Open access peer-reviewed chapter

Perspectives on RNA Interference in Immunopharmacology and Immunotherapy

Written By

Zhaohua Hou, Qiuju Han, Cai Zhang and Jian Zhang

Submitted: 02 May 2015 Reviewed: 21 September 2015 Published: 06 April 2016

DOI: 10.5772/61575

From the Edited Volume

RNA Interference

Edited by Ibrokhim Y. Abdurakhmonov

Chapter metrics overview

1,848 Chapter Downloads

View Full Metrics

Abstract

RNA interference (RNAi), mediated by short interfering RNA (siRNA), vector-derived short hairpin RNA (shRNA) and microRNA (miRNA), brings about revolutionary features to basic biomedical research and clinical application. New drugs based on RNAi have been developed for therapeutic applications. The family of RNAi molecules are efficient agents to modulate mammalian immune system, and many studies reported that these molecules could manipulate immune defence, surveillance and homeostasis. Both perfect match of siRNA/shRNA and non-perfect match of miRNA could be beneficial for designing RNAi-based drugs for treatment of tumour and viral infection. This chapter provides a view to control or utilize the immune regulation of various small RNAs that should help researchers to understand the successful clinical application of RNAi.

Keywords

  • RNA interference
  • siRNA
  • miRNA
  • immunopharmacology
  • immunotherapy

1. Introduction

RNA interference (RNAi) is a conserved mechanism against exogenous nucleic acid and transposon transcripts in plants and lower animals. No matter of transfected siRNA, vector-delivered shRNA or pre-miRNA (transcribed mainly by Pol II) , Dicer (DCLs) and Agronaute (AGO) family proteins efficiently process small RNAs into short double-stranded RNA(dsRNA). Further, dsRNAs assemble into the RNAi-induced silencing (protein) complex (RISC) to guide and cleave target mRNA, promote mRNA degradation or inhibit mRNA translation. The great potential of RNAi is to specifically repress the expression of disease-causing genes while avoiding undesirable effects.

It is well accepted that siRNA can be recognized by endosomal pathways, Toll-like receptor 3 (TLR3), TLR7, TLR8 and cytoplasmic pathways, retinoic acid-inducible gene I (RIG-I), melanoma differentiation-associated antigen 5 (MDA-5) and RNA-activated protein kinase (PKR), resulting in immune activation [15]. For example, it has been demonstrated that siRNA can cause activation of at least three key transcription factors, including NF-κB, interferon regulatory factor 3 (IRF-3) and IRF-7, and stimulate interferon (IFN) secretion. This activates T cells and dendritic cells (DCs) in the spleen in a TLR7-dependent manner [2,6]. Furthermore, 5′-triphosphate siRNA was demonstrated to activate RIG-I signal pathway, and then natural killer (NK) cells and DCs were activated [7,8]. In most circumstances, immune system stimulation is regarded as an unwanted side effect; therefore, siRNA-induced immune response should be controlled by using proper delivery system or chemical modification, although immune stimulation has been proved to be essential in cancer treatment and viral infection.

miRNAs are critical in regulating the development, differentiation, function and destiny of immune cells, including DCs, granulocytes, monocytes/macrophages, NK and natural killer T (NKT) cells and B and T lymphocytes. miRNAs influence both innate and adaptive immune defence and individual miRNAs may contribute their implications to various immune-mediated diseases. Furthermore, pattern recognition receptors (PRRs), kinases, adaptors, inflammatory factors and IFN could all be targets of miRNAs. Extra effort has been made to develop miRNA-based oligos or vectors for anti-infection purpose by manipulating corresponding immune genes.

In addition to silencing of targeted genes in a sequence-specific manner, components of RNAi technology often induce immune response. Several strategies were reported to design RNAi molecules with gene silencing and immune regulatory properties. Bifunctional molecules rely on the activation of PRRs such as TLR7/8, TLR9 or RIG-I, or just rely on down-regulation of target gene. This chapter summarizes RNAi-involved immune responses in the past 10 years and discusses the anticipated therapeutic application.

Advertisement

2. Chemically synthesized siRNA and vector-derived shRNA

2.1. RNAi drugs based on targeting specific immune genes

Immune disorders, both autoimmune diseases and immune defective or deficiency, are always caused by high-level overexpression of certain immune genes. A variety of immune inhibitory genes can serve as targets for RNAi-mediated gene silencing. Targeting specific immune suppressor could re-balance immune network and subsets.

Elevated activity of signal transducer and activator of transcription 3 (STAT3) has been found in several kinds of human tumours. Use of RNAi to knockdown STAT3 expression and inhibit its activation would reduce the tumour cell growth such as pancreatic cancer, colorectal cancer, melanoma and hepatocarcinoma cells. STAT 3 knockdown could induce bystanders immune response in vitro and in vivo, where CD4+, CD8+ and NKT cells were activated as well as the secretion of interferon-γ (IFN-γ), interleukin-12 (IL-12) and tumour necrosis factor alpha (TNF-α) was increased significantly [911]. siRNA-STAT3, synthetically linked to CpG (agonist of TLR9), was demonstrated to silence immune suppressor STAT3 gene in TLR9+ myeloid cells and B cells. This strategy of therapy leads to activation of various populations of immune cells, including DCs and macrophages, that ultimately induce potent anti-tumour immune responses [10,12]. Hossain DM et al. recently reported that CpG-siRNA-STAT3 conjugates could efficiently silence the target expression, and abrogate inhibition of CD8+ T cells in patients who received myeloid-derived suppressor cells (MDSCs) [13]. Researchers proved that immune-stimulation-inducing CpG(A)-STAT3-siRNA was non-toxic for normal human leukocytes [14]. In another experiment, Luo Z et al. [15] generated a nano-vaccine loaded with poly I:C (a TLR3 agonist) and STAT3 siRNA. Researchers found this kind of siRNA could promote the maturation of DC and reverse immunosuppression in the tumour micro-environment; the function of inhibitory cells in tumour-draining lymph nodes were inhibited; thus, anti-tumour immune responses were potently induced; and the survival were prolonged [15]. Therefore, STAT3 siRNAs are expected to be a promising immunomodulatory drugs to improve the treatment efficacy of cancer vaccines by abrogating tumour immunosuppression.

Suppressor of cytokine signalling 1 (SOCS1) is a negative regulator of antigen-presenting cell (APC)-based immune response. Silencing of SOCS1 gene expression by RNAi is essential for DCs to enhance Ag-specific anti-tumour immunity [16]. SOCS1-silenced bone marrow dendritic cells (BMDCs) were more potent in suppressing tumour growth [17]. When SOCS1 was silenced, maturation of DCs (i.e. expressions of CD80, CD40, CD86 and major histocompatibility complex II [MHC II]) was significantly accelerated. As a result, SOCS1 inhibition up-regulated the expression of IFN-γ and IL-12, and decreased IL-4 secretions, which induced Th1 cell differentiation and thereby affected the development of Th2 cell. The combined nanoparticle (NP) delivery, which can render both tumour antigen and siRNA-SOCS1 to BMDCs, simultaneously could enhance immunotherapeutic effects in BMDC-based cancer therapy [16,18]. DC-targeted delivery of SOCS1 siRNA has been shown to enhance antifungal immunity in response to Candida albicans in vitro and HIV-specific cytotoxic T cell in mice [16,19]. This evidence suggests the use of SOCS1–siRNA, as a potent adjuvant to improve immune response.

A20 is usually regarded as an attractive target for siRNA-mediated gene knockdown in DCs because it is a negative feedback regulator of multiple pro-inflammatory signal transduction. Several reports demonstrated that RNAi-mediated A20 silencing in DCs enhanced expression of co-stimulatory molecules (CD80, CD86, CD40 and MHC class II) and pro-inflammatory cytokines (IL-6 and TNF-α). Tumour-infiltrating cytotoxic T lymphocytes (CTLs), T helper cells that produced IL-6 and TNF-α were also activated by siA20-DC. A20 silencing in DCs can enhance the immune response against self-tumour-associated antigens [20,21]. Furthermore, A20-silenced DCs were proved to overcome CD4+CD25+regulatory T (Treg) cell suppression [21,22]. A20-silenced DCs could skew naive CD4+ T cells towards Th1 cell, but not Treg, Th2 or Th17 cells. Because a high amount of IL-10 was produced in A20-silenced DCs, simultaneous down-regulation of IL-10 and A20 resulted in enhanced T cell stimulatory capacity in DCs. A20 down-regulation resulted in enhanced CTLs immune response by the NF-κB and AP-1 pathways [20,23]. RNAi of A20 has enabled DCs to gain a potent ability to activate CTLs and Th cells, and inhibit Treg, providing a novel strategy to promote a tumour immune response.

