Tuberculosis (TB) is an infectious disease of extremely high epidemiological burden worldwide that is easily acquired through the inhalation of infected respiratory droplets. The complex pathogenesis of this infection spans from subjects never developing this disease despite intense exposure, to others in which immune containment fails catastrophically and severe or disseminated forms of disease ensue.
KEYWORDS: tuberculosis, miRNA, biomarker, Mycobacterium tuberculosis, infection, disease
ABSTRACT
Tuberculosis (TB) is an infectious disease of extremely high epidemiological burden worldwide that is easily acquired through the inhalation of infected respiratory droplets. The complex pathogenesis of this infection spans from subjects never developing this disease despite intense exposure, to others in which immune containment fails catastrophically and severe or disseminated forms of disease ensue. In recent decades, microRNAs (miRNAs) have gained increasing attention due to their role as gene silencers and because of their altered expression in diverse human diseases, including some infections. Recent research regarding miRNAs and TB has revealed that the expression profile for particular miRNAs clearly changes upon Mycobacterium tuberculosis infection and also varies in the different stages of this disease. However, despite the growing number of studies—some of which have even proposed some miRNAs as potential biomarkers—methodological variations and key differences in relevant factors, such as sex and age, cell type analyzed, M. tuberculosis strain, and antimicrobial therapy status, strongly hinder the comparison of data. In this review, we summarize and discuss the literature and highlight the role of selected miRNAs that have specifically and more consistently been associated with M. tuberculosis infection, together with a discussion of the possible gene and immune regulation pathways involved.
INTRODUCTION
Tuberculosis (TB), an infectious airborne disease caused by Mycobacterium tuberculosis, remains the leading cause of death among infectious diseases from a single agent as well as the ninth leading cause of death worldwide. In 2018, around 10 million people developed TB, and 1.2 million died as a result of this disease, exceeding the combined number of deaths from HIV and malaria during the same period (1). According to the World Health Organization (WHO), about a quarter of the global population is infected with this bacterium and about 5% to 15% will develop the disease in their lifetime, with an increasing risk in subjects with HIV infection or other conditions, such as undernourishment, diabetes, smoking, and heavy alcohol consumption (2).
M. tuberculosis infection is commonly acquired through inhalation of infected respiratory droplets that reach the alveolar space, where mycobacteria undergo phagocytosis by the alveolar macrophages (3–5). Once inside the macrophages, M. tuberculosis displays several mechanisms that avoid or counterfeit the immune response of the host, such as modulating the endosomal/phagosomal maturation pathway, blocking the fusion of the mycobacterial phagosome with the lysosome, and/or avoiding the bactericidal mechanisms of the host cells (3–5).
Currently, the diagnosis of active pulmonary TB (PTB) requires direct M. tuberculosis detection (microbiological or DNA amplification) and, therefore, is highly dependent on the availability of clinical samples, such as sputum, for analysis. Conversely, none of the currently available non-sputum-based tests to diagnose M. tuberculosis infection, such as the tuberculin skin test or immunoassays based on interferon gamma (IFN-γ) release, are able to discriminate between latent TB infection (LTBI) and active TB disease (6). Therefore, in order to improve early diagnosis and control transmission, there has been an intense global effort in the scientific community searching for new biomarkers that might distinguish between the different states of this infection that range from asymptomatic and latent to progressive and destructive active disease.
MicroRNAs (miRNAs) are small, noncoding, single-stranded RNA molecules that bind to specific mRNA to inhibit the translation of multiple genes into proteins (7). Compelling evidence shows that they are involved in a number of normal cellular processes, such as cell cycle control, apoptosis, and several developmental and physiological processes (8–11), and that their altered expression is associated with the pathogenesis of diverse diseases, including cancer and autoimmune conditions (12–16).
In recent years, it has also become evident that miRNAs have a role in the control of several infections (17, 18), mediating antiviral defenses (in plants and animals) (19, 20) and regulating the host cellular environment (21) as well as the immune response (22–24), with a critical role in adaptive immunity (25–28). Particularly, miRNAs can mediate the host response to M. tuberculosis infection mostly through the regulation of proteins involved in the innate and adaptive immune pathways (29).
In this review, recently discovered miRNAs that are related to M. tuberculosis infection are summarized, and we shed light on how these small molecules might be involved in TB pathogenesis and immune response.
miRNAs
miRNAs were first described as small noncoding RNA (ncRNA) with no biological role. However, in 1993, Ambros and Ruvkun discovered quite the opposite, when they found that lin-4 was essential for the normal temporal control of diverse postembryonic developmental events in the nematode Caenorhabditis elegans by negatively regulating the LIN-14 protein level (30, 31). This remarkable finding opened the door to the exploration of a novel cellular regulatory mechanism involving non-protein-coding transcripts and led, a decade later, to the discovery of the let-7 miRNA (32), which was found to be highly conserved across different species ranging from flies to humans (33). Since then, thousands of miRNAs have been widely studied, increasing our understanding of their biogenesis, target recognition, and role in gene regulation (34–38).
It is presently understood that the main biological role of miRNA—negative gene regulation—can be performed in the following three ways: mRNA degradation, translational repression, or miRNA‐mediated mRNA decay. These mechanisms depend on the level of complementarity between the miRNA and the mRNA target. Translation repression and miRNA‐mediated mRNA decay require a limited microRNA target base pairing, while mRNA cleavage requires a more extensive microRNA target base pairing (39).
Currently, the study of miRNA has been accelerated due to new available technologies, such as next-generation sequencing (NGS), specifically, RNA transcriptome sequencing (RNA-seq), allowing researchers to identify and determine the relative expression levels of all miRNA (closely related miRNA and isoforms) within a high number of samples (40). MicroRNA array, conversely, allows the study of multiple miRNA (41), while quantitative real-time PCR (qPCR) and multiplex miRNA profiling allow the validation of several miRNAs across a number of samples with high sensitivity (42, 43). Some of these technologies are considered supplementary to each other in the process of validation of findings, in which using an independent method helps to avoid misleading signals (e.g., RNA-seq can introduce bias through multiple PCR amplification and ligation steps during library preparation) (44).
Presently, more than 2,600 mature miRNA sequences have been identified within the human genome (http://www.mirbase.org/, v.22.1) (45–47), with more than 15,000 target genes and over 380,000 miRNA-target interactions (MTIs) identified and validated by Western blotting, luciferase assay, pulsed stable isotope labeling by/with amino acids in cell culture (pSILAC), microarray, and/or NGS (miRTarBase, v.7.0) (48).
miRNA EXPRESSION PROFILE UPON MYCOBACTERIUM TUBERCULOSIS INFECTION
Individuals with M. tuberculosis infection are usually classified binarily as having LTBI or active TB. However, a variety of stages exist between initial exposure and the development of active disease. After first exposure, some individuals may successfully eliminate the bacterium, and they would no longer carry viable bacteria but may still have immunological evidence of prior infection (49). When the immune system is unable to clear the infection but can control and contain it, the bacteria persist in a latent state of infection for years, may progress in a slow or fast manner to a primary active disease, or can cycle through incipient and subclinical states before developing into symptomatic disease or eventual disease resolution (49). Based on these facts, the fate of M. tuberculosis infection is highly dependent on individual immune response, and therefore genetic differences in the host’s immune regulatory mechanisms may influence the risk of developing clinical TB (50). Gene silencing via host miRNA has been proposed as one of the mechanisms by which human macrophages battle intracellular pathogens like M. tuberculosis (51). However, bacteria can also manipulate host cell pathways by regulating miRNA expression (52). Knowing how miRNAs modulate gene expression upon M. tuberculosis infection and identifying their target genes are critical in understanding the host response to infection. In this regard, a comparative study of three evolutionarily closely related Mycobacterium genomes (M. tuberculosis H37Rv, M. tuberculosis CDC1551, and Mycobacterium leprae TN), in which only conserved interactions between human miRNAs and putative M. tuberculosis target genes were considered, predicted 26 candidate M. tuberculosis genes that might be targeted by human miRNAs in lung and macrophages (51).
The miRNA expression profile can also be determined by phylogenetic differences in M. tuberculosis strains, as suggested by an in vitro model of THP-1 cells infected with Beijing/W versus the non-Beijing/W M. tuberculosis genotypes. Macrophages infected with the Beijing/W strain (more virulent) exhibited a repression of 13 miRNAs with respect to macrophages infected with the non-Beijing/W strain (53). Furthermore, profound differences in miRNA expression have also been described in relation to drug-resistant M. tuberculosis strains compared to drug-sensitive strains (54), a factor to be considered for study comparisons.
Interestingly, miRNA expression is also modified under the effect of successful antimicrobial therapy. Wang et al. (55) found that serum levels of miR-21-5p, miR-92a-3p, and miR-148b-3p distinguished between untreated, partially treated (for 2 months), and cured TB, in comparison with those with noninfection. Besides, these miRNAs were significantly reduced in patients with cured TB compared to those of patients with untreated TB, and miR-125a-5p levels were upregulated in the partially treated TB samples compared to those of untreated or cured TB. Also, significant increases in miR-21-5p, miR-92a-3p, and miR-148b-3p levels were found in patients with untreated TB compared to those of healthy controls, and miR-21-5p alone discriminated between cured and untreated TB (area under the receiver operating characteristic [ROC] curve [AUC] = 0.851). In a similar study, serum levels of miR-16 and miR-155 exhibited a down- and upregulation, respectively, after the completion of treatment for active TB, while no significant difference was observed when compared to levels in uninfected subjects (56). These studies suggest that miRNAs might be useful as biomarkers to distinguish between the different stages of TB infection or therapy responsiveness; however, none of them have been validated in prospective studies, and patients with other infections should be included.
