Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 23.
Published in final edited form as: Semin Cell Dev Biol. 2016 Jul 2;65:11–19. doi: 10.1016/j.semcdb.2016.06.016

Mammalian microRNAs and long noncoding RNAs in the host-bacterial pathogen crosstalk

Mélodie Duval a,b,c, Pascale Cossart a,b,c,*, Alice Lebreton d,e,**
PMCID: PMC7089780  EMSID: EMS85994  PMID: 27381344

Abstract

Gene expression regulation is a critical question in host-pathogen interactions, and RNAs act as key players in this process. In this review, we focus on the mammalian RNA response to bacterial infection, with a special interest on microRNAs and long non-coding RNAs. We discuss the role of cellular miRNAs in immunity, the implication of circulating miRNAs as well as the influence of the microbiome on the miRNA response. We also review how pathogens counteract the host miRNA expression. Interestingly, bacterial non-coding RNAs regulate host gene expression and conversely eukaryotic miRNAs may regulate bacterial gene expression. Overall, the characterization of RNA regulatory networks represents an emerging theme in the field of host pathogen interactions.

Keywords: Host-pathogen, Bacteria, microRNA, lncRNA, Regulatory network

1. Introduction

During infection, both bacteria and host undergo significant changes of environmental or physiological conditions, to which they can adapt or react. Extra- and intra-cellular pathogenic bacteria can adjust their metabolism and trigger the expression of virulence genes, which allow them to benefit from their host resources. Likewise, infected organisms are capable of sensing the intrusion by bacterial pathogens, and react by triggering innate or adaptive immune defences. The complex interplay of actions/counter-actions occurring between a bacterial invader and an infected organism is referred to as host-pathogen crosstalk. This crosstalk, on the bacterial or on the host side, involves the coordinated regulation of gene expression programmes with the phase of infection, controlled at the transcriptional or post-transcriptional level. For instance, the transcriptome of cells infected by intra-cellular bacteria is completely reshaped, as a result of pathogen detection by specific cellular sensors (reviewed in [1]), and of specific modulation by bacterial effectors (reviewed in [2]).

RNA itself represents a key regulatory molecule. Indeed, its physicochemical properties make non-coding RNAs (ncRNA) versatile tools for interfering with gene expression, independently of the coding function played by messenger RNAs (mRNA). Recognition by base-pairing allows one single ncRNA to bind multiple targets, and thereby to regulate several pathways simultaneously [3]. Conversely, a single gene can be regulated by a variety of ncRNAs, allowing its control in a broad range of conditions.

Over the past 10 years, mechanisms and effects of RNA-mediated regulation on gene expression have been broadly investigated in prokaryotes and eukaryotes. Various examples of RNA-mediated regulation in pathogenic bacteria have been recently reviewed [47]. Here, we focus on regulation involving mammalian ncRNAs during host-pathogen interaction, and more precisely on two classes of ncRNAs: microRNAs (miRNA) and long non-coding RNAs (lncRNA). These ncRNAs act at the transcriptional and post-transcriptional levels in the control of host gene expression, and are themselves the object of regulations.

The name lncRNA is given to a class of eukaryotic ncRNAs of more than 200 nucleotides transcribed by RNA polymerase II (PolII). Like messenger RNAs, most of these transcripts are poly-adenylated and spliced [8]. Even though the first mammalian lncRNA, XIST (X-inactive specific transcript), was discovered 25 years ago [9,10], the identification and functional characterization of these noncoding transcripts remain an emerging field. Several lncRNAs have been shown to regulate gene expression at the level of transcription or translation (for reviews see [1113]). The expression of most lncRNAs is tissue-specific, or restricted to precise developmental stages; in addition, some lncRNAs are involved in host response against viral infection (reviewed in [8]). Very recent studies show that lncRNAs are also involved in the response against pathogenic bacteria, which we will describe below.

miRNAs are a class of PolII-dependent transcripts that are processed into a short mature form (21–24 nucleotides). To date, more than 35,000 distinct microRNAs have been described in 223 organisms (miRBase, release 21), and are predicted to regulate 60% of human protein-coding genes [14]. They are involved in multiple processes including cell proliferation, differentiation, inflammation, etc. [1517]. After being transcribed, the canonical biogenesis of miRNAs involves the processing of a hairpin precursor into a double stranded duplex by two major nucleases, Drosha and Dicer (reviewed in [15]). After export to the cytoplasm, one strand of the duplex is loaded into an effector complex called RISC (RNA-induced silencing complex), which drives the microRNA to its target mRNA and mediates its function, usually by promoting RNA degradation or impeding translation, even though a few activation mechanisms have also been described [18]. In addition to transcription, the amount of available functional miRNAs in the cytoplasm can be regulated via their sequestration by binding proteins, or by a class of ncRNAs called miRNA sponges. Their deregulation was shown in some instances to promote diseases, such as cancer, auto-immunity or cardiovascular disorders [1921].

To counteract bacterial infection, host cells adjust their gene expression programme, among other pathways by using miRNA and lncRNA as regulatory molecules. Reciprocally, pathogens can escape host defence mechanisms using a variety of strategies, and in particular by targeting miRNA-mediated regulation. Here, we review the role and subversion of mammalian ncRNAs during host pathogen crosstalk.

2. Host cell microRNA response to bacterial infection

2.1. Discovery of microRNA response against pathogens

In 2006, the first studies describing a host miRNA response to challenges with bacterial components were published. Recognition of Pseudomonas syringae flagellin by Arabidopsis thaliana was shown to induce the expression of miR-393a and the subsequent inhibition of the expression of three F-box auxin receptors. This, in turn, decreases signalling by auxin, a plant hormone that modulates immune response, and limits bacterial spread within tissue [22]. In human monocytes, the involvement of miRNAs in innate immune response to lipopolysaccharide (LPS, the major component of the outer membrane of Gram negative bacteria) stimulation was investigated. This work has led to the characterization of miR-146 as an anti-inflammatory miRNA [23].

Following these two studies, the miRNA response upon bacterial infection in mammals has been widely investigated. Infection by different pathogens leads to the expression of specific microRNA sets that are regulated in a time-dependent manner [24]. Among them, miR-155, miR-146, let-7 and miR-29 play important roles in the host cells response to bacteria [2528]. These miRNAs regulate the immune responses in order to clear bacterial infection, while preserving the organism from deleterious effects of inflammation. They will be described below.

A recent analysis of the miRNAs expressed in dendritic cells in response to six different bacteria has revealed a core temporal response to infection comprising 49 miRNAs that may play an essential role in response to infection, in addition to subtle variability for each bacterium, that can be considered as a specific signature [29] (Fig. 1a). Moreover, infection can induce a switch in the relative abundance of mature miRNAs deriving from the 5′ and 3′ arms of the precursor (a process described as “arm-switching”, [30], Fig. 1b), and changes the proportion of miRNA variants (isomiRs, Fig. 1c). Such variability in miRNA identity is thought to affect their regulatory potential, but this aspect deserves more investigations [29].

Fig. 1. Host microRNA variation upon infection.

