermore, overexpression of miR-25 dramatically decreased the levels of mitochondrial calcium uniporter (MCU) expression, hindered mitochondrial Ca2+ uptake, and prevented apoptosis in cancer cells [149]. The control of mitochondrial calcium homeostasis, mitochondrial metabolism, and mitochondrial biogenesis are all critical processes that are impacted by microRNAs. Targeting the mitochondrial pathway may be an effective potential strategy for disease treatment.
RIG-I/MDA5 induces antitumor immune responses through the IFN-dependent activation of effector T cells. On the other hand, RIG-I/MDA5 also induces cancer cell apoptosis in an IFN-independent manner. Therefore, targeting RIG-I/MDA5 has several advantages over conventional cancer therapies. First, RIG-I/MDA5 may be activated using the corresponding ligands and, therefore, significant tumor-cell-specific apoptosis may be induced. Second, the expression of RIG-I/MDA5 is also associated with the prognosis of certain cancers. For instance, liver cancer patients with low RIG-I expression have a poor prognosis [150], while ovarian cancer patients with high RIG-I expression have a similar outcome [151]. Although several RIG-I/MDA5 ligands have been employed in clinical trials, challenges are encountered when attempting to target RIG-I/MDA5 for cancer treatment. For instance, one challenge is to further optimize the methods to be used for enabling these ligands to reach the site of the cancer. Another challenge is the difficulty of controlling the dosage of the ligand. While a high dosage of the ligand that activates RIG-I/MDA5 may kill the cancer cells, low doses of the ligand may result in low levels of interferons and pro-inflammatory cytokines, which could accelerate cancer growth. Moreover, the activation of RIG-I/MDA5 could activate the NF-κB signaling pathway, thereby aggravating inflammation. Activation of RIG-I/MDA5 through endogenous pathways, such as microRNAs and long noncoding RNAs (lncRNAs), might enable achieving better effects and could, therefore, serve as a novel strategy for cancer treatment. Unfortunately, studies so far have not reported any microRNAs that regulate LGP2, another member of the RLR family.
The emergence of drug resistance to anticancer medications is a major obstacle to successful treatment. MicroRNAs have gained attention due to their ability to regulate genes involved in the cellular responses to drugs. Several miRNAs have been identified to sensitize cancer cells to therapy and serve as biomarkers of drug resistance. For example, a study investigating the co-delivery of miR-345 and gemcitabine in pancreatic cancer demonstrated superior outcomes compared with treatment with either miR-345 or gemcitabine alone [152]. Similar positive results were observed when combining doxorubicin with miR-34a for breast cancer treatment [153]. Therefore, the combination of RLR agonists and microRNAs in tumor immunotherapy may significantly enhance treatment effectiveness, although further investigation is warranted.
Moreover, microRNAs and RLRs also have distinct roles in SARS-CoV-2 infection. While microRNAs such as miR-146a, let-7b, and miR-200c have been implicated in mediating RLR signaling during SARS-CoV-2 infection, it remains to be empirically validated whether these alterations in RLR signaling impact the infection itself.
MicroRNA-based therapies are promising. However, how to deliver microRNAs safely and effectively to target organs and target cells is the biggest challenge at present. Currently, the following major delivery modes have been developed, which may help miRNA therapeutics to move toward clinical application: (I) conjugation [154]; (II) virus-associated delivery [155]; (III) nanoparticles [156]; and (IV) exosome-associated delivery [157]. These delivery techniques are now being employed in therapies that have received clinical approval or are undergoing clinical studies. Due to their low cytotoxicity, low antigenicity, and capacity to avoid phagocytosis and the endocytosis route, exosomes may be the best delivery method of these [158,159]. However, dosage continues to be a key restriction in the area of miRNA therapy. Clinical trials have seldom been able to pinpoint the precise dose at which RNA treatment is successfully delivered to the cell type of interest.
In conclusion, the immune system utilizes various miRNAs to finely regulate its functional capacity, maintaining a delicate balance between activation and inhibition. However, the precise regulatory mechanisms of miRNAs in the RIG-I response are not fully understood, particularly how these miRNA networks collaboratively optimize the immune response. A deeper understanding of the miRNA-mediated regulation of RIG-I signaling will uncover novel therapeutic targets for inflammatory diseases and cancer.

Author Contributions

J.Z. (Jidong Zhang), Y.H. and T.S. contributed to the conception of the review; D.C. wrote the manuscript; Q.J., J.L. and F.C. performed manuscript preparation and contributed collections; J.Z. (Jishan Zheng) and Y.M. helped perform constructive discussions. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the College Students’ Innovation and Entrepreneurship Training Program of Zunyi Medical University (No. S202210661234, ZYDC202202147); the Health Commission of Guizhou Province, China (No. GZWKJ-2021-533); the National Nature Science Foundation of China (NSFC) (No. 31960156, 31660338); the Collaborative Innovation Center of the Chinese Ministry of Education (2020-39); the Guizhou University Dendrobium nobile industry development key technology engineering research center (No. QKJ-2022-048), and the Guizhou Provincial Department of Education “four new” and “four modernizations” science and technology research project (No. QJJ-2022-006).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef] [PubMed]
  2. Yoneyama, M.; Kikuchi, M.; Matsumoto, K.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Foy, E.; Loo, Y.-M.; Gale, M., Jr.; Akira, S.; et al. Shared and Unique Functions of the DExD/H-Box Helicases RIG-I, MDA5, and LGP2 in Antiviral Innate Immunity. J. Immunol. 2005, 175, 2851. [Google Scholar] [CrossRef]
  3. Hou, F.; Sun, L.; Zheng, H.; Skaug, B.; Jiang, Q.-X.; Chen, Z.J. MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell 2011, 146, 448–461. [Google Scholar] [CrossRef] [PubMed]
  4. Luthra, P.; Sun, D.; Silverman, R.H.; He, B. Activation of IFN-β expression by a viral mRNA through RNase L and MDA5. Proc. Natl. Acad. Sci. USA 2011, 108, 2118–2123. [Google Scholar] [CrossRef] [PubMed]