PITFALLS OF miRNA PROFILING IN MYCOBACTERIUM TUBERCULOSIS INFECTION AND DISEASE
A substantial number of studies have collected an enormous amount of data about miRNA expression profiles in different stages of M. tuberculosis infection and disease. Table 1 summarizes these results, compares different samples from healthy volunteers, subjects with LTBI, and patients with active TB, and shows the number of miRNAs found to be up- or downregulated. Discrepancies among studies due to methodological variations, choice of biological sample, differing stages of TB infection, and differences in RNA extraction processes, number of miRNAs obtained, cut-off selection criteria, and data analysis process (including data normalization) introduce a potential bias that influences these results and must be considered for accurate interpretation. For example, when comparing serum samples from patients with active TB and healthy controls, a study showed 59 upregulated and 33 downregulated miRNAs (57), while another report showed 10 and 25 miRNAs, respectively (58). Regardless of these variations, several miRNAs have been consistently shown to be either upregulated or downregulated across studies (Table 2). Particularly, hsa-let-7g, hsa-miR-30c, and hsa-miR-433 showed an upregulation in serum samples (57, 59, 60), and hsa-miR-1270, hsa-miR-371-3p, hsa-miR-380*, hsa-miR-582-3p, and hsa-miR-618 showed a downregulation in sputum (61) and serum samples (57). However, some other miRNAs presented a variable regulation depending on the study or sample analyzed (Table 3). As an example, hsa-miR-1 showed an upregulation in sputum samples (61), a downregulation in peripheral blood mononuclear cells (PBMCs) (62), and both an up- and downregulation in serum samples (60, 63) of patients with TB when compared to those of samples from healthy controls.
TABLE 1.
Differential miRNA expression profiles in different stages of tuberculosis
Samplea | No. of upregulated miRNAs | No. of downregulated miRNAs | No. of validated miRNAs | Participantsa | Reference |
---|---|---|---|---|---|
PBMC | 28 | 2 | 1 | PTB vs control | Liu et al., 2011 (78) |
PBMC | 9 | 1 | LTBI vs ATB | Wu et al., 2014 (62) | |
1 | ATB vs control | ||||
16 | 6 | LTBI vs control | |||
PBMC | 15 | 14 | 8 | ATB vs controlb | Zhou et al., 2016 (86) |
CD4+ T cells | 82 | 53 | 6 | ATB vs control | Fu et al., 2014 (117) |
33 | 46 | LTBI vs control | |||
62 | 62 | LTBI vs ATB | |||
Serum | 59 | 33 | 3 | ATB vs control | Fu et al., 2011 (57) |
Serum | 90 | 7 | 7 | PTB vs control | Qi et al., 2012 (59) |
Serum | 44 | 47 | 6 | PTB vs control | Zhang et al., 2013 (63) |
Serum | 62 | 33 | 2 | ATB vs LTBI | Zhang et al., 2014 (60) |
82 | 22 | ATB vs BCG+ | |||
37 | 65 | ATB vs BCG− | |||
24 | 6 | ATB vs three groups | |||
Serum | 10 | 25 | ATB vs control | Xu et al., 2015 (58) | |
Differentiated macrophages derived from PBMC | 6 | 3 | 11 | ATB vs control | Zheng et al., 2015 (53) |
8 | 1 | LTBI vs control | |||
4 | LTBI vs ATB | ||||
THP-1 cells | 1 | 13 | 14 | Beijing/W vs non-Beijing/W M. tuberculosis strains | |
Sputum | 43 | 52 | 3 | ATB vs control | Yi et al., 2012 (61) |
ATB, active tuberculosis; PTB, pulmonary tuberculosis; LTBI, latent tuberculosis infection; PBMC, peripheral blood mononuclear cells.
Samples obtained from children (2 to 14 years old).
TABLE 2.
miRNAs consistently upregulated and downregulated across studies analyzing miRNA expression profile
miRNAa | Samples | References |
---|---|---|
Upregulated miRNA | ||
hsa-let-7e | Serum/serum | Qi et al., 2012 (59)/Zhang et al., 2014 (60) |
hsa-miR-127-3p | ||
hsa-miR-145 | ||
hsa-miR-21 | ||
hsa-miR-24 | ||
hsa-miR-25 | ||
hsa-miR-26b | ||
hsa-miR-340 | ||
hsa-miR-432 | ||
hsa-miR-20a | Qi et al., 2012 (59)/Xu et al., 2015 (58) | |
hsa-miR-93 | Qi et al., 2012 (59)/Zhang et al., 2013 (63) | |
hsa-miR-191 | Fu et al., 2011 (57)/Zhang et al., 2014 (60) | |
hsa-miR-23b | ||
hsa-miR-146b-5p | Fu et al., 2011 (57)/Zhang et al., 2013 (63) | |
hsa-miR-483-5p | ||
hsa-miR-122 | Zhang et al., 2013 (63)/Zhang et al., 2014 (60) | |
hsa-miR-29c | ||
hsa-miR-320c | ||
hsa-miR-378 | Zhang et al., 2013 (63)/Xu et al., 2015 (58) | |
hsa-miR-193a-3p | Wu et al., 2014 (62)/Zhang et al., 2014 (60) | |
hsa-miR-143 | Serum/serum/serum | Fu et al., 2011 (57)/Zhang et al., 2014 (60)/Xu et al., 2015 (58) |
hsa-let-7g | Fu et al., 2011 (57)/Qi et al., 2012 (59)/Zhang et al., 2014 (60) | |
hsa-miR-30c | ||
hsa-miR-433 | ||
hsa-miR-22 | Fu et al., 2011 (57)/Zhang et al., 2013 (63)/Zhang et al., 2014 (60) | |
hsa-miR-151-3p | Serum/sputum | Qi et al., 2012 (59)/Yi et al., 2012 (61) |
hsa-miR-409-3p | ||
hsa-miR-1204 | Fu et al., 2011 (57)/Yi et al., 2012 (61) | |
hsa-miR-23a | Zhang et al., 2014 (60)/Yi et al., 2012 (61) | |
hsa-miR-548d-5p | Xu et al., 2015 (58)/Yi et al., 2012 (61) | |
hsa-miR-376c | Serum/serum/sputum | Qi et al., 2012 (59)/Zhang et al., 2014 (60)/Yi et al., 2012 (61) |
hsa-miR-29a | Serum/serum/serum/sputum | Fu et al., 2011 (57)/Qi et al., 2012 (59)/Zhang et al., 2014 (60)/Yi et al., 2012 (61) |
hsa-miR-144* | PBMC/serum | Liu et al., 2011 (78)/Qi et al., 2012 (59) |
hsa-miR-660 | ||
hsa-miR-296-5p | Liu et al., 2011 (78)/Zhang et al., 2014 (60) | |
hsa-miR-142-3p | Wu et al., 2014 (62)/Qi et al., 2012 (59) | |
Downregulated miRNA | ||
hsa-miR-1270 | Serum/sputum | Fu et al., 2011 (57)/Yi et al., 2012 (61) |
hsa-miR-371-3p | ||
hsa-miR-380* | ||
hsa-miR-582-3p | ||
hsa-miR-618 |
hsa, Homo sapiens.
TABLE 3.
miRNAs with variable expression (either upregulation or downregulation) across studies analyzing miRNA expression profile
miRNA | Upregulation sample (reference) | Downregulation sample (reference) |
---|---|---|
hsa-miR-1284 | Serum (Fu et al., 2011 [57]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-139-3p | Serum (Qi et al., 2012 [59]) | Serum (Zhang et al., 2014 [60]) |
hsa-miR-15b | ||
hsa-miR-330-3p | ||
hsa-miR-886-5p | ||
hsa-miR-889 | ||
hsa-miR-17 | Serum (Qi et al., 2012 [59]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-19b | ||
hsa-miR-28-3p | ||
hsa-miR-345 | ||
hsa-miR-206 | Serum (Zhang et al., 2014 [60]) | Serum (Fu et al., 2011 [57]) |
hsa-miR-221* | ||
hsa-miR-301a | Serum (Qi et al., 2012 [59]) | Serum (Zhang et al., 2013 [63]) |
hsa-miR-320b | Serum (Zhang et al., 2014 [60]) | Serum (Zhang et al., 2013 [63]) |
hsa-miR-320d | Serum (Zhang et al., 2014 [60]) | Serum (Fu et al., 2011 [57]) |
hsa-let-7c | Serum (Zhang et al., 2014 [60]) | Sputum (Yi et al., 2012 [61]) |
hsa-miR-100 | ||
hsa-miR-181a | ||
hsa-miR-1260b | Serum (Fu et al., 2011 [57]) | Sputum (Yi et al., 2012 [61]) |
hsa-miR-10a | Serum (Zhang et al., 2014 [60]) | PBMC (Wu et al., 2014 [62]) |
hsa-miR-31 | ||
hsa-miR-181a-2* | Serum (Zhang et al., 2013 [63]) | PBMC (Wu et al., 2014 [62]) |
hsa-miR-199a-5p | Serum (Zhang et al., 2014 [60]) | PBMC (Liu et al., 2011 [78]) |
hsa-miR-335 | Serum (Qi et al., 2012 [59]) | PBMC (Liu et al., 2011 [78]) |
hsa-miR-29b | PBMC (Wu et al., 2014 [62]) | Serum (Zhang et al., 2014 [60]) |
hsa-miR-503 | ||
hsa-miR-452 | PBMC (Liu et al., 2011 [78]) | Serum (Zhang et al., 2014 [60]) |
hsa-miR-532-5p | PBMC (Liu et al., 2011 [78]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-101 | Serum (Fu et al., 2011 [57]), serum (Zhang et al., 2014 [60]) | Serum (Zhang et al., 2013 [63]) |
hsa-miR-196b | Serum (Qi et al., 2012 [59]), serum (Zhang et al., 2014 [60]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-199a-3p | ||
hsa-miR-30d | ||
hsa-miR-20b | Serum (Qi et al., 2012 [59]), serum (Zhang et al., 2013 [63]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-26a | Serum (Qi et al., 2012 [59]), serum (Zhang et al., 2014 [60]) | Serum (Zhang et al., 2013 [63]) |
hsa-miR-744 | Serum (Fu et al., 2011 [57]), serum (Qi et al., 2012 [59]) | Sputum (Yi et al., 2012 [61]) |
hsa-miR-99b | Serum (Qi et al., 2012 [59]), serum (Zhang et al., 2014 [60]) | Sputum (Yi et al., 2012 [61]) |
hsa-let-7i | Serum (Fu et al., 2011 [57]), serum (Zhang et al., 2014 [60]) | PBMC (Zhou et al., 2016 [86]) |
hsa-miR-125b | Serum (Fu et al., 2011 [57]), serum (Zhang et al., 2014 [60]) | PBMC (Wu et al., 2014 [62]) |
hsa-miR-146a | Serum (Fu et al., 2011 [57]), serum (Qi et al., 2012 [59]) | PBMC (Wu et al., 2014 [62]) |
hsa-miR-103 | Serum (Fu et al., 2011 [57]), serum (Qi et al., 2012 [59]), serum (Zhang et al., 2014 [60]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-151-5p | Serum (Zhang et al., 2014 [60]), sputum (Yi et al., 2012 [61]) | Serum (Xu et al., 2015 [58]) |
hsa-miR-1 | Serum (Zhang et al., 2014 [60]), sputum (Yi et al., 2012 [61]) | PBMC (Wu et al., 2014 [62]), serum (Zhang et al., 2013 [63]) |
hsa-let-7b | Serum (Zhang et al., 2014 [60]) | Serum (Xu et al., 2015 [58]), sputum (Yi et al., 2012 [61]) |
Another very common problem encountered across miRNA TB biomarker studies including human samples is that the varying stages of TB infection are not always well defined, and recent infection, long-standing LTBI, newly diagnosed active TB, or treated TB are not clearly differentiated, hindering the group comparisons. In addition, very often, there is no differential analysis between males and females, even though this factor has been reported to define a different profile expression for several miRNAs (64).