Programmed death ligand (PD-L) is another exciting target on the surface of antigen-presenting cells (APCs); PD-L/PD-1 interactions were related to functional impairment and exhaustion of tumour antigen-specific CD8+ T cells. Although PD-L antibody exerts a potent anti-tumour effect, previous reports [24] have demonstrated that PD-L1-siRNA-PEI were preferentially and avidly engulfed by tumour-associated CD11c+PD-L1+ tolerogenic DCs at ovarian cancer locations. This kind of nanoparticle uptake stimulated multiple TLRs signalling, mainly via myeloid differentiation factor 88 (MyD88). Then, regulatory DCs activated into potent stimulators of CTLs that led to significant anti-tumour immunity in mouse models of ovarian cancer. Most importantly, PD-L knockdown DCs showed superior potential to expand minor histocompatibility antigen (MiHA)-specific CD8+ effector and memory T cells from leukaemia patients early after donor lymphocyte infusion and later during relapse. Combined PD-L1 and PD-L2 knockdown resulted in improved proliferation of CD4+ T cells and enhanced cytokine production [25,26]. In addition, another report demonstrated the improved effector functions of tumour-specific CD4+ and CD8+ human T cells by siRNA-mediated silencing of PD-1 ligands, PD-L1 or PD-L2 [27]. These results suggest that siRNA-mediated knockdown of PD-L is a fascinating strategy to inhibit a negative regulatory mechanism of tumour-specific T cells.

siRNA-CD40, delivered by a novel delivery system with a poly-dA extension at the 5'-end of the siRNA sense strand that was stably incorporated into 1,3-β-glucan, was captured and incorporated into DCs through its receptor, Dectin [28]. This strategy could induce antigen-specific Tregs, resulting in the permanent acceptance of mouse cardiac allografts. CD40 knockdown significantly suppressed Th1-type cytokines and induced Th2-type cytokines in rats with myocarditis. Knockdown of CD40 in experimental autoimmune myocarditis (EAM) rats promoted Foxp3 gene expression and increased Treg cells [29].

In addition, when silencing of CD40 or CD80/CD86, DCs exhibited suppressed allostimulatory activity with impaired APC function. In the well-established collagen-induced arthritis (CIA) model, multigene-silenced DCs were capable of delaying onset of joint pathology. Therapeutic effects of gene-silenced DCs were mediated by the inhibition of collagen II-specific Ab production and suppression of T cell recall responses. Also, multigene-silenced DCs inhibited Th1 and Th17 response, demonstrating IFN-γ and IL-2 inhibition [30]. Thus, inhibition of specific co-stimulatory molecules of DCs reveals a promising approach of suppressing immune responses in autoimmunity. These findings highlight the potential of immunomodulation of siRNA-CD40, and have important implications for developing RNAi-based clinical therapy in the transplantation field.

It is well documented that tumours could secrete immunosuppressive molecules, including the cytokines transforming growth factor β (TGF-β) and IL-10. This creates an immunosuppressive environment, which inhibits anti-tumour immunity. The suppression of Treg cell, induced by targeting TGF-β1 using siRNA, can enhance the efficacy of a DC vaccine against a poorly immunogenic tumour in mice [31]. Nanoparticle-delivered TGF-β siRNA enhances vaccination against advanced melanoma, and the tumour micro-environment was modified with increased levels of tumour-infiltrating CD8+ T cells and decreased level of regulatory T cells [32]. siRNA targeting IL-10 receptor α (siIL-10RA) initiated the significant antigen-specific CD8+ T cell immune responses. Concordantly, the combination of knockdown of IL-10RA and TGF-βR in DCs showed significant up-regulation of MHC I, enhancing co-stimulatory molecules CD40, CD80, CD86 and chemokine CCR7 after lipopolysaccharide (LPS) stimulation. It induced the strongest anti-tumour effects in the TC-1 P0 (a cervical cancer model expressing the human papillomavirus [HPV]-16 E7 antigen) tumour model, and even in the immune-resistant TC-1 (P3) ones [33]. These data revealed that siRNA co-targeting immunosuppressive molecules enhance the potency of DC-based immunotherapeutics.

High-mobility group box 1 (HMGB1) is highly expressed in tumour cells and increased levels of HMGB1 in tumour cells are usually associated with a greater tumour angiogenesis, growth, invasion and metastasis. Knockdown of tumour cell-derived HMGB1 by shRNA did not affect tumour cell growth, while naturally acquired long-lasting tumour-specific IFN-γ- or TNF-α-producing CD8+ T cell responses were induced, and ability to induce Treg was attenuated. This led to naturally acquired CD8 T cell-dependent anti-tumour immunity [34].

Foxp3, a master gene that controls the development and function of Treg cells, contributes to pathogenesis of several different tumours. Owing to the intracellular localization of Foxp3, RNAi technology was employed to knockdown its activation to suppress Treg activity in vivo. Tsai et al. [35] performed a study targeting silencing Foxp3 gene expression by shRNAs-mediated RNAi using a lentivirus vector in a murine model of leukaemia. The lentiviral vector was used to overcome poor transfection efficiency. Lentiviral-mediated Foxp3 RNAi showed suppressive effects on tumour growth and prolonged the survival of tumour-transplanted mice. Furthermore, Foxp3 knockdown mediated by siRNA increased the ratio of Th1/Th2 in chronic hepatitis B patients; transcription factors T-bet and GATA-3 may be partly involved in this progress [36]. This strategy provides a novel view about how to decrease the number of Treg cells and weaken its function.

Selective knockdown of CCL22 and CCL17 expression in monocyte-derived DC (MoDC) by siRNA decreased the ratios of CD4+ to CD8+ as well as lowered the frequency of Tregs recruited by MoDC. Furthermore, intratumoural injection of MoDC, which was transfected with siCCL22 and siCCL17, significantly reduced the number of Tregs while inducing CD8+ T cells infiltration in thymic nude mice with human tumour xenografts [37]. Using siRNA to selectively silence chemokines may lead to a new strategy for DC vaccine development to improve cancer biotherapy.

High expression of indoleamine 2, 3-dioxygenase (IDO) in DCs leads to the suppression of T cell responses. Gene silencing by siRNA or shRNA of IDO in DC would up-regulate IL-12 and IFN-γ and inhibit apoptosis in CD8 and CD4 T cells as well as Treg cells; IL-10 expression was significantly down-regulated, thus finally restraining tumour growth. DC-based vaccine with IDO silence was demonstrated to augment and enhance the anti-tumour response against breast cancer, melanoma, bladder tumour and liver cancer [3840]. A novel APC-targeted siRNA delivery system using mannosed liposomes (Man-lipo) with encapsulated siRNA-IDO (Man-lipo-siIDO) was demonstrated to preferentially silence IDO in APCs and efficiently enhance anti-tumour immune response [4143].

It was reported that natural killer group 2, member D (NKG2D) activation was involved in NK cell and CD8+ cell-mediated liver inflammation, and blockade of NKG2D by silencing of multiple NKG2D ligands on hepatocytes was considered efficient in liver disease intervention. Huang et al. [44] constructed a plasmid containing the three shRNA sequences (shRae1-shMult1-shH60). After hydrodynamic injection into mice, they found that the expression of all three NKG2D ligands on hepatocytes was down-regulated, and fulminant hepatitis mediated through NKG2D in NK cell was attenuated. Furthermore, simultaneous knockdown of multiple human NKG2D ligands (MHC class I polypeptide-related sequence B/A(MICA/B), UL16-binding protein 2 [ULBP2] and ULBP3) also significantly attenuated NK cell cytolysis. Simultaneous knockdown of multiple ligands of NKG2D is a potential therapeutic approach to treat liver diseases induced by NKG2D-expressing NK cells and CD8+ cells. Furthermore, inhibition of human leukocyte antigen-G (HLA-G) by siRNA boosted NK cell lytic function [45,46].

Among several molecules involved in immune response, the choice of targets should be carefully reviewed and validated comprehensively according to the emerging knowledge about their function.

2.2. Advantage of non-target immune effect of siRNA/shRNA drugs

siRNA/shRNA have the potential to recruit immune receptors specialized in RNA sensor, such as TLR3, TLR7/8 [2,3]. 5′-triphosphate siRNA (3p-siRNA) was demonstrated to be detected by RNA sensors RIG-I. These immunostimulatory siRNA or shRNA can non-specifically induce innate immune response, the so-called ‘off-target’ effects that have considerable implications for clinical application to cure cancer and infection disease.

IFN response is a common side effect of siRNAs and siRNAs with GU-rich sequences, which are very potent in inducing IFN-α response. A newly published report demonstrated that siRNA could induce IFN-α responses, and then induced the analgesic effects in the spinal cord. This off-target analgesia is dose- and sequence-dependent while non-GU-rich sequences also produced off-target analgesia at high doses, where pain relief by a designed siRNA may not be attributable to target gene knockdown but IFN response [47].

Early in 2004, Karikó et al. [48] demonstrated that siRNAs and shRNAs induce immune activation by signalling through TLR3 and activate sequence-independent inhibition of gene expression. Kleinman et al. [4] showed that non-targeted (against non-mammalian genes) and targeted (against vascular endothelial growth factor [VEGF] or VEGFR1) siRNAs suppressed choroidal neovascularization (CNV) via cell-surface TLR3 and its adaptor TIR-domain-containing adaptor-inducing interferon-β (TRIF), leading to the induction of IFN-α and IL-12. The effect of non-targeted siRNA to suppress dermal neovascularization in mice was as effective as vascular endothelial growth factor (VEGF) siRNA. This finding showed that two investigational siRNAs in clinical trials owe their anti-angiogenic effect in mice, which was not due to target knockdown but due to TLR3 activation. The efficiency of RNAi by siRNA is believed to be comparable with anti-VEGF antibodies. Kleinman’s group then concluded that a 21-nucleotide (nt) non-targeted siRNA suppresses both hemangiogenesis and lymphangiogenesis in mouse models of neovascularization, induced by corneal sutures or hindlimb ischemia, as efficiently as a 21-nt siRNA targeting VEGF-A [1].