  5. Marques, J.T.; Devosse, T.; Wang, D.; Zamanian-Daryoush, M.; Serbinowski, P.; Hartmann, R.; Fujita, T.; Behlke, M.A.; Williams, B.R. A structural basis for discriminating between self and nonself double-stranded RNAs in mammalian cells. Nat. Biotechnol. 2006, 24, 559–565. [Google Scholar] [CrossRef] [PubMed]
  6. Hornung, V.; Ellegast, J.; Kim, S.; Brzozka, K.; Jung, A.; Kato, H.; Poeck, H.; Akira, S.; Conzelmann, K.-K.; Schlee, M.; et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 2006, 314, 994–997. [Google Scholar] [CrossRef]
  7. Li, X.; Ranjith-Kumar, C.T.; Brooks, M.T.; Dharmaiah, S.; Herr, A.B.; Kao, C.; Li, P. The RIG-I-like receptor LGP2 recognizes the termini of double-stranded RNA. J. Biol. Chem. 2009, 284, 13881–13891. [Google Scholar] [CrossRef]
  8. Pichlmair, A.; Schulz, O.; Tan, C.P.; Naslund, T.I.; Liljestrom, P.; Weber, F.; Reis e Sousa, C. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5′-phosphates. Science 2006, 314, 997–1001. [Google Scholar] [CrossRef]
  9. Pichlmair, A.; Schulz, O.; Tan, C.P.; Rehwinkel, J.; Kato, H.; Takeuchi, O.; Akira, S.; Way, M.; Schiavo, G.; Reis e Sousa, C. Activation of MDA5 requires higher-order RNA structures generated during virus infection. J. Virol. 2009, 83, 10761. [Google Scholar] [CrossRef]
  10. Berke, I.C.; Li, Y.; Modis, Y. Structural basis of innate immune recognition of viral RNA. Cell Microbiol. 2013, 15, 386–394. [Google Scholar] [CrossRef]
  11. Rothenfusser, S.; Goutagny, N.; Diperna, G.; Gong, M.; Monks, B.G.; Schoenemeyer, A.; Yamamoto, M.; Akira, S.; Fitzgerald, K.A. The RNA Helicase Lgp2 Inhibits TLR-Independent Sensing of Viral Replication by Retinoic Acid-Inducible Gene-I. J. Immunol. 2005, 175, 5260–5268. [Google Scholar] [CrossRef] [PubMed]
  12. Saito, T.; Hirai, R.; Loo, Y.; Owen, D.; Johnson, C.; Sinha, S.; Yamamoto, M.; Akira, S.; Fitzgerald, K.A. Regulation of innate antiviral defenses through a shared repressor domain in RIG-I and LGP2. Proc. Natl. Acad. Sci. USA 2007, 104, 582–587. [Google Scholar] [CrossRef] [PubMed]
  13. Satoh, T. LGP2 is a positive regulator of RIG-I-and MDA5-mediated antiviral responses. Proc. Natl. Acad. Sci. USA 2010, 107, 1512–1517. [Google Scholar] [CrossRef]
  14. Stok, J.E.; Oosenbrug, T.; Ter Haar, L.R.; Gravekamp, D.; Bromley, C.P.; Zelenay, S.; e Sousa, C.R.; van der Veen, A.G. RNA sensing via the RIG-I-like receptor LGP2 is essential for the induction of a type I IFN response in ADAR1 deficiency. EMBO J. 2022, 41, e109760. [Google Scholar] [CrossRef] [PubMed]
  15. Takahashi, T.; Nakano, Y.; Onomoto, K.; Yoneyama, M.; Ui-Tei, K. Virus Sensor RIG-I Represses RNA Interference by Interacting with TRBP through LGP2 in Mammalian Cells. Genes 2018, 9, 511. [Google Scholar] [CrossRef]
  16. Lamb, R.A. Paramyxoviridae: The viruses and their replication. In Fields Virology; Lippincott, Williams, and Wilkins: Philadelphia, PA, USA, 2001; pp. 1305–1340. [Google Scholar]
  17. Weber, F.; Wagner, V.; Rasmussen, S.B.; Hartmann, R.; Paludan, S.R. Double-stranded RNA is produced by positive-strand RNA viruses and DNA viruses but not in detectable amounts by negative-strand RNA viruses. J. Virol. 2006, 80, 5059–5064. [Google Scholar] [CrossRef] [PubMed]
  18. Züst, R.; Cervantes-Barragan, L.; Habjan, M.; Maier, R.; Neuman, B.W.; Ziebuhr, J.; Szretter, K.J.; Baker, S.C.; Barchet, W.; Diamond, M.S.; et al. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 2011, 12, 137–143. [Google Scholar] [CrossRef]
  19. Li, Y.; Shi, X. MicroRNAs in the regulation of TLR and RIG-I pathways. Cell Mol. Immunol. 2013, 10, 65–71. [Google Scholar] [CrossRef]
  20. Lee, M.-K.; Kim, H.-E.; Park, E.-B.; Lee, J.; Kim, K.-H.; Lim, K.; Yum, S.; Lee, Y.-H.; Kang, S.-J.; Lee, J.-H.; et al. Structural features of influenza A virus panhandle RNA enabling the activation of RIG-I independently of 5′-triphosphate. Nucleic Acids Res. 2016, 44, 8407–8416. [Google Scholar] [CrossRef]
  21. Chen, Y.G.; Chen, R.; Ahmad, S.; Verma, R.; Kasturi, S.P.; Amaya, L.; Broughton, J.P.; Kim, J.; Cadena, C.; Pulendran, B.; et al. N6-Methyladenosine Modification Controls Circular RNA Immunity. Mol. Cell 2019, 76, 96–109.e9. [Google Scholar] [CrossRef]
  22. Ranoa, D.R.E.; Parekh, A.D.; Pitroda, S.P.; Huang, X.; Darga, T.; Wong, A.C.; Huang, L.; Andrade, J.; Staley, J.P.; Satoh, T.; et al. Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs. Oncotarget 2016, 7, 26496–26515. [Google Scholar] [CrossRef] [PubMed]
  23. Nallagatla, S.R.; Toroney, R.; Bevilacqua, P.C. A brilliant disguise for self RNA: 5′-end and internal modifications of primary transcripts suppress elements of innate immunity. RNA Biol. 2008, 5, 140–144. [Google Scholar] [CrossRef] [PubMed]
  24. Leung, D.W.; Amarasinghe, G.K. Structural insights into RNA recognition and activation of RIG-I-like receptors. Curr. Opin. Struct. Biol. 2012, 22, 297–303. [Google Scholar] [CrossRef] [PubMed]
  25. Kolakofsky, D.; Kowalinski, E.; Cusack, S. A structure-based model of RIG-I activation. RNA 2012, 18, 2118–2127. [Google Scholar] [CrossRef]
  26. Seth, R.B.; Sun, L.; Ea, C.K.; Chen, Z.J. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 2005, 122, 669–682. [Google Scholar] [CrossRef]
  27. Zhang, X.; Zhang, J.; Zhang, L.; Dam, H.V.; Dijke, P.T. UBE2O negatively regulates TRAF6-mediated NF-κB activation by inhibiting TRAF6 polyubiquitination. Cell Res. 2013, 23, 366–377. [Google Scholar] [CrossRef]
  28. Zhao, B.; Shu, C.; Gao, X.; Sankaran, B.; Du, F.; Shelton, C.L.; Herr, A.B.; Ji, J.-Y.; Li, P. Structural basis for concerted recruitment and activation of IRF-3 by innate immune adaptor proteins. Proc. Natl. Acad. Sci. USA 2016, 113, E3403–E3412. [Google Scholar] [CrossRef]
  29. Yanai, H.; Negishi, H.; Taniguchi, T. The IRF family of transcription factors: Inception, impact and implications in oncogenesis. Oncoimmunology 2012, 1, 1376–1386. [Google Scholar] [CrossRef]
  30. Richetta, C.; Faure, M. Autophagy in antiviral innate immunity. Cell Microbiol. 2013, 15, 368–376. [Google Scholar] [CrossRef]
  31. Lee, H.K.; Lund, J.M.; Ramanathan, B.; Mizushima, N.; Iwasaki, A. Autophagy-Dependent Viral Recognition by Plasmacytoid Dendritic Cells. Science 2007, 315, 1398–1401. [Google Scholar] [CrossRef]
  32. Jiang, M.; Zhang, S.; Yang, Z.; Lin, H.; Zhu, J.; Liu, L.; Wang, W.; Liu, S.; Liu, W.; Ma, Y.; et al. Self-Recognition of an Inducible Host lncRNA by RIG-I Feedback Restricts Innate Immune Response. Cell 2018, 173, 906–919.e13. [Google Scholar] [CrossRef] [PubMed]