Importantly, potential pitfalls can also be present in the current platforms used for miRNA expression profiling. The search for miRNAs as possible biomarkers usually involves studies of NGS or microRNA array, due to their advantages in allowing the identification and analysis of multiple targets (40, 41). However, both techniques may have disadvantages. NGS can produce misleading results by introducing bias during the multiple PCR amplification, where the expression levels of some RNAs can be artificially enhanced, diminished, or even undetectable. Also, during the ligation steps, the use of different adapters and barcodes influences cDNA synthesis efficacy, and the variable-specific RNA G+C content can be associated with unequal PCR amplification efficiency (44). Regarding microRNA arrays, this platform permits the study of a large number of miRNAs already identified; however, data comparison is hindered by the divergence in the stringency of the detection call criteria and the poor inter-platform concordance of microRNA expression values (65). Another step commonly involved in miRNA studies involves the validation of NGS or microarray findings by qPCR amplification. This technique allows miRNA profiling and validation with high sensitivity (42). However, qPCR data normalization remains one of the most challenging steps due to the lack of a reliable normalizer for miRNA quantification, hindering the accurate data normalization strategy (66, 67). Currently, small RNAs, in particular RNU6B, are widely used as a normalizer for miRNAs; however, it has been shown that it can experience changes in serum samples in a disease-specific manner. Therefore, it may not be a good normalizer for inflammatory conditions (68). In addition, small RNAs do not share the same biological and biochemical properties of miRNA molecules in terms of their transcription, processing, and tissue-specific expression patterns (68, 69). Therefore, it would be most suitable to use as a qPCR reference other miRNAs exhibiting stable expression under the same experimental conditions (70). Alternatively, another more rigorous strategy for selection of a good qPCR normalizer gene proposes the use of a global mean normalization of a set of reference genes (71, 72). According to this method, the geometric mean of a minimum of three stable housekeeping genes provided a more reliable normalization factor that can control for outliers and differences in abundance between genes (73).
ROLE OF VALIDATED miRNA IN DIFFERENT STAGES OF DISEASE
Active TB is believed to reflect a failure of the immune response restraining M. tuberculosis, with uncontrolled replication leading to progressive development of TB pathology and clinical manifestations. In this context, several investigators have looked for changes in molecular elements that might be related to TB infection development. When looking at miRNAs, a variable number of circulating or intracellular miRNAs have been found and described accordingly in the different stages of M. tuberculosis infection. Of these, only a small proportion has been further validated after discovery (Table 4). As an example, among validated miRNA, hsa-miR-196b and hsa-miR-376c exhibited a clear upregulation in serum from patients with active TB compared with that of the control group (LTBI and uninfected controls) (60).
TABLE 4.
miRNAs involved in different disease stages validateda in different samples and studies
Sample | Validated miRNA | Regulation | Individuals | Reference |
---|---|---|---|---|
PBMCs | hsa-miR-144* | Upregulated | PTB vs control | Liu et al., 2011 (78) |
hsa-miR‑29b | Upregulated | Zhou et al., 2016 (86)b | ||
hsa-miR‑1 | Downregulated | |||
hsa-miR‑150 | Downregulated | |||
hsa-miR‑155 | Downregulated | |||
hsa-miR‑31 | Downregulated | |||
hsa-miR‑146a | Downregulated | |||
hsa-miR‑10a | Downregulated | |||
hsa-miR‑125b | Downregulated | |||
Serum | hsa-miR-93* | Upregulated | PTB vs control | Fu et al., 2011 (57) |
hsa-miR-29a | Upregulated | |||
hsa-miR-3125 | Downregulated | |||
hsa-miR-361-5p | Upregulated | Qi et al., 2012 (59) | ||
hsa-miR-889 | Upregulated | |||
hsa-miR-576-3p | Upregulated | |||
hsa-miR-210 | Upregulated | |||
hsa-miR-26a | Upregulated | |||
hsa-miR-432 | Upregulated | |||
hsa-miR-134 | Upregulated | |||
hsa-miR-378 | Upregulated | Zhang et al., 2013 (63) | ||
hsa-miR-483-5p | Upregulated | |||
hsa-miR-22 | Upregulated | |||
hsa-miR-29c | Upregulated | |||
hsa-miR-320b | Downregulated | |||
hsa-miR-101 | Downregulated | |||
hsa-miR-196b | Upregulated | ATB vs control | Zhang et al., 2014 (60) | |
hsa-miR-376c | Upregulated | ATB vs LTBI/BCG− | ||
hsa-miR-16 | Upregulated | PTB vs control and TB treated | Wagh et al., 2017 (56) | |
hsa-miR-155 | Downregulated | |||
hsa-miR-148b-3p | Upregulated | Control vs ATB; ATB vs partially treated TB; ATB vs cured TB | Wang et al., 2018 (55) | |
hsa-miR-21-5p | Upregulated | |||
hsa-miR-92a-3p | Upregulated | Control vs ATB; ATB vs cured TB | ||
Has-miR-125a-5p | Upregulated | Partially treated TB vs untreated; partially treated TB vs cured TB | ||
hsa-miR-144 | Upregulated | ATB vs control | Lv et al., 2016 (74) | |
Sputum | hsa-miR-29a | Upregulated | PTB vs control | Fu et al., 2011 (57) |
hsa-miR-19b-2* | Downregulated | Yi et al., 2012 (61) | ||
hsa-miR-3179 | Upregulated | |||
hsa-miR-147 | Upregulated | |||
CD4+ cells | miR-451a | Upregulated | ATB vs control | Fu et al., 2014 (117) |
miR-340-5p | Upregulated | |||
miR-136-5p | Upregulated | |||
miR-4292 | Downregulated | |||
miR-29b | Upregulated | |||
THP-1 cells | hsa-miR-16 | Upregulated | ATB vs control | Zheng et al., 2015 (53) |
hsa-miR-137 | Upregulated | |||
hsa-miR-140-3p | Upregulated | |||
hsa-miR-193a-3p | Upregulated | |||
hsa-miR-501-5p | Upregulated | |||
hsa-miR-598 | Upregulated | |||
hsa-miR-95 | Downregulated |
Expression of miRNAs was confirmed by real-time PCR (qPCR) after sequencing.
Samples obtained from children (2 to 14 years old).
In the following sections, the analysis is focused on six miRNAs that have been specifically and consistently associated with TB disease (hsa-miR-144-3p, hsa-miR-144-5p, hsa-miR-146a, hsa-miR-155, hsa-miR-21, and hsa-miR-29a) according to the miRTarBase database (48), and evidence is shown that backs up this association as well as the possible gene regulation pathways involved.
miR-144-3p.
The expression of miR-144-3p in sputum and serum from patients with active TB was not only higher than that of healthy uninfected controls but was also able to discriminate patients that showed improvement after antibiotic treatment from those that did not (74). Accumulative evidence suggests that autophagy is a key process of innate immune response employed by the host against M. tuberculosis and other intracellular bacteria (75, 76). Autophagy is regulated by numerous distinct autophagy-related (ATG) proteins. Among the ATGs, ATG4 and microtubule-associated protein light chain 3 (LC3) are essential for autophagy induction. Interestingly, Mycobacterium bovis bacillus Calmette-Guérin (BCG) infection of murine macrophages induced increased expression of miR-144-3p, which targets ATG4a to inhibit autophagy activation and antimicrobial responses to BCG (Fig. 1A). Overexpression of miR-144-3p decreased both mRNA and protein levels of ATG4a as well as protein levels of p62 (molecular carrier of cargo to be degraded by autophagy) and LC3I/II, inhibiting maturation of autophagosome in macrophages, thus increasing intracellular survival of BCG. Conversely, when macrophage miR-144-3p expression was inhibited, ATG4a levels were increased, leading to an accelerated autophagic response and a decrease in BCG survival (77).