Among 15 siRNAs, Khairuddin et al. [49] identified an extremely immunostimulatory siRNAs, targeting the HPV, which exerted potent anti-tumoural function. This bifunctional siRNA could reduce growth of established TC-1 tumours in C57BL/6 mice, and its effect was TLR7 dependent, where ablation of TLR7 recruitment via 2′O-methyl modification of the oligo backbone reduced these anti-tumour effects. Flatekval et al. [40] designed either monofunctional siRNAs devoid of immunostimulation or bifunctional siRNAs with IDO silencing and immunostimulatory activities. They showed that bifunctional siRNAs were able to knockdown IDO expression and induce cytokine production through either endosomal TLR7/8 or RIG-I.

In the past 10 years, several studies reported that bifunctional 3p-siRNA (Exp:targeting Bcl2/TGF-β/Survivin/Glutaminase/IDO) with target silencing and an innate immunity stimulation via RIG-I activation could confer potent anti-tumour efficacy. This is illustrated for the first time by the work of Poeck et al. [8], who reported that bifunctional siRNAs, with 5′-triphosphate targeting Bcl2 (3p-siRNA), led to better melanoma tumour reduction than OH-siRNA or 5′-triphosphate siRNAs containing target mismatches. Poeck and his colleagues revealed that siRNA with 5′-triphosphate ends could be recognized by RIG-I and activate an innate immune cells such as DC; then, expression of IFNs was directly induced, leading to apoptosis in tumour cells. These bifunctional 3p-siRNAs with RIG-I activation and RNAi-mediated Bcl2 silencing could provoke massive apoptosis of tumour cells in lung metastases in vivo. This was the first report demonstrating that 3p-siRNA represents a single molecule-based approach in which RIG-I function activates immune cell and gene silencing, leading to a key molecular event. Researchers subsequently found that 3p-TGFβ1-siRNA combining RIG-I activation with gene silencing of TGF-β1 induced profound tumour cell apoptosis and revealed potent anti-tumour efficacy in pancreatic cancer. This kind of 3p-siRNA induces a Th1 cytokine profile, demonstrating IFN-γ induction and IL-4 inhibition. High level of IFN and CXCL10 recruited more activated CD8+ T cells to the tumour. Frequency of immunosuppressive CD11b+ Gr-1+ myeloid cells was reduced after 3p-TGFβ1-siRNA treatment [50].

In addition, 3p-siRNA against survivin gene was designed and generated. This finding demonstrated that 3p-survivin-siRNA inhibited lung cancer cell proliferation and induced a RIG-I-dependent type-I interferon response [7]. Recently, 5′-triphosphate siRNA combining glutaminase (GLS) silencing with RIG-I activation was demonstrated to induce more prominent anti-tumour responses than RIG-I ligand or GLS silencing capability alone. 3p-siRNA-GLS effectively induced intrinsic proapoptotic signalling, and GLS silence sensitized malignant cells to apoptosis induced by RIG-I activation. Moreover, cytotoxicity was enhanced, resulting from disturbed glutaminolysis induced by GLS silencing. Finally, RIG-I activation by 3p-siRNA-GLS blocked autophagic degradation, leading to dysfunction of mitochondria, whereas GLS silencing severely impaired reactive oxygen species (ROS) scavenging systems, leading to a vicious circle of ROS-mediated cytotoxicity [51]. Immature monocyte-derived DCs had been transfected with siRNA-bearing 5′-triphosphate-activated T cells [40].

In addition, 3p-siRNA can inhibit hepatitis B virus (HBV), Influenza A Virus and Coxsackievirus, by gene silencing and RIG-I activation. RNAi provides a promising approach for the specific treatment of HBV infection. Our laboratory has previously demonstrated that 3p-HBx-siRNA and shRNA-HBx not only directly inhibit HBV replication but also stimulate innate immunity against HBV, which are both beneficial for the inversion of HBV-induced immune tolerance [52]. In HBV-positive hepatoma HepG2.2.15 cells, 3p-HBx-siRNA combining RIG-I activation with HBx gene silencing induce stronger type I IFN response than non-target 3p-scramble-siRNA, indicating that a potent immunostimulatory effect may partly contribute to the reversal of immune tolerance through decreasing HBV load; 3p-HBx-siRNA more strongly inhibited HBV replication and promoted IFN production than HBx-siRNA in primary HBV(+) hepatocytes, and this effect was mediated by RIG-I activation [52]. This was consistent of the other two reports [53,54]. Our dually functional vector containing both an immunostimulatory single-stranded RNA (ssRNA) and an HBx-silencing shRNA could reverse HBV-induced hepatocyte-intrinsic immune tolerance; TLR7 signalling pathway was attributed to this progress [55].

Lin et al. [56] designed and tested a 3p-mNP1496-siRNA against influenza virus. They found that 3p-mNP1496-siRNA could activate the RIG-I-mediated IFN-β pathway and significantly reduce virus load and virus-induced pathogenesis. The inhibition effect was in an siRNA- and RIG-I-dependent manner, demonstrating siRNA playing dual antiviral roles: viral gene-specific silencing and non-gene-specific RIG-I activation. This strategy was also proved to elicit potent antiviral effects in coxsackievirus myocarditis, and virus-specific 3p-siRNA was superior to both conventional virus-specific siRNA and non-target 3p-siRNA in inhibiting viral replication and subsequent cytotoxicity [57].

In the attempt to inhibit the expression of woodchuck hepatitis virus (WHV), Meng et al. [58] found that innate immune responses could be enhanced by RNAi through the PKR- and TLR-dependent signalling pathways in primary hepatocytes. The immunostimulation by RNAi may contribute to the antiviral activity of siRNAs in vivo.

Furthermore, siRNA can also synergistically enhance DNA-mediated type III IFN (a newly characterized antiviral interferon) response in non-immune or primary immune cells. This enhancement is mediated by crosstalk signalling pathway between RIG-I (RNA sensor) and IFI16 (DNA sensor) [59].

Designing with GU sequences, addition of triphosphate motifs to siRNA, co-treatment with CpG oligos are believed to activate innate immunity when siRNA was applied in vitro and in vivo. Accumulating evidence suggests these bifunctional siRNAs could activate NK cells and CD8+ T cells in different models. Thus, specific clinical applications of RNAi can benefit from a concurrent activation of the immune system.

Advertisement

3. miRNA

It has been well discussed how miRNAs regulate signalling pathways, and the dynamics of the immune response, tolerance and homeostasis. Here we summarize and explore updated achievements of special miRNAs in immunopharmacology.

3.1. miRNAs as intrinsic targets in antiviral immunity

In addition to the conventional innate and adaptive immune responses, even in the earlier phase after virus invasion, the host cell suppresses viral replication by evolving the profile of special and constitutively expressed genes. These cell-intrinsic antiviral approaches based on host restriction factors may be no less important than in considerations of conventional immunity. At the same time, viruses also gain some countermeasures or adapt the unique phenotype of their hosts substantially to survive. Moreover, miRNAs may also be involved in the inextricably intertwined relationship between viruses and their hosts.

In 2005, a liver-specific miRNA, miR122, which is involved in cholesterol and lipid metabolism [60], was illustrated to be necessary for hepatitis C virus (HCV) accumulation in cultured liver cells [61]. Researchers found that miR122 directly binds to two close sites in the 5′ non-coding region of the HCV genome and promote HCV translation [6264]. This miRNA kept conserved among all HCV subtypes [65,66]. Even in non-hepatic cell, miR122 could boost HCV replication [67]. Moreover, miR122 was further proved to be significantly reduced after IFN-β treatment, and miR122 mimics neutralized IFN-induced anti-HCV effect [68]. Epidemiological and genomic researches further suggested that the level of miR122 in individuals with HCV might be an ’indicator’ for IFN therapy, and only those patients with high levels of miR122 responded well to IFN therapy [69,70]. Therefore, miR122 antagonist would also be called as IFN ’sensitizer‘ in HCV immune treatment.

Santaris Pharma designed and synthesized an LNA-based miR122 inhibitor, named Miravirsen (or SPC3649), to eradicate HCV. The product was first evaluated in preclinical studies in mice [71], cynomolgus monkeys [72], green African monkeys and chimpanzees [73,74]. Here the key concern is that whether miR122 inhibitor can effectively lower the level of free miR122 and inhibit HCV replication without disturbing normal cholesterol and lipid metabolism or without any potential chemical toxicity. Interestingly, although there was a reduction of cholesterol levels in plasma by nearly 40%, Miravirsen caused a dose-dependent reduction of miR122 and maintained ∼5-week-long half-life in the liver of monkeys and chimpanzees [73,74]. Moreover, in the high-dose treatment group, Miravirsen decreased HCV subtype 1a or 1b more than 2 orders of magnitude compared to control group. In all animal species, Miravirsen was reported to be safe, without serious adverse effects or dose-related toxicities in rats, monkeys and human [75,76].

In May 2008, Miravirsen was put into human clinical trials as the first miRNA-based drug (https://clinicaltrials.gov/ct2/show/NCT00979927). There was a significant, dose-dependent reduction and sustained decrease of HCV viremia after drug administration in human subjects, and several patients became even HCV undetectable during the study. At the same time, only infrequent and moderately adverse effects were caused to some volunteers and did not influence the trial process [77]. Because miR122 is only liver enriched in physiological conditions and there is high amount of miR122 in adult human liver, it may be an ideal target to design highly specific anti-HCV drugs with good resistance to HCV infected person, particularly to those who have no tolerance to traditional treatments. In the following years, miR196 [78], let-7b family [79] and some other miRNAs were then proved to influence HCV life cycle, providing new target to restrict hepatitis C infection and avoid chronic infection.