  33. Chang, M.X. The negative regulation of retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) signaling pathway in fish. Dev. Comp. Immunol. 2021, 119, 104038. [Google Scholar] [CrossRef] [PubMed]
  34. Malur, M.; Gale, M.; Krug, R.M. LGP2 downregulates interferon production during infection with seasonal human influenza A viruses that activate interferon regulatory factor 3. J. Virol. 2012, 86, 10733–10738. [Google Scholar] [CrossRef] [PubMed]
  35. Oshiumi, H.; Matsumoto, M.; Seya, T. Ubiquitin-mediated modulation of the cytoplasmic viral RNA sensor RIG-I. J. Biochem. 2012, 151, 5–11. [Google Scholar] [CrossRef]
  36. Martin-Vicente, M.; Medrano, L.M.; Resino, S.; Garcia-Sastre, A.; Martinez, I. TRIM25 in the Regulation of the Antiviral Innate Immunity. Front. Immunol. 2017, 8, 1187. [Google Scholar] [CrossRef]
  37. Gack, M.U.; Shin, Y.C.; Joo, C.H.; Urano, T.; Liang, C.; Sun, L.; Takeuchi, O.; Akira, S.; Chen, Z.; Inoue, S.; et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature 2007, 446, 916–920. [Google Scholar] [CrossRef]
  38. Hayman, T.J.; Hsu, A.C.; Kolesnik, T.B.; Dagley, L.F.; Willemsen, J.; Tate, M.D.; Baker, P.J.; Kershaw, N.J.; Kedzierski, L.; Webb, A.I.; et al. RIPLET, and not TRIM25, is required for endogenous RIG-I-dependent antiviral responses. Immunol. Cell Biol. 2019, 97, 840–852. [Google Scholar] [CrossRef]
  39. Cadena, C.; Ahmad, S.; Xavier, A.; Willemsen, J.; Park, S.; Park, J.W.; Oh, S.-W.; Fujita, T.; Hou, F.; Binder, M.; et al. Ubiquitin-Dependent and -Independent Roles of E3 Ligase RIPLET in Innate Immunity. Cell 2019, 177, 1187–1200.e1116. [Google Scholar] [CrossRef]
  40. Pepin, G.; Gantier, P.M. microRNA Decay: Refining microRNA Regulatory Activity. MicroRNA 2016, 5, 167–174. [Google Scholar] [CrossRef] [PubMed]
  41. Gregory, R.; Yan, K.; Amuthan, G.; Chendrimada, T.; Doratotaj, B.; Cooch, N.; Shiekhattar, R. The Microprocessor complex mediates the genesis of microRNAs. Nature 2004, 432, 235–240. [Google Scholar] [CrossRef] [PubMed]
  42. Jin, W.; Wang, J.; Liu, C.P.; Wang, H.W.; Xu, R.M. Structural Basis for pri-miRNA Recognition by Drosha—ScienceDirect. Mol. Cell 2020, 78, 423–433.e5. [Google Scholar] [CrossRef]
  43. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  44. Lee, R.C.; Ambros, V. An Extensive Class of Small RNAs in Caenorhabditis elegans. Science 2001, 294, 862–864. [Google Scholar] [CrossRef] [PubMed]
  45. Chirshev, E.; Oberg, K.C.; Ioffe, Y.J.; Unternaehrer, J.J. Let-7 as biomarker, prognostic indicator, and therapy for precision medicine in cancer. Clin. Transl. Med. 2019, 8, 24. [Google Scholar] [CrossRef]
  46. Huang, K.; Dong, S.; Li, W.; Xie, Z. The expression and regulation of microRNA-125b in cancers. Acta Biochim. Biophys. Sin. 2013, 45, 803–805. [Google Scholar] [CrossRef] [PubMed]
  47. Santovito, D.; Egea, V.; Bidzhekov, K.; Na Ta Relli, L.; Weber, C. Noncanonical inhibition of caspase-3 by a nuclear microRNA confers endothelial protection by autophagy in atherosclerosis. Sci. Transl. Med. 2020, 12, eaaz2294. [Google Scholar] [CrossRef]
  48. Jiang, F.; Chen, Q.; Wang, W.; Ling, Y.; Xia, P. Hepatocyte-Derived Extracellular Vesicles Promote Endothelial Inflammation and Atherogenesis via microRNA-1. J. Hepatol. 2019, 72, 156–166. [Google Scholar] [CrossRef]
  49. Mitra, R.; Adams, C.M.; Wei, J.; Greenawalt, E.J.; Eischen, C.M. Pan-cancer analysis reveals cooperativity of both strands of microRNA that regulate tumorigenesis and patient survival. Nat. Commun. 2020, 11, 968. [Google Scholar] [CrossRef] [PubMed]
  50. Konno, M.; Koseki, J.; Asai, A.; Yamagata, A.; Shimamura, T.; Motooka, D.; Okuzaki, D.; Kawamoto, K.; Mizushima, T.; Eguchi, H.; et al. Distinct methylation levels of mature microRNAs in gastrointestinal cancers. Nat. Commun. 2019, 10, 3888. [Google Scholar] [CrossRef]
  51. Zhu, S.; Liao, Z.; Hu, Y.; Chen, C.; Li, Y.; Liu, S.i.; Luo, J.; Xiong, S.; Xu, L. The effects of microRNA-7 on the metastasis of human lung cancer 95D cells in vitro and in vivo. J. Zunyi Med. Univ. 2013, 36, 5. [Google Scholar]
  52. Bijkerk, R.; Esguerra, J.; Ellenbroek, J.H.; Yu, W.A.; Zonneveld, A. In Vivo Silencing of MicroRNA-132 Reduces Blood Glucose and Improves Insulin Secretion. Nucleic Acid Ther. 2019, 29, 67–72. [Google Scholar] [CrossRef]
  53. Belgardt, B.F.; Ahmed, K.; Spranger, M.; Latreille, M.; Denzler, R.; Kondratiuk, N.; von Meyenn, F.; Villena, F.N.; Herrmanns, K.; Bosco, D.; et al. The microRNA-200 family regulates pancreatic beta cell survival in type 2 diabetes. Nat. Med. 2015, 21, 619–627. [Google Scholar] [CrossRef] [PubMed]
  54. Fatimy, R.E.; Li, S.; Chen, Z.; Mushannen, T.; Krichevsky, A.M. MicroRNA-132 provides neuroprotection for tauopathies via multiple signaling pathways. Acta Neuropathol. 2018, 136, 537–555. [Google Scholar] [CrossRef] [PubMed]
  55. Cao, J.; Huang, M.; Guo, L.; Zhu, L.; Cai, D. MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer’s disease pathogenesis. Mol. Psychiatry 2020, 26, 4687–4701. [Google Scholar] [CrossRef]
  56. Hu, R.; Kagele, D.; Huffaker, T.; Runtsch, M.; Alexander, M.; Liu, J.; Bake, E.; Su, W.; Williams, M.A.; Rao, D.S.; et al. miR-155 promotes T follicular helper cell accumulation during chronic, low-grade inflammation. Immunity 2014, 41, 605–619. [Google Scholar] [CrossRef] [PubMed]
  57. Mikami, Y.; Philips, R.L.; Sciume, G.; Petermann, F.; Meylan, F.; Nagashima, H.; Yao, C.; Davis, F.P.; Brooks, S.R.; Sun, H.-W.; et al. MicroRNA-221 and -222 modulate intestinal inflammatory Th17 cell response as negative feedback regulators downstream of interleukin-23. Immunity 2021, 54, 514–525 e516. [Google Scholar] [CrossRef] [PubMed]
  58. Mohamed, H.A.; Abdelkafy, A.E.; Khairy, R.M.M.; Abdelraheim, S.R.; Kamel, B.A.; Marey, H. MicroRNAs and cytokines as potential predictive biomarkers for COVID-19 disease progression. Sci. Rep. 2023, 13, 3531. [Google Scholar] [CrossRef] [PubMed]
  59. Fernández-Pato, A.; Virseda-Berdices, A.; Resino, S.; Ryan, P.; Martínez-González, O.; Pérez-García, F.; Martin-Vicente, M.; Valle-Millares, D.; Brochado-Kith, O.; Blancas, R.; et al. Plasma miRNA profile at COVID-19 onset predicts severity status and mortality. Emerg. Microbes Infect. 2022, 11, 676–688. [Google Scholar] [CrossRef]
  60. de Gonzalo-Calvo, D.; Benítez, I.D.; Pinilla, L.; Carratalá, A.; Moncusí-Moix, A.; Gort-Paniello, C.; Molinero, M.; González, J.; Torres, G.; Bernal, M.; et al. Circulating microRNA profiles predict the severity of COVID-19 in hospitalized patients. Transl. Res. 2021, 236, 147–159. [Google Scholar] [CrossRef]