FIG 1.
miRNA regulation of miR-144-3p and miR-144-5p upon mycobacterial infection. (A) After Mycobacterium bovis (BCG) infection in murine macrophages and dendritic cells, miR-144-3p is upregulated and targets ATG4a mRNA to inhibit autophagy enhancing mycobacterial survival. (B) In human monocytes infected with Mycobacterium tuberculosis, miR-144-5p targets DRAM2 mRNA to inhibit autophagy promoting mycobacterial survival.
miR-144-5p.
Several studies have shown that expression levels of miR-144-5p are increased in PBMCs from active TB patients compared to those of the same cells from uninfected healthy controls (78–80); however, it is worth noting that miR-144-5p expression might be under a compartmentally distinct type of regulatory process, since pleural fluid mononuclear cells exhibited lower levels of miR-144-5p than PBMCs from patients with pleural TB (80). Evidence from in vitro experiments using purified T cells overexpressing miR-144-5p revealed that this miRNA inhibited the proliferation rate of T cells and the production of crucial Th1 cytokines, such as IFN-γ and tumor necrosis factor alpha (TNF-α), in response to TB infection (78). Additionally, miR-144-5p was capable of downregulating DNA damage-regulated autophagy modulator 2 (DRAM2) and lysosome-associated membrane protein 2 (LAMP2), reducing their mRNA expression in human monocytes. Since DRAM2 and LAMP2 are important proteins in the initiation of autophagy and in the chaperone-mediated autophagy, respectively, these findings suggest that upon infection, miR-144-5p levels contribute to autophagy inhibition, allowing M. tuberculosis to escape the immune system (Fig. 1B) (79).
miR-146a.
Contradictory results have been found regarding miR-146a expression levels. Some studies showed that its expression was upregulated in murine peritoneal fluid after BCG challenge; in human THP-1 cells, murine bone marrow-derived macrophages (BMDMs), and human monocyte-derived macrophages (MDM) upon BCG infection; in lungs and MDM of experimentally M. tuberculosis-infected mice; and in human dendritic cells (DCs) infected with M. tuberculosis (81–85). Conversely, studies performed on a large range of patients with active TB, specifically in white blood cells (children with active TB), PBMCs (pulmonary and extra pulmonary TB), mononuclear cells of pleural fluid, and in plasma samples, showed a downregulation in miR-146a expression (80, 86, 87). Additionally, this downregulation was also observed in alveolar macrophages of bronchoalveolar lavage fluid from patients with active PTB, where miRNA levels in alveolar macrophages from smear-positive patients were lower than in those from smear-negative patients, and higher in healthy uninfected subjects (81). Moreover, miR-146a expression was also downregulated in CD4+ T cells and CD19+ B lymphocytes obtained from mice challenged with M. bovis BCG (83) as well as in liver tissue from BCG-infected mice (82).
The role of miR-146a in immune regulation involved and required the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway for its induction after BCG infection in vitro, and, in fact, mutation of NF-κB sites and pharmacological inhibition of NF-κB signaling significantly decreased miR-146a basal promoter activity and its expression in mycobacterium-challenged macrophages (81, 82). Interleukin-1 receptor-associated kinase 1 (IRAK1) (critical in initiating an innate immune response against foreign pathogens and important in leading to NF-κB nuclear translocation and activation), TNF receptor-associated factor 6 (TRAF6) (a signal transducer in the NF-κB pathway), and prostaglandin-endoperoxide synthase 2 (PTGS2) (responsible for the production of inflammatory prostaglandins) are potential regulatory targets of miR-146a since it downregulated their mRNA levels in THP-1 cells (Fig. 2A) (81). In addition, miR-146a inhibited NF-κB, c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK) pathways in RAW264.7 cells, impairing BCG-induced iNOS expression and nitric oxide (NO) production in macrophages by reducing the TRAF6 protein level (Fig. 2B) (82).
FIG 2.
Dual role of miR-146a in the survival of mycobacteria. (A) Upon Mycobacterium bovis (BCG) infection in macrophages differentiated from human THP-1 cells, miR-146a increases its expression and downregulates transcription factors to suppress the inflammatory response leading to a reduction in mycobacterial survival. (B and C) After BCG infection in murine macrophages, miR-146a increases its expression and downregulates transcription factors and cytokines to suppress the inflammatory response leading to an increase in mycobacterial survival.
Mycobacterial growth or bacterial fitness might also be regulated by miR-146a, as a lower mycobacterial colony count was found in macrophages differentiated from human THP-1 cells transfected with a miR-146a mimic (81). In contrast, in RAW264.7 cells, miR-146a promoted the survival of M. bovis BCG by suppressing the inflammatory response (Fig. 2C) (88) while miR-146a inhibition enhanced the mycobactericidal activity and, thus, mycobacterial clearance due to an increase in NO production (82). A possible explanation for the differences in mycobacterial survival found between these two studies could be the distinct origin of the cells analyzed, i.e., macrophages differentiated from the human monocytic THP-1 cell line (81) versus the mouse macrophage RAW264.7 cell line (88). Other methodological differences, such as the incubation period for the infection or the assay used to measure mycobacterial load, could also explain the differences found in these results.
Interestingly, a higher risk of developing active PTB has been related to miR-146 single-nucleotide polymorphisms (SNPs) in Tibetan, Kasak, and Han populations but not in others such as the Uygur (90, 91).
miR-155.
A few studies have shown that miR-155 levels were reduced in serum from patients with active TB (56, 92) and increased after TB therapy, although not reaching the same levels as those in uninfected subjects (56). Another study showed that human MDMs exposed to lipomannan from the M. tuberculosis H37Rv strain exhibited a lower miR-155 expression with correspondingly low TNF-α production (Fig. 3A) (93); this reduction in miR-155 levels was also present in human macrophages infected with a virulent M. tuberculosis strain, compared with macrophages infected with the avirulent strain or uninfected macrophages (Fig. 3B) (94). Interestingly, natural killer (NK) cells from TB patients that exhibited higher serum levels of miR-155 displayed lower cytotoxicity and lower TNF-α production in in vitro assays (92). Opposing these findings, a very large number of studies have shown miR-155 upregulation occurs in several types of cells and tissues in response to M. tuberculosis. These cases include RAW264.7 cells after M. tuberculosis infection (95), lung tissue from a PTB murine model at early time points of infection (30 days) (83), murine BMDMs and RAW264.7 cells challenged with M. bovis BCG (96), the J774A.1 mouse macrophage cell line infected with M. tuberculosis H37Rv and to a lesser extent when infected with M. bovis and M. bovis BCG (97), PBMCs from patients with active TB stimulated with purified protein derivatives of tuberculin (PPD) (98), and THP-1 cells infected with BCG (99).
FIG 3.
miRNA regulation of miR-155 upon mycobacterial infection and dual role of miR-155 in the survival of mycobacteria. (A and B) Upon lipomannan stimulation in human monocyte-derived macrophages (MDM) or Mycobacterium tuberculosis infection in human macrophages, miR-155 expression decreases. (C) Expression of miR-155 in macrophages stimulated with ESAT-6 increases and triggers a series of events to increase apoptosis and lead to a reduction of mycobacterial survival. (D) After M. tuberculosis infection in mice, miR-155 expression increases in liver and lungs, which is relevant to the immune response, since mice lacking miR-155 exhibit higher susceptibility and lower survival. (E) Upon Mycobacterium bovis (BCG) infection in macrophages, miR-155 expression increases and triggers a series of events to increase autophagy to reduce mycobacterial survival. (F to H) M. tuberculosis infection of human dendritic cells and murine macrophages or BCG infection of human monocytes promotes miR-155 expression that leads to downregulation of transcription factors that consequently favors the survival of the intracellular pathogens. (I) In mouse experiments, miR-155 expression induced by M. tuberculosis infection or ESAT-6 stimulation promotes Toll-like receptor pathway activation, upregulating the expression of certain cytokines that leads to renal injuries.
The upregulation of miR-155 in RAW264.7 cells induced by M. tuberculosis peptide ESAT-6 stimulation depended on the activation of the Toll-like receptor 2 (TLR2)/NF-κB pathway. In turn, the SOCS1 pathway was targeted by miR-155 for the ESAT-6-mediated protective immune response and macrophage apoptosis (Fig. 3C) (100). Moreover, experiments with mice lacking miR-155 (miR-155−/−) showed that they are more susceptible to M. tuberculosis infection and exhibited lower numbers of CD4+ T cells and an impaired M. tuberculosis clearance compared with the wild-type mice (Fig. 3D) (101). Additionally, IFN-γ levels measured in supernatants from cultures of splenocytes from M. tuberculosis-infected mice and stimulated in vitro with M. tuberculosis were lower in the miR-155−/− mice than in the wild type. These results suggest that miR-155 has an important role in the in vivo immune response against TB. Moreover, in infected RAW264.7 cells, miR-155 decreased the survival of intracellular BCG, M. tuberculosis H37Ra, and H37Rv. This was done targeting and downregulating posttranscriptionally the protein Rheb, consequently promoting the maturation of mycobacterial phagosomes and also activating autophagy (Fig. 3E) (96).
Virulent and live mycobacterium infection of human DCs upregulated miR-155 in opposition to BCG and heat-inactivated M. tuberculosis. The increased expression of miR-155 consequently decreased ATG3 protein levels as well as LC3-II and gamma-amino butyric acid receptor-associated protein (GABARAP) (involved in apoptosis and autophagy), thus inhibiting autophagosome formation (Fig. 3F) (85). In this same regard, human MDMs infected with M. tuberculosis H37Rv or stimulated with ESAT-6 showed an increase in miR-155 levels (102), as well as the human monocytic leukemia cell line (U937 cells) infected with BCG, where the upregulation was further enhanced by stimulating with Rv2346c, a putative ESAT-6-like protein of M. tuberculosis (103).