Besides HCV, some other kind of viruses also encode miRNAs or regulate the miRNAs expression in host cells to disturb the expression of many immune-associated genes directly and/or indirectly, so that they can be critical regulators for viral life cycle. For example, in HEK293T cell, prototype foamy virus I (PFV-1) encodes Tas protein to counteract cell-encoded miR32, which could inhibit PFV-1 gene expression and accumulation [80]. Kaposi’s sarcoma-associated herpesvirus (KSHV)-induced miR132 could silence p300 expression, which is critical for the transcription initialization of many antiviral genes, to help themselves maintain long-time latency [81]. The hematopoietic-cell-specific miR142-3p potently restricts the replication of eastern equine encephalitis virus in myeloid-lineage cells by binding to the 3′-untranslated region (UTR) of viral genome [82]. Even Drosha, the enzyme that processes miRNA biogenesis and maturation, was an independent factor for limiting RNA virus replication along with canonical type I IFN system in particular cell type [83]. Above of all, it is much likely that miRNA mimics (for viral inhibitory miRNAs) or antagonists (for viral beneficial miRNAs) can be effective antiviral strategies as intrinsic immune drugs.

3.2. miRNA regulation antimicrobial and anti-tumour immunity

3.2.1. miRNA in antimicrobial innate immunity

Of the known PRRs, TLRs and RIG-like receptors (RLRs) have been well studied in mediating antimicrobial and inflammatory responses during infections, which may be targets of pathogens or host-encoded miRNAs.

The first PRR targeting miRNA let7i was reported in 2007 [84], which targeted TLR4 mRNA in a MyD88/NF-κB-dependent way during Cryptosporidium parvum infection, controlling the production of inflammatory factors. During Bacillus Calmette-Guérin (BCG) infection, miR124 exerts its function by targeting multiple components of the TLR signalling pathway, including TLR6, MyD88, TNF receptor-associated factor 6 (TRAF6) and TNF-α in mouse lung cell [85]. After HCV infection, miR373 was induced and negatively regulated the type I IFN signalling pathway by suppressing Janus kinase 1(JAK1) and IRF9 in hepatocytes [86]. Experimental evaluation using miR124 inhibitors or miR373 knockout up-regulated BCG-induced pro-inflammatory factors or type I IFN and so as to inhibit BCG or HCV more efficiently. Besides using host miRNAs, human cytomegalovirus (HCMV) targeted TLR2 by encoding its own miRNA, miR-UL112-3p, and reduced the expression of IL-1β, IL-6 and IL-8 upon stimulation with a TLR2 agonist [87]. Neutralizing this miRNA might recover normal cytokines production.

Besides immune inhibitory miRNAs, dengue virus (DENV)-induced miR30e* up-regulated IFN-β and the downstream IFN-stimulated genes (ISGs) by suppressing IkBα and promoting NF-κB-dependent IFN production [88]. The transfection of miR30e* would increase the expression of 2′-5′-oligoadenylate synthetase 1(OAS-1), myxovirus resistance A (MxA) and interferon-induced transmembrane protein (IFITM). In 2014, miR526 [89] was proved to enhance RIG-I-induced viral replication by suppression of the expression of cylindromatosis (CYLD), which suppresses RIG-I K-63-linked polyubiquitin. Moreover, Enterovirus 71(EV71) inhibited miR526 transcription in an IRF-dependent way and so as to attenuate virus-triggered type I interferon production. These studies suggested that recruitment or increase of miR30e* or miR526 would stimulate type I IFN expression and inhibit virus more quickly.

3.2.2. ’Immune miRs‘ as immunopharmaceutic agents

With the general knowledge of immunologically relevant miRNAs established in the past 10 years, many miRNAs have been intensively investigated using gain- and loss-of-function methods, showing how this novel class of small non-coding RNA participates in mammalian immunity. And individual immune miRNA might contribute its implications to various immune-mediated diseases.

The role of miR125b in immune signalling may be paradoxical. After stimulation with LPS, miR125b was down-regulated and TNF-α, one of miR125b targets, was overexpressed in RAW264.7, which is essential for antimicrobial activity. Moreover, during M. tuberculosis infection, the overexpression of miR125a significantly attenuated the antimicrobial effects in macrophages through targeting UV radiation resistance–associated gene (UVRAG) [90]. Nevertheless, in diffuse large B cell lymphoma (DLBCL), miR125a and miR125b directly target a negative NF-κB regulator tumour necrosis factor alpha-induced protein 3(TNFAIP3) and present a positive self-regulatory property to maintain prolonged NF-κB activity. Taken together, whether overexpression or inhibition of miR125b in an anti-infection therapeutic study depends on concrete circumstances.

miR146a also acts as a negative regulator in immune sensing. Both in mouse and human, miR146a was always exploited by virus to attenuate innate and adaptive antiviral immunity mainly in DC [91], lymphocyte [92] and hepatocytes by inhibiting interleukin-1 receptor-associated kinase 1(IRAK1), TRAF6 [93], son of sevenless homolog 1 (SOS1) [94] and STAT1 [95]. Silencing of miR146a via the delivery of sponge or antagomiR could restore the expression of inflammatory factors, augment type I IFN production and promote clearance of vesicular stomatitis virus (VSV) [96], dengue virus [97], enterovirus 71 (EV71) [94,98] and HBV [95]. Because miR146a was also abnormally expressed in hepatocellular carcinoma (HCC) and exerted negative anti-tumour effects by up-regulation of immunoinhibitory cytokines such as TGF-β, IL-17, VEGF, miR146a may also be a novel immunotherapeutic target for HCC [99].

Unlike miR146a, miR155 always promotes immune signal transduction, enhances immune function or speeds lymphocyte proliferation. Mice lacking miR155 have impaired CTL cell responses to infections with lymphocytic choriomeningitis virus and the intracellular bacteria Listeria monocytogenes because of insufficient activation of Akt pathway after TCR cross-linking [100]. miR155 knockout mice died soon after Erdman (a variant from severe acute respiratory syndrome [SARS]) infection and held higher level of colony-forming units (CFU) in lungs than wild-type mice [101]. During HIV infection, miR155 inhibited the HIV-activating effects of tripartite motif-containing protein 32 (TRIM32), and therefore, it might promote a return to latency in CD4+ reservoir cells [102]. In addition, in NK cells, miR155 might regulate T cell immunoglobulin-3 (Tim-3)/T-bet/STAT-5-signalling axis, and following cytokine expression that balanced antiviral response and immune injury during chronic HCV infection [103]. A remarkably ectopic up-expression of miR155 can be observed by delivering hepatotropic adeno-associated virus 8 (AAV8) vectors to the liver of mice, and then high level of miR155-enhanced GAP’s protective capacity against parasite [104]. These studies imply miR155 as an immune-augmenting adjuvant in improving the antigenicity of vaccination.

miR223 was already proved to be of importance in myeloid progenitor cells proliferation and responsiveness to pathogenic stimuli in neutrophils by targeting myocyte-specific enhancer factor 2C (MEF2C), acting as a fine-tune regulator both in normal granulocytes generation and in preventing aberrant expansion and over-activated inflammatory responses. In recent years, miR223 was involved in inflammasome response by targeting NLR family pyrin domain containing 3 (NLRP3) in human [105]. Moreover, Epstein–Barr virus (EBV) encoded a mimic of hsa-miR223, called miR-BART15, targeting the same site within the NLRP3 3’-UTR to repress inflammasome activation. Furthermore, this miRNA can be secreted from EBV-infected B cells into exosomes to rheostat NLRP3 inflammasome activity in non-infected cells [106]. miR223 sponge would balance the amount of NLRP3 and ‘absorb’ EBV-miR-BART15 in macrophages and DCs.

It is noteworthy that two groups of miRNAs, which shaped NK-mediated cytotoxicity, have potent value for developing antiviral and anti-tumour biodrugs. First, NKG2D–NKG2D-L interaction plays a predominant role in ‘NK cell–abnormal cell’ recognition. MICB/A, ULBPs targeting miRNAs, are not only encoded by human genome as stress regulators but also synthesized by some virus (e.g. HCMV-miR-UL112 [107], EBV-miRBART2 [108], KSHV-miR-K12-7 [108] and BK virus (BKV)-miR-B1-3p, JC virus (JCV)-miR-J1-3p[109]), to escape from NK cell killing. Meanwhile, viral infected cell and tumour cell always express low MICA/B because of up-regulated MICB/MICA, targeting miRNAs such as miR20a, 93, 103, 106b [110] to maintain a compromised micro-environment. Furthermore, non-classical human leukocyte antigen G (HLA-G) is known as an inhibitory ligand, which suppresses the cytotoxic activity of T and NK cells. Studies demonstrated a strong post-transcriptional gene regulation of the HLA-G by miR148a, miR148b and miR152, and lower expression of these miRNAs in renal carcinoma [111] and placental choriocarcinoma cells [112]. Stable manipulation of these activating and inhibitory miRNAs may enhance NK and LAK cell-mediated cytotoxicity against infected and tumour cells. Therefore, it could be concluded that modulating the expression or inhibition of specific miRNAs could boost immune response during viral infections or against cancers.

3.3. miRNA in maintaining immune homeostasis

Because several miRNAs participate in immune cell development and differentiation, abnormal expression of miRNA may cause a disturbance of homeostasis by changing the ratio of helper and regulatory cell subsets, or perturb the functionality and survival of effect-memory cells that lead to lymphoproliferative disease. Utilization of miRNA interference techniques may recover regular immune balance.