  61. Liang, Y.; Fang, D.; Gao, X.; Deng, X.; Chen, N.; Wu, J.; Zeng, M.; Luo, M. Circulating microRNAs as emerging regulators of COVID-19. Theranostics 2023, 13, 125–147. [Google Scholar] [CrossRef]
  62. Samuel, C.E. Antiviral actions of interferons. Clin. Microbiol. Rev. 2001, 14, 778–809. [Google Scholar] [CrossRef]
  63. Raja, R.; Sen, G.C. The antiviral action of the RIG-I induced pathway of apoptosis (RIPA) is enhanced by its ability to degrade Otulin, which deubiquitinates IRF3. Cell Death Differ. 2022, 29, 504–513. [Google Scholar] [CrossRef] [PubMed]
  64. Heutinck, K.M.; Rowshani, A.T.; Kassies, J.; Claessen, N.; van Donselaar-van der Pant, K.A.M.I.; Bemelman, F.J.; Eldering, E.; van Lier, R.A.; Florquin, S.; Berge, I.J.T.; et al. Viral double-stranded RNA sensors induce antiviral, pro-inflammatory, and pro-apoptotic responses in human renal tubular epithelial cells. Kidney Int. 2012, 82, 664–675. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, M.J.; Hwang, S.Y.; Imaizumi, T.; Yoo, J.Y. Negative Feedback Regulation of RIG-I-Mediated Antiviral Signaling by Interferon-Induced ISG15 Conjugation. J. Virol. 2008, 82, 1474–1483. [Google Scholar] [CrossRef]
  66. Inn, K.S.; Gack, M.U.; Tokunaga, F.; Shi, M.; Jung, J.U. Linear ubiquitin assembly complex negatively regulates RIG-I- and TRIM25-mediated type I interferon induction. Mol. Cell 2011, 41, 354–365. [Google Scholar] [CrossRef]
  67. Hou, J.; Wang, P.; Lin, L.; Liu, X.; Ma, F.; An, H.; Wang, Z.; Cao, X. MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J. Immunol. 2009, 183, 2150–2158. [Google Scholar] [CrossRef]
  68. Li, Y.; Fan, X.; He, X.; Sun, H.; Zou, Z.; Yuan, H.; Xu, H.; Wang, C.; Shi, X. MicroRNA-466l inhibits antiviral innate immune response by targeting interferon-alpha. Cell Mol. Immunol. 2012, 9, 497–502. [Google Scholar] [CrossRef] [PubMed]
  69. Chen, G.; Yang, Y.; Wu, Q.-J.; Cao, L.; Ruan, W.; Shao, C.; Jiang, L.; Tang, P.; Ma, S.; Jiang, A.; et al. ILF3 represses repeat-derived microRNAs targeting RIG-I mediated type I interferon response. J. Mol. Biol. 2022, 434, 167469. [Google Scholar] [CrossRef]
  70. Sheng, Y.; Wang, Y.; Lu, W.; Zhou, Y.; Dong, G.; Ge, X.; Song, Y.; Zhang, Y. MicroRNA-92a inhibits macrophage antiviral response by targeting retinoic acid inducible gene-I. Microbiol. Immunol. 2018, 62, 585–593. [Google Scholar] [CrossRef]
  71. Zhao, L.; Zhang, X.; Wu, Z.; Huang, K.; Sun, X.; Chen, H.; Jin, M. The Downregulation of MicroRNA hsa-miR-340-5p in IAV-Infected A549 Cells Suppresses Viral Replication by Targeting RIG-I and OAS2. Mol. Ther. Nucleic Acids 2019, 14, 509–519. [Google Scholar] [CrossRef]
  72. Zhao, L.; Zhu, J.; Zhou, H.; Zhao, Z.; Zou, Z.; Liu, X.; Lin, X.; Zhang, X.; Deng, X.; Wang, R.; et al. Identification of cellular microRNA-136 as a dual regulator of RIG-I-mediated innate immunity that antagonizes H5N1 IAV replication in A549 cells. Sci. Rep. 2015, 5, 14991. [Google Scholar] [CrossRef] [PubMed]
  73. Qian, X.; Wu, B.; Xu, C.; Qi, Z. Hepatitis C Virus Infection Cycle-Specific MicroRNA Profiling Reveals Stage-Specific miR-4423-3p Targets RIG-I to Facilitate Infection. Front. Cell Infect. Microbiol. 2022, 12, 851917. [Google Scholar] [CrossRef] [PubMed]
  74. Ingle, H.; Kumar, S.; Raut, A.A.; Mishra, A.; Kulkarni, D.D.; Kameyama, T.; Takapka, A.; Akira, S.; Kumar, H. The microRNA miR-485 targets host and influenza virus transcripts to regulate antiviral immunity and restrict viral replication. Sci. Signal. 2015, 8, ra126. [Google Scholar] [CrossRef]
  75. Donyavi, T.; Bokharaei-Salim, F.; Baghi, H.B.; Khanaliha, K.; Kiani, S.J. Acute and Post-Acute Phase of Covid-19: Analyzing Expression patterns of miRNA-29a-3p, 146a-3p, 155–5p, and Let-7b-3p in PBMC. Int. Immunopharmacol. 2021, 97, 107641. [Google Scholar] [CrossRef] [PubMed]
  76. Hou, Z.; Zhang, J.; Han, Q.; Su, C.; Qu, J.; Xu, D.; Zhang, C.; Tian, Z. Hepatitis B virus inhibits intrinsic RIG-I and RIG-G immune signaling via inducing miR146a. Sci. Rep. 2016, 6, 26150. [Google Scholar] [CrossRef] [PubMed]
  77. Lu, Y.; Qin, Z.; Wang, J.; Zheng, X.; Lu, J.; Zhang, X.; Wei, L.; Peng, Q.; Zheng, Y.; Ou, C.; et al. Epstein-Barr Virus miR-BART6-3p Inhibits the RIG-I Pathway. J. Innate Immun. 2017, 9, 574–586. [Google Scholar] [CrossRef] [PubMed]
  78. Qiu, Y.; Geng, X.; Ban, J.; Liu, Y. MicroRNA-218 inhibits type I interferon production and facilitates virus immune evasion via targeting RIG-I. Biotechnol. Appl. Biochem. 2020, 67, 396–403. [Google Scholar] [CrossRef]
  79. Li, Z.; Luo, Q.; Xu, H.; Zheng, M.; Abdalla, B.A.; Feng, M.; Cai, B.; Zhang, X.; Nie, Q.; Zhang, X. MiR-34b-5p Suppresses Melanoma Differentiation-Associated Gene 5 (MDA5) Signaling Pathway to Promote Avian Leukosis Virus Subgroup J (ALV-J)-Infected Cells Proliferaction and ALV-J Replication. Front. Cell. Infect. Microbiol. 2017, 7, 17. [Google Scholar] [CrossRef]
  80. Chu, Q.; Han, J.; Sun, L.; Cui, J.; Xu, T. Characterization of MDA5 and microRNA-203 negatively regulates the RLR signaling pathway via targeting MDA5 in miiuy croaker. Dev. Comp. Immunol. 2022, 126, 104259. [Google Scholar] [CrossRef]
  81. Rayner, K.J.; Sheedy, F.J.; Esau, C.C.; Hussain, F.N.; Temel, R.E.; Parathath, S.; van Gils, J.M.; Rayner, A.J.; Chang, A.N.; Suarez, Y.; et al. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J. Clin. Investig. 2011, 121, 2921–2931. [Google Scholar] [CrossRef]
  82. Rayner, K.J.; Suárez, Y.; Dávalos, A.; Parathath, S.; Fitzgerald, M.L.; Tamehiro, N.; Fisher, E.A.; Moore, K.J.; Fernández-Hernando, C. MiR-33 contributes to the regulation of cholesterol homeostasis. Science 2010, 328, 1570–1573. [Google Scholar] [CrossRef]
  83. Liu, D.; Tan, Q.; Zhu, J.; Zhang, Y.; Xue, Y.; Song, Y.; Liu, Y.; Wang, Q.; Lai, L. MicroRNA-33/33* inhibit the activation of MAVS through AMPK in antiviral innate immunity. Cell Mol. Immunol. 2021, 18, 1450–1462. [Google Scholar] [CrossRef]
  84. Zhang, C.; Wang, Q.; Liu, A.-Q.; Zhang, C.; Liu, L.-H.; Lu, L.-F.; Tu, J.; Zhang, Y.-A. MicroRNA miR-155 inhibits cyprinid herpesvirus 3 replication via regulating AMPK-MAVS-IFN axis. Dev. Comp. Immunol. 2021, 129, 104335. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, B.; Han, J.; Chen, S.; Xie, R.; Xia, R. MicroLet-7b Dampens Neutrophilic Inflammation. Front. Immunol. 2021, 12, 653344. [Google Scholar] [CrossRef] [PubMed]