In contrast, opposing results found in M. tuberculosis-infected RAW264.7 cells have shown that miR-155 was involved in the downregulation of target proteins, such as SH2 domain-containing inositol 5′-phosphatase 1 (SHIP1) (affects cellular behaviors in innate and adaptive immune cells), transcription regulator protein BACH1 (negative regulator of hmox1 that increases its expression upon M. tuberculosis infection), Cox-2/PTGS2, and interleukin-6 (IL-6) (important in the differentiation of B-cells and required for the generation of Th17 cells), benefiting mycobacterial growth (Fig. 3G) (95). Other studies have reported that miR-155 contributed to mycobacterial survival by reducing apoptosis through modulation of Forkhead box O3 (FOXO3) (regulates apoptosis and autophagy) in the THP-1 human monocytic cell line (Fig. 3H) (99). Also, miR-155 increased mRNA expression of TLR4, MyD88, TNF-α, IL-17, and IFN-γ through the TLR4 signaling pathway, promoting the progression of infection, leading to renal injuries in extra PTB disease in mice (Fig. 3I) (104).
Furthermore, studies exploring the diagnostic value of specific miRNAs in TB have described that lower levels of miR-155 could be a potential biomarker for childhood TB (86) and in adults could discriminate between PTB and control groups with diagnostic AUCs consistently found above 0.9 (56, 86, 105).
miR-21.
The role of miR-21 in TB pathogenesis has been less studied; however, a lower expression was found in CD4+ T cells and plasma from patients with active TB than from subjects with LTBI (87, 106) but there was no difference between subjects with LTBI and healthy volunteers (106). Conversely, miR-21 expression was upregulated in M. tuberculosis-infected macrophages compared to uninfected macrophages and macrophages infected with M. bovis or M. bovis BCG (97). The same was observed in RAW264.7 and THP-1 cells where miR-21 enhanced M. tuberculosis survival and apoptosis and attenuated the secretion of inflammatory cytokines (IL-1β, IL-6, and TNF-α) by targeting B-cell lymphoma-2 (Bcl-2) and Toll-like receptor 4 (TLR4) (Fig. 4A) (107). Finally, miR-21 levels also increased in PPD-MPT64-stimulated RAW264.7 cells in contrast to PPD-stimulated cells, showing that M. tuberculosis immunogenic protein MPT64 upregulates miR-21 due to increased NF-κB expression, leading to the upregulation of Bcl-2, an antiapoptotic protein, thus decreasing cell death (Fig. 4B) (108). Possible explanations for the discrepancies related to the enhanced or decreased apoptosis might be due to the different stimulus performed (M. tuberculosis versus MPT64), which triggered different mechanisms leading to decreased or increased Bcl-2 levels that in turn correlate with the level of apoptosis.
FIG 4.
Dual role of miR-21 upon mycobacterial infection. (A) Expression of miR-21 is increased in human and murine macrophages infected with Mycobacterium tuberculosis, driving downregulation of Bcl-2 and TLR-4, suppression of inflammatory cytokines, and enhanced apoptosis and mycobacterial survival. (B) After MPT64 stimulation in murine macrophages, miR-21 is regulated by NF-κB to increase Bcl-2 expression, reducing apoptosis.
miR-29a.
A study shows that miR-29a expression depended on sex, where PBMCs from females with active TB exhibited a lower expression than PBMCs of male patients before beginning TB therapy and after up to 2 months of treatment. After completing 6 months of treatment, miR-29a levels increased in females, while showing a trend toward reduced levels in males (64). Discrepancies in miR-29a expression arose upon antimycobacterial therapy; while one study showed no significant difference in serum samples (56), other studies showed increased levels of miR-29a in CD4+ T cells (64, 109) as well as in plasma samples, where it was reduced to levels equivalent to healthy controls (87). Contradictory results in miR-29a expression were also found regarding the status of M. tuberculosis infection. In PBMCs from children with tuberculosis meningitis and in plasma from patients with active TB, miR-29a expression was found to be higher than that in healthy individuals (87, 110, 111). Additionally, human macrophages infected with M. tuberculosis exhibited an enhancement in miR-29a expression that led to the downregulation of caspase-7 (CASP7) (a key regulator of apoptosis), preventing cell death and allowing mycobacterial intracellular growth (Fig. 5A) (112). Conversely, in CD4+ T cells from patients with active TB and in whole blood from children with TB, miR-29a expression was low compared to that of their LTBI counterpart, and while no difference was exhibited between LTBI and healthy adults, miR-29a expression was found to be low in healthy children compared to that in children with LTBI (106). The expression of miR-29a was also reduced in CD4+ and CD8+ T cells from mice infected with M. bovis BCG; interestingly, both types of T cells showed an increase in IFN-γ concentration, suggesting an inverse correlation between miR-29a and IFN-γ expression during the infection (Fig. 5B) (113). Concordantly, in mice transfected with microRNA sponges—transcripts containing multiple miRNA-binding sites that sequester miRNAs inhibiting their function (114, 115)—and infected with M. bovis, the inverse correlation between miR-29a and IFN-γ was also observed. In addition, the challenged mice with M. tuberculosis PPD in the footpads exhibited more swelling in the infection site, more leukocyte aggregation, increased levels of IFN-γ, lower bacterial burden in the lungs, and a longer life expectancy than the nonchallenged control mice. Together, these results showed that miR-29a regulates the innate and adaptive immunity suppressing IFN-γ through its interaction with IFN-γ mRNA, protein Argonaute 2 (Ago2) (required for RNA-mediated gene silencing), and the RNA-induced silencing complex (RISC) (113). However, evidence also suggested that miR-29a might not alter IFN-γ production in CD4+ T cells (106, 109). Furthermore, miR-29a might also regulate other cytokines and chemokines since its inhibition in M. tuberculosis-infected DCs showed an enhanced response of 12 cytokines and chemokines, including IL-12B, IL-2RA, and CXCL10, predicted targets of miR-29a, compared to that of infected but noninhibited cells. Finally, M. tuberculosis-infected DCs transfected with a miR-29a mimic exhibited a reduced CXCL10 expression, suggesting a direct regulation of miR-29a on this chemokine in M. tuberculosis-infected DCs (116).
FIG 5.
Regulation of miR-29a upon mycobacterial infection. (A) Human macrophages infected with Mycobacterium tuberculosis show enhanced miR-29a expression that leads to downregulation of caspase-7, prevention of cell death, and mycobacterial survival. (B) CD4+ and CD8+ T cells isolated from mice infected with Mycobacterium bovis (BCG) exhibit a downregulation of miR-29a but increased levels of IFN-γ, showing an inverse dynamic between these two molecules.
CONCLUSIONS
Distinctive miRNAs, differentially expressed in the diverse stages of TB infection, might shed light on the nature of the immune response to this elusive pathogen. However, great variability in miRNA expression and its biological effects are dependent on the experimental model conditions, increasing difficulties to conclude any clear or unique effect on the immune response to TB, if any. Furthermore, although some miRNAs, such as miR-21-5p and miR-155, are more consistent as potential diagnostic biomarkers for TB in humans, it is clear that further validation is needed. Nevertheless, the following considerations can be drawn. (i) Different miRNA signatures can emerge from infection with different M. tuberculosis strains and degrees of virulence. (ii) The time and stage of M. tuberculosis infection play a key role in miRNA expression profile, with striking differences between LTBI and active TB. (iii) Ex vivo and in vitro experiments may provide different and even contradicting results on miRNA expression. (iv) Organ- or tissue-specific TB pathogenesis introduces variations into miRNA functions since pulmonary and extrapulmonary TB do not always provide the same miRNA expression profile. (v) Each specific miRNA may function in different cellular signal pathways and exerts diverse effects in macrophages during mycobacterial infection. (vi) Antimycobacterial therapy modifies the miRNA expression profile. (vii) The miRNA expression profile seems to be sex, age, species, and M. tuberculosis strain specific.
ACKNOWLEDGMENT
This work was supported by the Comisión Nacional de Investigación Científica y Tecnológica (CONICYT), FONDECYT (grant no. 1171570).
