ABSTRACT
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
INTRODUCTION
MicroRNAs (miRNAs) are a class of small noncoding RNAs (typically 20 to 22 nucleotides long) that posttranscriptionally regulate the expression of target mRNAs exhibiting partially complementary binding sites (1). miRNAs are found in a wide range of organisms, including animals, plants, and viruses. According to the latest release of miRBase (http://www.mirbase.org/; release 22 March 2018), a total of 48,885 mature miRNAs are currently annotated in 271 species; 2,694 mature miRNAs are annotated in the human genome.
miRNA biogenesis is a well-described multistep process (reviewed in reference 2) (Fig. 1). Typically, the primary miRNA transcripts (pri-miRNAs) are long, capped, and polyadenylated RNA molecules containing hairpin structures transcribed by RNA polymerase II. The pri-miRNA is then processed in the nucleus by the microprocessor complex (comprising the RNase III enzyme Drosha and the double-stranded RNA binding protein DGCR8) into a 60- to 100-nucleotide hairpin precursor miRNA. Exportin-5 mediates the transport of the precursor miRNA from the nucleus to the cytoplasm, where it is further processed into a miRNA duplex of approximately 20 bp by Dicer, an enzyme that also belongs to the RNase III family, in complex with the RNA binding protein TRBP. One of the strands of the miRNA is then loaded into the miRNA-induced silencing complex, which contains, among multiple components, an Argonaute (AGO) protein that binds to the mature miRNA. Target mRNAs are identified by base-pairing between the miRNA and mRNA, usually involving nucleotides 2 to 7/8 of the 5′ end of the miRNA (known as the seed-region). miRNAs repress target gene expression by a combination of mechanisms involving translation repression and target mRNA degradation following deadenylation and decapping (reviewed in reference 3).
FIGURE 1.
Overview of the canonical miRNA biogenesis pathway. miRNA genes are transcribed as pri-miRNAs by RNA polymerase II. The main proteins involved in the multistep miRNA processing are indicated. Repression of target gene expression occurs through inhibition of translation and mRNA degradation.
Based on the high number of miRNAs identified in the human genome, bioinformatic predictions, and experimental identification of hundreds of target mRNAs for a number of individual miRNAs, it has been suggested that approximately 60% of the human genome is under the regulation of miRNAs (4), though this may be an underestimation. Consistent with a pervasive role of miRNAs in the control of gene expression, miRNAs have been shown to regulate countless fundamental biological processes (e.g., cellular proliferation, differentiation, apoptosis [5–7]), and miRNA dysregulation has been implicated in a wide spectrum of human diseases (e.g., cancer, cardiovascular disorders [8, 9]).
In addition to these functions, it is now clear that miRNAs have a preponderant role during infections caused by viruses, parasites, fungi, and bacterial pathogens. Initial work addressing miRNA function in the context of infection focused on viruses (reviewed in reference 10). In these seminal studies, the expression of miRNAs encoded by DNA viruses (herpesvirus, polyomavirus, and adenovirus) was revealed; this was first shown for Epstein-Barr virus (11). Equally important, the repertoire of host miRNAs is vastly changed in response to viral infections. Indeed, viral and host miRNAs modulate multiple processes relevant to infection, ranging from virus replication and propagation to host antiviral responses and/or promotion of the viral life cycle through complex regulatory pathways.
During the past decade, miRNAs have also emerged as powerful players in the interaction of bacterial pathogens with the host. It has become clear that bacteria have evolved sophisticated mechanisms of harnessing host miRNAs to create an immune-tolerant environment and/or to modulate host pathways for their own benefit, promoting pathogen survival, replication, and latency/persistence. From the host perspective, miRNAs are an integral part of an effective immunological response, relevant for the control and clearance of infection.
In this article, we summarize a growing body of literature on the modulation and function of host miRNAs in the context of infection by bacterial pathogens.
miRNA RESPONSE TO BACTERIAL INFECTION
The first evidence of the regulatory role of miRNAs in response to bacterial infections was obtained in 2006 from studies performed with plants. In this seminal work, Navarro and colleagues described the increased expression of miR-393a as an important mediator of the resistance of Arabidopsis thaliana to infection by the extracellular pathogen Pseudomonas syringae (12). Specifically, the authors showed that recognition of a flagellin-derived peptide from P. syringae by the FLS2 receptor of A. thaliana induces the transcription of miR-393a, which in turn represses the expression of three F-box auxin receptors. This blunts signaling by auxin, a plant hormone that negatively regulates the plant immune system, ultimately restraining bacterial spreading and enhancing plant resistance to P. syringae infection (12). Interestingly, the authors later demonstrated that P. syringae is able to counteract this miRNA-mediated antibacterial response through the secretion into host cells of effector proteins that suppress transcription, biogenesis, stability, and activity of pathogen-associated molecular pattern-responsive miRNAs (13).
In mammalian cells, the first report implicating miRNAs in the innate immune response to bacterial components resulted from the pioneering study of Taganov and colleagues (14). These authors investigated the expression of a panel of 200 miRNAs in human monocytes stimulated with lipopolysaccharide (LPS), a major component of the outer membrane of Gram-negative bacteria that is sensed by the Toll-like receptor 4 (TLR4). This work led to the identification of the first three endotoxin-responsive miRNAs (miR-146a/b, miR-132, and miR-155). Interestingly, the characterization of miR-146a/b targets uncovered several components of the TLR4 signaling cascade, suggesting the existence of a negative-feedback loop that might protect host cells from an excessive inflammatory response.
Subsequent studies established miRNA regulation upon infection as a common phenomenon (summarized in Table 1), with implications for multiple host cell functions ranging from the control of the immune response and autophagy to cell cycle and cell death, among others (Fig. 2). The main findings obtained in this context are described below, grouped by bacterial pathogen.
TABLE 1.
Host cell miRNAs regulated upon infection by bacterial pathogensa
Bacterium | miRNA | Regulation | Cell/tissue | Reference |
---|---|---|---|---|
Salmonella enterica | let-7 family | Down | HeLa, RAW264.7 | 17 |
miR-1, miR-125a/b, miR-130, miR-148 | Down | Piglet MLN | 28 | |
miR-15 family | Down | HeLa | 22 | |
miR-21, miR-29a/b, miR-146a/b | Up | Zebrafish embryos | 25 | |
miR-21, miR-146a/b, miR-155 | Up | RAW264.7 | 17 | |
miR-26, miR-143 | Up | Pig whole blood | 30 | |
miR-29a | Up | Piglet ileal tissue | 27 | |
miR-30c/e | Up | HeLa, J774 | 24 | |
miR-34a-5p, miR-215-5p, miR-1662 | Down | Chicken cecum | 32 | |
miR-101-3p | Down | Chicken spleen tissue | 19 | |
miR-125b-5p, miR-1416-5p | Up | Chicken cecum | 32 | |
miR-128 | Up | HT-29, murine small intestine and colon tissue | 31 | |
miR-146a/b | Up | THP-1 | 18 | |
miR-155 | Up | Chicken spleen tissue | 19 | |
miR-193a-5p, miR-3525 | Up | Chicken cecum | 26 | |
miR-214 | Down | Piglet whole blood | 29 | |
miR-331-3p | Up | Piglet whole blood | 29 | |
miR-1308 | Up | HeLa | 17 | |
Helicobacter pylori | let-7 family, miR-103, miR-125a, miR-130a, miR-491-5p, miR-500, miR-532 | Down | AZ-521, human gastric mucosa | 58 |
let-7b | Down | AGS, GES-1, human gastric mucosa | 57 | |
miR-21 | Up | AGS, human gastric mucosa | 35 | |
miR-30b | Up | AGS, human gastric mucosa | 62 | |
miR-30d | Up | AGS, GES-1 | 63 | |
miR-101 | Down | GES-1, MKN-45, SGC-7901, primary gastric cells | 38 | |
miR-143-3p | Up | Human gastric mucosa | 47 | |
miR-146a | Up | MNK-45, GES-1, HGC-27, AGS, human gastric mucosa | 56 | |
miR-146a | Up | GES-1 | 55 | |
miR-146a | Up | HGC-27 | 54 | |
miR-146a, miR-16 | Up | GES-1 | 52 | |
miR-146a, miR-155 | Up | Human gastric mucosa | 50 | |
miR-152, miR-200b | Down | AGS, human gastric mucosa | 59 | |
miR-155 | Up | AGS, GES-1, MKN-45, human gastric mucosa | 52 | |
miR-155 | Up | Jurkat, CCRF-CEM, AGS, MKN-74, J774A, murine BMDMs, human gastric mucosa | 48 | |
miR-155 | Up | Human gastric mucosa | 56 | |
miR-155 | Up | Murine gastric mucosa | 51 | |
miR-155 | Up | AZ-521 | 58 | |
miR-155 | Up | J774A.1, murine BMDMs | 49 | |
miR-210 | Down | Mongolian gerbil and human gastric mucosa | 39 | |
miR-212-3p, miR-361-3p | Down | Human esophagus tissue, HET-1A, OE33 | 45 | |
miR-222 | Up | HGC-27, AGS, BGC-823, SGC-7901, GES-1 | 36 | |
miR-222 | Up | Human gastric mucosa | 37 | |
miR-223 | Up | Human gastric mucosa | 58 | |
miR-223 | Up | THP-1, AGS | 53 | |
miR-320 | Down | MKN-28, AGS | 40 | |
miR-370 | Down | AGS, human and murine gastric mucosa | 41 | |
miR-371, miR-372, miR-373 | Down | AGS | 46 | |
miR-584, miR-1290 | Up | AGS | 42 | |
miR-1289 | Up | AGS, human gastric mucosa | 64 | |
miR-4270, miR-4459 | Down | Human MDMs | 61 | |
Mycobacterium spp. | let-7e, miR-29a, miR-146a, miR-155, miR-886-5p | Up | Human MDMs | 75 |
let-7f | Down | RAW264.7; human MDMs; murine BMDMs; lung, spleen, and lymph node tissue | 79 | |
miR-15a, miR-21-3p, miR-22-3p, miR-23a, miR-30b-5p, miR-142-5p, miR-146a/b | Up | Bovine alveolar macrophages | 100 | |
miR-17-5p | Down | RAW264.7; murine BMDMs; lung, spleen, and lymph node tissue | 90 | |
miR-17-5p | Up | RAW264.7 | 91 | |
miR-20a | Up | RAW264.7 | 98 | |
miR-20b | Down | Human MDMs, murine lung tissue | 88 | |
miR-21 | Up | RAW264.7 | 86 | |
miR-21 | Up | Human PBMCs, skin samples | 82 | |
miR-21, miR-26a, miR-29a, miR-142-3p | Down | Human T-cells, peripheral blood | 84 | |
miR-26a | Down | RAW264.7; THP1; human MDMs; murine BMDMs; lung, spleen, and lymph nodes | 101 | |
miR-27a | Up | Human PBMCs, murine lung tissue, peritoneal macrophages | 95 | |
miR-27b | Up | RAW264.7, murine BMDMs, lung and spleen tissue | 78 | |
miR-29a | Up | Human serum, sputum | 83 | |
miR-29a/b | Down | Murine splenocytes, T-cells | 85 | |
miR-30a | Up | THP1, human alveolar macrophages | 92 | |
miR-30a/e, miR-155, miR-1275, miR-3178, miR-3665, miR-4484, miR-4497, miR-4668-5p | Up | THP1 | 68 | |
miR-33, miR-33* | Up | Murine peritoneal and alveolar macrophages, THP1, BMDMs | 94 | |
miR-106b-5p | Up | Human MDMs | 102 | |
miR-125a-3p | Up | RAW264.7, murine BMDMs | 93 | |
miR-125b, miR-146a, miR-155 | Up | RAW264.7, THP1, murine peritoneal macrophages | 72 | |
miR-125b, miR-155 | Up | Human MDMs | 73 | |
miR-142-3p | Up | J774A.1, human MDMs | 81 | |
miR-144-3p | Up | RAW264.7 | 97 | |
miR-144-5p | Up | THP1, human MDMs, lung and lymph nodes tissue | 96 | |
miR-155 | Up | Human PBMCs, murine spleen, lymph node and peritoneal macrophages | 66 | |
miR-155 | Up | RAW264.7, murine BMDMs | 67 | |
miR-155 | Up | RAW264.7, murine lung tissue and BMDMs | 76 | |
miR-155 | Up | THP1, human PMBCs | 71 | |
miR-155 | Up | Human dendritic cells | 70 | |
miR-155 | Up | Murine BMDMs | 74 | |
miR-155 | Up | RAW264.7 | 77 | |
miR-199a | Up | J774A.1, murine BMDMs, lung and spleen tissue | 99 | |
miR-223 | Up | Murine whole blood and lung tissue, human peripheral blood and lung tissue | 80 | |
miR-582-5p | Up | Human PBMCs | 87 | |
Listeria monocytogenes | miR-16, miR-146b, miR-155 | Up | Caco-2 | 107 |
miR-21 | Up | Murine BMDMs | 106 | |