3.3.1. miR17-92, miR146a and miR155 in Systemic Lupus Erythematosus (SLE)

In 2007, a unique mouse strain, ‘sanroque’, presented a pattern of lupus pathology, revealing the core role of T follicular helper (Tfh) in systemic autoimmunity [113]. miR17-92 was found to regulate Tfh cell differentiation, which is essential for maintenance of the germinal centre formation and sustained antibody responses. Overexpression of this miRNA in T cells would enhance Tfh cell proliferation and survive an autoantibody production [112]. Similarly, miR155 increased IL-21-mediated STAT3 signalling in T cell [114], which might accelerate Tfh differentiation and maturation as well. Moreover, miR155 deficiency ameliorates autoimmune inflammation of SLE by targeting s1pr1 in mice [115]. Therefore, miR17-92 and miR155 might be a new target to restrain aggressive autoimmune response in SLE.

3.3.2. miR29 and miR146 in type 1 diabetes

Type 1 diabetes (T1D) is a chronic autoimmune disease that results from the persisting destruction of pancreatic β-cells by autoreactive CD8+ T cell and Th1 cytokines. Dicer 1 deletion in β-cell would disrupt normal β-cell development and survival, lead to impairment of insulin secretion and diabetes development [116], apparently suggesting that miRNAs network is necessary for normal glycometabolism. Recently, endogenous miR29b released from pancreatic β-cells within exosomes stimulated TNF-α secretion in spleen cells isolated from diabetes-prone non-obese diabetic (NOD) mice. Delivery of miR29b to mice activated myeloid cell and pDCs to induce IFN-α, TNF-α and IL-6 production [117]. Abnormal expression of miR146 is associated with high serum titers of glutamic acid decarboxylase antibody in T1D patients, indicating the involvement of miR146 in the sustained immune imbalance during T1D progress [118]. These findings raised the possibility of developing a new clue for T1D immunotherapy using miRNA-based agents.

3.3.3. miR146a and miR155 in rheumatoid arthritis

The role of miR146a in controlling Treg-mediated decrease of Th1 responses has been demonstrated [119]. In contrast, miR155 promoted Th1 and Th17 differentiation and cell formation and lowered T cell sensitivity to IFN-γ-driven proliferation by targeting C-MAF and IFNγRα [120]. Therefore, imbalance of miR146a and miR155 may be an epigenetic phenotype for autoimmune response. In rheumatoid arthritis (RA), decreased expression of miR146a contributes to an abnormal Treg phenotype and allows Th1/Th17 skewing while low level of miR155 failed to support effective Th2 immunity [121]. Systemic administration of miR146a has potential therapeutic intervention for preventing bone destruction by inhibited Th1 and Th17 cells, as well as IL-1β, IL-6 and TNF-α [122].

3.3.4. miR15 and miR326 in multiple sclerosis

Multiple sclerosis (MS) is manifested by chronic and progressive inflammatory demyelination of the central nervous system and is one of the main causes of regressive neurological diseases. Study on MS animal model illustrated that mice with fewer Th17 cells were less susceptible to experimental autoimmune encephalomyelitis (EAE) [123]. Therefore, Th17-targeting biotherapeutic approaches may be a promising way to cure multiple sclerosis. Gang Pei’ s laboratory [124] found that miR326 promoted Th17 differentiation by targeting Ets-1 (a negative regulator of Th17 polarization) and antagonizing miR326 by sponge vector that resulted in fewer Th17 cells and Th17 cytokines and remitting EAE symptom. Inversely, increased miR155 in primary human microglia up-regulated pro-inflammatory cytokine secretion and co-stimulatory surface marker expression suggested that miR155 inhibition in myeloid cell might be useful to suppress allogeneic T cell responses [125]. In conclusion, reverse pathological expressed miRNAs and re-balance dysregulated immune genes are of consideration to treat multiple sclerosis.

Advertisement

4. Conclusion

RNAi technology holds promise for treating various human diseases. It is becoming apparent that clinical outcome of cancer immunotherapy and infectious diseases can be improved by targeted strategies to abrogate tumour-induced immunosuppression. Anti-tumour strategies using siRNA/shRNA/miRNA for both silencing of oncogenes and recruiting of innate receptors were designed. The present researches highlighted the potential therapeutic applications of this new generation of siRNAs in immunotherapy.

Additionally, but importantly, siRNA/shRNA or miRNA drugs with regard to pharmacodynamic difficulties and unwanted side effects are even more complicated compared to low molecular weight drugs and hard to be delivered into immune cells. This requires more extensive procedure than any other traditional drugs. Considering clinical challenges for RNA-based nucleic acid drugs, including barriers and RNases, the advanced tissue-directed delivery systems with safety, high efficiency and specificity, long-term function and controllability are required. Although the exploration of such tiny regulators causally bring pharmacists a considerable effort to draw up individualized and tailor-made strategies, we believe that immunoregulation triggered by siRNA/shRNA/miRNA can be used to regulate the host immunity against cancers or viruses. The development of multifunctional RNAi molecules will greatly contribute to the future arsenal of tools to combat not only microbial pathogens but also hard-to-treat cancer.

Advertisement

Acknowledgments

This work was supported by the Natural Science Foundation of China (81172789, 81373222, 31200651), National Basic Research Program of China (No. 2013CB531503) and National Mega Project on Major Infectious Diseases Prevention and Treatment (2012ZX10002006), The Special Foundation of Taishan Overseas Distinguished Experts and Scholars, The Priority Research Program of Shandong Academy of Sciences, Natural Science Foundation of Shandong Province (No.BS2015YY023), Natural Science Foundation of Shandong Academy of Sciences (No. 2014QN004) and Science and Technology Development Foundation of Shandong Analysis and Test Center.