Fig. 1

Upon pathogen exposure, miRNA variability occurs at different levels. (a) The abundance of miRNAs is modified to face infection, and miRNAs are up- or down-regulated early or late in response to pathogens, leading to the expression of a core temporal response common to all pathogens, as well as a specific response; (b) The choice of the arm of pre-miRNA loaded into the RISC complex may be changed by infection, in a process called “arm-switching”; (c) Generation of isomiRs in addition to canonical miRNAs is observed (inspired from [29]).

2.2. Key microRNAs involved in immunity

Upon infection, miR-155 and miR-146 are two miRNAs induced by the NF-κB pathway through pattern recognition receptors (PRR) sensing of pathogen motifs, in particular LPS (Fig. 2a). These miRNAs regulate distinct sets of genes [23,31].

Fig. 2. Mammalian non-coding RNAs at work during host-pathogen crosstalk.

Fig. 2

A complex interplay of microRNA and lncRNA occurs during bacterial infection. miRNAs miR-155 and −146 are induced by PRRs in a NF-κB-dependent pathway (a), and regulate different sets of transcripts (b,c,d,e) thus promoting or dampening inflammation. miR-155 is also induced by NOD2 cytoplasmic receptors (f). Exosomal transfer of miRNAs allows cell-to-cell communication (g) and extracellular miRNAs might regulate bacterial gene expression (h). To subvert host defences, bacteria can perturb inflammatory response (i,j). Bacteria might also generate miR-like molecules, and thereby interfere with host gene expression (k). lncRNAs are key players in inflammatory response, as they affect chromatin structure (l), thus leading to expression of antimicrobial molecules. They can also hamper inflammation, by sequestering RelA/p65 and thus preventing its DNA binding activity (m).

Expression of miR-146 is induced by sub-inflammatory levels of NF-κB activity. It acts as an anti-inflammatory regulator, by targeting TRAF6 (TNF Receptor-associated factor 6) and IRAK1 (IL-1R-associated kinase 1), which are involved in the NF-κB pathway (Fig. 2b), thus promoting tolerance to low doses of LPS [23,31]. This desensitized state is necessary to protect the organism against septic shock. A tolerance state is also crucial during the postnatal establishment of the intestinal microbiota in the newborn gut, where miR-146 prevents inappropriate inflammation (for a review, [32]).

In contrast, miR-155 is induced by higher doses of LPS, at levels which result in pro-inflammatory NF-κB activity, as well as by TNF-α and interferon β, via TAB2 [31,33,34]. miR-155 is known to amplify the expression of pro-inflammatory factors, thereby acting in defence against pathogens, and also to exert negative feedback on the immune system, thus protecting the host from potentially damaging overreaction [31]. The involvement of this miRNA in the pro-inflammatory response has been thoroughly investigated; among others, miR-155 targets SHIP1 (Fig. 2c), a negative regulator of the NF-κB pathway [35,36], and SOCS1 (suppressor of cytokine signalling 1), an effector involved in the homeostasis of Treg cells [37]. This in turn, stimulates the expression of the pro-inflammatory cytokines TNF-α, IL-6, IL-1β, IL-8 and IL-12, while it reduces the expression of the anti-inflammatory cytokine IL-10 [38,39]. A complex interplay occurs upon infection, as IL-10 itself can reduce miR-155 expression in response to LPS [40]. miR-155 also increases TNF-α production upon LPS stimulation (Fig. 2d), probably by stabilizing the transcript or by promoting its translation when targeting the expression of proteins that binds the transcript 3′-UTR [34]. Multiple miR-155 targets are also involved in T helper cell development, or promote autophagy by inhibiting the mTOR pathway [4143]. In line with these data, miR-155 is essential for an efficient immune response to several bacterial pathogens. Indeed, miR-155 null mice show a slower clearance upon Citrobacter rodentium infection [44], as well as an impaired CD8+ T-cell response to Listeria monocytogenes [45]. Moreover, miR-155 is essential in the vaccination process against Salmonella typhimurium [46]. However, this miRNA has also been shown to repress genes such as NIK, IKKε and TAB2, which encode proteins involved in the inflammatory pathway (Fig. 2e); accordingly, it is proposed that miR-155 could act as a limiter of inflammation [31,34,47]. Interestingly, miR-155 expression is also stimulated by the intracellular NOD2 receptor (Fig. 2f). In tolerant macrophages, where miR-146 prevents inflammation, stimulation of NOD2 can restore the NF-κB pathway activation, as well as the feedback control by miR-155 [31].

Another well-described miRNA family is the let-7 family, which targets different genes involved in immunity. This family is repressed upon infection by various pathogens as well as by exposure to LPS [4852]. Indeed, NF-κB activation induces Lin-28B expression, a protein that blocks let-7 maturation [53]. In addition, various studies have highlighted an active repression of these miRNAs mediated by the bacteria themselves [5456]. Several targets have been described for let-7 family miRNAs. let-7b targets the TLR4 transcript and as a consequence, bacterial infection-mediated let-7b repression promotes TLR sensing and subsequent NF-κB activation [51]. let-7a and let-7d target IL-6 and IL-10 transcripts; their down-regulation could contribute to maintaining a balanced immune response upon bacterial exposure [50]. let-7c targets the mTOR pathway, thus modulating T-cell activity [57]. let-7i reduces expression of SNAP23 protein, which is involved in exosome release, a process that is part of the antimicrobial response [52]. The abundant let-7f targets the deubiquinating enzyme A20, an inhibitor of the NF-κB pathway [56]; its downregulation by Mycobacterium tuberculosis favours bacterial survival in macrophages [56].

In murine leucocytes, miR-29 was shown to repress interferon (IFN)-γ expression, and thereby control immune responses to intracellular bacteria [58]. During infection of natural killer cells, CD4+ and CD8+ T-cells, by Listeria monocytogenes or Mycobacterium bovis bacillus Calmette-Guérin (BCG), miR-29 expression is down-regulated, thereby facilitating IFN-γ production and subsequent bacterial clearance. Strikingly, miR-29 is one of the microRNAs triggered during infection of macrophages by L. monocytogenes [59]. Even though the effect of this up-regulation is unknown, it suggests that infection can differentially control the expression of miRNAs, and as a consequence, that of their targets, depending on the affected cell type.

Altogether, these examples provide evidence that miRNAs act as major components of regulatory networks in immune pathways. Their production is temporally regulated in a dose-dependent manner, resulting in the induction or dampening of immunity, and allowing a rapid and potent response against pathogens.

2.3. Circulating microRNAs during infection

Aside from their activity inside the cell, miRNAs are powerful molecules for cell-to-cell communication. Indeed, miRNAs are found in many body fluids, including plasma, saliva, tears, urine, amniotic fluid, colostrum, breast milk, stool, etc. [60,61]. Exosomes, which are circulating small lipid vesicles formed from cell plasma membranes, can transport molecules. Exosomal transfer of miRNAs is a powerful mechanism to expand the host response to bacterial infection. Recently, it has been shown that upon LPS exposure, miR-155 and miR-146a are released from bone marrow-derived dendritic cells (BMDCs) within exosomes and are successfully transferred to recipient cells [62] (Fig. 2 g). Upon uptake, miRNAs regulate gene expression and reprogram the response of BMDCs to LPS. The authors propose several hypotheses; one of them is that miRNA release in exosomes could be a dynamically regulated process where the ratio of miR-155/miR-146a changes over time, thus enhancing the inflammation upon infection, and then dampening it during recovery.