miR-29a/b | Down | Murine splenocytes and NK cells | 85 | |
miR-125a-3p/-5p, miR-146a, miR-155, miR-149 | Up | Murine BMDMs | 104 | |
miR-143, miR-148a, miR-194, miR-200b/c, miR-378 | Down | Murine ileal tissue | 113 | |
miR-145, let-7a1 | Down | Caco-2 | 107 | |
miR-192, miR-200b, miR-215 | Down | Murine ileal tissue | 112 | |
Francisella spp. | miR-133a, miR-146a, miR-150, miR-155, miR-886-5p | Up | Human MDMs | 114 |
miR-155 | Up | Human PBMCs, murine lung, liver and spleen tissue | 115 | |
Citrobacter rodentium | miR-7a, miR-17, miR-20a, miR-21, miR-142-3p, miR-203 | Up | Murine colonic crypts | 117 |
Staphylococcus aureus | miR-15b-5p | Up | Human and porcine skin tissue | 121 |
miR-20b, miR-31, miR-155, miR-182, miR-222 | Up | Murine lung monocytes | 118 | |
miR-24 | Down | U937, RAW264.7 | 119 | |
Pseudomonas aeruginosa | miR-26a-5p, miR-155, miR-182-5p, miR-200c-3p, miR-294-3p, miR-302b-3p, miR-495-3p, miR-669k-3p | Up | MH-S | 124 |
miR-302b | Up | MLE-12, MH-S, murine lung tissue | 124 | |
Chlamydia spp. | miR-16, miR-23b, miR-30c/e, miR-125b-5p, miR-135a, miR-182, miR-183, miR-214 | Down | Murine genital tract tissue | 130 |
miR-30c-5p | Up | HUVEC, hFIMB, HFF | 127 | |
miR-100-5p, miR-200a-3p, miR-200b-3p/-5p, miR-411-5p | Down | Murine genital tract tissue | 131 | |
miR-132, miR-142-3p, miR-147-3p, miR-149-3p, miR-212-3p | Up | Murine genital tract tissue | 131 | |
miR-146, miR-451 | Up | Murine genital tract tissue | 130 | |
miR-155 | Up | Murine dendritic cells | 129 | |
miR-182, miR-183 | Up | Murine splenic T-cells | 129 | |
miR-214 | Down | Murine genital tract tissue | 128 | |
Brucella spp. | let-7b, miR-92a, miR-99a, miR-142-5p, miR-181b, miR-1981 | Up | RAW264.7 | 132 |
miR-93, miR-151-3p | Down | RAW264.7 | 132 | |
miR-125b-5p | Down | RAW264.7 | 133 | |
miR-130a-3p | Up | PAM | 134 | |
miR-146a, miR-181a/b, miR-301a-3p | Up | RAW264.7, PAM | 134 | |
miR-351-5p | Up | RAW264.7 | 134 | |
Shigella flexneri | miR-29b-2-5p | Down | HeLa | 135 |
Abbreviations: AGS, •••; BMDM, bone marrow-derived macrophages; CCRF-CEM, •••; HFF, primary human foreskin fibroblasts; hFIMB, human fallopian tube fimbriae cells; HUVEC, •••; MDM, monocyte-derived macrophages; MH-S, •••; MLN, mesenteric lymph node; PAM, •••; PBMC, peripheral blood mononuclear cells.
FIGURE 2.
Regulation of miRNAs upon infection by bacterial pathogens impacts multiple crucial host cell functions. miRNA modulation upon infection has been shown to be an integral part of the host response or a mechanism exploited by bacteria to promote infection.
Salmonella enterica
S. enterica remains a leading cause of gastroenteritis worldwide, with S. enterica subspecies enterica serovar Typhimurium (S. Typhimurium) being one of the most frequent serovars causing foodborne disease (15). Salmonella spp. can infect phagocytic and nonphagocytic cells in humans as well as in a wide range of domestic and wild animals (16).
Host miRNA regulation of S. Typhimurium infection was first shown in mouse macrophages, where miR-155, miR-146a/b, and miR-21 were strongly induced (17). Interestingly, regulation of these miRNAs was also observed with S. Typhimurium mutant strains defective in cell invasion (ΔSPI-1) and replication (ΔSPI-2), as well as upon treatment with purified Salmonella LPS, showing that sensing of extracellular stimuli triggers the regulation of these miRNAs. Similar results were obtained in human monocytes (18). These studies and additional findings have singled miR-155 as a key immune miRNA supporting the proinflammatory host response against infection (17, 19). However, miR-155 also participates in negative-feedback regulation of the proinflammatory signaling, ultimately protecting the host from a potentially damaging inflammation overreaction (20). Moreover, this miRNA was shown to be essential for normal immune function, particularly of dendritic cells and B and T lymphocytes, and as part of immunization with attenuated S. Typhimurium (21).
By comparing miRNome (the full spectrum of expressed miRNAs) changes in macrophages and epithelial cells, Schulte et al. showed that members of the let-7 miRNA family are downregulated in both cell types, revealing a common denominator of the phagocytic and nonphagocytic cell response to S. Typhimurium infection (17). Interestingly, the let-7 family was shown to target two major cytokines—the anti-inflammatory cytokine interleukin 10 (IL-10) and the proinflammatory cytokine IL-6. The decreased let-7 expression upon infection and consequent derepression of cytokines with opposing effects likely contributes to a balanced inflammatory response to S. Typhimurium infection.
In addition to participating in the host response to S. Typhimurium infection, miRNA regulation can be harnessed to modulate host physiology, ultimately rendering host cells more permissive to bacterial infection. By employing a high-content screening approach in which the individual effect of ca. 1,000 miRNAs was tested on S. Typhimurium replication in epithelial cells, our group has identified multiple miRNAs controlling bacterial interaction with host cells (22). Among the strongest inhibitors of S. Typhimurium infection, we identified the miR-15 family, which blocks host G1/S cell cycle transition through the repression of cyclin D1. Interestingly, expression of the miR-15 miRNA family is downregulated upon S. Typhimurium infection, thus favoring cell cycle progression and, ultimately, bacterial replication.
SUMOylation, a posttranslational modification pathway central for cell homeostasis (23), is essential for Salmonella intracellular survival, and it has also been shown to be subverted by modulation of host miRNAs. Indeed, S. Typhimurium survival inside host cells relies on the depletion of Ubc-9, a crucial enzyme of the SUMO pathway, which is achieved by the increased levels of miR-30c and miR-30e upon infection (24).
The effect of Salmonella infection on miRNAs has been also studied in vivo in different models of infection, including zebrafish, chicken, and especially pig. In zebrafish embryos, several miRNAs were shown to be upregulated upon S. Typhimurium infection, including the two miR-146 family members, miR-146a/b (25). Interestingly, combined knockdown of these miRNAs in zebrafish embryos infected with S. Typhimurium had a minor effect on the proinflammatory response, but it induced apolipoprotein-mediated lipid transport genes, suggesting a possible function of these miRNAs in regulating lipid metabolism during inflammation (25). In the chicken model, 14 miRNAs were shown to be differentially expressed upon S. Typhimurium infection. From these, the most strongly induced miRNAs were miR-3525 and miR-193a-5p, which might impact the immune response against S. Typhimurium by targeting IL-6 signal transducer and interferon-γ (IFN-γ), respectively (26). In the ileum of piglets, S. Typhimurium upregulated miR-29a expression (27). This miRNA was shown to target Caveolin-2, an inhibitor of the small Rho GTPase Cdc42, thus favoring S. Typhimurium invasion of epithelial cells. In a recent study, Herrera-Uribe et al. identified 110 dysregulated miRNAs in mesenteric lymph nodes of S. Typhimurium-infected piglets (28). Among these, the authors highlighted several miRNAs with predicted and validated targets among major histocompatibility complex class I and II antigen presentation pathways. The miRNA expression profile of whole-blood samples of pigs challenged with S. Typhimurium has also been analyzed, revealing 62 differentially expressed miRNAs (29). From these, the decrease of miR-214 was shown to increase expression of SLC11A1 and LILR-like expression, potentially regulating bacterial replication by actively removing iron from the phagosomal space and by negatively regulating TLR-mediated responses to maintain a balanced inflammatory response, respectively. Induction of miR-331-3p was also validated and suggested to contribute to blocking S. Typhimurium uptake by suppressing the activity of Rho GTPase family members (e.g., RhoA, Cdc42, and Rac1) through targeting of VAV2 (29). Similarly, Yao et al. demonstrated that miR-143, miR-26, and miR-4335 are differentially expressed in whole-blood samples of S. Typhimurium-infected pigs (30).
Although to a lesser extent, the miRNA regulation by other Salmonella serovars, specifically S. enterica subsp. enterica serovar Enteritidis (S. Enteritidis), has also been investigated. Analysis of the miRNome of colon epithelial cells infected with two strains of S. Enteritidis revealed 22 differentially regulated miRNAs (31). miR-128 was shown to be consistently upregulated in S. Enteritidis-infected samples in vitro and in vivo, through a mechanism likely involving activation of the p53 signaling pathway by bacterial secreted proteins. miR-128 was shown to target macrophage colony-stimulating factor (M-CSF), leading to impaired M-CSF-mediated macrophage recruitment and thus benefiting bacterial survival. In chicken, 37 and 32 miRNAs were shown to be differentially regulated between noninfected and S. Enteritidis-infected cecum (32) and spleen (19) samples, respectively. Through integrated analysis of miRNA and transcriptome datasets obtained from spleen samples, two miRNAs were identified in hub positions of the regulatory network—miR-155 and miR-101-3p—likely with important consequences to the immune response upon infection (19).
Helicobacter pylori
H. pylori is a Gram-negative bacterium estimated to chronically colonize the gastric mucosa of more than half of the human population. Unless treated, colonization persists lifelong and represents a key factor in the etiology of gastritis, peptic ulcer, and gastric cancer (33). Similar to other pathogens, H. pylori pathogenesis depends on its bacterial virulence factors, such as the cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), which affect a multitude of host cell pathways (34).
The first evidence that bacterial pathogens can affect the expression of mammalian host miRNAs was obtained in a pioneering study by Zhang and colleagues, who used H. pylori as a model pathogen (35). This study demonstrated that miR-21 is upregulated in gastric epithelium tissue samples of H. pylori-infected patients compared to noninfected samples; similar results were reported in vitro in the human gastric cancer cell line AGS. Interestingly, miR-21 overexpression was shown to promote cellular proliferation and invasion by targeting RECK, a known tumor suppressor. These results suggested a strong link between miR-21 regulation upon H. pylori infection and the development of gastric cancers (35). Other studies have further explored the connection between regulation of host miRNA expression upon H. pylori infection and the ability of this pathogen to induce tumorigenesis. In one of these studies, H. pylori infection was shown to induce the expression of miR-222, another oncomiR (oncogenic miRNA) that also targets RECK (36). Recently, miR-222 was also shown to directly target the homeodomain-interacting protein kinase-2, promoting cell proliferation and invasion and inhibiting apoptosis of gastric cancer cells (37). Along the same line, miR-101 was shown to be downregulated in H. pylori-infected samples and cultured cells, with similar consequences for cell growth and tumorigenesis, elicited by the derepression of its direct target, the oncogene SOCS2 (38). In addition, DNA methylation of the miR-210 gene promoter was shown to be increased in H. pylori-infected human gastric mucosa samples compared to negative controls, promoting cell proliferation through derepression of STMN1 (involved in the initiation of tumor development) and DIMT1 (a member of the S-adenosylmethionine-dependent methyltransferase superfamily) (39).