REFERENCES
- 1.World Health Organization. 2019. Global tuberculosis report 2019. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 2.Narasimhan P, Wood J, Macintyre CR, Mathai D. 2013. Risk factors for tuberculosis. Pulm Med 2013:828939. doi: 10.1155/2013/828939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bhatt K, Salgame P. 2007. Host innate immune response to Mycobacterium tuberculosis. J Clin Immunol 27:347–362. doi: 10.1007/s10875-007-9084-0. [DOI] [PubMed] [Google Scholar]
- 4.Liu PT, Modlin RL. 2008. Human macrophage host defense against Mycobacterium tuberculosis. Curr Opin Immunol 20:371–376. doi: 10.1016/j.coi.2008.05.014. [DOI] [PubMed] [Google Scholar]
- 5.Pieters J. 2008. Mycobacterium tuberculosis and the macrophage: maintaining a balance. Cell Host Microbe 3:399–407. doi: 10.1016/j.chom.2008.05.006. [DOI] [PubMed] [Google Scholar]
- 6.Frahm M, Goswami ND, Owzar K, Hecker E, Mosher A, Cadogan E, Nahid P, Ferrari G, Stout JE. 2011. Discriminating between latent and active tuberculosis with multiple biomarker responses. Tuberculosis (Edinb) 91:250–256. doi: 10.1016/j.tube.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bartel DP. 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297. doi: 10.1016/S0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
- 8.Xu P, Vernooy SY, Guo M, Hay BA. 2003. The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol 13:790–795. doi: 10.1016/s0960-9822(03)00250-1. [DOI] [PubMed] [Google Scholar]
- 9.Hatfield SD, Shcherbata HR, Fischer KA, Nakahara K, Carthew RW, Ruohola-Baker H. 2005. Stem cell division is regulated by the microRNA pathway. Nature 435:974–978. doi: 10.1038/nature03816. [DOI] [PubMed] [Google Scholar]
- 10.Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D, Wilson M, Wang X, Shelton J, Shingara J, Chin L, Brown D, Slack FJ. 2007. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 67:7713–7722. doi: 10.1158/0008-5472.CAN-07-1083. [DOI] [PubMed] [Google Scholar]
- 11.Thatcher EJ, Flynt AS, Li N, Patton JR, Patton JG. 2007. miRNA expression analysis during normal zebrafish development and following inhibition of the Hedgehog and Notch signaling pathways. Dev Dyn 236:2172–2180. doi: 10.1002/dvdy.21211. [DOI] [PubMed] [Google Scholar]
- 12.Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, Golub TR, Pieske B, Pu WT. 2007. Altered microRNA expression in human heart disease. Physiol Genomics 31:367–373. doi: 10.1152/physiolgenomics.00144.2007. [DOI] [PubMed] [Google Scholar]
- 13.Agirre X, Jiménez-Velasco A, San José-Enériz E, Garate L, Bandrés E, Cordeu L, Aparicio O, Saez B, Navarro G, Vilas-Zornoza A, Pérez-Roger I, García-Foncillas J, Torres A, Heiniger A, Calasanz MJ, Fortes P, Román-Gómez J, Prósper F. 2008. Down-regulation of hsa-miR-10a in chronic myeloid leukemia CD34+ cells increases USF2-mediated cell growth. Mol Cancer Res 6:1830–1840. doi: 10.1158/1541-7786.MCR-08-0167. [DOI] [PubMed] [Google Scholar]
- 14.Johnson R, Zuccato C, Belyaev ND, Guest DJ, Cattaneo E, Buckley NJ. 2008. A microRNA-based gene dysregulation pathway in Huntington’s disease. Neurobiol Dis 29:438–445. doi: 10.1016/j.nbd.2007.11.001. [DOI] [PubMed] [Google Scholar]
- 15.Sun R, Fu X, Li Y, Xie Y, Mao Y. 2009. Global gene expression analysis reveals reduced abundance of putative microRNA targets in human prostate tumours. BMC Genomics 10:93. doi: 10.1186/1471-2164-10-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Almeida MI, Reis RM, Calin GA. 2011. MicroRNA history: discovery, recent applications, and next frontiers. Mutat Res 717:1–8. doi: 10.1016/j.mrfmmm.2011.03.009. [DOI] [PubMed] [Google Scholar]
- 17.Skalsky RL, Cullen BR. 2010. Viruses, microRNAs, and host interactions. Annu Rev Microbiol 64:123–141. doi: 10.1146/annurev.micro.112408.134243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Staedel C, Darfeuille F. 2013. MicroRNAs and bacterial infection. Cell Microbiol 15:1496–1507. doi: 10.1111/cmi.12159. [DOI] [PubMed] [Google Scholar]
- 19.Vance V, Vaucheret H. 2001. RNA silencing in plants–defense and counterdefense. Science 292:2277–2280. doi: 10.1126/science.1061334. [DOI] [PubMed] [Google Scholar]
- 20.Watanabe Y, Kishi A, Yachie N, Kanai A, Tomita M. 2007. Computational analysis of microRNA-mediated antiviral defense in humans. FEBS Lett 581:4603–4610. doi: 10.1016/j.febslet.2007.08.049. [DOI] [PubMed] [Google Scholar]
- 21.Gottwein E, Mukherjee N, Sachse C, Frenzel C, Majoros WH, Chi JTA, Braich R, Manoharan M, Soutschek J, Ohler U, Cullen BR. 2007. A viral microRNA functions as an orthologue of cellular miR-155. Nature 450:1096–1099. doi: 10.1038/nature05992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lindsay MA. 2008. microRNAs and the immune response. Trends Immunol 29:343–351. doi: 10.1016/j.it.2008.04.004. [DOI] [PubMed] [Google Scholar]
- 23.Pedersen I, David M. 2008. MicroRNAs in the immune response. Cytokine 43:391–394. doi: 10.1016/j.cyto.2008.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tsitsiou E, Lindsay MA. 2009. microRNAs and the immune response. Curr Opin Pharmacol 9:514–520. doi: 10.1016/j.coph.2009.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li QJ, Chau J, Ebert PJR, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P, Klein LO, Davis MM, Chen CZ. 2007. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129:147–161. doi: 10.1016/j.cell.2007.03.008. [DOI] [PubMed] [Google Scholar]
- 26.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. doi: 10.1126/science.1139253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Thai T, Calado D, Casola S, Ansel K, Xiao C, Xue Y, Murphy A, Frendewey D, Valenzuela D, Kutok J, Schmidt-Supprian M, Rajewsky N, Yancopoulos G, Rao A, Rajewsky K. 2007. Regulation of the germinal center response by microRNA-155. Science 316:604–609. doi: 10.1126/science.1141229. [DOI] [PubMed] [Google Scholar]
- 28.Lu L, Boldin MP, Chaudhry A, Lin L, Konstantin D, Hanada T, Yoshimura A, Baltimore D, Rudensky AY. 2010. Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell 142:914–929. doi: 10.1016/j.cell.2010.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.O'Connell RM, Rao DS, Chaudhuri AA, Baltimore D. 2010. Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol 10:111–122. doi: 10.1038/nri2708. [DOI] [PubMed] [Google Scholar]
- 30.Lee RC, Feinbaum RL, Ambros V. 1993. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75:843–854. doi: 10.1016/0092-8674(93)90529-Y. [DOI] [PubMed] [Google Scholar]
- 31.Wightman B, Ha I, Ruvkun G. 1993. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 75:855–862. doi: 10.1016/0092-8674(93)90530-4. [DOI] [PubMed] [Google Scholar]
- 32.Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G. 2000. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403:901–906. doi: 10.1038/35002607. [DOI] [PubMed] [Google Scholar]
- 33.Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B, Hayward DC, Ball EE, Degnan B, Müller P, Spring J, Srinivasan A, Fishman M, Finnerty J, Corbo J, Levine M, Leahy P, Davidson E, Ruvkun G. 2000. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 408:86–89. doi: 10.1038/35040556. [DOI] [PubMed] [Google Scholar]
- 34.Treiber T, Treiber N, Meister G. 2019. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol 20:5–20. doi: 10.1038/s41580-019-0106-6. [DOI] [PubMed] [Google Scholar]
- 35.Saliminejad K, Khorram Khorshid HR, Soleymani Fard S, Ghaffari SH. 2019. An overview of microRNAs: biology, functions, therapeutics, and analysis methods. J Cell Physiol 234:5451–5465. doi: 10.1002/jcp.27486. [DOI] [PubMed] [Google Scholar]
- 36.O’Brien J, Hayder H, Zayed Y, Peng C. 2018. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol 9:402. doi: 10.3389/fendo.2018.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Michlewski G, Cáceres JF. 2019. Post-transcriptional control of miRNA biogenesis. RNA 25:1–16. doi: 10.1261/rna.068692.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pu M, Chen J, Tao Z, Miao L, Qi X, Wang Y, Ren J. 2019. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci 76:441–451. doi: 10.1007/s00018-018-2940-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhang B, Wang Q, Pan X. 2007. MicroRNAs and their regulatory roles in animals and plants. J Cell Physiol 210:279–289. doi: 10.1002/jcp.20869. [DOI] [PubMed] [Google Scholar]
- 40.Morin RD, Bainbridge M, Fejes A, Hirst M, Krzywinski M, Pugh TJ, McDonald H, Varhol R, Jones SJM, Marra MA. 2008. Profiling the HeLa S3 transcriptome using randomly primed cDNA and massively parallel short-read sequencing. Biotechniques 45:81–94. doi: 10.2144/000112900. [DOI] [PubMed] [Google Scholar]
- 41.Thomson JM, Parker J, Perou CM, Hammond SM. 2004. A custom microarray platform for analysis of microRNA gene expression. Nat Methods 1:47–53. doi: 10.1038/nmeth704. [DOI] [PubMed] [Google Scholar]
- 42.Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, Barbisin M, Xu NL, Mahuvakar VR, Andersen MR, Lao KQ, Livak KJ, Guegler KJ. 2005. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res 33:e179. doi: 10.1093/nar/gni178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lao K, Xu NL, Yeung V, Chen C, Livak KJ, Straus NA. 2006. Multiplexing RT-PCR for the detection of multiple miRNA species in small samples. Biochem Biophys Res Commun 343:85–89. doi: 10.1016/j.bbrc.2006.02.106. [DOI] [PubMed] [Google Scholar]
- 44.Raabe CA, Tang TH, Brosius J, Rozhdestvensky TS. 2014. Biases in small RNA deep sequencing data. Nucleic Acids Res 42:1414–1426. doi: 10.1093/nar/gkt1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Griffiths-Jones S. 2004. The microRNA registry. Nucleic Acids Res 32:D109–D111. doi: 10.1093/nar/gkh023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kozomara A, Griffiths-Jones S. 2014. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res 42:D68–D73. doi: 10.1093/nar/gkt1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kozomara A, Birgaoanu M, Griffiths-Jones S. 2019. miRBase: from microRNA sequences to function. Nucleic Acids Res 47:D155–D162. doi: 10.1093/nar/gky1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, Huang WC, Sun TH, Tu SJ, Lee WH, Chiew MY, Tai CS, Wei TY, Tsai TR, Huang HT, Wang CY, Wu HY, Ho SY, Chen PR, Chuang CH, Hsieh PJ, Wu YS, Chen WL, Li MJ, Wu YC, Huang XY, Ng FL, Buddhakosai W, Huang PC, Lan KC, Huang CY, Weng SL, Cheng YN, Liang C, Hsu WL, Huang HD. 2018. miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res 46:D296–D302. doi: 10.1093/nar/gkx1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Drain PK, Bajema KL, Dowdy D, Dheda K, Naidoo K, Schumacher SG, Ma S, Meermeier E, Lewinsohn DM, Sherman DR. 2018. Incipient and subclinical tuberculosis: a clinical review of early stages and progression of infection. Clin Microbiol Rev 31:e00021-18. doi: 10.1128/CMR.00021-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Abel L, Fellay J, Haas DW, Schurr E, Srikrishna G, Urbanowski M, Chaturvedi N, Srinivasan S, Johnson DH, Bishai WR. 2018. Genetics of human susceptibility to active and latent tuberculosis: present knowledge and future perspectives. Lancet Infect Dis 18:e64–e75. doi: 10.1016/S1473-3099(17)30623-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Guo W, Li JT, Pan X, Wei L, Wu JY. 2010. Candidate Mycobacterium tuberculosis genes targeted by human microRNAs. Protein Cell 1:419–421. doi: 10.1007/s13238-010-0056-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Maudet C, Mano M, Eulalio A. 2014. MicroRNAs in the interaction between host and bacterial pathogens. FEBS Lett 588:4140–4147. doi: 10.1016/j.febslet.2014.08.002. [DOI] [PubMed] [Google Scholar]