Exosomal miRNAs could give rise to therapeutic perspectives: miR-146-containing exosomes could be used to treat inflammatory diseases, while miR-155-containing exosomes could be used as an adjuvant to improve vaccine efficacy, or to fight infections as it has been proposed for Helicobacter pylori [63]. In addition, these circulating miRNAs are now used or proposed as biomarkers in diagnostic tools for diseases, including sepsis and tuberculosis [6467]. For instance, the signature pattern of circulating miRNAs in the blood of mice shows a differential response to either LPS [68] or lipoteichoic acids (LTA, a major constituent of the cell wall of Gram positive bacteria) [69]. Nine specific circulating miRNAs were also identified in the serum of mice infected by Staphylococcus aureus, but not by Escherichia coli [70].

Finally, a recent article relates the in vitro ability of miRNAs isolated from faeces to penetrate inside E. coli and Fusobacterium nucleatum and to regulate the expression of bacterial genes in a sequence-specific manner (Fig. 2 h). Whether this phenomenon occurs in vivo remains to be determined, nevertheless this discovery opens new perspectives about the possible roles of miRNAs in host-pathogen interaction [60].

2.4. Influence of commensals on the microRNA response to a food-borne pathogen

The resident intestinal microbiota has been long known to play a major role in intestinal homeostasis by shaping the gut transcriptome. An increasing number of studies indicate that miRNAs are participating in this host/commensal crosstalk, and largely contribute to regulatory networks controlling tissue integrity, immunity and metabolism (reviewed in [71]). For instance, miRNA expression influences the microbiota composition as well as host resistance to dextran sulphate sodium (DSS)-induced colitis [60]. The presence and nature of the intestinal flora can also tune infection by a bacterial pathogen such as L. monocytogenes, as well as the host mRNA and miRNA responses [72,73]. Oral pre-treatment with lactobacilli of otherwise germ-free mice significantly reduces their sensitivity to orally-acquired listeriosis and reshapes host gene expression, notably of genes coding for proteins in the interferon pathway. Furthermore, lactobacilli oral pre-treatment can counteract the down-regulation of miR-192, miR-200b, and miR-215 induced by L. monocytogenes infection [73]. A study comparing the intestinal response to Listeria infection in conventional versus germ-free mice has additionally highlighted five microR-NAs down-regulated by Listeria infection only in the presence of the microbiota, and not affected in germ-free mice (miR-143, miR-148a, miR-200b, miR-200c, and miR-378), suggesting that commensals can prime the host microRNA response to infection [72]. This work has also outlined a network of putative regulatory interactions between host microRNAs and their target mRNAs impacted by infection, paving the way to future mechanistic studies aiming at understanding how these microRNAs modulate the intestinal response to infection.

3. Subversion of microRNA pathways by bacterial pathogens

3.1. Manipulation of cell physiology

During infection, bacteria can interfere with many cellular processes using effector molecules [74]. In particular, by modulating the production of miRNAs, they manipulate their host cell physiology and defences, with favourable consequences for pathogen survival (Table 1). For example, Helicobacter pylori induces the increase of miR-1289, which represses HKα, a component of the gastric H+/K+ ATPase. This results in a transient decrease of gastric acidity that promotes H. pylori colonization. The mechanism is dependent on CagA, a bacterial effector known to activate the NF-κB pathway [75]. H. pylori also interferes with autophagy by inducing the expression of miR-30b, which targets BECN1 and ATG12 transcripts that encode proteins involved in the formation and maturation of autophagosomes [76].

Table 1. Bacteria subvert host defences by modulating microRNA expression.

Bacteria miRNA Up/down-regulation Targeted cellular function Cell lines References
Helicobacter pylori miR-1289 up gastric acidity AGS [76]
miR-30b up autophagy AGS and human gastric tissues [77]
miR-372 and miR-373 down cell cycle AGS [80]
miR-584 and miR-1290 up epithelial-mesenchymal transition AGS, SGC7901 and 293T [81]
miR-21 and miR-222 up carcinogenesis AGS, SGC7901, MKN28, MKN45, HGC-27, BGC-823, GES-1 [82,83]
Salmonella spp. miR-30c and miR-30e up SUMOylation HCT-8, HeLa, J774A.2 [78]
miR-15 family down cell cycle HeLa, HT-29, RAW 264.7 [79]
miR-128 up macrophage recruitment HT29 [84]
Mycobacterium tuberculosis miR-132, miR-26a up macrophage response to IFN-γ hMDM [85]
miR-125b, miR-99b, miR-155 up, up, down production of TNF-α hMDM, mDC [86,87]
let-7f down NF-κB pathway RAW264.7, BMDM, and hMDM [57]
miR-155 up NF-κB pathway RAW264.7 and BMDM [88]

Bacteria manipulate the expression of various miRNAs in order to modulate cellular processes, favouring their survival and/or proliferation. AGS, SGC7901, MKN28, MKN45, HGC-27, BGC-823, GES-1 and 293T: human gastric cell lines; HCT-8 and HT-29: human colonic cell lines; HeLa: human cervix cell line; J774A.2 and RAW 264.7: murine macrophage cell lines; hMDM: human monocyte-derived macrophage cell line; BMDM: murine bone marrow-derived macrophage cell line; mDC: mice dendritic cell line.

Another example of this subversion is the reduction of host SUMOylation by Salmonella, via the stimulation of two miR-30 family miRNAs, miR-30c and miR-30e, and subsequent down-regulation of their target Ubc9, the only cellular E2 SUMO-conjugating enzyme [77]. The mechanism used by Salmonella to up-regulate these microRNAs is unknown.

The cell cycle is an important target for pathogens, as it influences their ability to survive and/or proliferate. The perturbation of specific miRNAs is used to this end by Salmonella. Indeed, by inhibiting the production of the transcriptional factor E2F1, Salmonella reduces the expression of the miR-15 family [78]. This induces the de-repression of cyclin D1 and thus promotes the G1/S cell cycle transition, which is favouring bacterial intracellular replication (Fig. 3a). A similar mechanism has been described for H. pylori, which down-regulates miR-372 and miR-373 expression in a CagA-dependent manner. This induces a cell cycle arrest at the G1/S transition and inhibits renewal of the gastric epithelium (Fig. 3b), a major host defence mechanism against bacterial infection [79]. CagA also promotes the expression of miR-584 and miR-1290, which target FOXA1, a negative regulator of the epithelial-mesenchymal transition [80]. In addition to short-term, acute bacterial infections, pathogens also induce long-term effects, as it is exemplified by H. pylori in carcinogenesis. The investigation of the roles of miRNAs in this process has revealed that this bacterium induces the expression of miR-21 and miR-222, two miRNAs that target the tumor suppressor RECK [81,82].

Fig. 3. Modulation of microRNA by pathogens can perturb the cell cycle.