Mechanistically, the H. pylori virulence factor CagA was shown to be an important player in H. pylori-induced miRNA expression changes, with 61 miRNAs differentially expressed in a CagA-dependent manner (40). CagA-dependent downregulation of miR-320 upon H. pylori infection results in increased expression of its target MCL1, an antiapoptotic gene, likely contributing to an increased risk of tumorigenesis (40). Similarly, the downregulation of miR-370, in a CagA-dependent manner, leads to increased expression of FoxM1, a transcription factor involved in promoting cell cycle progression (41). CagA was also shown to stimulate expression of miRNAs, specifically of miR-584 and miR-1290. These miRNAs have as a common target FOXA1, which is an important negative regulator of epithelial-mesenchymal transition, critical for the development of cancer metastasis (42).
Although H. pylori primarily affects the stomach mucosa, its presence has also been associated with other pathological conditions, such as Barrett’s esophageal disease and esophagus adenocarcinoma (43, 44). In the context of esophageal epithelial cells, Teng et al. showed that H. pylori decreases miR-212-3p and miR-361-3p expression, leading to derepression of their targets, COX2 and CDX2, respectively, two oncoproteins associated with transformation of esophageal epithelial cells (45).
Although a large body of literature coherently supports the involvement of miRNA regulation during H. pylori infection to promote cell proliferation and, ultimately, favor tumorigenesis, H. pylori has also been shown to block cell cycle progression and cell proliferation in gastric epithelial cells through the regulation of miRNAs (46, 47). Indeed, CagA-dependent downregulation of miR-372 and miR-373 in H. pylori-infected cells increases expression of their target LATS2, a tumor suppressor, leading to G1 cell cycle arrest (46). Recently, Wang et al. analyzed miRNA expression in gastric cancer patients, comparing H. pylori-positive and -negative subgroups. Among the 53 miRNAs differentially expressed in these samples, the authors focused on miR-143-3p, which was the most upregulated miRNA in H. pylori-positive gastric cancer tissues. The authors demonstrated that high levels of miR-143-3p dampen cell growth, migration, and invasion through the direct targeting of AKT2 (47), a pro-survival protein that is frequently overexpressed in cancer. Although these studies reveal an unexpected facet of H. pylori, i.e., bacterial-induced host cell cycle arrest and inhibition of cell proliferation, it is conceivable that this can be relevant to inhibit gastric epithelium renewal and therefore constitute a host defense mechanism against infection.
H. pylori infection elicits a strong immune response, triggering the expression of diverse cytokines and chemokines by gastric epithelial cells. Along the same line, several studies have reported the deregulation of immune-related miRNAs in response to H. pylori infection. For example, miR-155 is strongly upregulated upon H. pylori infection, both in vitro (epithelial cells, macrophages, and lymphocytes) and in vivo (human biopsies from infected gastric mucosa) (48–52). Upregulation of miR-155 was shown to be dependent on sensing of H. pylori LPS by TLR4 and subsequent NF-κB pathway activation (49, 52), as well as on the secretion of bacterial proteins by the type-IV secretion system, particularly VacA and γ-glutamyl transpeptidase (48). miR-155 inhibits the release of the proinflammatory cytokines IL-8 and GRO-α, resulting in attenuated NF-κB activity and weakened inflammatory response against H. pylori (52). In macrophages, upregulation of miR-155 was also shown to inhibit DNA damage-induced apoptosis, a mechanism that might increase cell survival upon DNA damage induced by H. pylori (49). Interestingly, miR-155 knockout mice are unable to control H. pylori infection, owing to a deficient pathogen-specific Th1 and Th17 response (51). Pachathundikandi et al. showed that in infected THP-1 monocytes, the inflammasome-forming NLRP3 protein is downregulated upon H. pylori infection through the upregulation of miR-223-3p (53). The immune-related miRNA miR-146a is also upregulated by H. pylori infection, both in vitro and in vivo (52, 54–56). miR-146a negatively regulates TRAF6, PTGS2, and IRAK1 and, consequently, blunts the inflammatory response against H. pylori (54–56). Similar to Salmonella spp., H. pylori infection downregulates the let-7 miRNA family both in vitro and in vivo through its major virulence factor CagA (57, 58). Decreased expression of let-7 family members leads to the derepression of their target TLR4 and to the consequent activation of the NF-κB-dependent inflammatory response. In addition to classical immune-related miRNAs, other miRNAs have been implicated in the regulation of immune function in the context of H. pylori infection. In a recent study, Xie and colleagues showed that H. pylori induces the expression of B7-H1 by decreasing the levels of miR-152 and miR-200b (59). Indeed, B7-H1 functions as a negative regulator of cell-mediated immune response by inhibiting proliferation and inducing apoptosis of activated T-cells (60). In a recent study, Pagliari et al. described that H. pylori can alter miR-4270 expression as a strategy to persist in macrophages and thus evade the immune system (61). miR-4270 controls the expression of the plasma membrane receptor CD300E, compromising the ability of macrophages to expose major histocompatibility complex class II molecules, ultimately compromising recognition by T-cells, and providing a survival niche for the bacterium.
Beyond immune-related functions, H. pylori subverts other cellular processes for its own benefit through miRNA regulation. Upregulation of miR-30b was shown to occur in AGS cells and gastric tissues infected with H. pylori (62). miR-30b negatively regulates the autophagy pathway by targeting Beclin-1 and ATG12, two proteins involved in the formation and maturation of autophagosomes, likely contributing to preventing H. pylori clearance by autophagy. The expression of these two autophagy proteins (along with ATG2B, ATG5, and BNIP3L) was also shown to be repressed by the upregulation of miR-30d induced during H. pylori infection (63). H. pylori can also decrease the gastric acidity by inducing the expression of miR-1289 in a CagA- and soluble lytic transglycolase-dependent manner. miR-1289 represses HKα, a subunit of the gastric H+/K+ ATPase (64), leading to a transient hypochlorhydria that favors H. pylori colonization of the gastric mucosa.
Mycobacterium Species
The genus Mycobacterium includes highly pathogenic species responsible for diseases that remain major public health challenges, such as tuberculosis (caused by Mycobacterium tuberculosis) and leprosy (caused by Mycobacterium leprae). In addition, it also encompasses opportunistic pathogens, such as Mycobacterium avium, that can infect immunocompromised patients. Not surprisingly, miRNAs play relevant roles in the modulation of the host response to mycobacterial infection (reviewed in reference 65).
Several studies have examined the regulation and role of miR-155 in mycobacterial infection using different cellular models, experimental conditions, and Mycobacterium species (66–77). These studies have generated divergent results, particularly regarding the function of miR-155 during infection, though the majority underlined the recurrent function of miR-155 in the regulation of the innate immune response. Adding to the body of knowledge on the role of miR-155 in infection, a recent study by Rothchild and colleagues applied an integrative approach to analyze the miRNA network in infected macrophages, revealing a critical and dual role for miR-155 in M. tuberculosis infection (74): on the one hand, miR-155 promotes the survival of infected macrophages, providing a bacterial niche during the early stages of infection, while on the other hand, it promotes the survival and function of specific T-cells, enabling an effective adaptive immune response. miR-27b, another miRNA with a dual role in mycobacterial infection, has recently been shown to be induced upon infection. miR-27b targets the Bcl-2-associated athanogene 2 protein (Bag2)—which inhibits the production of proinflammatory factors by blunting NF-κB activity but also increases p53-dependent apoptosis and reactive oxygen species production—thus increasing bacterial clearance (78). Other miRNAs have been shown to blunt the proinflammatory response against Mycobacterium to favor bacterial survival. For example, downregulation of let-7f in macrophages infected with M. tuberculosis was shown to derepress the expression of A20 deubiquitinase, a negative regulator of the NF-κB pathway (79). Along the same line, induction of miR-223 reduces the expression of the cytokine IL6 and chemokines CXCL2 and CCL3 upon infection, inhibiting proper recruitment of neutrophils to the infection site and increasing bacterial survival (80).
The miRNA response against Mycobacterium spp. has been shown to be highly dependent on the cell context and bacterial species. For example, while miR-21, miR-29a, and miR-142-3p levels were shown to increase in infected macrophages and serum of tuberculosis patients (75, 81–83), they are downregulated in CD4+ T-cells and samples of peripheral blood of tuberculosis patients (84). Similar to the observations in Listeria (see “Listeria monocytogenes” below), miR-29 is downregulated in natural killer (NK) cells upon M. bovis infection (85), which was shown to enhance bacterial clearance by derepressing IFN-γ expression. In contrast, in macrophages infected with M. avium, miR-29a expression, along with let-7e, is upregulated, resulting in inhibition of apoptosis through a decrease of caspase-3 and caspase-7 (75). Regulation of host cell death is emerging as a recurrent target of miRNAs regulated during mycobacterial infection. Along this line, upon M. tuberculosis infection, upregulation of miR-582-5p, miR-155, and miR-21 was also shown to inhibit apoptosis, by targeting FOX1, FOX3, and NF-κB/Bcl-2, respectively (71, 86, 87). On the other hand, a decrease of miR-20b in M. tuberculosis-infected macrophages derepresses NLRP3, leading to the activation of the NRLP3/caspase-1/IL-1β pathway and, consequently, enhancing inflammation and pyroptosis (88).
Autophagy is a process by which intracellular components are targeted for degradation, and it plays a crucial role in the host defense against intracellular pathogens, including mycobacteria (89). Mycobacterium spp. have evolved strategies based on host miRNA modulation to subvert the autophagy pathway and ensure intracellular survival. Figure 3 illustrates the impact of mycobacteria-regulated miRNAs on autophagy and lysosomal trafficking. The role of miR-155 as a positive or negative regulator of autophagy in the context of mycobacterial infection is still controversial, and it likely depends on cell context and bacterial strain. A proautophagic function of miR-155 through the repression of the negative autophagy regulator RHEB was shown in macrophages (76), compromising M. tuberculosis survival. Conversely, upon M. tuberculosis infection of dendritic cells, upregulated miR-155 was shown to directly target ATG3 and reduce the lipidated form of LC3 in the autophagosomes, indicating negative regulation of autophagy (70). In addition to miR-155, other miRNAs were shown to impinge on autophagic regulation during M. tuberculosis infection. Downregulation of miR-17 leads to an increase of STAT3, a transcriptional activator of the autophagy regulator MCL-1 (90), impairing autophagy and contributing to mycobacterial survival. In the context of M. bovis bacillus Calmette-Guérin (BCG) infection, upregulation of miR-17 has been reported to repress ULK1, an essential autophagy initiation protein, ultimately favoring BCG growth (91). This apparently contradictory result on miR-17 expression might be linked to the different Mycobacterium spp. used. Mycobacterium was also shown to block autophagy by increasing the levels of miR-30a and miR-125a-3p, which target Beclin-1 and UVRAG, respectively, two essential players in autophagy induction (92, 93). M. tuberculosis also induces the expression of miR-33 and miR-33*, which target several players in the autophagy and lysosomal pathways (ATG5, ATG12, LC3B, LAMP1, AMPK, FOXO3, and TFEB) (94). Recently, Liu et al. showed that induction of miR-27a upon infection represses the expression of the endoplasmic reticulum-located calcium transporter CAC-NA2D3, impairing the calcium signaling required for autophagosome formation (95). miR-144-5p and miR-144-3p have also been implicated in the control of autophagy: miR-144-5p is upregulated during M. tuberculosis infection, suppressing autophagy by targeting the autophagy regulator DRAM2 (96), while an increase of miR-144-3p upon BCG infection inhibits autophagy by targeting ATG4a (97). miR-20a and miR-199a are also upregulated upon M. bovis infection and inhibit autophagy through downregulation of ATG7, ATG16L1 (98), and TBK1 (99), respectively. Overall, the majority of the studies reported above support a model in which mycobacteria-modulated host miRNAs have a predominant effect on dampening the autophagic process, with positive consequences for bacterial survival.
FIGURE 3.
Mycobacterium spp.-induced miRNA changes have a strong impact on autophagy. The autophagic flux is controlled by multiple miRNAs that are regulated as a consequence of mycobacterial infection. Most studies report that miRNA modulation inhibits specific steps of the autophagy pathway, thus impairing bacterial degradation.