- 53.Zheng L, Leung E, Lee N, Lui G, To KF, Chan RCY, Ip M. 2015. Differential microRNA expression in human macrophages with Mycobacterium tuberculosis infection of Beijing/W and non-Beijing/W strain types. PLoS One 10:e0126018. doi: 10.1371/journal.pone.0126018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ren N, Gao G, Sun Y, Zhang L, Wang H, Hua W, Wan K, Li X. 2015. MicroRNA signatures from multidrug-resistant Mycobacterium tuberculosis. Mol Med Rep 12:6561–6567. doi: 10.3892/mmr.2015.4262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang C, Yang S, Liu CM, Jiang TT, Chen ZL, Tu HH, Mao LG, Li ZJ, Li JC. 2018. Screening and identification of four serum miRNAs as novel potential biomarkers for cured pulmonary tuberculosis. Tuberculosis (Edinb) 108:26–34. doi: 10.1016/j.tube.2017.08.010. [DOI] [PubMed] [Google Scholar]
- 56.Wagh V, Urhekar A, Modi D. 2017. Levels of microRNA miR-16 and miR-155 are altered in serum of patients with tuberculosis and associate with responses to therapy. Tuberculosis (Edinb) 102:24–30. doi: 10.1016/j.tube.2016.10.007. [DOI] [PubMed] [Google Scholar]
- 57.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. doi: 10.1128/JCM.05459-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Xu Z, Zhou A, Ni J, Zhang Q, Wang Y, Lu J, Wu W, Karakousis PC, Lu S, Yao Y. 2015. Differential expression of miRNAs and their relation to active tuberculosis. Tuberculosis (Edinb) 95:395–403. doi: 10.1016/j.tube.2015.02.043. [DOI] [PubMed] [Google Scholar]
- 59.Qi Y, Cui L, Ge Y, Shi Z, Zhao K, Guo X, Yang D, Yu H, Cui L, Shan Y, Zhou M, Wang H, Lu Z. 2012. Altered serum microRNAs as biomarkers for the early diagnosis of pulmonary tuberculosis infection. BMC Infect Dis 12:384. doi: 10.1186/1471-2334-12-384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhang H, Sun Z, Wei W, Liu Z, Fleming J, Zhang S, Lin N, Wang M, Chen M, Xu Y, Zhou J, Li C, Bi L, Zhou G. 2014. Identification of serum microRNA biomarkers for tuberculosis using RNA-seq. PLoS One 9:e88909. doi: 10.1371/journal.pone.0088909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yi Z, Fu Y, Ji R, Li R, Guan Z. 2012. Altered microRNA signatures in sputum of patients with active pulmonary tuberculosis. PLoS One 7:e43184. doi: 10.1371/journal.pone.0043184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wu LSH, Lee SW, Huang KY, Lee TY, Hsu PWC, Weng J. 2014. Systematic expression profiling analysis identifies specific microRNA-gene interactions that may differentiate between active and latent tuberculosis infection. Biomed Res Int 2014:895179. doi: 10.1155/2014/895179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang X, Guo J, Fan S, Li Y, Wei L, Yang X, Jiang T, Chen Z, Wang C, Liu J, Ping Z, Xu D, Wang J, Li Z, Qiu Y, Li JC. 2013. Screening and identification of six serum microRNAs as novel potential combination biomarkers for pulmonary tuberculosis diagnosis. PLoS One 8:e81076. doi: 10.1371/journal.pone.0081076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Corral-Fernández NE, Cortes-García JD, Bruno RS, Romano-Moreno S, Medellín-Garibay SE, Magaña-Aquino M, Salazar-González RA, González-Amaro R, Portales-Pérez DP. 2017. Analysis of transcription factors, microRNAs and cytokines involved in T lymphocyte differentiation in patients with tuberculosis after directly observed treatment short-course. Tuberculosis (Edinb) 105:1–8. doi: 10.1016/j.tube.2017.03.007. [DOI] [PubMed] [Google Scholar]
- 65.Sato F, Tsuchiya S, Terasawa K, Tsujimoto G. 2009. Intra-platform repeatability and inter-platform comparability of microRNA microarray technology. PLoS One 4:e5540. doi: 10.1371/journal.pone.0005540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Schwarzenbach H, da Silva AM, Calin G, Pantel K. 2015. Data normalization strategies for microRNA quantification. Clin Chem 61:1333–1342. doi: 10.1373/clinchem.2015.239459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Faraldi M, Gomarasca M, Sansoni V, Perego S, Banfi G, Lombardi G. 2019. Normalization strategies differently affect circulating miRNA profile associated with the training status. Sci Rep 9:1584. doi: 10.1038/s41598-019-38505-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Benz F, Roderburg C, Cardenas DV, Vucur M, Gautheron J, Koch A, Zimmermann H, Janssen J, Nieuwenhuijsen L, Luedde M, Frey N, Tacke F, Trautwein C, Luedde T. 2013. U6 is unsuitable for normalization of serum miRNA levels in patients with sepsis or liver fibrosis. Exp Mol Med 45:e42. doi: 10.1038/emm.2013.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gee HE, Buffa FM, Camps C, Ramachandran A, Leek R, Taylor M, Patil M, Sheldon H, Betts G, Homer J, West C, Ragoussis J, Harris AL. 2011. The small-nucleolar RNAs commonly used for microRNA normalisation correlate with tumour pathology and prognosis. Br J Cancer 104:1168–1177. doi: 10.1038/sj.bjc.6606076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F. 2002. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3:RESEARCH0034. doi: 10.1186/gb-2002-3-7-research0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shen Y, Tian F, Chen Z, Li R, Ge Q, Lu Z. 2015. Amplification-based method for microRNA detection. Biosens Bioelectron 71:322–331. doi: 10.1016/j.bios.2015.04.057. [DOI] [PubMed] [Google Scholar]
- 72.Mestdagh P, Van Vlierberghe P, De Weer A, Muth D, Westermann F, Speleman F, Vandesompele J. 2009. A novel and universal method for microRNA RT-qPCR data normalization. Genome Biol 10:R64. doi: 10.1186/gb-2009-10-6-r64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chugh P, Dittmer DP. 2012. Potential pitfalls in microRNA profiling. Wiley Interdiscip Rev RNA 3:601–616. doi: 10.1002/wrna.1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lv Y, Guo S, Li XG, Chi JY, Qu YQ, Zhong HL. 2016. Sputum and serum microRNA-144 levels in patients with tuberculosis before and after treatment. Int J Infect Dis 43:68–73. doi: 10.1016/j.ijid.2015.12.014. [DOI] [PubMed] [Google Scholar]
- 75.Krakauer T. 2019. Inflammasomes, autophagy, and cell death: the trinity of innate host defense against intracellular bacteria. Mediators Inflamm 2019:2471215. doi: 10.1155/2019/2471215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Siqueira MDS, Ribeiro RM, Travassos LH. 2018. Autophagy and its interaction with intracellular bacterial pathogens. Front Immunol 9:935. doi: 10.3389/fimmu.2018.00935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.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. doi: 10.1371/journal.pone.0179772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Liu Y, Wang X, Jiang J, Cao Z, Yang B, Cheng X. 2011. Modulation of T cell cytokine production by miR-144* with elevated expression in patients with pulmonary tuberculosis. Mol Immunol 48:1084–1090. doi: 10.1016/j.molimm.2011.02.001. [DOI] [PubMed] [Google Scholar]
- 79.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. doi: 10.1080/15548627.2016.1241922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Spinelli SV, Diaz A, D'Attilio L, Marchesini MM, Bogue C, Bay ML, Bottasso OA. 2013. Altered microRNA expression levels in mononuclear cells of patients with pulmonary and pleural tuberculosis and their relation with components of the immune response. Mol Immunol 53:265–269. doi: 10.1016/j.molimm.2012.08.008. [DOI] [PubMed] [Google Scholar]
- 81.Liu Z, Zhou G, Deng X, Yu Q, Hu Y, Sun H, Wang Z, Chen H, Jia C, Wang D. 2014. Analysis of miRNA expression profiling in human macrophages responding to Mycobacterium infection: induction of the immune regulator miR-146a. J Infect 68:553–561. doi: 10.1016/j.jinf.2013.12.017. [DOI] [PubMed] [Google Scholar]
- 82.Li M, Wang J, Fang Y, Gong S, Li M, Wu M, Lai X, Zeng G, Wang Y, Yang K, Huang X. 2016. MicroRNA-146a promotes mycobacterial survival in macrophages through suppressing nitric oxide production. Sci Rep 6:24555. doi: 10.1038/srep24555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Malardo T, Gardinassi LG, Moreira BP, Padilha É, Lorenzi JCC, Soares LS, Gembre AF, Fontoura IC, de Almeida LP, de Miranda Santos IKF, Silva CL, Coelho-Castelo A. 2016. MicroRNA expression signatures in lungs of mice infected with Mycobacterium tuberculosis. Tuberculosis (Edinb) 101:151–159. doi: 10.1016/j.tube.2016.09.003. [DOI] [PubMed] [Google Scholar]
- 84.Furci L, Schena E, Miotto P, Cirillo DM. 2013. Alteration of human macrophages microRNA expression profile upon infection with Mycobacterium tuberculosis. Int J Mycobacteriol 2:128–134. doi: 10.1016/j.ijmyco.2013.04.006. [DOI] [PubMed] [Google Scholar]
- 85.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. doi: 10.1371/journal.ppat.1006790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhou M, Yu G, Yang X, Zhu C, Zhang Z, Zhan X. 2016. Circulating microRNAs as biomarkers for the early diagnosis of childhood tuberculosis infection. Mol Med Rep 13:4620–4626. doi: 10.3892/mmr.2016.5097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Barry SE, Ellis M, Yang YR, Guan G, Wang X, Britton WJ, Saunders BM. 2018. Identification of a plasma microRNA profile in untreated pulmonary tuberculosis patients that is modulated by anti-mycobacterial therapy. J Infect 77:341–348. doi: 10.1016/j.jinf.2018.03.006. [DOI] [PubMed] [Google Scholar]
- 88.Li S, Yue Y, Xu W, Xiong S. 2013. MicroRNA-146a represses mycobacteria-induced inflammatory response and facilitates bacterial replication via targeting IRAK-1 and TRAF-6. PLoS One 8:e81438. doi: 10.1371/journal.pone.0081438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Reference deleted.