  86. Blanco, L.P.; Wang, X.; Carlucci, P.M.; Torres-Ruiz, J.J.; Kaplan, M.J. RNA externalized by neutrophil extracellular traps promotes inflammatory pathways in endothelial cells. Arthritis Rheumatol. 2021, 73, 2282–2292. [Google Scholar] [CrossRef]
  87. Tang, H.; Ma, M.; Dai, J.; Cui, C.; Si, L.; Sheng, X.; Chi, Z.; Xu, L.; Yu, S.; Xu, T.; et al. miR-let-7b and miR-let-7c suppress tumourigenesis of human mucosal melanoma and enhance the sensitivity to chemotherapy. J. Exp. Clin. Cancer Res. 2019, 38, 212. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, Q.; Pan, J.; Xiong, D.; Wang, Y.; Miller, M.S.; Sei, S.; Shoemaker, R.H.; Izzotti, A.; You, M. Pulmonary Aerosol Delivery of Let-7b microRNA Confers a Striking Inhibitory Effect on Lung Carcinogenesis through Targeting the Tumor Immune Microenvironment. Adv. Sci. 2021, 8, e2100629. [Google Scholar] [CrossRef]
  89. Serino, G.; Pesce, F.; Sallustio, F.; De Palma, G.; Cox, S.; Curci, C.; Zaza, G.; Lai, K.N.; Leung, J.C.; Tang, S.C.; et al. In a retrospective international study, circulating miR-148b and let-7b were found to be serum markers for detecting primary IgA nephropathy. Kidney Int. 2016, 89, 683–692. [Google Scholar] [CrossRef]
  90. Serino, G.; Sallustio, F.; Curci, C.; Cox, S.N.; Pesce, F.; De Palma, G.; Schena, F.P. Role of let-7b in the regulation of N-acetylgalactosaminyltransferase 2 in IgA nephropathy. Nephrol Dial. Transplant. 2015, 30, 1132–1139. [Google Scholar] [CrossRef]
  91. Yeh, Y.J.; Tseng, C.P.; Hsu, S.D.; Huang, H.Y.; Lai, M.M.; Huang, H.D.; Cheng, J.C. Effects of Let-7b in the Early Stage of Hepatitis C Virus Infection. J. Virol. 2020, 95, e01800-20. [Google Scholar] [CrossRef]
  92. Xu, S.; Han, L.; Wei, Y.; Zhang, B.; Wang, Q.; Liu, J.; Liu, M.; Chen, Z.; Wang, Z.; Chen, H.; et al. MicroRNA-200c-targeted contactin 1 facilitates the replication of influenza A virus by accelerating the degradation of MAVS. PLoS Pathog. 2022, 18, e1010299. [Google Scholar] [CrossRef]
  93. Xu, T.; Chu, Q.; Cui, J.; Bi, D. Inducible MicroRNA-3570 Feedback Inhibits the RIG-I-Dependent Innate Immune Response to Rhabdovirus in Teleost Fish by Targeting MAVS/IPS-1. J. Virol. 2017, 92, e01594-17. [Google Scholar] [CrossRef] [PubMed]
  94. You, J.; Wu, W.; Lu, M.; Xie, Y.; Miao, R.; Gu, M.; Xi, D.; Yan, W.; Wu, D.; Wang, X.; et al. Hepatic exosomes with declined MiR-27b-3p trigger RIG-I/TBK1 signal pathway in macrophages. Liver Int. 2022, 42, 1676–1691. [Google Scholar] [CrossRef]
  95. Xu, C.; He, X.; Zheng, Z.; Zhang, Z.; Wei, C.; Guan, K.; Hou, L.; Zhang, B.; Zhu, L.; Cao, Y.; et al. Downregulation of MicroRNA miR-526a by Enterovirus Inhibits RIG-I-Dependent Innate Immune Response. J. Virol. 2014, 88, 11356–11368. [Google Scholar] [CrossRef]
  96. Mo, L.; Zeng, Z.; Deng, R.; Li, Z.; Hu, Y. Hepatitis A virus-induced hsa-miR-146a-5p attenuates IFN-β signaling by targeting adaptor protein TRAF6. Arch. Virol. 2021, 166, 789–799. [Google Scholar] [CrossRef] [PubMed]
  97. Li, B.; Zheng, J. MicroR-9-5p suppresses EV71 replication through targeting NFκB of the RIG-I-mediated innate immune response. FEBS Open Bio 2018, 8, 1457–1470. [Google Scholar] [CrossRef] [PubMed]
  98. Li, J.; Xie, Y.; Li, L.; Li, X.; Shen, L.; Gong, J.; Zhang, R. MicroRNA-30a Modulates Type I Interferon Responses to Facilitate Coxsackievirus B3 Replication Via Targeting Tripartite Motif Protein 25. Front. Immunol. 2020, 11, 603437. [Google Scholar] [CrossRef]
  99. Liu, W.; Jin, Y.; Zhang, W.; Xiang, Y.; Jia, P.; Yi, M.; Jia, K. MiR-202-5p Inhibits RIG-I-Dependent Innate Immune Responses to RGNNV Infection by Targeting TRIM25 to Mediate RIG-I Ubiquitination. Viruses 2020, 12, 261. [Google Scholar] [CrossRef]
  100. Guo, M.; Gan, L.; Si, J.; Zhang, J.; Liu, Z.; Zhao, J.; Gou, Z.; Zhang, H. Role of miR-302/367 cluster in human physiology and pathophysiology. Acta Biochim. Biophys. Sin. 2020, 52, 791–800. [Google Scholar] [CrossRef]
  101. Liu, J.; Wang, Y.; Ji, P.; Jin, X. Application of the microRNA-302/367 cluster in cancer therapy. Cancer Sci. 2020, 111, 1065–1075. [Google Scholar] [CrossRef]
  102. Yasukawa, K.; Kinoshita, D.; Yaku, K.; Nakagawa, T.; Koshiba, T. The microRNAs miR-302b and miR-372 regulate mitochondrial metabolism via the SLC25A12 transporter, which controls MAVS-mediated antiviral innate immunity. J. Biol. Chem. 2020, 295, 444–457. [Google Scholar] [CrossRef] [PubMed]
  103. Gui, S.; Chen, X.; Zhang, M.; Zhao, F.; Wan, Y.; Wang, L.; Xu, G.; Zhou, L.; Yue, X.; Zhu, Y.; et al. Mir-302c mediates influenza A virus-induced IFNβ expression by targeting NF-κB inducing kinase. FEBS Lett. 2015, 589, 4112–4118. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, X.; Zhou, L.; Peng, N.; Yu, H.; Li, M.; Cao, Z.; Lin, Y.; Wang, X.; Li, Q.; Wang, J.; et al. MicroRNA-302a suppresses influenza A virus-stimulated interferon regulatory factor-5 expression and cytokine storm induction. J. Biol. Chem. 2017, 292, 21291–21303. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, K.; Xiao, F.; Hu, D.; Ge, W.; Tian, M.; Wang, W.; Pan, P.; Wu, K.; Wu, J. SARS-CoV-2 Nucleocapsid Protein Interacts with RIG-I and Represses RIG-Mediated IFN-beta Production. Viruses 2020, 13, 47. [Google Scholar] [CrossRef]
  106. Park, A.; Iwasaki, A. Type I and Type III Interferons—Induction, Signaling, Evasion, and Application to Combat COVID-19. Cell Host Microbe 2020, 27, 870–878. [Google Scholar] [CrossRef]
  107. Zheng, Y.; Zhuang, M.W.; Han, L.; Zhang, J.; Wang, P.H. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) membrane (M) protein inhibits type I and III interferon production by targeting RIG-I/MDA-5 signaling. Signal Transduct. Target. Ther. 2020, 5, 299. [Google Scholar] [CrossRef]
  108. Chen, Y.; Liu, Q.; Guo, D. Emerging coronaviruses: Genome structure, replication, and pathogenesis. J. Med. Virol. 2020, 92, 2249. [Google Scholar] [CrossRef]
  109. Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science 2020, 368, 409–412. [Google Scholar] [CrossRef]
  110. Zheng, Y.; Deng, J.; Han, L.; Zhuang, M.-W.; Xu, Y.; Zhang, J.; Nan, M.-L.; Xiao, Y.; Zhan, P.; Liu, X.; et al. SARS-CoV-2 NSP5 and N protein counteract the RIG-I signaling pathway by suppressing the formation of stress granules. Signal Transduct. Target. Ther. 2022, 7, 22. [Google Scholar] [CrossRef]
  111. Cullen, B.R. Viral and cellular messenger RNA targets of viral microRNAs. Nature 2009, 457, 421–425. [Google Scholar] [CrossRef]
  112. Zhang, X.; Zhang, E.; Ma, Z.; Pei, R.; Jiang, M.; Schlaak, J.F.; Roggendorf, M.; Lu, M. Modulation of hepatitis B virus replication and hepatocyte differentiation by MicroRNA-1. Hepatology 2011, 53, 1476–1485. [Google Scholar] [CrossRef] [PubMed]