Mycobacterium spp. can also modulate phagosomal trafficking for their own benefit by modulating several miRNAs. Vegh et al. found a set of miRNAs regulated in bovine macrophages upon infection with M. bovis (6 miRNAs at 24 h postinfection and 40 miRNAs at 48 h postinfection); analysis of the predicted targets of the upregulated miRNAs showed an enrichment for genes related to the lysosome and endocytosis, including members of the membrane trafficking Rab family (100). Downregulation of miR-26a during M. tuberculosis infection was shown to derepress KLF4, preventing bacterial trafficking to lysosomes and reducing inducible nitric oxide synthase production (101). Recently, it was also shown that M. tuberculosis upregulates miR-106b-5p expression, which directly represses cathepsin-S, decreasing the lysosomal enzymatic activity and consequently increasing bacterial intracellular survival (102). miRNAs have also been shown to negatively modulate bacterial uptake, likely as part of the host response to counteract infection. Specifically, an increase of miR-142-3p in response to M. tuberculosis infection represses N-WASP, an actin binding protein essential for phagocytosis (81).
Listeria monocytogenes
L. monocytogenes is a Gram-positive facultative intracellular bacterium that is the causative agent of human listeriosis, a foodborne disease. Listeria can cause gastroenteritis in healthy adults and severe illness in children, the elderly, and other immunocompromised individuals (103).
Genome-wide miRNA profiling in murine bone marrow-derived macrophages infected with L. monocytogenes identified 13 miRNAs that were significantly upregulated, including miRNAs known to regulate the inflammatory response (e.g., miR-155 and miR-146a) (104). In addition, miR-155 has been shown to be essential for the CD8+ T-cell response to L. monocytogenes (105). Recently, the immune-related miRNA miR-21 was also shown to be upregulated following L. monocytogenes infection of bone marrow-derived macrophages (106). Interestingly, miR-21 knockout macrophages showed an increased bacterial burden at early times postinfection (30 min). The identification of the actin-modulating proteins RHOB and MARCKS as putative miR-21 targets suggests a possible role for this miRNA in the negative regulation of phagocytosis during infection.
The effect of L. monocytogenes infection on host miRNA expression has also been addressed in human intestinal epithelial Caco-2 cells (107); in this study, five miRNAs (miR-146b, miR-16, let-7a1, miR-145, and miR-155) were found to be dysregulated upon L. monocytogenes infection. Similar to what has been described in macrophages (104), in Caco-2 cells the expression of miR-155 was induced to a comparable extent by infection with wild-type L. monocytogenes or with a mutant deficient for listeriolysin, a secreted toxin that is essential for bacterial vacuolar escape, among other functions, in L. monocytogenes virulence. Strikingly, upregulation of miR-155 also occurs following incubation of cells with purified listeriolysin (107). Among the miRNAs dysregulated by L. monocytogenes in Caco-2 cells, Izar and colleagues showed the downregulation of miR-145 (107). This miRNA exerts a proinflammatory effect by potentiating the production of the proinflammatory cytokines IL-5 and IL-13 (108) and by decreasing the levels of the anti-inflammatory cytokine IFN-β (109), a cytokine shown to be beneficial to L. monocytogenes infection (reviewed in 110). However, a recent study by Li and colleagues revealed an anti-inflammatory effect of miR-145 by reducing ARF6 expression and downstream signal transduction via NF-κB (111). Taken together, these studies suggest that downregulation of miR-145 could play an essential role in balancing the inflammatory response during Listeria infection.
In vivo, challenge with L. monocytogenes has also been shown to induce significant host miRNome changes. Systemic infection of mice with L. monocytogenes downregulates miR-29 expression in NK cells, CD4+ T-cells, and CD8+ T-cells (85). Ma et al. (85) suggested that decreased miR-29 expression upon infection facilitates IFN-γ production, promoting bacterial clearance and host resistance to L. monocytogenes infection. miR-192, miR-215, and miR-200b were shown to be downregulated in the ileum of orally infected gnotobiotic humanized mice (112). Interestingly, pretreatment with Lactobacillus casei prevented the decrease of expression of these miRNAs upon L. monocytogenes infection. In another study, Archambaud et al. reported the decreased expression of six miRNAs (miR-143, miR-148a, miR-194, miR-200b, miR-200c, and miR-378) in the ileum of conventional mice infected with L. monocytogenes (113). Interestingly, regulation of four of these miRNAs (miR-143, miR-148a, miR-200b, and miR-200c) occurs only in the presence of a normal microbiota, whereas miR-194 is decreased in both the presence and absence of gut microbiota and miR-378 expression is increased in germ-free mice. Overall, these studies reveal an interesting microbiota-dependent regulation of miRNA expression during Listeria infection, suggesting that the microbiota plays an important role in the miRNA host cell response, with relevant implications for infection with bacterial pathogens.
Other Bacterial Pathogens
As described above, analysis of miRNA expression and the characterization of the role of miRNAs are well under way for certain bacterial pathogens. For most bacterial pathogens, however, this analysis has not yet been performed or is just at its inception. We provide below a nonexhaustive review of what is known concerning the role of miRNAs in infection by other bacterial pathogens. Of note, most of these studies have focused on immune response-related miRNAs, most prominently miR-155. For example, miR-155 was shown to be differentially induced upon infection with Francisella tularensis (114, 115), a Gram-negative facultative intracellular bacterium that causes turalemia. Interestingly, miR-155 is strongly induced in an LPS-dependent manner upon infection with F. tularensis subsp. novicida, a low-virulence F. tularensis subspecies, while infection with the highly virulent F. tularensis subsp. tularensis leads to significantly lower miR-155 induction, probably due to the low bioactivity of its LPS. This might contribute to explaining the higher bacterial dissemination and more severe disease caused by F. tularensis subsp. tularensis (114, 115). Indeed, the increase of miR-155 upon F. tularensis subsp. novicida infection represses SHIP (a key negative regulator of the PI3K/Akt pathway), similarly to what was described for Mycobacterium infection (67), thus enhancing the proinflammatory cytokine response and contributing to infection clearance (115). Clare and colleagues demonstrated that miR-155 knockout mice are unable to clear infection by Citrobacter rodentium (116), a murine pathogen that is used as a model for understanding enteropathogenic and enterohemorrhagic Escherichia coli in vivo since it shares several pathogenic mechanisms with these two important human gastrointestinal pathogens. In addition to miR-155, the increased expression of miR-203 in colonic crypts of C. rodentium-infected mice was detected and shown to deregulate Wnt/β-catenin signaling through the direct repression of the Wnt antagonist WIF1 (117). This may contribute to the observed Wnt/β-catenin-dependent crypt hyperplasia in response to C. rodentium infection.
Staphylococcus aureus is another example of a pathogen for which the role of miRNAs during infection is starting to be addressed. Initial reports investigated the impact of staphylococcal enterotoxin B (SEB), an exotoxin responsible for food poisoning and toxic shock, on miRNA expression in lung-infiltrating mononuclear cells isolated from mice exposed to SEB (118). miR-155, the most upregulated miRNA, was shown to enhance the accumulation of IFN-γ by targeting SOCS1, leading to an exacerbated inflammatory response. Interestingly, miR-155 knockout mice are protected against the overinflammation and lung injury elicited by SEB (118), further supporting the relevance of this miRNA on SEB toxicity and, arguably, on S. aureus pathogenicity. Decreased expression of miR-24 has been described in S. aureus-infected macrophages (119). In these cells, reduced miR-24 levels were shown to contribute to a macrophage M1 polarization phenotype via increased CHI3L1 expression. Colonization by S. aureus is frequently linked to chronic skin wounds, particularly in diabetic patients and obese or immunosuppressed individuals (120). The presence of S. aureus in wounds increases miR-15b-5p expression, impairing DNA repair and the inflammatory response, with likely negative consequences to wound healing (121). Using an miR-142 knockout mouse, miR-142-3p and miR-142-5p were also shown to contribute to healing of S. aureus-infected skin wounds (122). Indeed, this phenotype was related to impaired neutrophil chemotactic and phagocytic behavior due to abnormal expression of Rac and Rho GTPases. Recently, the therapeutic potential of miRNA modulation was shown in a model of S. aureus-infected wounds (123). Mice with a specific deletion of miR-223 showed enhanced repair of S. aureus-infected wounds compared to wild-type mice. Similar improvement of wound healing was observed in wild-type mice transplanted with neutrophils lacking miR-223 or treated with inhibitors of miR-223. Interestingly, miR-223 was identified as one of the most highly expressed miRNAs at wound sites during the inflammatory phase.
More systematic studies of miRNA expression have also been performed for a number of pathogens. For example, infection of mouse alveolar macrophages with the opportunistic pathogen Pseudomonas aeruginosa was shown to upregulate eight miRNAs, miR-302b being the most prominently increased (124). Upregulation of this miRNA results in a dampened inflammatory response via targeting of IRAK4, a component of the MyD88 signaling complex critical for NF-κB activation.
The effect of infection with Chlamydia trachomatis, an obligate intracellular pathogen that is the causative agent of trachoma and sexually transmitted infections, on host miRNA expression has been analyzed (reviewed in reference 125). Derrick et al. analyzed miRNA expression profiles of trachoma disease patients, including conjunctival scarring with and without inflammation, revealing 82 differentially expressed miRNAs (126). Pathway analysis revealed an enrichment of genes related to fibrosis and epithelial cell differentiation among the predicted targets of these miRNAs. A recent study by Chowdhury and colleagues analyzed differentially expressed miRNAs in primary human umbilical vein endothelial cells infected with C. trachomatis, revealing miR-30c-5p as one of the most upregulated miRNAs (127). miR-30c-5p upregulation leads to p53-mediated downregulation of Drp1, a mitochondrial fission regulator, thereby inhibiting stress-induced mitochondrial fission and promoting cell survival during infection. In addition, several groups used the mouse pathogen Chlamydia muridarum, a model organism to study human C. trachomatis urogenital tract infections, to investigate miRNA expression changes (128–131). Interestingly, Yeruva et al. showed that C. muridarum infection decreases the protein levels of two components of the miRNA machinery, Dicer and Ago2 (131). Although the consequence of these changes to infection is not yet clear, this suggests a broad effect of C. muridarum in blunting host miRNA responses.
In macrophages, infection by the Gram-negative bacterium Brucella melitensis, the causative agent of brucellosis, was shown to regulate the expression of 57 miRNAs (132). Putative targets of the six most abundant differentially expressed miRNAs (miR-92a, miR-93, miR-151-3p, miR-181b, miR-1981, and let7-b) revealed enrichment for apoptosis, autophagy, and immune response-related pathways. Recent studies have demonstrated that the related pathogen Brucella abortus is also able to modulate host miRNA expression to increase its intracellular survival in macrophages. Indeed, downregulation of miR-125b-5p by B. abortus was shown to diminish the proinflammatory host response via derepression of the inhibitor of NF-κB activation A20 (133). miRNA expression upon Brucella suis infection of porcine and murine macrophages revealed that a common set of five miRNAs (miR-130a-3p, miR-146a, miR-181a, miR-301a-3p, and miR-351-5p) are upregulated. By controlling these miRNAs, B. suis negatively regulates TNFα to promote bacterial intracellular survival (134).
High-throughput functional screenings have recently started to be applied to identify miRNAs controlling different biological processes. We have pioneered the application of this approach to infections by bacterial pathogens, specifically S. Typhimurium (cf. section above [22]) and Shigella flexneri (135). S. flexneri is a major causative agent of shigellosis in humans (136). Using an unbiased functional screening approach, we identified miR-29b-2-5p as a miRNA with a dual regulatory effect on S. flexneri infection, enhancing both bacterial binding to host cells and intracellular bacterial replication (135). miR-29b-2-5p leads to increased filopodia formation through the direct regulation of UNC5C, ultimately enhancing S. flexneri binding to host cells. Interestingly, S. flexneri intracellular replication decreases miR-29b-2-5p expression at late times postinfection by specific degradation of the mature miRNA by the exonuclease PNPT1. A decrease of miR-29b-2-5p may constitute a bacterial strategy to promote balanced intracellular replication, avoiding premature host cell death and favoring spreading to neighboring cells or, alternatively, may be part of the host response to counteract Shigella infection.