References

  1. 1. Cho WG, Albuquerque RJ, Kleinman ME: Small interfering RNA-induced TLR3 activation inhibits blood and lymphatic vessel growth. Proc Natl Acad Sci U S A. 2009; 106: 7137–7142. DOI: 10.1073/pnas.08123171060812317106
  2. 2. Forsbach A, Nemorin JG, Volp K: Characterization of conserved viral leader RNA sequences that stimulate innate immunity through TLRs. Oligonucleotides. 2007; 17: 405–417. DOI: 10.1089/oli.2007.0098
  3. 3. Judge AD, Sood V, Shaw JR: Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol. 2005; 23: 457–462. DOI: 10.1038/nbt1081
  4. 4. Kleinman ME, Yamada K, Takeda A: Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008; 452: 591–597. DOI: 10.1038/nature06765
  5. 5. Melchjorsen J, Jensen SB, Malmgaard L: Activation of innate defense against a paramyxovirus is mediated by RIG-I and TLR7 and TLR8 in a cell-type-specific manner. J Virol. 2005; 79: 12944–12951. DOI: 10.1128/JVI.79.20.12944-12951.2005
  6. 6. Hornung V, Guenthner-Biller M, Bourquin C: Sequence-specific potent induction of IFN-alpha by short interfering RNA in plasmacytoid dendritic cells through TLR7. Nat Med. 2005; 11: 263-270. DOI: 10.1038/nm1191
  7. 7. Wang K, Chen X, Yan F: 5'-triphosphate-siRNA against survivin gene induces interferon production and inhibits proliferation of lung cancer cells in vitro. J Immunother. 2013; 36: 294–304. DOI: 10.1097/CJI.0b013e318294183b
  8. 8. Poeck H, Besch R, Maihoefer C: 5'-Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma. Nat Med. 2008; 14: 1256–1263. DOI: 10.1038/nm.1887
  9. 9. Alshamsan A, Hamdy S, Haddadi A: STAT3 knockdown in B16 melanoma by siRNA lipopolyplexes induces bystander immune response in vitro and in vivo. DOI: Transl Oncol. 2011; 4: 178–188. DOI: 10.1593/tlo.11100
  10. 10. Herrmann A, Kortylewski M, Kujawski M: Targeting Stat3 in the myeloid compartment drastically improves the in vivo antitumor functions of adoptively transferred T cells. Cancer Res. 2010; 70: 7455–7464. DOI: 10.1158/0008-5472.CAN-10-0736
  11. 11. Alshamsan A, Haddadi A, Hamdy S: STAT3 silencing in dendritic cells by siRNA polyplexes encapsulated in PLGA nanoparticles for the modulation of anticancer immune response. Mol Pharm. 2010; 7: 1643–1654. DOI: 10.1021/mp100067u
  12. 12. Kortylewski M, Swiderski P, Herrmann A: In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses. Nat Biotechnol. 2009; 27: 925–932. DOI: 10.1038/nbt.1564
  13. 13. Hossain DM, Pal SK, Moreira D: TLR9-targeted STAT3 silencing abrogates immunosuppressive activity of myeloid-derived suppressor cells from prostate cancer patients. Clin Cancer Res. 2015; 21: 3771–3782. DOI: 10.1158/1078-0432.CCR-14-3145
  14. 14. Zhang Q, Hossain DM, Nechaev S: TLR9-mediated siRNA delivery for targeting of normal and malignant human hematopoietic cells in vivo. Blood. 2013; 121: 1304–1315. DOI: 10.1182/blood-2012-07-442590b
  15. 15. Luo Z, Wang C, Yi H: Nanovaccine loaded with poly I:C and STAT3 siRNA robustly elicits anti-tumor immune responses through modulating tumor-associated dendritic cells in vivo. Biomaterials. 2015; 38: 50–60. DOI: 10.1016/j.biomaterials.2014.10.050
  16. 16. Shi D, Li D, Yin Q: Silenced suppressor of cytokine signaling 1 (SOCS1) enhances the maturation and antifungal immunity of dendritic cells in response to Candida albicans in vitro. Immunol Res. 2015; 61: 206-218. DOI: 10.1007/s12026-014-8562-8
  17. 17. Akita H, Kogure K, Moriguchi R: Nanoparticles for ex vivo siRNA delivery to dendritic cells for cancer vaccines: programmed endosomal escape and dissociation. J Control Release. 2010; 143: 311–317. DOI: 10.1016/j.jconrel.2010.01.012
  18. 18. Heo MB, Cho MY, Lim YT: Polymer nanoparticles for enhanced immune response: combined delivery of tumor antigen and small interference RNA for immunosuppressive gene to dendritic cells. Acta Biomater. 2014; 10: 2169–2176. DOI: 10.1016/j.actbio.2013.12.050
  19. 19. Subramanya S, Armant M, Salkowitz JR: Enhanced induction of HIV-specific cytotoxic T lymphocytes by dendritic cell-targeted delivery of SOCS-1 siRNA. Mol Ther. 2010; 18: 2028–2037. DOI: 10.1038/mt.2010.148
  20. 20. Zhang X, Su Y, Song H: Attenuated A20 expression of acute myeloid leukemia-derived dendritic cells increased the anti-leukemia immune response of autologous cytolytic T cells. Leuk Res. 2014; 38: 673–681. DOI: 10.1016/j.leukres.2014.03.011
  21. 21. Warashina S, Nakamura T, Harashima H: A20 silencing by lipid envelope-type nanoparticles enhances the efficiency of lipopolysaccharide-activated dendritic cells. Biol Pharm Bull. 2011; 34: 1348–1351. DOI: 10.1248/bpb.34.1348
  22. 22. Song XT, Evel-Kabler K, Shen L: A20 is an antigen presentation attenuator, and its inhibition overcomes regulatory T cell-mediated suppression. Nat Med. 2008; 14: 258–265. DOI: 10.1038/nm1721
  23. 23. Breckpot K, Aerts-Toegaert C, Heirman C: Attenuated expression of A20 markedly increases the efficacy of double-stranded RNA-activated dendritic cells as an anti-cancer vaccine. J Immunol. 2009; 182: 860–870. DOI: 10.4049/jimmunol.182.2.860
  24. 24. Cubillos-Ruiz JR, Engle X, Scarlett UK: Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J Clin Invest. 2009; 119: 2231–2244. DOI: 10.1172/JCI3771637716 [pii]
  25. 25. Hobo W, Maas F, Adisty N: siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells. Blood. 2010; 116: 4501–4511. DOI: 10.1182/blood-2010-04-278739
  26. 26. van der Waart AB, Fredrix H, van der Voort R: siRNA silencing of PD-1 ligands on dendritic cell vaccines boosts the expansion of minor histocompatibility antigen-specific CD8(+) T cells in NOD/SCID/IL2Rg(null) mice. Cancer Immunol Immunother. 2015; 64: 645–654. DOI: 10.1007/s00262-015-1668-6
  27. 27. Iwamura K, Kato T, Miyahara Y: siRNA-mediated silencing of PD-1 ligands enhances tumor-specific human T-cell effector functions. Gene Ther. 2012; 19: 959–966. DOI: 10.1038/gt.2011.185
  28. 28. Zhang Q, Ichimaru N, Higuchi S: Permanent acceptance of mouse cardiac allografts with CD40 siRNA to induce regulatory myeloid cells by use of a novel polysaccharide siRNA delivery system. Gene Ther. 2015; 22: 1–10. DOI: 10.1038/gt.2014.119
  29. 29. Gong X, Han B, Zou Y: Attenuation of experimental autoimmune myocarditis by si-RNA mediated CD40 silencing. Int Heart J. 2014; 55: 539–545. DOI: 10.1536/ihj.14-125
  30. 30. Zheng X, Suzuki M, Ichim TE: Treatment of autoimmune arthritis using RNA interference-modulated dendritic cells. J Immunol. 2010; 184: 6457–6464. DOI: 10.4049/jimmunol.0901717
  31. 31. Conroy H, Galvin KC, Higgins SC: Gene silencing of TGF-beta1 enhances antitumor immunity induced with a dendritic cell vaccine by reducing tumor-associated regulatory T cells. Cancer Immunol Immunother. 2012; 61: 425–431. DOI: 10.1007/s00262-011-1188-y
  32. 32. Xu Z, Wang Y, Zhang L: Nanoparticle-delivered transforming growth factor-beta siRNA enhances vaccination against advanced melanoma by modifying tumor microenvironment. ACS Nano. 2014; 8: 3636–3645. DOI: 10.1021/nn500216y
  33. 33. Ahn YH, Hong SO, Kim JH: The siRNA cocktail targeting interleukin 10 receptor and transforming growth factor-beta receptor on dendritic cells potentiates tumour antigen-specific CD8(+) T cell immunity. Clin Exp Immunol. 2015; 181: 164–178. DOI: 10.1111/cei.12620
  34. 34. Liu Z, Falo LD, Jr., You Z: Knockdown of HMGB1 in tumor cells attenuates their ability to induce regulatory T cells and uncovers naturally acquired CD8 T cell-dependent antitumor immunity. J Immunol. 2011; 187: 118–125. DOI: 10.4049/jimmunol.1003378
  35. 35. Tsai BY, Suen JL, Chiang BL: Lentiviral-mediated Foxp3 RNAi suppresses tumor growth of regulatory T cell-like leukemia in a murine tumor model. Gene Ther. 2010; 17: 972–979. DOI: 10.1038/gt.2010.38
  36. 36. Yongsheng Y, Xiaoliang L, Zhenghao T: siRNA-mediated knockdown of FoxP3 promotes the ratio of T-helper 1 (Th1) to Th2 in chronic hepatitis B patients. Turk J Gastroenterol. 2011; 22: 587–593. DOI: 10.4318/tjg.2011.0251
  37. 37. Kang S, Xie J, Ma S: Targeted knock down of CCL22 and CCL17 by siRNA during DC differentiation and maturation affects the recruitment of T subsets. Immunobiology. 2010; 215: 153–162. DOI: 10.1016/j.imbio.2009.03.001
  38. 38. Zheng X, Koropatnick J, Chen D: Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int J Cancer. 2013; 132: 967–977. DOI: 10.1002/ijc.27710
  39. 39. Zheng X, Koropatnick J, Li M: Reinstalling antitumor immunity by inhibiting tumor-derived immunosuppressive molecule IDO through RNA interference. J Immunol. 2006; 177: 5639–5646. DOI: 10.4049/jimmunol.177.8.5639
  40. 40. Flatekval GF, Sioud M: Modulation of dendritic cell maturation and function with mono- and bifunctional small interfering RNAs targeting indoleamine 2,3-dioxygenase. Immunology. 2009; 128: e837–848. DOI: 10.1111/j.1365-2567.2009.03093.x