Fig. 3

By inhibiting different sets of miRNAs, Salmonella (a) and Helicobacter pylori (b) perturb the host cell cycle, thus promoting their survival and proliferation.

3.2. Effect on immune response

Bacteria deploy diverse mechanisms to counteract macrophage recruitment and/or activation, as well as induction of pro-inflammatory factors. For instance, secreted effectors from Salmonella stimulate, via the p53 signalling pathway, the expression of miR-128, which targets the macrophage colony-stimulating factor (M-CSF) and thereby leads to impaired M-CSF–mediated macrophage recruitment [83]. Likewise, M. tuberculosis can limit macrophage response to IFN-γ by up-regulating miR-132 and miR-26a [84]. These two miRNAs down-regulate the transcriptional coactivator p300, a component of the IFN-γ signalling cascade. Mycobacterium also reduces the production of TNF-α, a potent mediator of macrophage activation and bacterial clearance, by regulating the abundance of miR-125b, miR-155 and miR-99b [85,86] (Fig. 2i). This leads to the destabilization of TNF-α mRNA transcripts and an increase of SHIP1 production, subsequently reducing the production of TNF-α. In addition, the secreted effector from M. tuberculosis ESAT-6 down-regulates the expression of let-7f in macrophages, which leads to increased expression of the deubiquitinating enzyme A20, a negative regulator of the NF-κB pathway [56]. M. tuberculosis ESAT-6 also stimulates miR-155 expression in macrophages, thus repressing BACH1 and SHIP1. This, in turn, induces the expression of the heme oxygenase 1, and activates the serine/threonine kinase AKT, which respectively promote bacterial dormancy and survival [87] (Fig. 2j). However, the ESAT-6-dependent induction of miR-155 also induces macrophage apoptosis by down regulating SOCS1 protein [88] and targeting Rheb [43] which is deleterious for the pathogen.

Taken together, these studies indicate that bacteria can control miRNA expression in order to subvert host defences.

3.3. Regulation of host gene expression by bacterial non-coding RNAs

In addition to deregulation of endogenous microRNAs, emerging data suggest that bacteria could also produce regulatory RNAs that would modulate the host gene expression, as previously shown for viruses (reviewed in [89]). A typical example of this system is the E. coli OxyS and DsrA ncRNAs which, after ingestion by Caenorhabditis elegans, modulate che-2 and F42G9.6 gene expression, probably by promoting the degradation of the host transcripts [90]. Recently, two studies have investigated bacterial microRNA-like molecules. First, an in silico search for bacterial ncRNAs harbouring a secondary structure that might generate miRNA if processed by the host has identified 68 candidate bacterial RNAs from 28 bacterial genomes [91]. These putative bacterial miRNAs are predicted to target mRNAs involved in 47 different human diseases, including cancer and diabetes, thus providing a new perspective for bacterial influence on health and disease. In an independent study, a pre-microRNA was identified in Mycobacterium marinum, for which a mature 23 nucleotide-long form was found associated with the host RISC complex upon infection (Fig. 2k) [92]. While the abundance of the endogenous pre-microRNA was too low to induce any detectable effect, their overexpression efficiently decreased mRNA target expression. This type of bacterial-derived miRNA molecules may thus represent a novel class of regulatory factors in the host pathogen crosstalk.

4. Long noncoding RNAs at play in response to bacterial infection

The involvement of lncRNAs in defence against viruses has been well described (reviewed in [8]), and recent publications show that several lncRNAs are involved in inflammatory response to LPS and live bacteria. Among them, LINoCR (for LPS Inducible NonCoding RNA) activates the expression of lysozyme in a chicken macrophage cell line challenged with LPS, by remodelling the chromatin (Fig. 2l) [93]. More recently, lncRNA profiles in response to infection or LPS stimulation have been analysed in several mammalian cell lines, and the mechanism of action for some of these lncRNAs has been elucidated.

In human monocytes, 76 enhancer RNAs (eRNA), 40 canonical lncRNAs, 65 antisense lncRNAs and 35 regions of bidirectional transcription are differently expressed upon LPS stimulation, [94]. Knockdowns of IL1β-eRNA and of a region of bidirectional transcription (IL1 β-RBT46) attenuate LPS-induced mRNA transcription and release of proinflammatory mediators, highlighting their implication in the inflammatory response. One lncRNA, lnc-IL7R, has been well characterized: this lncRNA is a negative regulator of proinflammatory mediators such as IL-8, IL-6, E-selectin and VCAM-1, by maintaining a repressive chromatin mark, trimethylation of histone H3 at lysine 27 (H3K27me3) at their gene promoters [95]. In contrast, NeST lncRNA expressed in mice CD4+/CD8+ T cells promotes IFN-γ production by methylating lysine 4 of histone H3 – a mark of active transcription – at the IFN-γ locus [96]. In mouse bone-marrow derived macrophages (BMDMs), 27 lncRNAs are differentially expressed upon LPS stimulation, and their de-regulation is associated with histone trimethylation or acetylation of neighbouring genes, suggesting a possible regulatory role in innate-immune response [97]. In HeLa cells infected by Salmonella, a variation of 44% of total lncRNAs has been monitored, leading the authors to propose that lncRNAs could be used as sensitive markers for pathogen activity in the early infection phase [98]. Last, HOTAIR, a lncRNA first shown to participate in the transcriptional repression of HOX genes [99], is also a positive regulator of inflammation [100]. Indeed, HOTAIR up-regulation in mice cardiomyocytes after LPS-induced sepsis induces TNF-α production by promoting phosphorylation of p65 protein and NF-κB activation.

A subtype of lncRNAs named lincRNAs (long intergenic non-coding RNAs), which are expressed from intergenic regions, have also been studied. In bone marrow dendritic cells, LPS stimulation induces the expression of about 20 lincRNAs, most of them being dependent of NF-κB. Among them, lincRNA-Cox2 is the most expressed [101]. This lincRNA is also up-regulated in mouse bone-marrow derived macrophages upon stimulation by TLR ligands, as well as by L. monocytogenes [102], and modulates the expression of many genes involved in inflammation. More precisely, the authors could identify that lincRNA-Cox2 forms a complex with hnRNP-A/B and −A2/B1, two nuclear RNA binding proteins, and thereby repress the transcription of immune genes [102].

LncRNAs can also arise from the expression of pseudogenes. Among them Lethe, which is induced by NF-κB upon TNF-α stimulation, was recently found to contribute to a negative feedback regulation of this pathway, by sequestering the RelA/p65 subunit and thus inactivating its DNA binding activity [103] (Fig. 2m). The active regulation of pseudogene lncRNA expression can thus constitute a means to control inflammatory signalling.

The above-reported studies highlight the involvement of lncRNAs in inflammatory response. Even though some of them were performed using live bacteria, whether bacterial pathogens can subvert lncRNA-mediated regulation remains to be further explored. A first hint of bacterial subversion of lncRNA was obtained in macrophages infected by BCG, showing that 11 lncRNAs were less expressed during a challenge with live bacteria compared to heat inactivated bacteria [104].