PERSPECTIVES
Advances in RNA sequencing have contributed to the extensive analysis of the host miRNome and contributed to demonstrating that infection by various bacterial pathogens induces major miRNome changes. Of note, these studies highlighted that even closely related species (e.g., M. tuberculosis versus M. bovis versus M. avium, or F. tularensis subsp. tularensis versus F. tularensis subsp. novicida) lead to widely different host miRNA profiles. However, a common set of miRNAs regulated as part of the host immune response (e.g., miR-155, miR-146, and let-7) has been described for a wide spectrum of pathogens. Along this line, a recent analysis of miRNA response to infection of dendritic cells by six bacteria revealed a core of 49 miRNAs with consistently altered expression (137). Interestingly, this study also showed that the relative abundance of miRNA duplex arms (–3p and –5p miRNAs) and the expression dynamics of miRNA isoforms (isomiRs) are strongly impacted by bacterial infection, although the consequences of these phenomena to infection are yet to be determined.
In addition to the identity of the pathogen, the regulation of host miRNAs upon infection is dependent on the cellular context (e.g., epithelial cells versus macrophages). Added to this, the majority of the miRNA expression analysis in the context of infection has been performed in bulk cell populations comprising both cells with internalized bacteria and bystander cells. The heterogeneity of the miRNA response of individual cells to infection and its dependency on the extent of bacterial internalization and/or replication still require further investigation. With the improvement of single-cell RNA sequencing, reliable analysis of miRNA profiles with sufficient coverage will be attainable (138). One additional interesting point concerns differences in the response of cells with internalized bacteria and bystander cells. Initial reports in sorted populations of infected and bystander cells suggest that although miRNA changes in the bystander population generally echo those observed in infected cells, a portion of the changes appear to be specific to bystander cells (22). The relevance of the miRNA reprogramming in bystander cells and their consequences to infection clearly deserve further investigation. Importantly, the molecular mechanisms underlying most of the observed miRNA regulations, irrespective of whether these occur in infected or bystander cells, remain poorly understood. In this regard, the direct manipulation of the miRNA pathway by secreted bacterial factors remains a fascinating hypothesis prompted by the discovery of RNA silencing suppressor proteins in viruses and plant bacterial pathogens.
The impacts of microbiota on host miRNA expression and their implications for organ/tissue homeostasis and/or infections are also an exciting subject. In this regard, the resident gut microbiota has been shown to modulate host miRNA expression (139, 140), and a recent report suggests that the host might influence the gut microbiome through miRNAs (141). Moreover, analysis of the host miRNA response to L. monocytogenes infection in conventional versus germ-free mice identified five miRNAs that were downregulated upon infection in a microbiota-dependent manner (113).
Overall, miRNAs are now considered major players in the infection process. As summarized in this article, a growing number of studies are uncovering host miRNAs as part of the defense strategies mounted by the host to fight infection but also as pathogen strategies to subvert host functions and promote virulence. The study of miRNAs in the context of infection and, equally importantly, the identification and characterization of downstream targets and underlying mechanisms of action will continue to provide important insights into the intricate interplay between pathogens and host.
ACKNOWLEDGMENTS
Work in the M.M. and A.E. laboratories on microRNAs and infection was supported by the ERA-NET Infect-ERA CampyRNA and grants from the Portuguese Science Foundation (FCT, #POCI-01-0145-FEDER-029999 and IF/01105/2015).
Contributor Information
Carmen Aguilar, Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany.
Miguel Mano, Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
Ana Eulalio, Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
Pascale Cossart, Institut Pasteur, Paris, France.
Craig R. Roy, Yale University School of Medicine, New Haven, Connecticut
Philippe Sansonetti, Institut Pasteur, Paris, France.
REFERENCES
- 1.Bartel DP. 2018. Metazoan MicroRNAs. Cell 173:20–51 10.1016/j.cell.2018.03.006. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Krol J, Loedige I, Filipowicz W. 2010. The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 11:597–610 10.1038/nrg2843. [PubMed] [DOI] [PubMed] [Google Scholar]
- 3.Jonas S, Izaurralde E. 2015. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet 16:421–433 10.1038/nrg3965. [PubMed] [DOI] [PubMed] [Google Scholar]
- 4.Friedman RC, Farh KK, Burge CB, Bartel DP. 2009. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19:92–105 10.1101/gr.082701.108. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bueno MJ, Pérez de Castro I, Malumbres M. 2008. Control of cell proliferation pathways by microRNAs. Cell Cycle 7:3143–3148 10.4161/cc.7.20.6833. [PubMed] [DOI] [PubMed] [Google Scholar]
- 6.Jovanovic M, Hengartner MO. 2006. miRNAs and apoptosis: RNAs to die for. Oncogene 25:6176–6187 10.1038/sj.onc.1209912. [PubMed] [DOI] [PubMed] [Google Scholar]
- 7.Shenoy A, Blelloch RH. 2014. Regulation of microRNA function in somatic stem cell proliferation and differentiation. Nat Rev Mol Cell Biol 15:565–576 10.1038/nrm3854. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Croce CM. 2009. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 10:704–714 10.1038/nrg2634. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Small EM, Olson EN. 2011. Pervasive roles of microRNAs in cardiovascular biology. Nature 469:336–342 10.1038/nature09783. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bruscella P, Bottini S, Baudesson C, Pawlotsky JM, Feray C, Trabucchi M. 2017. Viruses and miRNAs: more friends than foes. Front Microbiol 8:824 10.3389/fmicb.2017.00824. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pfeffer S, Zavolan M, Grässer FA, Chien M, Russo JJ, Ju J, John B, Enright AJ, Marks D, Sander C, Tuschl T. 2004. Identification of virus-encoded microRNAs. Science 304:734–736 10.1126/science.1096781. [PubMed] [DOI] [PubMed] [Google Scholar]
- 12.Navarro L, Dunoyer P, Jay F, Arnold B, Dharmasiri N, Estelle M, Voinnet O, Jones JD. 2006. A plant miRNA contributes to antibacterial resistance by repressing auxin signaling. Science 312:436–439 10.1126/science.1126088. [PubMed] [DOI] [PubMed] [Google Scholar]
- 13.Navarro L, Jay F, Nomura K, He SY, Voinnet O. 2008. Suppression of the microRNA pathway by bacterial effector proteins. Science 321:964–967 10.1126/science.1159505. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Taganov KD, Boldin MP, Chang KJ, Baltimore D. 2006. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A 103:12481–12486 10.1073/pnas.0605298103. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Majowicz SE, Musto J, Scallan E, Angulo FJ, Kirk M, O’Brien SJ, Jones TF, Fazil A, Hoekstra RM, International Collaboration on Enteric Disease ‘Burden of Illness’ Studies. 2010. The global burden of nontyphoidal Salmonella gastroenteritis. Clin Infect Dis 50:882–889 10.1086/650733. [PubMed] [DOI] [PubMed] [Google Scholar]
- 16.Herrero-Fresno A, Olsen JE. 2018. Salmonella Typhimurium metabolism affects virulence in the host: a mini-review. Food Microbiol 71:98–110 10.1016/j.fm.2017.04.016. [PubMed] [DOI] [PubMed] [Google Scholar]
- 17.Schulte LN, Eulalio A, Mollenkopf HJ, Reinhardt R, Vogel J. 2011. Analysis of the host microRNA response to Salmonella uncovers the control of major cytokines by the let-7 family. EMBO J 30:1977–1989 10.1038/emboj.2011.94. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sharbati S, Sharbati J, Hoeke L, Bohmer M, Einspanier R. 2012. Quantification and accurate normalisation of small RNAs through new custom RT-qPCR arrays demonstrates Salmonella-induced microRNAs in human monocytes. BMC Genomics 13:23 10.1186/1471-2164-13-23. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li P, Fan W, Li Q, Wang J, Liu R, Everaert N, Liu J, Zhang Y, Zheng M, Cui H, Zhao G, Wen J. 2017. Splenic microRNA expression profiles and integration analyses involved in host responses to Salmonella enteritidis infection in chickens. Front Cell Infect Microbiol 7:377 10.3389/fcimb.2017.00377. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Schulte LN, Westermann AJ, Vogel J. 2013. Differential activation and functional specialization of miR-146 and miR-155 in innate immune sensing. Nucleic Acids Res 41:542–553 10.1093/nar/gks1030. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR, van Dongen S, Grocock RJ, Das PP, Miska EA, Vetrie D, Okkenhaug K, Enright AJ, Dougan G, Turner M, Bradley A. 2007. Requirement of bic/microRNA-155 for normal immune function. Science 316:608–611 10.1126/science.1139253. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Maudet C, Mano M, Sunkavalli U, Sharan M, Giacca M, Förstner KU, Eulalio A. 2014. Functional high-throughput screening identifies the miR-15 microRNA family as cellular restriction factors for Salmonella infection. Nat Commun 5:4718 10.1038/ncomms5718. [PubMed] [DOI] [PubMed] [Google Scholar]
- 23.Flotho A, Melchior F. 2013. Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem 82:357–385 10.1146/annurev-biochem-061909-093311. [PubMed] [DOI] [PubMed] [Google Scholar]
- 24.Verma S, Mohapatra G, Ahmad SM, Rana S, Jain S, Khalsa JK, Srikanth CV. 2015. Salmonella engages host microRNAs to modulate SUMOylation: a new arsenal for intracellular survival. Mol Cell Biol 35:2932–2946 10.1128/MCB.00397-15. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ordas A, Kanwal Z, Lindenberg V, Rougeot J, Mink M, Spaink HP, Meijer AH. 2013. MicroRNA-146 function in the innate immune transcriptome response of zebrafish embryos to Salmonella Typhimurium infection. BMC Genomics 14:696 10.1186/1471-2164-14-696. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chen Q, Tong C, Ma S, Zhou L, Zhao L, Zhao X. 2017. Involvement of MicroRNAs in probiotics-induced reduction of the cecal inflammation by Salmonella Typhimurium. Front Immunol 8:704 10.3389/fimmu.2017.00704. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hoeke L, Sharbati J, Pawar K, Keller A, Einspanier R, Sharbati S. 2013. Intestinal Salmonella Typhimurium infection leads to miR-29a induced caveolin 2 regulation. PLoS One 8:e67300 10.1371/journal.pone.0067300. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Herrera-Uribe J, Zaldívar-López S, Aguilar C, Luque C, Bautista R, Carvajal A, Claros MG, Garrido JJ. 2018. Regulatory role of microRNA in mesenteric lymph nodes after Salmonella Typhimurium infection. Vet Res (Faisalabad) 49:9 10.1186/s13567-018-0506-1. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bao H, Kommadath A, Liang G, Sun X, Arantes AS, Tuggle CK, Bearson SM, Plastow GS, Stothard P, Guan L. 2015. Genome-wide whole blood microRNAome and transcriptome analyses reveal miRNA-mRNA regulated host response to foodborne pathogen Salmonella infection in swine. Sci Rep 5:12620 10.1038/srep12620. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yao M, Gao W, Tao H, Yang J, Liu G, Huang T. 2016. Regulation signature of miR-143 and miR-26 in porcine Salmonella infection identified by binding site enrichment analysis. Mol Genet Genomics 291:789–799 10.1007/s00438-015-1146-z. [PubMed] [DOI] [PubMed] [Google Scholar]