  113. Roganovi, J.R. microRNA-146a and -155, upregulated by periodontitis and type 2 diabetes in oral fluids, are predicted to regulate SARS-CoV-2 oral receptors genes. J. Periodontol. 2021, 92, 35–43. [Google Scholar] [CrossRef] [PubMed]
  114. Firoozi, Z.; Mohammadisoleimani, E.; Shahi, A.; Naghizadeh, M.M.; Mirzaei, E.; Asad, A.G.; Salmanpour, Z.; Nouri, S.M.J.; Mansoori, Y. Hsa_circ_0000479/Hsa-miR-149-5p/RIG-I, IL-6 Axis: A Potential Novel Pathway to Regulate Immune Response against COVID-19. Can. J. Infect. Dis. Med. Microbiol. 2022, 2022, 2762582. [Google Scholar] [CrossRef] [PubMed]
  115. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef]
  116. Cheng, J.; Li, G.; Wang, W.; Stovall, D.B.; Sui, G.; Li, D. Circular RNAs with protein-coding ability in oncogenesis. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188909. [Google Scholar] [CrossRef]
  117. Ho-Xuan, H.; Glažar, P.; Latini, C.; Heizler, K.; Haase, J.; Hett, R.; Anders, M.; Weichmann, F.; Bruckmann, A.; Berg, D.V.D.; et al. Comprehensive analysis of translation from overexpressed circular RNAs reveals pervasive translation from linear transcripts. Nucleic Acids Res. 2020, 48, 10368–10382. [Google Scholar] [CrossRef]
  118. Bertolazzi, G.; Cipollina, C.; Benos, P.V.; Tumminello, M.; Coronnello, C. miR-1207-5p Can Contribute to Dysregulation of Inflammatory Response in COVID-19 via Targeting SARS-CoV-2 RNA. Front. Cell. Infect. Microbiol. 2020, 10, 673. [Google Scholar] [CrossRef]
  119. Duecker, R.P.; Adam, E.H.; Wirtz, S.; Gronau, L.; Khodamoradi, Y.; Eberhardt, F.J.; Donath, H.; Gutmann, D.; Vehreschild, M.J.G.T.; Zacharowski, K.; et al. The MiR-320 Family Is Strongly Downregulated in Patients with COVID-19 Induced Severe Respiratory Failure. Int. J. Mol. Sci. 2021, 22, 351. [Google Scholar] [CrossRef]
  120. Pimenta, R.; Viana, N.I.; Santos, G.D.; Candido, P.; Reis, S.T. MiR-200c-3p expression may be associated with worsening of the clinical course of patients with COVID-19. Mol. Biol. Res. Commun. 2021, 10, 141–147. [Google Scholar] [CrossRef]
  121. Niemiec, S.M.; Hilton, S.A.; Wallbank, A.; Azeltine, M.; Liechty, K.W. Cerium oxide nanoparticle delivery of microRNA-146a for local treatment of acute lung injury. Nanomed. Nanotechnol. Biol. Med. 2021, 34, 102388. [Google Scholar] [CrossRef]
  122. Abdolahi, S.; Hosseini, M.; Rezaei, R.; Mohebbi, S.R.; Rostami-Nejad, M.; Mojarad, E.N.; Mirjalali, H.; Yadegar, A.; Aghdaei, H.A.; Zali, M.R.; et al. Evaluation of miR-200c-3p and miR-421-5p levels during immune responses in the admitted and recovered COVID-19 subjects. Infect. Genet. Evol. 2022, 98, 105207. [Google Scholar] [CrossRef] [PubMed]
  123. Jafarinejad-Farsangi, S.; Jazi, M.M.; Rostamzadeh, F.; Hadizadeh, M. High affinity of host human microRNAs to SARS-CoV-2 genome: An in silico analysis. Non-Coding RNA Res. 2020, 5, 222–231. [Google Scholar] [CrossRef] [PubMed]
  124. Zhu, Y.; Zhang, Z.; Song, J.; Qian, W.; Gu, X.; Yang, C.; Shen, N.; Xue, F.; Tang, Y. SARS-CoV-2-Encoded MiRNAs Inhibit Host Type I Interferon Pathway and Mediate Allelic Differential Expression of Susceptible Gene. Front. Immunol. 2021, 12, 767726. [Google Scholar] [CrossRef] [PubMed]
  125. Hasan, M.; Roy, S.; Sharma, B.; Ishtiaque, M.; Azim, F. Identification and Host Response Interaction Study of SARS-CoV-2 encoded miRNA- Like Sequences: An in silico Approach. Comput. Biol. Med. 2021, 134, 104451. [Google Scholar] [CrossRef]
  126. Liu, Z.; Wang, J.; Ge, Y.; Xu, Y.; Guo, M.; Mi, K.; Xu, R.; Pei, Y.; Zhang, Q.; Luan, X.; et al. SARS-CoV-2 encoded microRNAs are involved in the process of virus infection and host immune response. J. Biomed. Res. 2021, 35, 12. [Google Scholar] [CrossRef]
  127. Wu, Y.; Wu, X.; Wu, L.; Wang, X.; Liu, Z. The anticancer functions of RIG-I–like receptors, RIG-I and MDA5, and their applications in cancer therapy. Transl. Res. 2017, 190, 51–60. [Google Scholar] [CrossRef]
  128. Elion, D.L.; Jacobson, M.E.; Hicks, D.J.; Rahman, B.; Cook, R.S. Therapeutically Active RIG-I Agonist Induces Immunogenic Tumor Cell Killing in Breast Cancers. Cancer Res. 2018, 78, 6183–6195. [Google Scholar] [CrossRef]
  129. Hou, P.; Yang, K.; Jia, P.; Liu, L.; Lin, Y.; Li, Z.; Li, J.; Chen, S.; Guo, S.; Pan, J.; et al. A novel selective autophagy receptor, CCDC50, delivers K63 polyubiquitination-activated RIG-I/MDA5 for degradation during viral infection. Cell Res. 2021, 31, 62–79. [Google Scholar] [CrossRef]
  130. Wu, J.; Xu, W.; Ma, L.; Sheng, J.; Chen, H. Formononetin relieves the facilitating effect of lncRNA AFAP1-AS1-miR-195/miR-545 axis on progression and chemo-resistance of triple-negative breast cancer. Aging 2021, 13, 18191. [Google Scholar] [CrossRef]
  131. Luo, L.; Miao, P.; Ming, Y.; Tao, J.; Shen, H. Circ-ZFR Promotes Progression of Bladder Cancer by Upregulating WNT5A Via Sponging miR-545 and miR-1270. Front. Oncol. 2021, 10, 596623. [Google Scholar] [CrossRef]
  132. Zhang, H.; Zhang, K.; Xu, Z.; Chen, Z.; Cui, J. MicroRNA-545 suppresses progression of ovarian cancer through mediating PLK1 expression by a direct binding and an indirect regulation involving KDM4B-mediated demethylation. BMC Cancer 2021, 21, 163. [Google Scholar] [CrossRef] [PubMed]
  133. Hu, C.; Wang, Y.; Li, A.; Zhang, J.; Xue, F.; Zhu, L. Overexpressed circ_0067934 acts as an oncogene to facilitate cervical cancer progression via the miR-545/EIF3C axis. J. Cell. Physiol. 2018, 234, 9225–9232. [Google Scholar] [CrossRef]
  134. He, M.; Feng, L.; Qi, L.; Rao, M.; Zhu, Y. Long Noncoding RNASBF2-AS1 Promotes Gastric Cancer Progression via Regulating miR-545/EMS1 Axis. BioMed Res. Int. 2020, 2020, 6590303. [Google Scholar] [CrossRef] [PubMed]
  135. Li, L.; Kong, X.A.; Zang, M.; Dong, J.; Hu, Y. Hsa_circ_0003732 promotes osteosarcoma cells proliferation via miR-545/CCNA2 axis. Biosci. Rep. 2020, 40, BSR20191552. [Google Scholar] [CrossRef] [PubMed]
  136. Du, B.; Wang, Z.; Zhang, X.; Feng, S.; Wang, G.; He, J.; Zhang, B. MicroRNA-545 Suppresses Cell Proliferation by Targeting Cyclin D1 and CDK4 in Lung Cancer Cells. PLoS ONE 2014, 9, e88022. [Google Scholar] [CrossRef]
  137. Song, B.; Ji, W.; Guo, S.; Liu, A.; Jing, W.; Shao, C.; Li, G.; Jin, G. miR-545 inhibited pancreatic ductal adenocarcinoma growth by targeting RIG-I. FEBS Lett. 2014, 588, 4375–4381. [Google Scholar] [CrossRef]
  138. Liu, Z.; Dou, C.; Yao, B.; Xu, M.; Ding, L.; Wang, Y.; Jia, Y.; Li, Q.; Zhang, H.; Tu, K.; et al. Ftx non coding RNA-derived miR-545 promotes cell proliferation by targeting RIG-I in hepatocellular carcinoma. Oncotarget 2016, 7, 25350–25365. [Google Scholar] [CrossRef]