- 90.Li D, Wang T, Song X, Qucuo M, Yang B, Zhang J, Wang J, Ying B, Tao C, Wang L. 2011. Genetic study of two single nucleotide polymorphisms within corresponding microRNAs and susceptibility to tuberculosis in a Chinese Tibetan and Han population. Hum Immunol 72:598–602. doi: 10.1016/j.humimm.2011.03.004. [DOI] [PubMed] [Google Scholar]
- 91.Zhang X, Li Y, Li X, Zhang W, Pan Z, Wu F, Wang C, Chen Z, Jiang T, Xu D, Ping Z, Liu J, Liu C, Li Z, Li JC. 2015. Association of the miR-146a, miR-149, miR-196a2 and miR-499 polymorphisms with susceptibility to pulmonary tuberculosis in the Chinese Uygur, Kazak and Southern Han populations. BMC Infect Dis 15:41. doi: 10.1186/s12879-015-0771-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zhang C, Xi X, Wang Q, Jiao J, Zhang L, Zhao H, Lai Z. 2015. The association between serum miR-155 and natural killer cells from tuberculosis patients. Int J Clin Exp Med 8:9168–9172. [PMC free article] [PubMed] [Google Scholar]
- 93.Rajaram MVS, 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. doi: 10.1073/pnas.1112660108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Das K, Saikolappan S, Dhandayuthapani S. 2013. Differential expression of miRNAs by macrophages infected with virulent and avirulent Mycobacterium tuberculosis. Tuberculosis 93:S47–S50. doi: 10.1016/S1472-9792(13)70010-6. [DOI] [PubMed] [Google Scholar]
- 95.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. doi: 10.1111/j.1462-5822.2012.01827.x. [DOI] [PubMed] [Google Scholar]
- 96.Wang J, Yang K, Zhou L, Minhao W, 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. doi: 10.1371/journal.ppat.1003697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Elizabeth MC, Hernández de la Cruz ON, Mauricio CA. 2016. Infection of J774A.1 with different Mycobacterium species induces differential immune and miRNA-related responses. Microbiol Immunol 60:356–363. doi: 10.1111/1348-0421.12380. [DOI] [PubMed] [Google Scholar]
- 98.Wu J, Lu C, Diao N, Zhang S, Wang S, Wang F, Gao Y, Chen J, Shao L, Lu J, Zhang X, Weng X, Wang H, Zhang W, Huang Y. 2012. Analysis of microRNA expression profiling identifies miR-155 and miR-155* as potential diagnostic markers for active tuberculosis: a preliminary study. Hum Immunol 73:31–37. doi: 10.1016/j.humimm.2011.10.003. [DOI] [PubMed] [Google Scholar]
- 99.Huang J, Jiao J, Xu W, Zhao H. 2015. miR-155 is upregulated in patients with active tuberculosis and inhibits apoptosis of monocytes by targeting FOXO3. Mol Med Rep 12:7102–7108. doi: 10.3892/mmr.2015.4250. [DOI] [PubMed] [Google Scholar]
- 100.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. doi: 10.1159/000373950. [DOI] [PubMed] [Google Scholar]
- 101.Iwai H, Funatogawa K, Matsumura K, Kato-Miyazawa M, Kirikae F, Kiga K, Sasakawa C, Miyoshi-Akiyama T, Kirikae T. 2015. MicroRNA-155 knockout mice are susceptible to Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 95:246–250. doi: 10.1016/j.tube.2015.03.006. [DOI] [PubMed] [Google Scholar]
- 102.Bonilla-Muro MG, Hernández de la Cruz ON, Gonzalez-Barrios JA, Alcaráz-Estrada SL, Castañón-Arreola M. 13 September 2019. EsxA mainly contributes to the miR-155 overexpression in human monocyte-derived macrophages and potentially affect the immune mechanism of macrophages through miRNA dysregulation. J Microbiol Immunol Infect doi: 10.1016/j.jmii.2019.07.007. [DOI] [PubMed] [Google Scholar]
- 103.Yao J, Du X, Chen S, Shao Y, Deng K, Jiang M, Liu J, Shen Z, Chen X, Feng G. 2018. Rv2346c enhances mycobacterial survival within macrophages by inhibiting TNF-α and IL-6 production via the p38/miRNA/NF-κB pathway. Emerg Microbes Infect 7:1–16. doi: 10.1038/s41426-018-0162-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhou Z-Q, Wang Z-K, Zhang L, Ren Y-Q, Ma Z-W, Zhao N, Sun F-Y. 2017. Role of ESAT-6 in renal injury by regulating microRNA-155 expression via TLR4/MyD88 signaling pathway in mice with Mycobacterium tuberculosis infection. Biosci Rep 37:BSR20170021. doi: 10.1042/BSR20170021. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 105.Zheng M, Zhou N, Luo C. 2016. MiRNA-155 and miRNA-132 as potential diagnostic biomarkers for pulmonary tuberculosis: a preliminary study. Microb Pathog 100:78–83. doi: 10.1016/j.micpath.2016.09.005. [DOI] [PubMed] [Google Scholar]
- 106.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. doi: 10.1371/journal.pone.0061609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zhao Z, Hao J, Li X, Chen Y, Qi X. 2019. MiR-21-5p regulates mycobacterial survival and inflammatory responses by targeting Bcl-2 and TLR4 in Mycobacterium tuberculosis-infected macrophages. FEBS Lett 593:1326–1335. doi: 10.1002/1873-3468.13438. [DOI] [PubMed] [Google Scholar]
- 108.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. doi: 10.1371/journal.pone.0100949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Afum-Adjei Awuah A, Ueberberg B, Owusu-Dabo E, Frempong M, Jacobsen M. 2014. Dynamics of T-cell IFN-γ and miR-29a expression during active pulmonary tuberculosis. Int Immunol 26:579–582. doi: 10.1093/intimm/dxu068. [DOI] [PubMed] [Google Scholar]
- 110.Pan D, Pan M, Xu YM. 2017. Mir-29a expressions in peripheral blood mononuclear cell and cerebrospinal fluid: diagnostic value in patients with pediatric tuberculous meningitis. Brain Res Bull 130:231–235. doi: 10.1016/j.brainresbull.2017.01.013. [DOI] [PubMed] [Google Scholar]
- 111.Yi Z, Gao K, Li R, Fu Y. 2018. Dysregulated circRNAs in plasma from active tuberculosis patients. J Cell Mol Med 22:4076–4084. doi: 10.1111/jcmm.13684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.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. doi: 10.1371/journal.pone.0020258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.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-gamma. Nat Immunol 12:861–869. doi: 10.1038/ni.2073. [DOI] [PubMed] [Google Scholar]
- 114.Ebert MS, Neilson JR, Sharp PA. 2007. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods 4:721–726. doi: 10.1038/nmeth1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kluiver J, Gibcus JH, Hettinga C, Adema A, Richter MKS, Halsema N, Slezak-Prochazka I, Ding Y, Kroesen BJ, van den Berg A. 2012. Rapid generation of microRNA sponges for microRNA inhibition. PLoS One 7:e29275. doi: 10.1371/journal.pone.0029275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Siddle KJ, Deschamps M, Tailleux L, Nédélec Y, Pothlichet J, Lugo-Villarino G, Libri V, Gicquel B, Neyrolles O, Laval G, Patin E, Barreiro LB, Quintana-Murci L. 2014. A genomic portrait of the genetic architecture and regulatory impact of microRNA expression in response to infection. Genome Res 24:850–859. doi: 10.1101/gr.161471.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Fu Y, Yi Z, Li J, Li R. 2014. Deregulated microRNAs in CD4+ T cells from individuals with latent tuberculosis versus active tuberculosis. J Cell Mol Med 18:503–513. doi: 10.1111/jcmm.12205. [DOI] [PMC free article] [PubMed] [Google Scholar]