- 31.Zhang T, Yu J, Zhang Y, Li L, Chen Y, Li D, Liu F, Zhang CY, Gu H, Zen K. 2014. Salmonella enterica serovar Enteritidis modulates intestinal epithelial miR-128 levels to decrease macrophage recruitment via macrophage colony-stimulating factor. J Infect Dis 209:2000–2011 10.1093/infdis/jiu006. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wu G, Qi Y, Liu X, Yang N, Xu G, Liu L, Li X. 2017. Cecal MicroRNAome response to Salmonella enterica serovar Enteritidis infection in white leghorn layer. BMC Genomics 18:77 10.1186/s12864-016-3413-8. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cover TL, Blaser MJ. 2009. Helicobacter pylori in health and disease. Gastroenterology 136:1863–1873 10.1053/j.gastro.2009.01.073. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jones KR, Whitmire JM, Merrell DS. 2010. A tale of two toxins: Helicobacter pylori CagA and VacA modulate host pathways that impact disease. Front Microbiol 1:115 10.3389/fmicb.2010.00115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, Xiao S, Lu H. 2008. miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest 88:1358–1366 10.1038/labinvest.2008.94. [PubMed] [DOI] [PubMed] [Google Scholar]
- 36.Li N, Tang B, Zhu ED, Li BS, Zhuang Y, Yu S, Lu DS, Zou QM, Xiao B, Mao XH. 2012. Increased miR-222 in H. pylori-associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK. FEBS Lett 586:722–728 10.1016/j.febslet.2012.01.025. [PubMed] [DOI] [PubMed] [Google Scholar]
- 37.Tan X, Tang H, Bi J, Li N, Jia Y. 2018. MicroRNA-222-3p associated with Helicobacter pylori targets HIPK2 to promote cell proliferation, invasion, and inhibits apoptosis in gastric cancer. J Cell Biochem 119:5153–5162 10.1002/jcb.26542. [PubMed] [DOI] [PubMed] [Google Scholar]
- 38.Zhou X, Xia Y, Li L, Zhang G. 2015. MiR-101 inhibits cell growth and tumorigenesis of Helicobacter pylori related gastric cancer by repression of SOCS2. Cancer Biol Ther 16:160–169 10.4161/15384047.2014.987523. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kiga K, Mimuro H, Suzuki M, Shinozaki-Ushiku A, Kobayashi T, Sanada T, Kim M, Ogawa M, Iwasaki YW, Kayo H, Fukuda-Yuzawa Y, Yashiro M, Fukayama M, Fukao T, Sasakawa C. 2014. Epigenetic silencing of miR-210 increases the proliferation of gastric epithelium during chronic Helicobacter pylori infection. Nat Commun 5:4497 10.1038/ncomms5497. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Noto JM, Piazuelo MB, Chaturvedi R, Bartel CA, Thatcher EJ, Delgado A, Romero-Gallo J, Wilson KT, Correa P, Patton JG, Peek RM Jr. 2013. Strain-specific suppression of microRNA-320 by carcinogenic Helicobacter pylori promotes expression of the antiapoptotic protein Mcl-1. Am J Physiol Gastrointest Liver Physiol 305:G786–G796 10.1152/ajpgi.00279.2013. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Feng Y, Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, Chen C, Jia J. 2013. FoxM1 is overexpressed in Helicobacter pylori-induced gastric carcinogenesis and is negatively regulated by miR-370. Mol Cancer Res 11:834–844 10.1158/1541-7786.MCR-13-0007. [PubMed] [DOI] [PubMed] [Google Scholar]
- 42.Zhu Y, Jiang Q, Lou X, Ji X, Wen Z, Wu J, Tao H, Jiang T, He W, Wang C, Du Q, Zheng S, Mao J, Huang J. 2012. MicroRNAs up-regulated by CagA of Helicobacter pylori induce intestinal metaplasia of gastric epithelial cells. PLoS One 7:e35147 10.1371/journal.pone.0035147. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chu YX, Wang WH, Dai Y, Teng GG, Wang SJ. 2014. Esophageal Helicobacter pylori colonization aggravates esophageal injury caused by reflux. World J Gastroenterol 20:15715–15726 10.3748/wjg.v20.i42.15715. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu FX, Wang WH, Wang J, Li J, Gao PP. 2011. Effect of Helicobacter pylori infection on Barrett’s esophagus and esophageal adenocarcinoma formation in a rat model of chronic gastroesophageal reflux. Helicobacter 16:66–77 10.1111/j.1523-5378.2010.00811.x. [PubMed] [DOI] [PubMed] [Google Scholar]
- 45.Teng G, Dai Y, Chu Y, Li J, Zhang H, Wu T, Shuai X, Wang W. 2018. Helicobacter pylori induces caudal-type homeobox protein 2 and cyclooxygenase 2 expression by modulating microRNAs in esophageal epithelial cells. Cancer Sci 109:297–307 10.1111/cas.13462. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Belair C, Baud J, Chabas S, Sharma CM, Vogel J, Staedel C, Darfeuille F. 2011. Helicobacter pylori interferes with an embryonic stem cell micro RNA cluster to block cell cycle progression. Silence 2:7 10.1186/1758-907X-2-7. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang F, Liu J, Zou Y, Jiao Y, Huang Y, Fan L, Li X, Yu H, He C, Wei W, Wang H, Sun G. 2017. MicroRNA-143-3p, up-regulated in H. pylori-positive gastric cancer, suppresses tumor growth, migration and invasion by directly targeting AKT2. Oncotarget 8:28711–28724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fassi Fehri L, Koch M, Belogolova E, Khalil H, Bolz C, Kalali B, Mollenkopf HJ, Beigier-Bompadre M, Karlas A, Schneider T, Churin Y, Gerhard M, Meyer TF. 2010. Helicobacter pylori induces miR-155 in T cells in a cAMP-Foxp3-dependent manner. PLoS One 5:e9500 10.1371/journal.pone.0009500. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Koch M, Mollenkopf HJ, Klemm U, Meyer TF. 2012. Induction of microRNA-155 is TLR- and type IV secretion system-dependent in macrophages and inhibits DNA-damage induced apoptosis. Proc Natl Acad Sci U S A 109:E1153–E1162 10.1073/pnas.1116125109. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lario S, Ramírez-Lázaro MJ, Aransay AM, Lozano JJ, Montserrat A, Casalots Á, Junquera F, Álvarez J, Segura F, Campo R, Calvet X. 2012. microRNA profiling in duodenal ulcer disease caused by Helicobacter pylori infection in a Western population. Clin Microbiol Infect 18:E273–E282 10.1111/j.1469-0691.2012.03849.x. [PubMed] [DOI] [PubMed] [Google Scholar]
- 51.Oertli M, Engler DB, Kohler E, Koch M, Meyer TF, Müller A. 2011. MicroRNA-155 is essential for the T cell-mediated control of Helicobacter pylori infection and for the induction of chronic gastritis and colitis. J Immunol 187:3578–3586 10.4049/jimmunol.1101772. [PubMed] [DOI] [PubMed] [Google Scholar]
- 52.Xiao B, Liu Z, Li BS, Tang B, Li W, Guo G, Shi Y, Wang F, Wu Y, Tong WD, Guo H, Mao XH, Zou QM. 2009. Induction of microRNA-155 during Helicobacter pylori infection and its negative regulatory role in the inflammatory response. J Infect Dis 200:916–925 10.1086/605443. [PubMed] [DOI] [PubMed] [Google Scholar]
- 53.Pachathundikandi SK, Backert S. 2018. Helicobacter pylori controls NLRP3 expression by regulating hsa-miR-223-3p and IL-10 in cultured and primary human immune cells. Innate Immun 24:11–23 10.1177/1753425917738043. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Li N, Xu X, Xiao B, Zhu ED, Li BS, Liu Z, Tang B, Zou QM, Liang HP, Mao XH. 2012. H. pylori related proinflammatory cytokines contribute to the induction of miR-146a in human gastric epithelial cells. Mol Biol Rep 39:4655–4661 10.1007/s11033-011-1257-5. [PubMed] [DOI] [PubMed] [Google Scholar]
- 55.Liu Z, Wang D, Hu Y, Zhou G, Zhu C, Yu Q, Chi Y, Cao Y, Jia C, Zou Q. 2013. MicroRNA-146a negatively regulates PTGS2 expression induced by Helicobacter pylori in human gastric epithelial cells. J Gastroenterol 48:86–92 10.1007/s00535-012-0609-9. [PubMed] [DOI] [PubMed] [Google Scholar]
- 56.Liu Z, Xiao B, Tang B, Li B, Li N, Zhu E, Guo G, Gu J, Zhuang Y, Liu X, Ding H, Zhao X, Guo H, Mao X, Zou Q. 2010. Up-regulated microRNA-146a negatively modulate Helicobacter pylori-induced inflammatory response in human gastric epithelial cells. Microbes Infect 12:854–863 10.1016/j.micinf.2010.06.002. [PubMed] [DOI] [PubMed] [Google Scholar]
- 57.Teng GG, Wang WH, Dai Y, Wang SJ, Chu YX, Li J. 2013. Let-7b is involved in the inflammation and immune responses associated with Helicobacter pylori infection by targeting Toll-like receptor 4. PLoS One 8:e56709 10.1371/journal.pone.0056709. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, Nakao K, Hirayama T, Kohno S. 2011. MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer 128:361–370 10.1002/ijc.25348. [PubMed] [DOI] [PubMed] [Google Scholar]
- 59.Xie G, Li W, Li R, Wu K, Zhao E, Zhang Y, Zhang P, Shi L, Wang D, Yin Y, Deng R, Tao K. 2017. Helicobacter pylori promote B7-H1 expression by suppressing miR-152 and miR-200b in gastric cancer cells. PLoS One 12:e0168822 10.1371/journal.pone.0168822. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chen J, Li G, Meng H, Fan Y, Song Y, Wang S, Zhu F, Guo C, Zhang L, Shi Y. 2012. Upregulation of B7-H1 expression is associated with macrophage infiltration in hepatocellular carcinomas. Cancer Immunol Immunother 61:101–108 10.1007/s00262-011-1094-3. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pagliari M, Munari F, Toffoletto M, Lonardi S, Chemello F, Codolo G, Millino C, Della Bella C, Pacchioni B, Vermi W, Fassan M, de Bernard M, Cagnin S. 2017. Helicobacter pylori affects the antigen presentation activity of macrophages modulating the expression of the immune receptor CD300E through miR-4270. Front Immunol 8:1288 10.3389/fimmu.2017.01288. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Tang B, Li N, Gu J, Zhuang Y, Li Q, Wang HG, Fang Y, Yu B, Zhang JY, Xie QH, Chen L, Jiang XJ, Xiao B, Zou QM, Mao XH. 2012. Compromised autophagy by MIR30B benefits the intracellular survival of Helicobacter pylori. Autophagy 8:1045–1057 10.4161/auto.20159. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yang XJ, Si RH, Liang YH, Ma BQ, Jiang ZB, Wang B, Gao P. 2016. Mir-30d increases intracellular survival of Helicobacter pylori through inhibition of autophagy pathway. World J Gastroenterol 22:3978–3991 10.3748/wjg.v22.i15.3978. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang YM, Noto JM, Hammond CE, Barth JL, Argraves WS, Backert S, Peek RM Jr, Smolka AJ. 2014. Helicobacter pylori-induced posttranscriptional regulation of H-K-ATPase α-subunit gene expression by miRNA. Am J Physiol Gastrointest Liver Physiol 306:G606–G613 10.1152/ajpgi.00333.2013. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Abdalla AE, Duan X, Deng W, Zeng J, Xie J. 2016. MicroRNAs play big roles in modulating macrophages response toward mycobacteria infection. Infect Genet Evol 45:378–382 10.1016/j.meegid.2016.09.023. [PubMed] [DOI] [PubMed] [Google Scholar]
- 66.Ghorpade DS, Leyland R, Kurowska-Stolarska M, Patil SA, Balaji KN. 2012. MicroRNA-155 is required for Mycobacterium bovis BCG-mediated apoptosis of macrophages. Mol Cell Biol 32:2239–2253 10.1128/MCB.06597-11. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kumar R, Halder P, Sahu SK, Kumar M, Kumari M, Jana K, Ghosh Z, Sharma P, Kundu M, Basu J. 2012. Identification of a novel role of ESAT-6-dependent miR-155 induction during infection of macrophages with Mycobacterium tuberculosis. Cell Microbiol 14:1620–1631 10.1111/j.1462-5822.2012.01827.x. [PubMed] [DOI] [PubMed] [Google Scholar]