5. Conclusion

The major property of ncRNAs is certainly their ability to regulate several targets by specific base-pairing; thereby, they constitute a potent tool allowing cells to tune the expression of a whole regulon with one simple mechanism. As described above, ncRNAs can affect all steps in the gene expression process. When affecting the translation and stability of mature mRNAs, their action takes place at a downstream step in the pathway; as a consequence, their effects are more immediate and flexible than those of transcriptional regulators. Regulation by ncRNAs thus provide a diversity of responses by the host cell during the early stages of bacterial infection, when cells readjust promptly their gene expression programme to face damage and organize immune defences. Given the various microRNAs and lncRNAs that are differentially regulated in response to bacterial pathogens, in diverse cell types and tissues, they constitute a reservoir of gene expression that can fine-tune an impressive number of putative targets.

In addition to their direct regulatory activity on target gene expression, lncRNA and miRNA may also regulate each other; lncRNAs can act as miRNA sponges, in a process described as “competitive endogenous RNA” (ceRNA, reviewed in [105]). Reciprocally, miRNAs may indirectly influence gene expression by targeting lncRNAs [104]. Thus, a complex regulatory network is at play in the host cell to tightly modulate gene expression. This type of interplay might be involved in host response against bacterial infection, as recently shown for cancer [106]. Given the arising data on the ability of regulatory RNAs produced by pathogens to regulate host gene expression and also the possible regulatory function of cellular RNAs on the pathogen, the role of RNA in the host-pathogen crosstalk might be even more sophisticated than previously anticipated (Fig. 4).

Fig. 4. The coding/non-coding RNA interplay in host-pathogen crosstalk.

Fig. 4

During host-pathogen interaction, RNA is a key regulatory molecule allowing bacterial adaptation, subversion and survival, as well as host response and immunity. Eukaryotic ncRNAs are able to cross-regulate themselves, and could even regulate bacterial genes. In addition, bacterial regulatory RNAs as well as secreted effectors are able to modulate the host transcriptome. Thus, a complex RNA network is at play in the host-pathogen crosstalk.

Acknowledgements

We would like to thank Olivier Dussurget for his helpful comments on the manuscript. Work in the laboratory of P.C. received financial support from European Research Council advanced grant BacCellEpi-670823, ERA-NET Infect-ERA ProAntiLis, Investissements d’Avenir BACNET (ANR-10-BINF-0201) and IBEID (ANR-10-LABX-62-01), Human Frontier Science Program (RGP0011/2013), and Fondation le Roch Les Mousquetaires. The laboratory of A.L. is supported by Fondation pour la Recherche Médicale (FRM-AJE20131128944), Inserm ATIP-Avenir, Mairie de Paris (Programme Émergences–Recherche médicale) and Investissements d’Avenir MemoLife (ANR-10-LABX-54). P.C. is a Senior International Research Scholar of the Howard Hughes Medical Institute.