  139. Yuan, G.; Wu, H.; Du, Y.; He, F. Tumor suppressor role of microRNA-545 in oral squamous cell carcinoma. Oncol. Lett. 2018, 17, 2063–2068. [Google Scholar] [CrossRef]
  140. Zhang, B. Systematic analysis of intronic microRNAs reveals cooperativity within the multi-component FTX locus to promote colon cancer development. Cancer Res. 2020, 81, 1038–1320. [Google Scholar] [CrossRef]
  141. Nam, R.K.; Benatar, T.; Amemiya, Y.; Seth, A. MiR-139 Induces an Interferon-beta Response in Prostate Cancer Cells by Binding to RIG-1. Cancer Genom. Proteom. 2021, 18, 197–206. [Google Scholar] [CrossRef]
  142. Jang, S.I.; Tandon, M.; Teos, L.; Zheng, C.; Warner, B.M.; Alevizos, I. Dual function of miR-1248 links interferon induction and calcium signaling defects in Sjogren’s syndrome. EBioMedicine 2019, 48, 526–538. [Google Scholar] [CrossRef] [PubMed]
  143. Karlsen, T.A.; Brinchmann, J.E. Liposome delivery of microRNA-145 to mesenchymal stem cells leads to immunological off-target effects mediated by RIG-I. Mol. Ther. 2013, 21, 1169–1181. [Google Scholar] [CrossRef] [PubMed]
  144. Han, J.; Sun, Y.; Song, W.; Xu, T. microRNA-145 regulates the RLR signaling pathway in miiuy croaker after poly(I:C) stimulation via targeting MDA5. Dev. Comp. Immunol. 2017, 68, 79–86. [Google Scholar] [CrossRef] [PubMed]
  145. Han, J.; Chu, Q.; Huo, R.; Xu, T. Inducible microRNA-122 modulates RIG-I signaling pathway via targeting DAK in miiuy croaker after poly(I:C) stimulation. Dev. Comp. Immunol. 2018, 78, 52–60. [Google Scholar] [CrossRef] [PubMed]
  146. Gao, W.; Zheng, W.; Sun, Y.; Xu, T. microRNA-489 negatively modulates RIG-I signaling pathway via targeting TRAF6 in miiuy croaker after poly(I:C) stimulation. Fish Shellfish. Immunol. 2021, 113, 61–68. [Google Scholar] [CrossRef]
  147. Lemecha, M.; Morino, K.; Imamura, T.; Iwasaki, H.; Ohashi, N.; Ida, S.; Sato, D.; Sekine, O.; Ugi, S.; Maegawa, H. MiR-494-3p regulates mitochondrial biogenesis and thermogenesis through PGC1-α signalling in beige adipocytes. Sci. Rep. 2018, 8, 15096. [Google Scholar] [CrossRef]
  148. Lo, Y.L.; Wang, C.S.; Chen, Y.C.; Wang, T.Y.; Chang, Y.H.; Chen, C.-J.; Yang, C.-P. Mitochondrion-Directed Nanoparticles Loaded with a Natural Compound and a microRNA for Promoting Cancer Cell Death via the Modulation of Tumor Metabolism and Mitochondrial Dynamics. Pharmaceutics 2020, 12, 756. [Google Scholar] [CrossRef] [PubMed]
  149. Marchi, S.; Lupini, L.; Patergnani, S.; Rimessi, A.; Missiroli, S.; Bonora, M.; Bononi, A.; Corrà, F.; Giorgi, C.; De Marchi, E.; et al. Downregulation of the mitochondrial calcium uniporter by cancer-related miR-25. Curr. Biol. 2013, 23, 58–63. [Google Scholar] [CrossRef]
  150. Hou, J.; Zhou, Y.; Zheng, Y.; Fan, J.; Zhou, W.; Ng, I.L.; Sun, H.; Qin, L.; Qiu, S.; Lee, J.M.; et al. Hepatic RIG-I Predicts Survival and Interferon-α Therapeutic Response in Hepatocellular Carcinoma. Cancer Cell 2014, 25, 49–63. [Google Scholar] [CrossRef]
  151. Wolf, D.; Fiegl, H.; Zeimet, A.G.; Wieser, V.; Marth, C.; Sprung, S.; Sopper, S.; Hartmann, G.; Reimer, D.; Boesch, M. High RIG-I expression in ovarian cancer associates with an immune-escape signature and poor clinical outcome. Int. J. Cancer 2020, 146, 2007–2018. [Google Scholar] [CrossRef]
  152. Uz, M.; Kalaga, M.; Pothuraju, R.; Ju, J.; Junker, W.M.; Batra, S.K.; Mallapragada, S.; Rachagani, S. Dual delivery nanoscale device for miR-345 and gemcitabine co-delivery to treat pancreatic cancer. J. Control Release 2019, 294, 237–246. [Google Scholar] [CrossRef] [PubMed]
  153. Deng, X.; Cao, M.; Zhang, J.; Hu, K.; Yin, Z.; Zhou, Z.; Xiao, X.; Yang, Y.; Sheng, W.; Wu, Y.; et al. Hyaluronic acid-chitosan nanoparticles for co-delivery of MiR-34a and doxorubicin in therapy against triple negative breast cancer. Biomaterials 2014, 35, 4333–4344. [Google Scholar] [CrossRef] [PubMed]
  154. Xiao, X.; Wang, X.; Gao, H.; Chen, X.; Li, J.; Zhang, Y. Cell-Selective Delivery of MicroRNA with a MicroRNA-Peptide Conjugate Nanocomplex. Chem. Asian J. 2018, 13, 3845–3849. [Google Scholar] [CrossRef] [PubMed]
  155. Scholz, J.; Weil, P.P.; Pembaur, D.; Koukou, G.; Aydin, M.; Hauert, D.; Postberg, J.; Kreppel, F.; Hagedorn, C. An Adenoviral Vector as a Versatile Tool for Delivery and Expression of miRNAs. Viruses 2022, 14, 1952. [Google Scholar] [CrossRef]
  156. Ganju, A.; Khan, S.; Hafeez, B.B.; Behrman, S.W.; Yallapu, M.M.; Chauhan, S.C.; Jaggi, M. miRNA nanotherapeutics for cancer. Drug Discov. Today 2017, 22, 424–432. [Google Scholar] [CrossRef]
  157. Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 2020, 18, 10. [Google Scholar] [CrossRef]
  158. Zhang, Y.; Liu, Q.; Zhang, X.; Huang, H.; Tang, S.; Chai, Y.; Xu, Z.; Li, M.; Chen, X.; Liu, J.; et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J. Nanobiotechnol. 2022, 20, 279. [Google Scholar] [CrossRef]
  159. Barile, L.; Vassalli, G. Exosomes: Therapy delivery tools and biomarkers of diseases. Pharmacol. Ther. 2017, 174, 63–78. [Google Scholar] [CrossRef]
Figure 1. RIG-I-like receptor protein domain structure. The three RLRs are DExD/H-box helicase proteins of type SF2 that contain the conserved domains Hel1 and Hel2 in their core DExD/H-box helicase domain. The Hel2i insertion domain, essential for RIG-I autoregulation, is located between the two helicase domains. A broadly conserved C-terminal region is also present in all RLRs (CTD). A positively charged binding pocket is present only in the CTD of RIG-I to identify 5′-PPP or 5′PP RNA substrates. Two N-terminal caspase activation and recruitment domains are shared by RIG-I and MDA5 (CARDs). For interactions with MAVS and the start of downstream antiviral signaling, several CARDs must be aligned.
Figure 1. RIG-I-like receptor protein domain structure. The three RLRs are DExD/H-box helicase proteins of type SF2 that contain the conserved domains Hel1 and Hel2 in their core DExD/H-box helicase domain. The Hel2i insertion domain, essential for RIG-I autoregulation, is located between the two helicase domains. A broadly conserved C-terminal region is also present in all RLRs (CTD). A positively charged binding pocket is present only in the CTD of RIG-I to identify 5′-PPP or 5′PP RNA substrates. Two N-terminal caspase activation and recruitment domains are shared by RIG-I and MDA5 (CARDs). For interactions with MAVS and the start of downstream antiviral signaling, several CARDs must be aligned.