- 68.Das K, Saikolappan S, Dhandayuthapani S. 2013. Differential expression of miRNAs by macrophages infected with virulent and avirulent Mycobacterium tuberculosis. Tuberculosis (Edinb) 93(Suppl):S47–S50 10.1016/S1472-9792(13)70010-6. [DOI] [PubMed] [Google Scholar]
- 69.Ahluwalia PK, Pandey RK, Sehajpal PK, Prajapati VK. 2017. Perturbed microRNA expression by Mycobacterium tuberculosis promotes macrophage polarization leading to pro-survival foam cell. Front Immunol 8:107 10.3389/fimmu.2017.00107. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Etna MP, Sinigaglia A, Grassi A, Giacomini E, Romagnoli A, Pardini M, Severa M, Cruciani M, Rizzo F, Anastasiadou E, Di Camillo B, Barzon L, Fimia GM, Manganelli R, Coccia EM. 2018. Mycobacterium tuberculosis-induced miR-155 subverts autophagy by targeting ATG3 in human dendritic cells. PLoS Pathog 14:e1006790 10.1371/journal.ppat.1006790. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Huang J, Jiao J, Xu W, Zhao H, Zhang C, Shi Y, Xiao Z. 2015. MiR-155 is upregulated in patients with active tuberculosis and inhibits apoptosis of monocytes by targeting FOXO3. Mol Med Rep 12:7102–7108 10.3892/mmr.2015.4250. [PubMed] [DOI] [PubMed] [Google Scholar]
- 72.Qin Y, Wang Q, Zhou Y, Duan Y, Gao Q. 2016. Inhibition of IFN-γ-induced nitric oxide dependent antimycobacterial activity by miR-155 and C/EBPβ. Int J Mol Sci 17:535 10.3390/ijms17040535. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rajaram MV, Ni B, Morris JD, Brooks MN, Carlson TK, Bakthavachalu B, Schoenberg DR, Torrelles JB, Schlesinger LS. 2011. 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 108:17408–17413 10.1073/pnas.1112660108. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rothchild AC, Sissons JR, Shafiani S, Plaisier C, Min D, Mai D, Gilchrist M, Peschon J, Larson RP, Bergthaler A, Baliga NS, Urdahl KB, Aderem A. 2016. MiR-155-regulated molecular network orchestrates cell fate in the innate and adaptive immune response to Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 113:E6172–E6181 10.1073/pnas.1608255113. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sharbati J, Lewin A, Kutz-Lohroff B, Kamal E, Einspanier R, Sharbati S. 2011. Integrated microRNA-mRNA-analysis of human monocyte derived macrophages upon Mycobacterium avium subsp. hominissuis infection. PLoS One 6:e20258 10.1371/journal.pone.0020258. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wang J, Yang K, Zhou L, Minhaowu, Wu Y, Zhu M, Lai X, Chen T, Feng L, Li M, Huang C, Zhong Q, Huang X. 2013. MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog 9:e1003697 10.1371/journal.ppat.1003697. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Yang S, Li F, Jia S, Zhang K, Jiang W, Shang Y, Chang K, Deng S, Chen M. 2015. Early secreted antigen ESAT-6 of Mycobacterium tuberculosis promotes apoptosis of macrophages via targeting the microRNA155-SOCS1 interaction. Cell Physiol Biochem 35:1276–1288 10.1159/000373950. [PubMed] [DOI] [PubMed] [Google Scholar]
- 78.Liang S, Song Z, Wu Y, Gao Y, Gao M, Liu F, Wang F, Zhang Y. 2018. MicroRNA-27b modulates inflammatory response and apoptosis during Mycobacterium tuberculosis infection. J Immunol 200:3506–3518 10.4049/jimmunol.1701448. [PubMed] [DOI] [PubMed] [Google Scholar]
- 79.Kumar M, Sahu SK, Kumar R, Subuddhi A, Maji RK, Jana K, Gupta P, Raffetseder J, Lerm M, Ghosh Z, van Loo G, Beyaert R, Gupta UD, Kundu M, Basu J. 2015. MicroRNA let-7 modulates the immune response to Mycobacterium tuberculosis infection via control of A20, an inhibitor of the NF-κB pathway. Cell Host Microbe 17:345–356 10.1016/j.chom.2015.01.007. [PubMed] [DOI] [PubMed] [Google Scholar]
- 80.Dorhoi A, Iannaccone M, Farinacci M, Faé KC, Schreiber J, Moura-Alves P, Nouailles G, Mollenkopf HJ, Oberbeck-Müller D, Jörg S, Heinemann E, Hahnke K, Löwe D, Del Nonno F, Goletti D, Capparelli R, Kaufmann SH. 2013. MicroRNA-223 controls susceptibility to tuberculosis by regulating lung neutrophil recruitment. J Clin Invest 123:4836–4848 10.1172/JCI67604. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bettencourt P, Marion S, Pires D, Santos LF, Lastrucci C, Carmo N, Blake J, Benes V, Griffiths G, Neyrolles O, Lugo-Villarino G, Anes E. 2013. Actin-binding protein regulation by microRNAs as a novel microbial strategy to modulate phagocytosis by host cells: the case of N-Wasp and miR-142-3p. Front Cell Infect Microbiol 3:19 10.3389/fcimb.2013.00019. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Liu PT, Wheelwright M, Teles R, Komisopoulou E, Edfeldt K, Ferguson B, Mehta MD, Vazirnia A, Rea TH, Sarno EN, Graeber TG, Modlin RL. 2012. MicroRNA-21 targets the vitamin D-dependent antimicrobial pathway in leprosy. Nat Med 18:267–273 10.1038/nm.2584. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fu Y, Yi Z, Wu X, Li J, Xu F. 2011. Circulating microRNAs in patients with active pulmonary tuberculosis. J Clin Microbiol 49:4246–4251 10.1128/JCM.05459-11. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kleinsteuber K, Heesch K, Schattling S, Kohns M, Sander-Jülch C, Walzl G, Hesseling A, Mayatepek E, Fleischer B, Marx FM, Jacobsen M. 2013. Decreased expression of miR-21, miR-26a, miR-29a, and miR-142-3p in CD4+ T cells and peripheral blood from tuberculosis patients. PLoS One 8:e61609 10.1371/journal.pone.0061609. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ma F, Xu S, Liu X, Zhang Q, Xu X, Liu M, Hua M, Li N, Yao H, Cao X. 2011. The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-γ. Nat Immunol 12:861–869 10.1038/ni.2073. [PubMed] [DOI] [PubMed] [Google Scholar]
- 86.Wang Q, Liu S, Tang Y, Liu Q, Yao Y. 2014. MPT64 protein from Mycobacterium tuberculosis inhibits apoptosis of macrophages through NF-kB-miRNA21-Bcl-2 pathway. PLoS One 9:e100949 10.1371/journal.pone.0100949. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu Y, Jiang J, Wang X, Zhai F, Cheng X. 2013. miR-582-5p is upregulated in patients with active tuberculosis and inhibits apoptosis of monocytes by targeting FOXO1. PLoS One 8:e78381 10.1371/journal.pone.0078381. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lou J, Wang Y, Zhang Z, Qiu W. 2017. MiR-20b inhibits mycobacterium tuberculosis induced inflammation in the lung of mice through targeting NLRP3. Exp Cell Res 358:120–128 10.1016/j.yexcr.2017.06.007. [PubMed] [DOI] [PubMed] [Google Scholar]
- 89.Jo EK, Yuk JM, Shin DM, Sasakawa C. 2013. Roles of autophagy in elimination of intracellular bacterial pathogens. Front Immunol 4:97 10.3389/fimmu.2013.00097. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kumar R, Sahu SK, Kumar M, Jana K, Gupta P, Gupta UD, Kundu M, Basu J. 2016. MicroRNA 17-5p regulates autophagy in Mycobacterium tuberculosis-infected macrophages by targeting Mcl-1 and STAT3. Cell Microbiol 18:679–691 10.1111/cmi.12540. [PubMed] [DOI] [PubMed] [Google Scholar]
- 91.Duan X, Zhang T, Ding S, Wei J, Su C, Liu H, Xu G. 2015. microRNA-17-5p modulates bacille Calmette-Guerin growth in RAW264.7 cells by targeting ULK1. PLoS One 10:e0138011 10.1371/journal.pone.0138011. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Chen Z, Wang T, Liu Z, Zhang G, Wang J, Feng S, Liang J. 2015. Inhibition of autophagy by MiR-30A induced by Mycobacteria tuberculosis as a possible mechanism of immune escape in human macrophages. Jpn J Infect Dis 68:420–424 10.7883/yoken.JJID.2014.466. [PubMed] [DOI] [PubMed] [Google Scholar]
- 93.Kim JK, Yuk JM, Kim SY, Kim TS, Jin HS, Yang CS, Jo EK. 2015. MicroRNA-125a inhibits autophagy activation and antimicrobial responses during mycobacterial infection. J Immunol 194:5355–5365 10.4049/jimmunol.1402557. [PubMed] [DOI] [PubMed] [Google Scholar]
- 94.Ouimet M, Koster S, Sakowski E, Ramkhelawon B, van Solingen C, Oldebeken S, Karunakaran D, Portal-Celhay C, Sheedy FJ, Ray TD, Cecchini K, Zamore PD, Rayner KJ, Marcel YL, Philips JA, Moore KJ. 2016. Mycobacterium tuberculosis induces the miR-33 locus to reprogram autophagy and host lipid metabolism. Nat Immunol 17:677–686 10.1038/ni.3434. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Liu F, Chen J, Wang P, Li H, Zhou Y, Liu H, Liu Z, Zheng R, Wang L, Yang H, Cui Z, Wang F, Huang X, Wang J, Sha W, Xiao H, Ge B. 2018. MicroRNA-27a controls the intracellular survival of Mycobacterium tuberculosis by regulating calcium-associated autophagy. Nat Commun 9:4295 10.1038/s41467-018-06836-4. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kim JK, Lee HM, Park KS, Shin DM, Kim TS, Kim YS, Suh HW, Kim SY, Kim IS, Kim JM, Son JW, Sohn KM, Jung SS, Chung C, Han SB, Yang CS, Jo EK. 2017. MIR144* inhibits antimicrobial responses against Mycobacterium tuberculosis in human monocytes and macrophages by targeting the autophagy protein DRAM2. Autophagy 13:423–441 10.1080/15548627.2016.1241922. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Guo L, Zhou L, Gao Q, Zhang A, Wei J, Hong D, Chu Y, Duan X, Zhang Y, Xu G. 2017. MicroRNA-144-3p inhibits autophagy activation and enhances bacillus Calmette-Guérin infection by targeting ATG4a in RAW264.7 macrophage cells. PLoS One 12:e0179772 10.1371/journal.pone.0179772. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Guo L, Zhao J, Qu Y, Yin R, Gao Q, Ding S, Zhang Y, Wei J, Xu G. 2016. microRNA-20a inhibits autophagic process by targeting ATG7 and ATG16L1 and favors mycobacterial survival in macrophage cells. Front Cell Infect Microbiol 6:134 10.3389/fcimb.2016.00134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wang J, Hussain T, Yue R, Liao Y, Li Q, Yao J, Song Y, Sun X, Wang N, Xu L, Sreevatsan S, Zhao D, Zhou X. 2018. MicroRNA-199a inhibits cellular autophagy and downregulates IFN-β expression by targeting TBK1 in Mycobacterium bovis infected cells. Front Cell Infect Microbiol 8:238 10.3389/fcimb.2018.00238. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Vegh P, Magee DA, Nalpas NC, Bryan K, McCabe MS, Browne JA, Conlon KM, Gordon SV, Bradley DG, MacHugh DE, Lynn DJ. 2015. MicroRNA profiling of the bovine alveolar macrophage response to Mycobacterium bovis infection suggests pathogen survival is enhanced by microRNA regulation of endocytosis and lysosome trafficking. Tuberculosis (Edinb) 95:60–67 10.1016/j.tube.2014.10.011. [PubMed] [DOI] [PubMed] [Google Scholar]
- 101.Sahu SK, Kumar M, Chakraborty S, Banerjee SK, Kumar R, Gupta P, Jana K, Gupta UD, Ghosh Z, Kundu M, Basu J. 2017. MicroRNA 26a (miR-26a)/KLF4 and CREB-C/EBPβ regulate innate immune signaling, the polarization of macrophages and the trafficking of Mycobacterium tuberculosis to lysosomes during infection. PLoS Pathog 13:e1006410 10.1371/journal.ppat.1006410. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Pires D, Bernard EM, Pombo JP, Carmo N, Fialho C, Gutierrez MG, Bettencourt P, Anes E. 2017. Mycobacterium tuberculosis modulates miR-106b-5p to control cathepsin S expression resulting in higher pathogen survival and poor T-cell activation. Front Immunol 8:1819 10.3389/fimmu.2017.01819. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Cossart P. 2011. Illuminating the landscape of host-pathogen interactions with the bacterium Listeria monocytogenes. Proc Natl Acad Sci U S A 108:19484–19491 10.1073/pnas.1112371108. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schnitger AK, Machova A, Mueller RU, Androulidaki A, Schermer B, Pasparakis M, Krönke M, Papadopoulou N. 2011. Listeria monocytogenes infection in macrophages induces vacuolar-dependent host miRNA response. PLoS One 6:e27435 10.1371/journal.pone.0027435. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lind EF, Elford AR, Ohashi PS. 2013. Micro-RNA 155 is required for optimal CD8+ T cell responses to acute viral and intracellular bacterial challenges. J Immunol 190:1210–1216 10.4049/jimmunol.1202700. [PubMed] [DOI] [PubMed] [Google Scholar]