  41. 41. Chen D, Koropatnick J, Jiang N: Targeted siRNA silencing of indoleamine 2, 3-dioxygenase in antigen-presenting cells using mannose-conjugated liposomes: a novel strategy for treatment of melanoma. J Immunother. 2014; 37: 123–134. DOI: 10.1097/CJI.0000000000000022
  42. 42. Yen MC, Lin CC, Chen YL: A novel cancer therapy by skin delivery of indoleamine 2,3-dioxygenase siRNA. Clin Cancer Res. 2009; 15: 641–649. DOI: 10.1158/1078-0432.CCR-08-1988
  43. 43. Huang TT, Yen MC, Lin CC: Skin delivery of short hairpin RNA of indoleamine 2,3 dioxygenase induces antitumor immunity against orthotopic and metastatic liver cancer. Cancer Sci. 2011; 102: 2214–2220. DOI: 10.1111/j.1349-7006.2011.02094.x
  44. 44. Huang M, Sun R, Wei H: Simultaneous knockdown of multiple ligands of innate receptor NKG2D prevents natural killer cell-mediated fulminant hepatitis in mice. Hepatology. 2013; 57: 277–288. DOI: 10.1002/hep.25959
  45. 45. Zeng XC, Zhang T, Huang DH: RNA interfering targeting human leukocyte antigen-G enhanced immune surveillance mediated by the natural killer cells on hepatocellular carcinoma. Ann Clin Lab Sci. 2013; 43: 135–144.
  46. 46. Chen LJ, Han ZQ, Zhou H: Inhibition of HLA-G expression via RNAi abolishes resistance of extravillous trophoblast cell line TEV-1 to NK lysis. Placenta. 2010; 31: 519–527. DOI: 10.1016/j.placenta.2010.03.008
  47. 47. Tan PH, Gao YJ, Berta T: Short small-interfering RNAs produce interferon-alpha-mediated analgesia. Br J Anaesth. 2012; 108: 662–669. DOI: 10.1093/bja/aer492
  48. 48. Kariko K, Bhuyan P, Capodici J: Small interfering RNAs mediate sequence-independent gene suppression and induce immune activation by signaling through toll-like receptor 3. J Immunol. 2004; 172: 6545–6549. DOI: 10.4049/jimmunol.172.11.6545
  49. 49. Khairuddin N, Gantier MP, Blake SJ: siRNA-induced immunostimulation through TLR7 promotes antitumoral activity against HPV-driven tumors in vivo. Immunol Cell Biol. 2012; 90: 187–196. DOI: 10.1038/icb.2011.19
  50. 50. Ellermeier J, Wei J, Duewell P: Therapeutic efficacy of bifunctional siRNA combining TGF-beta1 silencing with RIG-I activation in pancreatic cancer. Cancer Res. 2013; 73: 1709–1720. DOI: 10.1158/0008-5472.CAN-11-3850
  51. 51. Meng G, Xia M, Xu C: Multifunctional antitumor molecule 5'-triphosphate siRNA combining glutaminase silencing and RIG-I activation. Int J Cancer. 2014; 134: 1958–1971. DOI: 10.1002/ijc.28416
  52. 52. Han Q, Zhang C, Zhang J: Reversal of hepatitis B virus-induced immune tolerance by an immunostimulatory 3p-HBx-siRNAs in a retinoic acid inducible gene I-dependent manner. Hepatology. 2011; 54: 1179–1189. DOI: 10.1002/hep.24505
  53. 53. Chen X, Qian Y, Yan F: 5'-triphosphate-siRNA activates RIG-I-dependent type I interferon production and enhances inhibition of hepatitis B virus replication in HepG2.2.15 cells. Eur J Pharmacol. 2013; 721: 86–95. DOI: 10.1016/j.ejphar.2013.09.050
  54. 54. Ebert G, Poeck H, Lucifora J: 5' Triphosphorylated small interfering RNAs control replication of hepatitis B virus and induce an interferon response in human liver cells and mice. Gastroenterology. 2011; 141: 696–706, 706 e691-693. DOI: 10.1053/j.gastro.2011.05.001
  55. 55. Lan P, Zhang C, Han Q: Therapeutic recovery of hepatitis B virus (HBV)-induced hepatocyte-intrinsic immune defect reverses systemic adaptive immune tolerance. Hepatology. 2013; 58: 73–85. DOI: 10.1002/hep.26339
  56. 56. Lin L, Liu Q, Berube N: 5'-Triphosphate-short interfering RNA: potent inhibition of influenza A virus infection by gene silencing and RIG-I activation. J Virol. 2012; 86: 10359–10369. DOI: 10.1128/JVI.00665-12
  57. 57. Ahn J, Ko A, Jun EJ: Antiviral effects of small interfering RNA simultaneously inducing RNA interference and type 1 interferon in coxsackievirus myocarditis. Antimicrob Agents Chemother. 2012; 56: 3516–3523. DOI: 10.1128/AAC.06050-12
  58. 58. Meng Z, Zhang X, Wu J: RNAi induces innate immunity through multiple cellular signaling pathways. PLoS One. 2013; 8: e64708. DOI: 10.1371/journal.pone.0064708
  59. 59. Sui H, Zhou M, Chen Q: siRNA enhances DNA-mediated interferon lambda-1 response through crosstalk between RIG-I and IFI16 signalling pathway. Nucleic Acids Res. 2014; 42: 583–598. DOI: 10.1093/nar/gkt844
  60. 60. Esau C, Davis S, Murray SF: miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006; 3: 87–98. DOI: 10.1016/j.cmet.2006.01.005
  61. 61. Jopling CL, Yi M, Lancaster AM: Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005; 309: 1577–1581. DOI: 10.1126/science.1113329
  62. 62. Jopling CL, Schutz S, Sarnow P: Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell Host Microbe. 2008; 4: 77–85. DOI: 10.1016/j.chom.2008.05.013
  63. 63. Henke JI, Goergen D, Zheng J: microRNA-122 stimulates translation of hepatitis C virus RNA. EMBO J. 2008; 27: 3300–3310. DOI: 10.1038/emboj.2008.244
  64. 64. Randall G, Panis M, Cooper JD: Cellular cofactors affecting hepatitis C virus infection and replication. Proc Natl Acad Sci U S A. 2007; 104: 12884–12889. DOI: 10.1073/pnas.0704894104
  65. 65. Machlin ES, Sarnow P, Sagan SM: Masking the 5' terminal nucleotides of the hepatitis C virus genome by an unconventional microRNA-target RNA complex. Proc Natl Acad Sci U S A. 2011; 108: 3193–3198. DOI: 10.1073/pnas.1012464108
  66. 66. Li YP, Gottwein JM, Scheel TK: MicroRNA-122 antagonism against hepatitis C virus genotypes 1-6 and reduced efficacy by host RNA insertion or mutations in the HCV 5' UTR. Proc Natl Acad Sci U S A. 2011; 108: 4991–4996. DOI: 10.1073/pnas.1016606108
  67. 67. Chang J, Guo JT, Jiang D: Liver-specific microRNA miR-122 enhances the replication of hepatitis C virus in nonhepatic cells. J Virol. 2008; 82: 8215–8223. DOI: 10.1128/JVI.02575-07
  68. 68. Pedersen IM, Cheng G, Wieland S: Interferon modulation of cellular microRNAs as an antiviral mechanism. Nature. 2007; 449: 919–922. DOI: 10.1038/nature06205
  69. 69. Sarasin-Filipowicz M, Krol J, Markiewicz I: Decreased levels of microRNA miR-122 in individuals with hepatitis C responding poorly to interferon therapy. Nat Med. 2009; 15: 31–33. DOI: 10.1038/nm.1902
  70. 70. Murakami Y, Tanaka M, Toyoda H: Hepatic microRNA expression is associated with the response to interferon treatment of chronic hepatitis C. BMC Med Genomics. 2010; 3: 48. DOI: 10.1186/1755-8794-3-48
  71. 71. Elmen J, Lindow M, Silahtaroglu A: Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res. 2008; 36: 1153–1162. DOI: 10.1093/nar/gkm1113
  72. 72. Hildebrandt-Eriksen ES, Aarup V, Persson R: A locked nucleic acid oligonucleotide targeting microRNA 122 is well-tolerated in cynomolgus monkeys. Nucleic Acid Ther. 2012; 22: 152–161. DOI: 10.1089/nat.2011.0332
  73. 73. Lanford RE, Hildebrandt-Eriksen ES, Petri A: Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science. 2010; 327: 198–201. DOI: 10.1126/science.1178178
  74. 74. Elmen J, Lindow M, Schutz S: LNA-mediated microRNA silencing in non-human primates. Nature. 2008; 452: 896–899. DOI: 10.1038/nature06783
  75. 75. van der Ree MH, van der Meer AJ, de Bruijne J: Long-term safety and efficacy of microRNA-targeted therapy in chronic hepatitis C patients. Antiviral Res. 2014; 111: 53–59. DOI: 10.1016/j.antiviral.2014.08.015
  76. 76. Masaki T, Arend KC, Li Y: miR-122 stimulates hepatitis C virus RNA synthesis by altering the balance of viral RNAs engaged in replication versus translation. Cell Host Microbe. 2015; 17: 217–228. DOI: 10.1016/j.chom.2014.12.014
  77. 77. Janssen HL, Kauppinen S, Hodges MR: HCV infection and miravirsen. N Engl J Med. 2013; 369: 878. DOI: 10.1056/NEJMc1307787
  78. 78. Hou W, Tian Q, Zheng J: MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expression in human hepatoma cells expressing hepatitis C viral proteins. Hepatology. 2010; 51: 1494–1504. DOI: 10.1002/hep.23401
  79. 79. Cheng JC, Yeh YJ, Tseng CP: Let-7b is a novel regulator of hepatitis C virus replication. Cell Mol Life Sci. 2012; 69: 2621–2633. DOI: 10.1007/s00018-012-0940-6
  80. 80. Lecellier CH, Dunoyer P, Arar K: A cellular microRNA mediates antiviral defense in human cells. Science. 2005; 308: 557–560. DOI: 10.1126/science.1108784
  81. 81. Lagos D, Pollara G, Henderson S: miR-132 regulates antiviral innate immunity through suppression of the p300 transcriptional co-activator. Nat Cell Biol. 2010; 12: 513–519. DOI: 10.1038/ncb2054
  82. 82. Trobaugh DW, Gardner CL, Sun C: RNA viruses can hijack vertebrate microRNAs to suppress innate immunity. Nature. 2014; 506: 245–248. DOI: 10.1038/nature12869
  83. 83. Shapiro JS, Schmid S, Aguado LC: Drosha as an interferon-independent antiviral factor. Proc Natl Acad Sci U S A. 2014; 111: 7108–7113. DOI: 10.1073/pnas.1319635111