References

  • [1].Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev. 2009;22:240–273. doi: 10.1128/CMR.00046-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Niller HH, Minarovits J. Patho-epigenetics of infectious diseases caused by intracellular bacteria. Patho-Epigenetics Infect Dis. 2016:107–130. doi: 10.1007/978-3-319-24738-0. [DOI] [PubMed] [Google Scholar]
  • [3].Peterson SM, Thompson JA, Ufkin ML, Sathyanarayana P, Liaw L, Congdon CB. Common features of microRNA target prediction tools. Front Genet. 2014;5:1–10. doi: 10.3389/fgene.2014.00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Bouloc P, Repoila F. Fresh layers of RNA-mediated regulation in Gram-positive bacteria. Curr Opin Microbiol. 2016;30:30–35. doi: 10.1016/j.mib.2015.12.008. [DOI] [PubMed] [Google Scholar]
  • [5].Caldelari I, Chao Y, Romby P, Vogel J. RNA-mediated regulation in pathogenic bacteria. Cold Spring Harb Perspect Biol. 2013;5:1–22. doi: 10.1101/cshperspect.a010298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Sesto N, Koutero M, Cossart P. Bacterial and cellular RNAs at work during Listeria infection. Future Microbiol. 2014;9:1025–1037. doi: 10.2217/fmb.14.79. [DOI] [PubMed] [Google Scholar]
  • [7].Wagner EGH, Romby P. Small RNAs in bacteria and archaea: who they are what they do, and how they do it. Adv Genet. 2015 doi: 10.1016/bs.adgen.2015.05.001. [DOI] [PubMed] [Google Scholar]
  • [8].Ding Y, Zhang Z, Liu Y, Shi C, Zhang J, Zhang Y. Relationship of long noncoding RNA and viruses. Genomics. 2016:1–5. doi: 10.1016/j.ygeno.2016.01.007. [DOI] [PubMed] [Google Scholar]
  • [9].Brown CJ, Ballabio a, Rupert JL, Lafreniere RG, Grompe M, Tonlorenzi R, et al. A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome. Nature. 1991;349:38–44. doi: 10.1038/349038a0. [DOI] [PubMed] [Google Scholar]
  • [10].Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–526. doi: 10.1016/0092-8674(92)90519-I. [DOI] [PubMed] [Google Scholar]
  • [11].Quan M, Chen J, Zhang D. Exploring the secrets of long noncoding RNAs. Int J Mol Sci. 2015;16:5467–5496. doi: 10.3390/ijms16035467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Geisler S, Coller J. RNA in unexpected places: long non-coding RNA functions in diverse cellular contexts. Nat Rev Mol Cell Biol. 2013;14:699–712. doi: 10.1038/nrm3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482:339–346. doi: 10.1038/nature10887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Friedman RC, Farh KKH, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19:92–105. doi: 10.1101/gr.082701.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].O’Connell RM, Rao DS, Chaudhuri AA, Baltimore D. Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 2010;10:111–122. doi: 10.1038/nri2708. [DOI] [PubMed] [Google Scholar]
  • [16].Bueno MJ, De Castro IP, Malumbres M. Control of cell proliferation pathways by microRNAs. Cell Cycle. 2008;7:3143–3148. doi: 10.4161/cc.7.20.6833. [DOI] [PubMed] [Google Scholar]
  • [17].Kedde M, Agami R. Interplay between microRNAs and RNA-binding proteins determines developmental processes. Cell Cycle. 2008;7:899–903. doi: 10.4161/cc.7.7.5644. (5644 [pii]) [DOI] [PubMed] [Google Scholar]
  • [18].Orang AV, Safaralizadeh R, Kazemzadeh-Bavili M. Mechanisms of miRNA-mediated gene regulation from common downregulation to mRNA-specific upregulation. Int J Genom. 2014;2014 doi: 10.1155/2014/970607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Parkes JE, Day PJ, Chinoy H, Lamb JA. The role of microRNAs in the idiopathic inflammatory myopathies. Curr Opin Rheumatol. 2015;27:608–615. doi: 10.1097/BOR.0000000000000225. [DOI] [PubMed] [Google Scholar]
  • [20].Gurha P. MicroRNAs in cardiovascular disease. Curr Opin Cardiol. 2016;31:1–6. doi: 10.3410/M3-10. [DOI] [PubMed] [Google Scholar]
  • [21].Huang W, Lin J, Zhang H. miR-126: a novel regulator in colon cancer (Review) Biomed Rep. 2015:131–134. doi: 10.3892/br.2015.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Navarro L, Dunoyer P, Jay F, Arnold B, Dharmasiri N, Estelle M, et al. A plant miRNA contributes to antibacterial resistance by repressing auxin signaling. Science. 2006;3121(80):436–439. doi: 10.1126/science.1126088. [DOI] [PubMed] [Google Scholar]
  • [23].Taganov KD, Boldin MP, Chang K-J Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 2006;103:12481–12486. doi: 10.1073/pnas.0605298103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Jin W, Ibeagha-Awemu EM, Liang G, Beaudoin F, Zhao X, Guan LL. Transcriptome microRNA profiling of bovine mammary epithelial cells challenged with Escherichia coli or Staphylococcus aureus bacteria reveals pathogen directed microRNA expression profiles. BMC Genom. 2014;15:181. doi: 10.1186/1471-2164-15-181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Zeng FR, Tang LJ, He Y, Garcia RC. An update on the role of miRNA-155 in pathogenic microbial infections. Microbes Infect. 2015;17:613–621. doi: 10.1016/j.micinf.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • [26].Staedel C, Darfeuille F. MicroRNAs and bacterial infection. Cell Microbiol. 2013;15:1496–1507. doi: 10.1111/cmi.12159. [DOI] [PubMed] [Google Scholar]
  • [27].Eulalio A, Schulte L, Vogel J. The mammalian microRNA response to bacterial infections. RNA Biol. 2012;9:742–750. doi: 10.4161/rna.20018. [DOI] [PubMed] [Google Scholar]
  • [28].Maudet C, Mano M, Eulalio A. MicroRNAs in the interaction between host and bacterial pathogens. FEBS Lett. 2014;588:4140–4147. doi: 10.1016/j.febslet.2014.08.002. [DOI] [PubMed] [Google Scholar]
  • [29].Siddle KJ, Tailleux L, Deschamps M, Loh YHE, Deluen C, Gicquel B, et al. Bacterial infection drives the expression dynamics of microRNAs and their isomiRs. PLoS Genet. 2015;11:1–25. doi: 10.1371/journal.pgen.1005064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Griffiths-Jones S, Hui JHL, Marco A, Ronshaugen M. MicroRNA evolution by arm switching. EMBO Rep. 2011;12:172–177. doi: 10.1038/embor.2010.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Schulte LN, Westermann AJ, Vogel J. Differential activation and functional specialization of miR-146 and miR-155 in innate immune sensing. Nucleic Acids Res. 2013;41:542–553. doi: 10.1093/nar/gks1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Duerr CU, Hornef MW. The mammalian intestinal epithelium as integral player in the establishment and maintenance of host-microbial homeostasis. Semin Immunol. 2012;24:25–35. doi: 10.1016/j.smim.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • [33].O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA. 2007;104:1604–1609. doi: 10.1073/pnas.0610731104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Tili E, Michaille J-J, Cimino A, Costinean S, Dumitru CD, Adair B, et al. Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimulation and their possible roles in regulating the response to endotoxin shock. J Immunol. 2007;179:5082–5089. doi: 10.4049/jimmunol.179.8.5082. [DOI] [PubMed] [Google Scholar]