Biomolecules 13 01344 g001
Figure 2. Biological processes of miRNAs. Pri-miRNAs can be formed via the intronic pathway and the intergenic pathway. Processing of pri-miRNAs produces pre-miRNAs. Pre-miRNAs are cleaved by the enzyme Dicer and then re-digested to generate mature miRNAs, which can target mRNAs, resulting in mRNA degradation or translational repression.
Figure 2. Biological processes of miRNAs. Pri-miRNAs can be formed via the intronic pathway and the intergenic pathway. Processing of pri-miRNAs produces pre-miRNAs. Pre-miRNAs are cleaved by the enzyme Dicer and then re-digested to generate mature miRNAs, which can target mRNAs, resulting in mRNA degradation or translational repression.
Biomolecules 13 01344 g002
Figure 3. The hsa_circ_0000479/hsa-miR-149/RIG-I, IL-6 axis regulates SARS-CoV-2 infection. Up-expressed hsa_circ_0000479 could modulate the expression of IL-6 and RIG-I through sponging of hsa-miR-149–5p. On the one hand, activation of the RIG-I signaling pathway promotes the release of IFN-I, which exerts antiviral effects. On the other hand, the expression level of IL-6 is associated with lung inflammation and extensive lung injury.
Figure 3. The hsa_circ_0000479/hsa-miR-149/RIG-I, IL-6 axis regulates SARS-CoV-2 infection. Up-expressed hsa_circ_0000479 could modulate the expression of IL-6 and RIG-I through sponging of hsa-miR-149–5p. On the one hand, activation of the RIG-I signaling pathway promotes the release of IFN-I, which exerts antiviral effects. On the other hand, the expression level of IL-6 is associated with lung inflammation and extensive lung injury.
Biomolecules 13 01344 g003
Figure 4. MicroRNAs regulate the RIG-I-like receptor signaling pathway. RIG-I and MDA5 recognize a complementary set of viral cytosolic dsRNA ligands. Their activation is tightly regulated by phosphorylation, ubiquitination, and host proteins such as LGP2. RIG-I and MDA5 signal to MAVS, and then induce NF-κB and IRF3/7 to enter the nucleus, which promotes the release of type I IFNs and inflammatory cytokines. The activity of RIG-I and MDA5 can be regulated by several E3 ubiquitin ligases, including TRIM25 and RNF123. The circled ‘P’ indicates phosphorylation, and the circled ‘Ub’ indicates ubiquitination. MiR-BART6–3p, miR-4423–3p, miR-340–5p, miR-545, miR-145, miR-146a, miR-218, miR-485, miR-149, miR-92a, and miR-582–3p target RIG-I; miR-34b-5p, miR-203, and miR-145-5p target MDA5; miR-122 targets DAK to inhibit the expression of MDA5; miR-30a and miR-202-5p target TRIM25; miR-155 and miR-33 target AMPK; miR-200c targets CNTN1 to regulate MAVS, while miR-3570 directly targets MAVS; miR-489 and miR-146a-5p target TRAF6; and miR-9–5p targets NF-κB. MicroRNA let-7b targets IKKα. These microRNAs directly or indirectly regulate the RIG-I-like receptor signaling pathway to play antiviral, antitumor, or immunomodulatory roles.
Figure 4. MicroRNAs regulate the RIG-I-like receptor signaling pathway. RIG-I and MDA5 recognize a complementary set of viral cytosolic dsRNA ligands. Their activation is tightly regulated by phosphorylation, ubiquitination, and host proteins such as LGP2. RIG-I and MDA5 signal to MAVS, and then induce NF-κB and IRF3/7 to enter the nucleus, which promotes the release of type I IFNs and inflammatory cytokines. The activity of RIG-I and MDA5 can be regulated by several E3 ubiquitin ligases, including TRIM25 and RNF123. The circled ‘P’ indicates phosphorylation, and the circled ‘Ub’ indicates ubiquitination. MiR-BART6–3p, miR-4423–3p, miR-340–5p, miR-545, miR-145, miR-146a, miR-218, miR-485, miR-149, miR-92a, and miR-582–3p target RIG-I; miR-34b-5p, miR-203, and miR-145-5p target MDA5; miR-122 targets DAK to inhibit the expression of MDA5; miR-30a and miR-202-5p target TRIM25; miR-155 and miR-33 target AMPK; miR-200c targets CNTN1 to regulate MAVS, while miR-3570 directly targets MAVS; miR-489 and miR-146a-5p target TRAF6; and miR-9–5p targets NF-κB. MicroRNA let-7b targets IKKα. These microRNAs directly or indirectly regulate the RIG-I-like receptor signaling pathway to play antiviral, antitumor, or immunomodulatory roles.
Biomolecules 13 01344 g004
Table 1. MicroRNAs involved in the regulation of RLR signaling pathway, their targets, and results mentioned in this paper.
Table 1. MicroRNAs involved in the regulation of RLR signaling pathway, their targets, and results mentioned in this paper.
MicroRNAsTargetsResultsReference
miR-92aRIG-ILowering VSV-induced production of type I IFN and promoting viral replication in macrophages[70]
miR-218RIG-IImpairing interferon production to promote viral immune escape[78]
miR-4423-3pRIG-IPromoting HCV replication[73]
miR-BART6-3pRIG-IPromoting EBV infection Limiting influenza virus infection and avoiding erroneous antiviral signaling activation[77]
miR-485RIG-IInhibiting pancreatic ductal adenocarcinoma growth[76]
miR-545RIG-IPromoting the proliferation of hepatocellular carcinoma cells; inhibiting the proliferation and migration of oral squamous cell carcinoma[137,138,139]
miR-145RIG-IInducing immune responses by targeting RIG-I expression[143]
miR-582-3pRIG-IControlling VSV replication[69]
miR-340-5pRIG-I,
OAS2
Inhibiting IAV replication[71]
miR-146aRIG-I, RIG-GAggravating HBV infection[76]
miR-34b-5pMDA5Promoting the proliferation and migration of DF-1 cells and ALV-J replication[79]
miR-203MDA5Negatively regulating RLR signaling pathway[80]
miR-145-5pMDA5Regulating RLR signaling pathway[144]
miR-122DAKInhibiting the expression of MDA5[145]
miR-30aTRIM25Regulating type I IFN response and promoting CVB3 replication[98]
miR-202-5pTRIM25Inhibiting RIG-I-dependent innate immune responses to RGNNV infection[99]
miR-3570MAVSInhibiting the expression of MAVS and promoting the replication of rhabdovirus[93]
miR-200cCNTN1Inhibiting the expression of MAVS and promoting the replication of IAV[92]
miR-155AMPKInhibiting cyprinid herpesvirus 3 replication via regulating the AMPK-MAVS-IFN axis[84]
miR-33AMPKInhibiting the activation of MAVS through AMPK[83]
miR-489TRAF6Negatively regulating TRAF6 and involved in the immune response to poly(I:C) stimulation[146]
miR-146a-5pTRAF6,
IRAK1,
IRAK2
Negatively regulating VSV-triggered type I IFN
production, thus promoting VSV replication in
macrophages,
[96]
let-7bIKKaleading to an increase in IFN-I expression[91]
miR-9-5pNF-κBRestraining EV71 replication[97]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chen, D.; Ji, Q.; Liu, J.; Cheng, F.; Zheng, J.; Ma, Y.; He, Y.; Zhang, J.; Song, T. MicroRNAs in the Regulation of RIG-I-like Receptor Signaling Pathway: Possible Strategy for Viral Infection and Cancer. Biomolecules 2023, 13, 1344. https://doi.org/10.3390/biom13091344

AMA Style

Chen D, Ji Q, Liu J, Cheng F, Zheng J, Ma Y, He Y, Zhang J, Song T. MicroRNAs in the Regulation of RIG-I-like Receptor Signaling Pathway: Possible Strategy for Viral Infection and Cancer. Biomolecules. 2023; 13(9):1344. https://doi.org/10.3390/biom13091344

Chicago/Turabian Style

Chen, Dengwang, Qinglu Ji, Jing Liu, Feng Cheng, Jishan Zheng, Yunyan Ma, Yuqi He, Jidong Zhang, and Tao Song. 2023. "MicroRNAs in the Regulation of RIG-I-like Receptor Signaling Pathway: Possible Strategy for Viral Infection and Cancer" Biomolecules 13, no. 9: 1344. https://doi.org/10.3390/biom13091344

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

Article Metrics

Back to TopTop