  84. 84. Chen XM, Splinter PL, O'Hara SP: A cellular micro-RNA, let-7i, regulates toll-like receptor 4 expression and contributes to cholangiocyte immune responses against Cryptosporidium parvum infection. J Biol Chem. 2007; 282: 28929–28938. DOI: 10.1074/jbc.M702633200
  85. 85. Ma C, Li Y, Zeng J: Mycobacterium bovis BCG triggered MyD88 induces miR-124 feedback negatively regulates immune response in alveolar epithelial cells. PLoS One. 2014; 9: e92419. DOI: 10.1371/journal.pone.0092419
  86. 86. Mukherjee A, Di Bisceglie AM, Ray RB: Hepatitis C virus-mediated enhancement of microRNA miR-373 impairs the JAK/STAT signaling pathway. J Virol. 2015; 89: 3356–3365. DOI: 10.1128/JVI.03085-14
  87. 87. Landais I, Pelton C, Streblow D: Human cytomegalovirus miR-UL112-3p targets TLR2 and modulates the TLR2/IRAK1/NFkappaB signaling pathway. PLoS Pathog. 2015; 11: e1004881. DOI: 10.1371/journal.ppat.1004881
  88. 88. Michael SF, Zhu X, He Z: MicroRNA-30e* suppresses dengue virus replication by promoting NF-κB–dependent IFN production. PLoS Neglected Tropical Diseases. 2014; 8: e3088. DOI: 10.1371/journal.pntd.0003088
  89. 89. Xu C, He X, Zheng Z: Downregulation of microRNA miR-526a by enterovirus inhibits RIG-I-dependent innate immune response. J Virol. 2014; 88: 11356–11368. DOI: 10.1128/JVI.01400-14
  90. 90. Kim JK, Yuk JM, Kim SY: MicroRNA-125a inhibits autophagy activation and antimicrobial responses during mycobacterial infection. J Immunol. 2015; 194: 5355–5365. DOI: 10.4049/jimmunol.1402557
  91. 91. Park H, Huang X, Lu C: MicroRNA-146a and microRNA-146b regulate human dendritic cell apoptosis and cytokine production by targeting TRAF6 and IRAK1 proteins. J Biol Chem. 2015; 290: 2831–2841. DOI: 10.1074/jbc.M114.591420
  92. 92. Wang S, Zhang X, Ju Y: MicroRNA-146a feedback suppresses T cell immune function by targeting Stat1 in patients with chronic hepatitis B. J Immunol. 2013; 191: 293–301. DOI: 10.4049/jimmunol.1202100
  93. 93. Taganov KD, Boldin MP, Chang KJ: 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. 2006; 103: 12481–12486. DOI: 10.1073/pnas.0605298103
  94. 94. Chang YL, Ho BC, Sher S: miR-146a and miR-370 coordinate enterovirus 71-induced cell apoptosis through targeting SOS1 and GADD45beta. Cell Microbiol. 2015; 17: 802–818. DOI: 10.1111/cmi.12401
  95. 95. Hou ZH, Han QJ, Zhang C: miR146a impairs the IFN-induced anti-HBV immune response by downregulating STAT1 in hepatocytes. Liver Int. 2014; 34: 58–68. DOI: 10.1111/liv.12244
  96. 96. Hou J, Wang P, Lin L: MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J Immunol. 2009; 183: 2150–2158. DOI: 10.4049/jimmunol.0900707
  97. 97. Wu S, He L, Li Y: miR-146a facilitates replication of dengue virus by dampening interferon induction by targeting TRAF6. J Infect. 2013; 67: 329–341. DOI: 10.1016/j.jinf.2013.05.003
  98. 98. Ho BC, Yu IS, Lu LF: Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon. Nat Commun. 2014; 5: 3344. DOI: 10.1038/ncomms4344
  99. 99. Sun X, Zhang J, Hou Z: miR-146a is directly regulated by STAT3 in human hepatocellular carcinoma cells and involved in anti-tumor immune suppression. Cell Cycle. 2015; 14: 243–252. DOI: 10.4161/15384101.2014.977112
  100. 100. Lind EF, Elford AR, Ohashi PS: Micro-RNA 155 is required for optimal CD8+ T cell responses to acute viral and intracellular bacterial challenges. J Immunol. 2013; 190: 1210–1216. DOI: 10.4049/jimmunol.1202700
  101. 101. Iwai H, Funatogawa K, Matsumura K: MicroRNA-155 knockout mice are susceptible to mycobacterium tuberculosis infection. Tuberculosis (Edinb). 2015; 95: 246–250. DOI: 10.1016/j.tube.2015.03.006
  102. 102. Ruelas DS, Chan JK, Oh E: MicroRNA-155 Reinforces HIV Latency. J Biol Chem. 2015; 290: 13736–13748. DOI: 10.1074/jbc.M115.641837
  103. 103. Cheng YQ, Ren JP, Zhao J: MicroRNA-155 regulates interferon-gamma production in natural killer cells via Tim-3 signalling in chronic hepatitis C virus infection. Immunology. 2015; 145: 485–497. DOI: 10.1111/imm.12463
  104. 104. Hentzschel F, Hammerschmidt-Kamper C, Borner K: AAV8-mediated in vivo overexpression of miR-155 enhances the protective capacity of genetically attenuated malarial parasites. Mol Ther. 2014; 22: 2130–2141. DOI: 10.1038/mt.2014.172
  105. 105. Bauernfeind F, Rieger A, Schildberg FA: NLRP3 inflammasome activity is negatively controlled by miR-223. J Immunol. 2012; 189: 4175–4181. DOI: 10.4049/jimmunol.1201516
  106. 106. Haneklaus M, Gerlic M, Kurowska-Stolarska M: Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1beta production. J Immunol. 2012; 189: 3795-3799. DOI: 10.4049/jimmunol.1200312
  107. 107. Stern-Ginossar N, Elefant N, Zimmermann A: Host immune system gene targeting by a viral miRNA. Science. 2007; 317: 376–381. DOI: 10.1126/science.1140956
  108. 108. Nachmani D, Stern-Ginossar N, Sarid R: Diverse herpesvirus microRNAs target the stress-induced immune ligand MICB to escape recognition by natural killer cells. Cell Host Microbe. 2009; 5: 376–385. DOI: 10.1016/j.chom.2009.03.003
  109. 109. Bauman Y, Nachmani D, Vitenshtein A: An identical miRNA of the human JC and BK polyoma viruses targets the stress-induced ligand ULBP3 to escape immune elimination. Cell Host Microbe. 2011; 9: 93–102. DOI: 10.1016/j.chom.2011.01.008
  110. 110. Nachmani D, Lankry D, Wolf DG: The human cytomegalovirus microRNA miR-UL112 acts synergistically with a cellular microRNA to escape immune elimination. Nat Immunol. 2010; 11: 806–813. DOI: 10.1038/ni.1916
  111. 111. Jasinski-Bergner S, Stoehr C, Bukur J: Clinical relevance of miR-mediated HLA-G regulation and the associated immune cell infiltration in renal cell carcinoma. Oncoimmunology. 2015; 4: e1008805. DOI: 10.1080/2162402X.2015.1008805
  112. 112. Zhu XM, Han T, Wang XH: Overexpression of miR-152 leads to reduced expression of human leukocyte antigen-G and increased natural killer cell mediated cytolysis in JEG-3 cells. Am J Obstet Gynecol. 2010; 202: 592 e591–597. DOI: 10.1016/j.ajog.2010.03.002
  113. 113. Vinuesa CG, Cook MC, Angelucci C: A RING-type ubiquitin ligase family member required to repress follicular helper T cells and autoimmunity. Nature. 2005; 435: 452–458. DOI: 10.1038/nature03555
  114. 114. Rasmussen TK, Andersen T, Bak RO: Overexpression of microRNA-155 increases IL-21 mediated STAT3 signaling and IL-21 production in systemic lupus erythematosus. Arthritis Res Ther. 2015; 17: 154. DOI: 10.1186/s13075-015-0660-z
  115. 115. Xin Q, Li J, Dang J: miR-155 Deficiency Ameliorates Autoimmune Inflammation of Systemic Lupus Erythematosus by Targeting S1pr1 in Faslpr/lpr Mice. J Immunol. 2015; 194: 5437–5445. DOI: 10.4049/jimmunol.1403028
  116. 116. Kalis M, Bolmeson C, Esguerra JL: Beta-cell specific deletion of Dicer1 leads to defective insulin secretion and diabetes mellitus. PLoS One. 2011; 6: e29166. DOI: 10.1371/journal.pone.0029166
  117. 117. Salama A, Fichou N, Allard M: MicroRNA-29b modulates innate and antigen-specific immune responses in mouse models of autoimmunity. PLoS One. 2014; 9: e106153. DOI: 10.1371/journal.pone.0106153
  118. 118. Yang M, Ye L, Wang B: Decreased miR-146 expression in peripheral blood mononuclear cells is correlated with ongoing islet autoimmunity in type 1 diabetes patients 1miR-146. J Diabetes. 2015; 7: 158–165. DOI: 10.1111/1753-0407.12163
  119. 119. Lu LF, Boldin MP, Chaudhry A: Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell. 2010; 142: 914–929. DOI: 10.1016/j.cell.2010.08.012
  120. 120. Banerjee A, Schambach F, DeJong CS: Micro-RNA-155 inhibits IFN-gamma signaling in CD4+ T cells. Eur J Immunol. 2010; 40: 225–231. DOI: 10.1002/eji.200939381
  121. 121. Okoye IS, Czieso S, Ktistaki E: Transcriptomics identified a critical role for Th2 cell-intrinsic miR-155 in mediating allergy and antihelminth immunity. Proc Natl Acad Sci U S A. 2014; 111: E3081–3090. DOI: 10.1073/pnas.1406322111
  122. 122. Zhou Q, Haupt S, Kreuzer JT: Decreased expression of miR-146a and miR-155 contributes to an abnormal Treg phenotype in patients with rheumatoid arthritis. Ann Rheum Dis. 2015; 74: 1265–1274. DOI: 10.1136/annrheumdis-2013-204377
  123. 123. Ivanov II, McKenzie BS, Zhou L: The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006; 126: 1121–1133. DOI: 10.1016/j.cell.2006.07.035
  124. 124. Du C, Liu C, Kang J: MicroRNA miR-326 regulates TH-17 differentiation and is associated with the pathogenesis of multiple sclerosis. Nat Immunol. 2009; 10: 1252–1259. DOI: 10.1038/ni.1798
  125. 125. Moore CS, Rao VT, Durafourt BA: miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization. Ann Neurol. 2013; 74: 709–720. DOI: 10.1002/ana.23967

Written By

Zhaohua Hou, Qiuju Han, Cai Zhang and Jian Zhang

Submitted: 02 May 2015 Reviewed: 21 September 2015 Published: 06 April 2016