- 106.Johnston DGW, Kearney J, Zasłona Z, Williams MA, O’Neill LAJ, Corr SC. 2017. MicroRNA-21 limits uptake of Listeria monocytogenes by macrophages to reduce the intracellular niche and control infection. Front Cell Infect Microbiol 7:201 10.3389/fcimb.2017.00201. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Izar B, Mannala GK, Mraheil MA, Chakraborty T, Hain T. 2012. microRNA response to Listeria monocytogenes infection in epithelial cells. Int J Mol Sci 13:1173–1185 10.3390/ijms13011173. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Collison A, Mattes J, Plank M, Foster PS. 2011. Inhibition of house dust mite-induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment. J Allergy Clin Immunol 128:160–167e164. [PubMed] [DOI] [PubMed] [Google Scholar]
- 109.Witwer KW, Sisk JM, Gama L, Clements JE. 2010. MicroRNA regulation of IFN-beta protein expression: rapid and sensitive modulation of the innate immune response. J Immunol 184:2369–2376 10.4049/jimmunol.0902712. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Dussurget O, Bierne H, Cossart P. 2014. The bacterial pathogen Listeria monocytogenes and the interferon family: type I, type II and type III interferons. Front Cell Infect Microbiol 4:50 10.3389/fcimb.2014.00050. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li R, Shen Q, Wu N, He M, Liu N, Huang J, Lu B, Yao Q, Yang Y, Hu R. 2018. MiR-145 improves macrophage-mediated inflammation through targeting Arf6. Endocrine 60:73–82 10.1007/s12020-018-1521-8. [PubMed] [DOI] [PubMed] [Google Scholar]
- 112.Archambaud C, Nahori MA, Soubigou G, Bécavin C, Laval L, Lechat P, Smokvina T, Langella P, Lecuit M, Cossart P. 2012. Impact of lactobacilli on orally acquired listeriosis. Proc Natl Acad Sci U S A 109:16684–16689 10.1073/pnas.1212809109. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Archambaud C, Sismeiro O, Toedling J, Soubigou G, Bécavin C, Lechat P, Lebreton A, Ciaudo C, Cossart P. 2013. The intestinal microbiota interferes with the microRNA response upon oral Listeria infection. MBio 4:e00707-13 10.1128/mBio.00707-13. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bandyopadhyay S, Long ME, Allen LA. 2014. Differential expression of microRNAs in Francisella tularensis-infected human macrophages: miR-155-dependent downregulation of MyD88 inhibits the inflammatory response. PLoS One 9:e109525 10.1371/journal.pone.0109525. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Cremer TJ, Ravneberg DH, Clay CD, Piper-Hunter MG, Marsh CB, Elton TS, Gunn JS, Amer A, Kanneganti TD, Schlesinger LS, Butchar JP, Tridandapani S. 2009. 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 4:e8508 10.1371/journal.pone.0008508. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Clare S, John V, Walker AW, Hill JL, Abreu-Goodger C, Hale C, Goulding D, Lawley TD, Mastroeni P, Frankel G, Enright AJ, Vigorito E, Dougan G. 2013. Enhanced susceptibility to Citrobacter rodentium infection in microRNA-155-deficient mice. Infect Immun 81:723–732 10.1128/IAI.00969-12. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Roy BC, Subramaniam D, Ahmed I, Jala VR, Hester CM, Greiner KA, Haribabu B, Anant S, Umar S. 2015. Role of bacterial infection in the epigenetic regulation of Wnt antagonist WIF1 by PRC2 protein EZH2. Oncogene 34:4519–4530 10.1038/onc.2014.386. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rao R, Rieder SA, Nagarkatti P, Nagarkatti M. 2014. Staphylococcal enterotoxin B-induced microRNA-155 targets SOCS1 to promote acute inflammatory lung injury. Infect Immun 82:2971–2979 10.1128/IAI.01666-14. (Erratum, 82:3986.) [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Jingjing Z, Nan Z, Wei W, Qinghe G, Weijuan W, Peng W, Xiangpeng W. 2017. MicroRNA-24 modulates Staphylococcus aureus-induced macrophage polarization by suppressing CHI3L1. Inflammation 40:995–1005 10.1007/s10753-017-0543-3. [PubMed] [DOI] [PubMed] [Google Scholar]
- 120.Wolcott RD, Hanson JD, Rees EJ, Koenig LD, Phillips CD, Wolcott RA, Cox SB, White JS. 2016. Analysis of the chronic wound microbiota of 2,963 patients by 16S rDNA pyrosequencing. Wound Repair Regen 24:163–174 10.1111/wrr.12370. [PubMed] [DOI] [PubMed] [Google Scholar]
- 121.Ramirez HA, Pastar I, Jozic I, Stojadinovic O, Stone RC, Ojeh N, Gil J, Davis SC, Kirsner RS, Tomic-Canic M. 2018. Staphylococcus aureus triggers induction of miR-15B-5P to diminish DNA repair and deregulate inflammatory response in diabetic foot ulcers. J Invest Dermatol 138:1187–1196. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Tanaka K, Kim SE, Yano H, Matsumoto G, Ohuchida R, Ishikura Y, Araki M, Araki K, Park S, Komatsu T, Hayashi H, Ikematsu K, Tanaka K, Hirano A, Martin P, Shimokawa I, Mori R. 2017. MiR-142 is required for Staphylococcus aureus clearance at skin wound sites via small GTPase-mediated regulation of the neutrophil actin cytoskeleton. J Invest Dermatol 137:931–940 10.1016/j.jid.2016.11.018. [PubMed] [DOI] [PubMed] [Google Scholar]
- 123.de Kerckhove M, Tanaka K, Umehara T, Okamoto M, Kanematsu S, Hayashi H, Yano H, Nishiura S, Tooyama S, Matsubayashi Y, Komatsu T, Park S, Okada Y, Takahashi R, Kawano Y, Hanawa T, Iwasaki K, Nozaki T, Torigoe H, Ikematsu K, Suzuki Y, Tanaka K, Martin P, Shimokawa I, Mori R. 2018. Targeting miR-223 in neutrophils enhances the clearance of Staphylococcus aureus in infected wounds. EMBO Mol Med 10:e9024 10.15252/emmm.201809024. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhou X, Li X, Ye Y, Zhao K, Zhuang Y, Li Y, Wei Y, Wu M. 2014. MicroRNA-302b augments host defense to bacteria by regulating inflammatory responses via feedback to TLR/IRAK4 circuits. Nat Commun 5:3619 10.1038/ncomms4619. (Erratum, 6:8679. .) [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Eledge MR, Yeruva L. 2018. Host and pathogen interface: microRNAs are modulators of disease outcome. Microbes Infect 20:410–415 10.1016/j.micinf.2017.08.002. [PubMed] [DOI] [PubMed] [Google Scholar]
- 126.Derrick T, Roberts C, Rajasekhar M, Burr SE, Joof H, Makalo P, Bailey RL, Mabey DC, Burton MJ, Holland MJ. 2013. Conjunctival MicroRNA expression in inflammatory trachomatous scarring. PLoS Negl Trop Dis 7:e2117 10.1371/journal.pntd.0002117. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Chowdhury SR, Reimer A, Sharan M, Kozjak-Pavlovic V, Eulalio A, Prusty BK, Fraunholz M, Karunakaran K, Rudel T. 2017. Chlamydia preserves the mitochondrial network necessary for replication via microRNA-dependent inhibition of fission. J Cell Biol 216:1071–1089 10.1083/jcb.201608063. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Arkatkar T, Gupta R, Li W, Yu JJ, Wali S, Neal Guentzel M, Chambers JP, Christenson LK, Arulanandam BP. 2015. Murine MicroRNA-214 regulates intracellular adhesion molecule (ICAM1) gene expression in genital Chlamydia muridarum infection. Immunology 145:534–542 10.1111/imm.12470. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Gupta R, Arkatkar T, Keck J, Koundinya GK, Castillo K, Hobel S, Chambers JP, Yu JJ, Guentzel MN, Aigner A, Christenson LK, Arulanandam BP. 2016. Antigen specific immune response in Chlamydia muridarum genital infection is dependent on murine microRNAs-155 and -182. Oncotarget 7:64726–64742 10.18632/oncotarget.11461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Gupta R, Arkatkar T, Yu JJ, Wali S, Haskins WE, Chambers JP, Murthy AK, Bakar SA, Guentzel MN, Arulanandam BP. 2015. Chlamydia muridarum infection associated host MicroRNAs in the murine genital tract and contribution to generation of host immune response. Am J Reprod Immunol 73:126–140 10.1111/aji.12281. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yeruva L, Pouncey DL, Eledge MR, Bhattacharya S, Luo C, Weatherford EW, Ojcius DM, Rank RG. 2016. MicroRNAs modulate pathogenesis resulting from chlamydial infection in mice. Infect Immun 85:e00768-16. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zheng K, Chen DS, Wu YQ, Xu XJ, Zhang H, Chen CF, Chen HC, Liu ZF. 2012. MicroRNA expression profile in RAW264.7 cells in response to Brucella melitensis infection. Int J Biol Sci 8:1013–1022 10.7150/ijbs.3836. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Liu N, Wang L, Sun C, Yang L, Sun W, Peng Q. 2016. MicroRNA-125b-5p suppresses Brucella abortus intracellular survival via control of A20 expression. BMC Microbiol 16:171 10.1186/s12866-016-0788-2. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Luo X, Zhang X, Wu X, Yang X, Han C, Wang Z, Du Q, Zhao X, Liu SL, Tong D, Huang Y. 2018. Brucella downregulates tumor necrosis factor-α to promote intracellular survival via Omp25 regulation of different MicroRNAs in porcine and murine macrophages. Front Immunol 8:2013 10.3389/fimmu.2017.02013. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Sunkavalli U, Aguilar C, Silva RJ, Sharan M, Cruz AR, Tawk C, Maudet C, Mano M, Eulalio A. 2017. Analysis of host microRNA function uncovers a role for miR-29b-2-5p in Shigella capture by filopodia. PLoS Pathog 13:e1006327 10.1371/journal.ppat.1006327. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kotloff KL, Riddle MS, Platts-Mills JA, Pavlinac P, Zaidi AKM. 2018. Shigellosis. Lancet 391:801–812 10.1016/S0140-6736(17)33296-8. [DOI] [PubMed] [Google Scholar]
- 137.Siddle KJ, Tailleux L, Deschamps M, Loh YH, Deluen C, Gicquel B, Antoniewski C, Barreiro LB, Farinelli L, Quintana-Murci L. 2015. Bacterial infection drives the expression dynamics of microRNAs and their isomiRs. PLoS Genet 11:e1005064 10.1371/journal.pgen.1005064. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Faridani OR, Abdullayev I, Hagemann-Jensen M, Schell JP, Lanner F, Sandberg R. 2016. Single-cell sequencing of the small-RNA transcriptome. Nat Biotechnol 34:1264–1266 10.1038/nbt.3701. [PubMed] [DOI] [PubMed] [Google Scholar]
- 139.Dalmasso G, Nguyen HT, Yan Y, Laroui H, Charania MA, Ayyadurai S, Sitaraman SV, Merlin D. 2011. Microbiota modulate host gene expression via microRNAs. PLoS One 6:e19293 10.1371/journal.pone.0019293. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Singh N, Shirdel EA, Waldron L, Zhang RH, Jurisica I, Comelli EM. 2012. The murine caecal microRNA signature depends on the presence of the endogenous microbiota. Int J Biol Sci 8:171–186 10.7150/ijbs.8.171. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu S, da Cunha AP, Rezende RM, Cialic R, Wei Z, Bry L, Comstock LE, Gandhi R, Weiner HL. 2016. The host shapes the gut microbiota via fecal MicroRNA. Cell Host Microbe 19:32–43 10.1016/j.chom.2015.12.005. [PubMed] [DOI] [PMC free article] [PubMed] [Google Scholar]