  • [35].O’Connell RM, Chaudhuri AA, Rao DS, Baltimore D. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc Natl Acad Sci USA. 2009;106:7113–7118. doi: 10.1073/pnas.0902636106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Cremer TJ, Ravneberg DH, Clay CD, Piper-Hunter MG, Marsh CB, Elton TS, et al. MiR-155 induction by F. novicida but not the virulent F. tularensis results in SHIP down-regulation and enhanced pro-inflammatory cytokine response. PLoS One. 2009;4 doi: 10.1371/journal.pone.0008508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Lu LF, Thai TH, Calado DP, Chaudhry A, Kubo M, Tanaka K, et al. Foxp3-dependent MicroRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein. Immunity. 2009;30:80–91. doi: 10.1016/j.immuni.2008.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Billeter AT, Hellmann J, Roberts H, Druen D, Gardner SA, Sarojini H, et al. MicroRNA-155 potentiates the inflammatory response in hypothermia by suppressing IL-10 production. FASEB J. 2014;28:5322–5336. doi: 10.1096/fj.14-258335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Kurowska-Stolarska M, Alivernini S, Ballantine LE, Asquith DL, Millar NL, Gilchrist DS, et al. MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proc Natl Acad Sci USA. 2011;108:11193–11198. doi: 10.1073/pnas.1019536108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].McCoy CE, Sheedy FJ, Qualls JE, Doyle SL, Quinn SR, Murray PJ, et al. IL-10 inhibits miR-155 induction by toll-like receptors. J Biol Chem. 2010;285:20492–20498. doi: 10.1074/jbc.M110.102111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Hu R, Kagele DA, Huffaker TB, Runtsch MC, Alexander M, Liu J, et al. MiR-155 promotes T follicular helper cell accumulation during chronic, low-grade inflammation. Immunity. 2014;41:605–619. doi: 10.1016/j.immuni.2014.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Wan G, Xie W, Liu Z, Xu W, Lao Y, Huang N, et al. Hypoxia-induced MIR155 is a potent autophagy inducer by targeting multiple players in the MTOR pathway. Autophagy. 2014;10:70–79. doi: 10.4161/auto.26534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Wang J, Yang K, Zhou L, Wu M, Wu Y, Zhu M, et al. MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog. 2013;9 doi: 10.1371/journal.ppat.1003697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Clare S, John V, Walker AW, Hill JL, Abreu-Goodger C, Hale C, et al. Enhanced susceptibility to citrobacter rodentium infection in microRNA-155-deficient mice. Infect Immun. 2013;81:723–732. doi: 10.1128/IAI.00969-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Lind EF, Elford AR, Ohashi PS. Micro-RNA 155 is required for optimal CD8+ T cell responses to acute viral and intracellular bacterial challenges. J Immunol. 2013;190:1210–1216. doi: 10.4049/jimmunol.1202700. [DOI] [PubMed] [Google Scholar]
  • [46].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;981(80):608–611. doi: 10.1126/science.1139253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Imaizumi T, Tanaka H, Tajima A, Yokono Y, Matsumiya T, Yoshida H, et al. IFN-g and TNF-a synergistically induce microRNA-155 which regulates TAB2/IP-10 expression in human mesangial cells. Am J Nephrol. 2010;32:462–468. doi: 10.1159/000321365. [DOI] [PubMed] [Google Scholar]
  • [48].Chen XM, Splinter PL, O’Hara SP, LaRusso NF. A cellular micro-RNA, let-7i, regulates toll-like receptor 4 expression and contributes to cholangiocyte immune responses against Cryptosporidium parvum infection. J Biol Chem. 2007;282:28929–28938. doi: 10.1074/jbc.M702633200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Izar B, Mannala GK, Mraheil MA, Chakraborty T, Hain T. MicroRNA response to listeria monocytogenes infection in epithelial cells. Int J Mol Sci. 2012;13:1173–1185. doi: 10.3390/ijms13011173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Schulte LN, Eulalio A, Mollenkopf H-J, Reinhardt R, Vogel J. Analysis of the host microRNA response to Salmonella uncovers the control of major cytokines by the let-7 family. EMBO J. 2011;30:1977–1989. doi: 10.1038/emboj.2011.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Teng G gen, Wang W hong, Dai Y, Wang S jun, Chu Y xiang, Li J. Let-7b Is involved in the inflammation and immune responses associated with Helicobacter pylori infection by targeting toll-like receptor 4. PLoS One. 2013;8 doi: 10.1371/journal.pone.0056709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Hu G, Gong AY, Roth AL, Huang BQ, Ward HD, Zhu G, et al. Release of luminal exosomes contributes to TLR4-mediated epithelial antimicrobial defense. PLoS Pathog. 2013;9 doi: 10.1371/journal.ppat.1003261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].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;139:693–706. doi: 10.1016/j.cell.2009.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, et al. MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer. 2011;128:361–370. doi: 10.1002/ijc.25348. [DOI] [PubMed] [Google Scholar]
  • [55].Hayashi Y, Tsujii M, Wang J, Kondo J, Akasaka T, Jin Y, et al. CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis. Gut. 2012:1536–1546. doi: 10.1136/gutjnl-2011-301625. [DOI] [PubMed] [Google Scholar]
  • [56].Kumar M, Sahu SK, Kumar R, Subuddhi A, Maji RK, Jana K, et al. MicroRNA let-7 modulates the immune response to mycobacterium tuberculosis infection via control of A20, an inhibitor of the NF-kB pathway. Cell Host Microbe. 2015;17:345–356. doi: 10.1016/j.chom.2015.01.007. [DOI] [PubMed] [Google Scholar]
  • [57].Marcais A, Blevins R, Graumann J, Feytout A, Dharmalingam G, Carroll T, et al. microRNA-mediated regulation of mTOR complex components facilitates discrimination between activation and anergy in CD4 T cells. J Exp Med. 2014;211:2281–2295. doi: 10.1084/jem.20132059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Ma F, Xu S, Liu X, Zhang Q, Xu X, Liu M, et al. The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-gamma. Nat Immunol. 2011;12:861–869. doi: 10.1038/ni.2073. [DOI] [PubMed] [Google Scholar]
  • [59].Schnitger AKD, Machova A, Mueller RU, Androulidaki A, Schermer B, Pasparakis M, et al. Listeria monocytogenes infection in macrophages induces vacuolar-dependent host miRNA response. PLoS One. 2011;6 doi: 10.1371/journal.pone.0027435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Liu S, da Cunha AP, Rezende RM, Cialic R, Wei Z, Bry L, et al. The host shapes the gut microbiota via fecal microRNA. Cell Host Microbe. 2016;19:32–43. doi: 10.1016/j.chom.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56:1733–1741. doi: 10.1373/clinchem.2010.147405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Alexander M, Hu R, Runtsch MC, Kagele DA, Mosbruger TL, Tolmachova T, et al. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat Commun. 2015;6 doi: 10.1038/ncomms8321. 7321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Wu K, Zhu C, Yao Y, Wang X, Song J, Zhai J. MicroRNA-155-enhanced autophagy in human gastric epithelial cell in response to Helicobacter pylori. Saudi J Gastroenterol. 2016;22:30–36. doi: 10.4103/1319-3767.173756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Benz F, Roy S, Trautwein C, Roderburg C, Luedde T. Circulating MicroRNAs as biomarkers for sepsis. Int J Mol Sci. 2016 doi: 10.3390/ijms17010078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Chakraborty C, Das S. Profiling cell-free and circulating miRNA: a clinical diagnostic tool for different cancers. Tumor Biol. 2016 doi: 10.1007/s13277-016-4907-3. [DOI] [PubMed] [Google Scholar]
  • [66].Guay C, Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nat Rev Endocrinol. 2013;9:513–521. doi: 10.1038/nrendo.2013.86. [DOI] [PubMed] [Google Scholar]
  • [67].Ueberberg B, Kohns M, Mayatepek E, Jacobsen M. Are microRNAs suitable biomarkers of immunity to tuberculosis? Mol Cell Pediatr. 2014;1:8. doi: 10.1186/s40348-014-0008-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Hsieh C-H, Rau C-S, Jeng JC, Chen Y-C, Lu T-H, Wu C-J, et al. Whole blood-derived microRNA signatures in mice exposed to lipopolysaccharides. J Biomed Sci. 2012;19:69. doi: 10.1186/1423-0127-19-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Hsieh C-H, Yang JC-S, Jeng JC, Chen Y-C, Lu T-H, Tzeng S-L, et al. Circulating microRNA signatures in mice exposed to lipoteichoic acid. J Biomed Sci. 2013;20:2. doi: 10.1186/1423-0127-20-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Rau C-S, Wu S-C, Yang J, Lu T-H, Wu Y-C, Chen Y-C, et al. Profiling the circulating miRNAs in mice exposed to gram-positive and gram-negative bacteria by Illumina small RNA deep sequencing. J Biomed Sci. 2015;22:1. doi: 10.1186/s12929-014-0106-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Runtsch MC, Round JL, O’Connell RM. MicroRNAs and the regulation of intestinal homeostasis. Front Genet. 2014;5:1–10. doi: 10.3389/fgene.2014.00347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Archambaud C, Sismeiro O, Toedling J, Soubigou G, Bécavin C, Lechat P. The intestinal microbiota interferes with the microRNA response upon oral listeria infection. MBio. 2013;4:200707–200713. doi: 10.1128/mBio.00707-13. Editor. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Archambaud C, Nahori M, Soubigou G, Bécavin C, Laval L, Lechat P, et al. Impact of lactobacilli on orally acquired listeriosis. Proc Natl Acad Sci U S A. 2012;109:16684–1668. doi: 10.1073/pnas.1212809109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Reddick LE, Alto NM. Bacteria fighting back: how pathogens target and subvert the host innate immune system. Mol Cell. 2014;54:321–328. doi: 10.1016/j.molcel.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Zhang Y-M, Noto JM, Hammond CE, Barth JL, Argraves WS, Backert S, et al. Helicobacter pylori-induced posttranscriptional regulation of H-K-ATPase—subunit gene expression by miRNA. AJP Gastrointest Liver Physiol. 2014;306:G606–G613. doi: 10.1152/ajpgi.00333.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Tang B, Li N, Gu J, Zhuang Y, Li Q, Wang HG, et al. Compromised autophagy by MIR30B benefits the intracellular survival of Helicobacter pylori. Autophagy. 2012;8:1045–1057. doi: 10.4161/auto.20159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Verma S, Mohapatra G, Ahmed SM, Rana S, Jain S, Khalsa JK, et al. Salmonella engages host microRNAs to modulate SUMOylation: a new arsenal for intracellular survival. Mol Cell Biol. 2015 doi: 10.1128/mcb.00397-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Maudet C, Mano M, Sunkavalli U, Sharan M, Giacca M, Förstner KU, et al. Functional high-throughput screening identifies the miR-15 microRNA family as cellular restriction factors for Salmonella infection. Nat Commun. 2014;5 doi: 10.1038/ncomms5718. 4718. [DOI] [PubMed] [Google Scholar]
  • [79].Belair C, Baud J, Chabas S, Sharma CM, Vogel J, Staedel C, et al. Helicobacter pylori interferes with an embryonic stem cell micro RNA cluster to block cell cycle progression. Silence. 2011;2:7. doi: 10.1186/1758-907x-2-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Zhu Y, Jiang Q, Lou X, Ji X, Wen Z, Wu J, et al. MicroRNAs up-regulated by CagA of helicobacter pylori induce intestinal metaplasia of gastric epithelial cells. PLoS One. 2012;7:1–10. doi: 10.1371/journal.pone.0035147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Li N, Tang B, Zhu ED, Li BS, Zhuang Y, Yu S, et al. Increased miR-222 in H. pylori-associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK. FEBS Lett. 2012;586:722–728. doi: 10.1016/j.febslet.2012.01.025. [DOI] [PubMed] [Google Scholar]
  • [82].Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, et al. miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest. 2008;88:1358–1366. doi: 10.1038/labinvest.2008.94. [DOI] [PubMed] [Google Scholar]
  • [83].Zhang T, Yu J, Zhang Y, Li L, Chen Y, Li D, et al. Salmonella enterica serovar enteritidis modulates intestinal epithelial mir-128 levels to decrease macrophage recruitment via macrophage colony-stimulating factor. J Infect Dis. 2014;209:2000–2011. doi: 10.1093/infdis/jiu006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis decreases human macrophage IFN-γ responsiveness through miR-132 and miR-26a. J Immunol. 2014;193:4537–4547. doi: 10.4049/jimmunol.1400124. [DOI] [PubMed] [Google Scholar]
  • [85].Rajaram MVS, Ni B, Morris JD, Brooks MN, Carlson TK, Bakthavachalu B, et al. Mycobacterium tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-activated protein kinase 2 (MK2) and microRNA miR-125b. Proc Natl Acad Sci U S A. 2011;108:17408–17413. doi: 10.1073/pnas.1112660108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Singh Y, Kaul V, Mehra A, Chatterjee S, Tousif S, Dwivedi VP, et al. Mycobacterium tuberculosis controls MicroRNA-99b (miR-99b) expression in infected murine dendritic cells to modulate host immunity. J Biol Chem. 2013;288:5056–5061. doi: 10.1074/jbc.C112.439778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Kumar R, Halder P, Sahu SK, Kumar M, Kumari M, Jana K, et al. Identification of a novel role of ESAT-6-dependent miR-155 induction during infection of macrophages with mycobacterium tuberculosis. Cell Microbiol. 2012;14:1620–1631. doi: 10.1111/j.1462-5822.2012.01827.x. [DOI] [PubMed] [Google Scholar]
  • [88].Yang S, Li F, Jia S, Zhang K, Jiang W, Shang Y, et al. Early secreted antigen ESAT-6 of mycobacterium tuberculosis promotes apoptosis of macrophages via targeting the microrna155-SOCS1 interaction. Cell Physiol Biochem. 2015;35:1276–1288. doi: 10.1159/000373950. [DOI] [PubMed] [Google Scholar]
  • [89].Grundhoff A, Sullivan CS. Virus-encoded microRNAs. Virology. 2011;411:325–343. doi: 10.1016/j.virol.2011.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Liu H, Wang X, Wang H-D, Wu J, Ren J, Meng L, et al. Escherichia coli noncoding RNAs can affect gene expression and physiology of Caenorhabditis elegans. Nat Commun. 2012;3 doi: 10.1038/ncomms2071. 1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Shmaryahu A, Carrasco M, Valenzuela PDT. Prediction of Bacterial microRNAs and possible targets in human cell transcriptome. J Microbiol. 2014;52:482–489. doi: 10.1007/s12275-014-3658-3. [DOI] [PubMed] [Google Scholar]
  • [92].Furuse Y, Finethy R, Saka HA, Xet-Mull AM, Sisk DM, Jurcic Smith KL, et al. Search for MicroRNAs expressed by intracellular bacterial pathogens in infected mammalian cells. PLoS One. 2014;9 doi: 10.1371/journal.pone.0106434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lefevre P, Witham J, Lacroix CE, Cockerill PN, Bonifer C. The LPS-induced transcriptional upregulation of the chicken lysozyme locus involves CTCF eviction and noncoding RNA transcription. Mol Cell. 2008;32:129–139. doi: 10.1016/j.molcel.2008.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Ilott NE, Heward JA, Roux B, Tsitsiou E, Fenwick PS, Lenzi L, et al. Long non-coding RNAs and enhancer RNAs regulate the lipopolysaccharide-induced inflammatory response in human monocytes. Nat Commun. 2014;5 doi: 10.1038/ncomms4979. 3979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Cui H, Xie N, Tan Z, Banerjee S, Thannickal VJ, Abraham E, et al. The human long noncoding RNA, lnc-IL7R, regulates inflammatory response. Eur J Immunol. 2014;44:2085–2095. doi: 10.1097/SHK.0b013e31828fad82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Gomez JA, Wapinski OL, Yang YW, Gopinath S, Monack DM, Chang HY, et al. The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-g locus. Cell. 2013;152:743–754. doi: 10.1016/j.cell.2013.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Mao A-P, Shen J, Zuo Z. Expression and regulation of long noncoding RNAs in TLR4 signaling in mouse macrophages. BMC Genomics. 2015;16:45. doi: 10.1186/s12864-015-1270-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Westermann AJ, Förstner KU, Amman F, Barquist L, Chao Y, Schulte LN, et al. Dual RNA-seq unveils noncoding RNA functions in host-pathogen interactions. Nature. 2016:1–23. doi: 10.1038/nature16547. [DOI] [PubMed] [Google Scholar]
  • [99].Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311–1323. doi: 10.1016/j.cell.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Wu H, Liu J, Li W, Liu G, Li Z. LncRNA-HOTAIR promotes TNF-α production in cardiomyocytes of LPS-induced sepsis mice by activating NF-κB pathway. Biochem Biophys Res Commun. 2016;471:240–246. doi: 10.1016/j.bbrc.2016.01.117. [DOI] [PubMed] [Google Scholar]
  • [101].Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–227. doi: 10.1038/nature07672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341:789–792. doi: 10.1126/science.1240925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM, Chang HY. A mammalian pseudogene lncRNA at the interface of inflammation and antiinflammatory therapeutics. Elife. 2013;2013:1–16. doi: 10.7554/eLife.00762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Pawar K, Hanisch C, Eliseo S, Vera P, Einspanier R, Sharbati S. Down regulated lncRNA MEG3 eliminates mycobacteria in macrophages via autophagy. Sci Rep. 2016:1–13. doi: 10.1038/srep19416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the rosetta stone of a hidden RNA language? Cell. 2011;146:353–358. doi: 10.1016/j.cell.2011.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Du Z, Sun T, Hacisuleyman E, Fei T, Wang X, Brown M, et al. Integrative analyses reveal a long noncoding RNA-mediated sponge regulatory network in prostate cancer. Nat Commun. 2016;7 doi: 10.1038/ncomms10982. 10982. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES