Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2018 Sep 21;286(9):1624–1637. doi: 10.1111/febs.14654

Advances and challenges in targeting IRF5, a key regulator of inflammation

Hannah Almuttaqi 1, Irina A Udalova 1,
PMCID: PMC6563445  PMID: 30199605

Abstract

Interferon regulatory factor 5 (IRF5) belongs to a family of transcription factors, originally implicated in antiviral responses and interferon production. However, studies conducted in different laboratories over the last decade have placed IRF5 as a central regulator of the inflammatory response. It has become clear that IRF5 contributes to the pathogenesis of many inflammatory and autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease and systemic lupus erythematosus. Given the role of IRF5 in physiology and disease, IRF5 represents a potential therapeutic target. However, despite a significant interest from the pharmaceutical industry, inhibitors that interfere with the IRF5 pathway remain elusive. Here, we review the advances made by various studies in targeting multiple steps of signalling leading to IRF5 activation with their therapeutic potential, and the possible complications of such strategies are discussed.

Keywords: autoimmune diseases, inflammation, interferon regulatory factor 5, therapy, transcription factor


Abbreviations

DBD

DNA‐binding domain

IAD

IRF‐associated domain

IBD

inflammatory bowel disease

IFN

interferon

iPSdMs

induced pluripotent stem cells

IRF5

interferon regulatory factor 5

ISREs

interferon‐stimulated response elements

LNA

locked nucleic acid

MHC

major histocompatibility complex

MI

myocardial infarction

MS

multiple sclerosis

ODNs

oligonucleotides

RA

rheumatoid arthritis

siRNA

small interfering RNA

SLE

systemic lupus erythematosus

TLR

toll‐like receptor

Introduction

The interferon regulatory factor (IRF) family of transcription factors were first characterized for mediating antiviral responses and type I interferon (IFN) production. They have since been demonstrated to play diverse functions in apoptosis, the cell cycle, oncogenesis and gene regulation in response to pathogen‐derived signals 1. The mammalian IRF family comprise nine members (IRFs 1–9) with conserved multidomains 2. The N‐terminal DNA‐binding domain (DBD) recognizes a core DNA sequence within interferon‐stimulated response elements 3 and the C‐terminal IRF‐associated domain (IAD) mediates protein–protein interactions between IRFs and other proteins to form transcriptional complexes 4.

Among the IRF family members, interferon regulatory factor 5 (IRF5) plays a central role in inflammation. IRF5 mediates induction of proinflammatory cytokines such as interleukin‐6 (IL‐6), IL‐12, IL‐23 and tumour‐necrosis factor‐alpha (TNF‐α) 5, 6, and its recruitment to promoters of inflammatory genes is assisted by the NF‐kB p65 subunit RelA 7. IRF5 is a key factor in defining the inflammatory macrophage phenotype. IRF5 is highly expressed in not only monocytes and macrophages but also in B cells and dendritic cells. Its expression in macrophages can be upregulated in response to the inflammatory environment and, in particular, to the stimulation with GM‐CSF and IFN‐gamma 6, 8.

In humans, IRF5 exists as multiple distinct isoforms (V1–V11) that are generated by alternatively spliced transcripts. Each isoform utilizes one of three promoters, giving rise to transcripts containing exon 1A, 1B or 1C, and displays differences in composition of other exons. These isoforms show cell‐type specific expression, subcellular localization and function 9. Multiple GWAS studies have identified polymorphisms in the Irf5 locus, leading to expression of alternatively spliced isoforms of Irf5 that are associated with risk of autoimmune diseases such as systemic lupus erythematosus (SLE) in humans 10, 11. For example, IRF5 isoforms generated from exon 1B (v2, v9, v10) are strongly linked to overexpression of IRF5 and to susceptibility to SLE, whereas elevated expression of IRF5 in the absence of exon 1B does not confer risk 10. Several IRF5 isoforms including isoform v2 contain splicing variations in and around exon 6, which encodes for a proline‐, glutamic acid‐, serine‐ and threonine‐rich (PEST) domain thought to be important for protein stability in the IRF family of proteins 12.

IRF5 as an attractive therapeutic target

There is overwhelming evidence that IRF5 plays a key role in numerous conditions based on the phenotype of IRF5 knockout mice in disease models. Mice lacking Irf5 are resistant to lethal endotoxin‐induced shock with reduced expression of proinflammatory cytokines 5, 13. Irf5‐deficient mice are also protected from arthritis and lupus in murine models of inflammatory arthritis and pristane‐induced lupus respectively 14, 15, 16. For example, Irf5 −/− mice exhibit reduced knee swelling when challenged with methylated BSA in the acute antigen‐induced arthritis murine model 16. Irf5 −/− mice demonstrate impaired expression of IL‐12b and enhanced expression of IL‐10 in their affected joints 8. Moreover, proinflammatory monocyte‐derived macrophages with IRF5 expression are specifically detected in the affected knees. Somewhat contradictory to these results was a report describing no differences between WT and Irf5 −/− mice in a model of collagen‐induced arthritis (CIA) 17. The mice on C57BL/6 background express the b haplotype of the major histocompatibility complex (MHC) class II and need MHC class II A(q) to develop CIA dependent on autoreactive T cells 18, 19. When C57BL/6 Irf5 −/− mice were crossed with the strain carrying MHC class II Aq, a significant reduction in the number of mice developing the pathology was observed (H. Eames, unpublished data), suggesting that the conclusion of no role for IRF5 in the CIA induced pathologies needs to be revisited.

Increased IRF5 levels are associated with better prognosis of pulmonary disease 20. In murine asthma models with house dust mite (HDM) exposure, Irf5 −/− demonstrate impaired lung function and extracellular matrix deposition, but mice overexpressing IRF5 were protected from allergic inflammation 20. Recent studies have also highlighted important contributions of IRF5 to neuropathic pain 21, vascular diseases 22, 23, 24, obesity 25 and hepatic and skin fibrosis 26, 27. For example, Irf5 −/− mice on a high fat display beneficial expansion of subcutaneous adipose tissue and retain their insulin sensitivity 25. Other reported roles for IRF5 include cell cycle arrest and apoptosis 28, microbial infection 29, 30, and glycolysis 31. Several studies have also found that autoantibodies against IRF5 were able to cross react with homologous peptides from Mycobacterium avium subsp. Paratuberculosis and Epstein–Barr virus 32, 33, 34. Furthermore, antibodies against these peptides were significantly higher in the cerebrospinal fluid and serum of multiple sclerosis (MS) patients due to molecular mimicry 32, 33.

From these studies and given the fact that in humans IRF5 gene polymorphisms related to higher IRF5 expression 10, 35, 36, 37, 38, 39 have been associated with susceptibility to inflammatory and autoimmune diseases including rheumatoid arthritis (RA), inflammatory bowel disease, SLE, MS, and Sjörgens syndrome, IRF5 has emerged as an attractive target for therapeutic intervention.

A benefit of targeting IRF5, is that it acts in a cell‐type and activity‐specific manner. Both IRF5 and NF‐kB transcription factors are essential for the induction of proinflammatory genes 7. Due to the broader functional activities of NF‐kB and its ubiquitous nature, there are concerns of the detrimental effects that might result from blockage of NF‐kB activity. In comparison, targeting IRF5 may well be more beneficial and offer less adverse effects to general cell function.

Despite the strong rationale for targeting IRF5, inhibitors that interfere with the IRF5‐specific pathway remain elusive. This review provides an overview of some of the approaches currently used to target IRF5 and their potential as a therapeutic agent.

Strategies in targeting IRF5

Given the complexity of IRF5 signalling, finding an effective and feasible way of targeting IRF5 function presents a challenge. Strategies for modulation of IRF5 activity and expression which will be discussed in further detail and include (a) Modulating IRF5 expression, (b) Interfering with the post‐translational modifications that modulate IRF5 function including phosphorylation and ubiquitination, and (c) Interfering with IRF5 association with protein partners, disrupting dimer formation or DNA binding. This review will describe the different approaches, the mechanism by which they affect IRF5 levels, considerations for their uses in therapeutic settings, and discuss their advantages and limitations (Table 1).

Table 1.

Summary of strategies to study and modulate interferon regulatory factor 5 (IRF5) function

Mode of action Class of drug Advantages Disadvantages
Inhibition of IRF5 gene expression Small interfering RNA Selectivity Off‐target effects
Delivery
Inhibition of IRF5 gene expression CRISPR/Cas9 Easy design
High efficiency
Off‐target effects
IRF5 overexpression Adenoviral vector High transduction efficiency
High levels of transgene expression
Transient transfer and expression
Disruption of IRF5–protein interactions Peptide inhibitors Selectivity Low proteolytic stability
Low conformational stability
Disruption of IRF5–DNA interactions Decoy oligonucleotides Selectivity Delivery
Inhibition of IRF5 kinases Kinase inhibitors Targetable by small molecule inhibitors May interfere with other signalling pathways

Modulating IRF5 expression

Several genetic manipulation technologies exist with the potential to control IRF5 expression levels and help reduce the severity of IRF5‐affected conditions (Fig. 1). Strategies for reducing IRF5 expression include small interfering RNA (siRNA) and locked nucleic acid (LNA) oligonucleotides (ODNs) for genetic knockdowns, or clustered regularly interspaced short palindromic repeats‐associated Cas9 nuclease (CRISPR‐Cas9) technology for genetic knockouts. These strategies are particularly useful for mechanistic and target validation experiments that will provide a platform for the development of specific therapeutic agents targeted towards IRF5, but could also be explored as therapeutic strategies themselves.

Figure 1.

Figure 1

Strategies to modulate interferon regulatory factor 5 (IRF5) expression. (A) Small interfering RNA (siRNA)‐mediated therapy using lipidoid nanoparticles (LNP) delivery to modulate the expression of IRF5 through RNA interference (RNAi). (B) IRF5 knockouts in human‐induced pluripotent stem cell‐derived macrophages using the CRISPR/Cas9 system. (C) Top – splice variants of IRF5 including isoforms v1/v2/v3/v5. Each isoform includes exon 1A, 1B or 1C. Some isoforms have 30/48 nucleotide deletions in exon 6. Bottom – upon toll‐like receptor 7 (TLR7) stimulation TRIpartite motif 21 (TRIM21) regulates IRF5 stability and activity in an isoform‐specific manner. (D) IRF5 gene delivery with an adenoviral vector to increase levels of IRF5.

One approach to target IRF5 is to inhibit the expression of IRF5 using siRNA that are specific for sequences in the irf5 mRNA (Fig. 1A). An in vivo study in an animal model of myocardial infarction (MI), demonstrated the use of siRNA to suppress IRF5 expression in cardiac macrophages 40. Intravenous administration of siRNA packaged lipidoid nanoparticles (LNP) in mice, attenuated M1 macrophage polarization with a decrease in M1 macrophage‐associated genes without increasing M2 genes. Furthermore, siRNA treatment ameliorated inflammation following MI, and improved infarct healing. Given that the delivery of siRNA‐encapsulated LNPs has proven safe and effective in humans, RNAi drugs could be a therapeutic prospect 41, 42, 43. Despite this, a number of hurdles must be overcome before RNAi drugs can be therapeutically used including off‐target effects, toxicity and delivery. To date, RNAi‐based therapeutics to modulate the expression of target proteins are currently at the clinical trial stages 44.

Related to siRNA‐mediated interference with target mRNA expression is the use of LNA‐based antisense ODNs. Although LNA to IRF5 is yet to be developed, of specific interest to this review is an attempt to target another transcription factor, HIF‐1α, in patients with refractory solid tumours which resulted in preliminary proof‐of‐concept results 45. The use of LNA antisense ODNs in human clinical studies have demonstrated proof‐of‐pharmacology for a number of targets 45, 46, 47, but have shown also some side effects 46.

A recent study utilized CRISPR/Cas9 to study the role of IRF5 in Chlamydia infection by generating IRF5 knockout mutations in macrophages derived from human‐induced pluripotent stem cells (iPSdMs; Fig. 1B) 48. IRF5 iPSdM knockouts were more susceptible to Chlamydia infection, highlighting a role for IRF5 in limiting Chlamydia infection. Although the CRISPR‐Cas9 approach was used for mechanistic validation, the present study opens the possibility of achieving therapeutic genome editing of IRF5 in human macrophages. While still in its infancy, therapeutics using CRISPR/Cas9 are being developed. For example, genome editing by CRISPR to delete the PD‐1 gene in T cells from lung cancer patients is reportedly underway 49.

The E3‐ubiquitin ligase TRIM21 has been demonstrated to target IRF5 for degradation and may potentially present another strategy based on modulation of IRF5 levels 50, 51. Following toll‐like receptor 7 (TLR7) activation, TRIM21 mediates degradation of IRF5 in an isoform specific manner, that is, it targets isoforms v1/v5 but isoforms v2/v3 are resistant (Fig. 1C). Increasing E3 activity of TRIM21 in macrophages may help to reducing the level of IRF5 in some but not all patients, as it will be ineffective against the isoform v2 linked to overexpression of IRF5 and to susceptibility to SLE 52.

Interferon regulatory factor 5 overexpression in the airway lumen was shown to enhance immune responses in the lung following allergen exposure 20. In this study, an adenoviral vector expressing IRF5 (AdIRF5) was used to increase IRF5 expression in the lungs (Fig. 1D). In response to HDM, IRF5 overexpression ameliorated airway hyper‐responsiveness, reduced mucus production and reduced goblet cell hyperplasia. Moreover, overexpression of IRF5 resulted in diminished production of type 2 cytokines and decreased eosinophilia. Thus, localized adenoviral delivery of IRF5 (e.g., via intranasal administration 20) to enhance IRF5 expression in macrophages and stimulate their immune potential could be a promising therapeutic strategy in eosinophilic asthma and would circumvent a concern related to the systemic administration of adenoviruses which results in hepatic tropism independent of the primary receptors 53.

Adenovirus‐based vectors are a widely used therapeutic platform for gene delivery, especially in the field of cancer gene therapy, where they demonstrate a good safety record and a great promise in preclinical studies 54. IRF5 overexpression could represent a promising approach to enhance cancer therapies through the reprogramming of tumour‐infiltrating macrophages, which are primarily M2‐polarized.

Inhibiting IRF5 post‐translational modifications

Other than strategies to module IRF5 expression, for example, blocking the enzymatic action of regulators of IRF5 signalling including kinases and E3 ubiquitin ligases, could provide attractive targets for therapeutic intervention. The regulation of IRF5 has been reviewed in detail recently 55, and involves various signalling cascades that converge on IRF5 activation by ubiquitination, and phosphorylation (Fig. 2).

Figure 2.

Figure 2

A schematic representation of the proposed mechanisms of IRF5 activation. The stimulation of toll‐like receptors (TLRs) on cell membranes by pathogen‐derived products or RIG‐I‐like receptors (RLRs) in the cytosol by viral PAMPs trigger signalling cascades. IRAK4 regulates the TLR/MyD88 response via TAK1 and IKKβ. Interferon regulatory factor 5 (IRF5) is activated from its latent state by post‐translational modifications that include ubiquitination by the ubiquitin ligase TRAF6 and phosphorylation by IKKβ. These modifications trigger IRF5 homodimerization, translocation to the nucleus and binding to gene promoters along with coactivator proteins (p300). Alternatively, DNA damage agents (CPT) can induce IRF5 phosphorylation and transcriptional activation. Another model proposes that the Dectin‐1‐induced IFN‐B production is mediated by a Syk‐Card9‐IRF5‐dependent pathway. In another model, IRF5 interacts with the COP9 signalosome (CSN). Upon stimulation with agonists such as TRAIL, an unknown kinase phosphorylates IRF5 which leads to its dissociation from CSN. Loss of the CSN–IRF5 interaction leads to K48‐linked ubiquitination of IRF5 and degradation by the ubiquitin–proteasome pathway. Some activated IRF5 migrates to the nucleus and activates target genes. An alternative model suggests that upon NOD2 stimulation IRF5 associates with RIP2, IRAK1, TRAF6 and IKKb to increase glycolysis through Akt2 activation.

Probably most explored is the activation of IRF5 via the TLR‐MyD88 pathway 56. In this pathway, TLR activation by TLR ligands (LPS, R848, and CpG) induces a signalling cascade whereby IRF5 binds to TRAF6 and the adaptor protein MyD88 5, 57. Following TLR7/8 stimulation, IRAK4 kinase acts through TAK1 and IKKβ to activate IRF5 in human monocytes 58. A recent study found that Lyn kinase binds to and inhibits the activity of IRF5 in the TLR‐MyD88 pathway by blocking the post‐translational modification step in a kinase‐independent manner 59. Other modes of IRF5 activation include viral infection by Newcastle disease virus (NDV), vesicular stomatis virus (VSV) and herpes simplex virus type 1 (HSV‐1) 13, 60, 61, 62, as well as DNA‐damaging agents (CPT) 63.

Another study demonstrated a role for IRF5 in induction of IFN‐β following fungal infection by the pathogen Candida albicans, whereby the C‐type lectin receptor Dectin‐1 activation of IRF5 was dependent on the tyrosine kinase Syk and the adaptor protein Card9 30. An alternative model suggests that the COP9 signalosome (CSN) interacts with and stabilizes IRF5 64. Stimulation of the death receptor by TRAIL ligand leads to phosphorylation of the complex by an unknown kinase and IRF5 degradation through a ubiquitin–proteasome pathway. Some of the phosphorylated IRF5 migrates to the nucleus and transactivates target genes. IRF5 has been shown to associate with RIP2, IRAK1 and TRAF6 in human monocyte‐derived macrophages (hMDMs) 31. Each of these molecules along with IKKβ is required for NOD2‐induced IRF5 serine phosphorylation, which increases glycolysis through Akt2 activation 31.

During IRF5 signalling, IRF5 is subjected to TRAF6‐mediated K63‐linked ubiquitination 57. However, to date, there is some controversy in the literature on the functional consequence of this ubiquitination. Mutagenesis studies suggest ubiquitination of IRF5 is essential for nuclear translocation and target gene regulation 57. However, several lines of evidence using double KK/RR mutants, and the A20‐K63 ubiquitination enzyme have indicated that ubiquitination is not essential for IRF5 transcriptional activity 65. Instead, the carboxyl terminal phosphorylation of IRF5 is thought to be the critical modification that determines IRF5 transcriptional activity and thus a better therapeutic strategy is to target the kinase responsible for IRF5 phosphorylation.

Based on the crystal structure of the transactivation domain of pseudophosphorylated human IRF‐5, phosphorylation is thought to induce a conformational change in a C‐terminal autoinhibitory region, to enable dimerization 66. Phosphorylation sites towards the C‐terminal serine‐rich region of IRF5 include S425, S427, S430 and S436 (human isoform v3/v4); phosphorylation of S436 contributes to the stabilization of the activated dimer, whereas phosphorylation of S425, S427 and S430 are essential for release of the C‐terminal autoinhibitory conformation (Fig. 3A) 60, 66. Mass spectrometry analysis confirmed phosphorylation of S425 and S436 and revealed their contribution to IRF5 activity 65. Phosphomimetic S425, and S436D displayed an increase in IL‐12p40 promoter‐controlled luciferase activity. In contrast, the loss of function mutation with S436A reduced RIP2‐induced activation of the luciferase reporter, and the double mutant S427A, S436A also failed to induce promoter activity in cells stimulated with peptidoglycan (PGN) or muramyl dipeptide (MDP). Together, these studies indicate that phosphorylation of carboxyl serine residues are essential for IRF5 function, and provide a rationale for targeting the kinases involved.

Figure 3.

Figure 3

Modulation of interferon regulatory factor 5 (IRF5) via post‐translational modification. (A) Schematics of putative IRF5 phosphorylation and ubiquitination sites (human isoform v3/v4) and their positions with respect to functional domains. Suggested targets and functions of mapped sites are also shown (B) Inhibition of IRF5 phosphorylation through the use of kinase inhibitors. Kinases reported to phosphorylate IRF5 include IRAK4, TAK1, RIP2, IKKα, IKKβ, IKKε and TBK1.

Kinase inhibitors

Targeting kinases in a signalling pathway with small molecule inhibitors is typically the most straightforward approach to block a protein. Kinases have been established as an important drug target. Despite a high degree of similarity in their catalytic core with the ATP‐binding pocket, kinases are amendable to blockade by small molecule agents with high selectivity. Moreover, high‐throughput kinase inhibitor libraries can identify potential candidate kinases. Several kinases including RIP2 from the receptor‐interacting protein (RIP) kinase family, and TAK1, IKKα, IKKβ, IKKε and TBK1 from the IKK family, are thought to phosphorylate IRF5 (Fig. 3B) 56, 67, 68. Phosphorylation of IRF5 by TBK1 or IKKε fail to induce nuclear translocation of IRF5 69. Furthermore, macrophages from TBK1‐deficient mice produce normal levels of proinflammatory cytokines in response to LPS 70. Thus, these kinases can be excluded as suitable IRF5 candidate kinases. Stimulation of the NOD2 intracellular receptor with MDP or PGN, can induce IFNB expression via IRF5 and RIP2 71. Using overexpression systems, a later study identified S436 on IRF5 as a RIP2 target residue 65. More recently, endogenous IRF5 serine phosphorylation in response to MDP was shown to be dependent on RIP2 and IKKβ, where IRF5 associates with RIP2, IRAK1 and TRAF6 31. IRF5 and IKKβ cross‐regulate each other's phosphorylation, but the kinase in the IRF5 complex responsible for IKKβ activation is unknown in this model 31. Since IKKB phosphorylation of IRF5 is known, it could be an interesting pharmacological target.

Earlier studies based on the use of IKKβ loss of function mutations, indicate that IKKβ activity is required for the activation of IRF5. Two groups have shown that phosphorylation by IKKβ on S436 induces IRF5 nuclear translocation and an IFN response 72, 73. Numerous inhibitors which are specific to IKKβ, and do not inhibit other IKK‐related kinases are available, but as IKKβ is involved in the activation of several targets its inhibition is unlikely to result in the specific inhibition of IRF5 function. Besides, the well documented role of IKKβ in phosphorylating IkB in the NF‐kB pathway, other reported substrates include forkhead transcription factor (FOXO3a), 14‐3‐3b, insulin receptor substrate 1 (ISR1) and docking protein 1 (DOK1) 74, 75, 76, 77. IKKα is another kinase from the IKK family reported to phosphorylate IRF5. Unlike IKKβ, IKKα‐mediated phosphorylation of IRF5 negatively regulates IRF5 in the MyD88‐IRF5 pathway 68. IKKα could represent a therapeutic target in conditions with low IRF5 activity. However, no potent commercially available IKKα‐specific inhibitors have been reported to date. Another reported kinase involved in IRF5 activation is TGF‐B‐activated kinase 1 (TAK1). Upstream of IKKβ, TAK1 can induce IFN‐B and IL‐12 in response to Staphylococcus aureus RNA 78. Several inhibitors for TAK1 reported to inhibit IRF5 include NG‐25, and 5Z‐7‐oxozeaenol, which both block nuclear translocation of IRF5 in monoyctes stimulated with TLR8 ligands 78. However, as TAK1 plays an essential role in several signalling pathways including the MAPK cascade and IKKs 79, inhibition of TAK1 might not offer the best approach for inhibiting IRF5 function.

In a recent report, IRAK4 was identified as a kinase that activates IRF5 via an IRAK4‐TAK1‐IKKβ axis in human monocytes 58. Chemical inhibition of IRAK4 with a potent and selective IRAK4 inhibitor blocked IRF5 nuclear translocation and IRF5 transcriptional activity at the promoters of inflammatory cytokines, such as IL‐1, Il‐6, TNF, in human monocytes. Despite blocking IKKβ activation, the IRAK4 inhibitor had no effect on NF‐kB nuclear translocation and transcriptional activity. In their proposed model, the kinase activity of IRAK4 kinase regulates the IRF5 pathway, whereas the scaffolding function of IRAK4 regulates the NF‐kB pathway. The requirement of IRAK4 kinase activity in regulating IRF5 supports the rationale for IRAK4 inhibitor use. Inhibition with IRAK4‐specific inhibitors could be sufficient to block IRF5 activity without having an impact on NF‐kB activation. This is a promising study and future studies will have to establish the efficiency of the inhibitor in disease mouse models. Mice harbouring the IRAK4 kinase‐dead mutant are protected from disease models of RA 80, emphasizing IRAK4 as an attractive therapeutic target in the context of IRF5.

Interfering with IRF5‐interacting partners

Transcription factors are traditionally considered as ‘non‐druggable’ targets. Blocking their activity with small molecules presents a challenging task, given the large surface areas for binding proteins or DNA, the lack of hydrophobic pockets and the absence of enzymatic activity 81. Despite these obstacles, therapeutic modulation of transcription factor activity has been well documented by interfering with their ability to bind to DNA, partner proteins or their ability to dimerize. For example, the small molecule agent Nutlin can disrupt the p53–MDM2 interaction, thereby increasing p53 levels and p53 target gene expression 82. Another example is the MAML1‐derived stapled peptide which disrupts the NOTCH transcription complex and represses Notch transcriptional function 83.

One such method for blocking protein–protein interactions is the use of peptide inhibitors. These consist of short amino acid sequences that competitively inhibit the interaction between proteins. Several advantages that peptide inhibitors offer are their affordable synthesis, specificity, potency and activity 84. However, several drawbacks to consider are their low proteolytic stability and low conformational stability, which could decrease target binding 85. Despite these issues, ongoing efforts have been undertaken to develop effective peptide inhibitors to disrupt protein–protein interactions.

With regard to IRF5, peptide‐based inhibitors to decrease myocardial inflammation and fibrosis have been developed. In a murine model of systemic scleroderma, IRF5 bound to an apoAI mimic 4F, a peptide that inhibits myocardial inflammation 86. Hearts from 4F‐treated Tsk2/+ mice are less inflamed with a decrease in IRF5 expression, phosphoserine levels and nuclear localization. Whether these effects are mediated in part by the ability of 4F to directly bind to IRF5 or to other targets is unclear. It is also noted in the study that the changes observed may be due to differences in immune cell content. Nevertheless, the possible involvement of 4F binding to IRF5 warrants further exploration.

A more recent promising study developed a peptide inhibitor specifically designed from the peptide sequence of IRF5 23. The synthesized decoy peptide, termed IRF5D is derived from the IRF5 C‐terminal dimerization domain with the original sequence ELSWSADSIRLQISNPD replaced by the 17 amino acid‐long sequence ELDWDADDIRLQIDNPD. The aspartate substitution mimics activated IRF5, whereby IRF5D associates with IRF5 and prevents nuclear translocation of IRF5. In the study, Tsk/+ mice were used as a murine model of myocardial inflammation and fibrosis. IRF5D‐treated Tsk/+ mice produced reduced levels of ICAM‐1 and IRF5 expression in the heart, reduced leucocyte infiltration of the myocardium, improved endothelial vasodilation and IRF5 nuclear translocation was reduced in cultured Tsk/+ myocytes. Moreover, IRF5D treatment had no effect on endothelial cell (EC) proliferation and apoptosis. Thus, this study demonstrates that IRF5 is a druggable target and based on the published 3D structure of IRF5 66, it is possible to develop IRF5‐specific peptide inhibitors.

Alternatively, peptide inhibitors may target other interfaces such as the IRF5/RelA‐binding interaction. A previous study has demonstrated that IRF5 is recruited to the TNFα gene via interaction with NF‐kB subunit RelA 7, 87. The requirement for RelA for induction of proinflammatory genes with IRF5 supports the concept of developing inhibitors to block the IRF5–RelA interaction. This association has been mapped to the IAD of IRF5 and the dimerization domain (DD) of RelA 7.

Based on this information, along with the crystal structure of the IRF5 IAD 66, peptide inhibitors have been developed to sterically inhibit the IRF5/RelA interface but showed no significant inhibitory effects on IRF5‐dependent gene expression [88]. Further work to optimize peptide delivery and modifications to protect the peptide from degradation are worth pursuing to develop effective IRF5 inhibitors.

Other IRF5‐interacting proteins thought to be involved in IRF5 activation include CBP/p300 and histone deacetylases 67, 89, as well as signalling molecules KAP1, RIP2, TRAF6, MyD88, IRAK1 and IRAK4 5, 31, 57, 90 (Table 2). Thus, binding sites on IRF5 could be a potential peptide target and would require identifying the peptide sequences that are crucial for the IRF5–protein interactions. Another IRF5‐binding partner that has been shown to interact with and stabilize IRF5 is the CSN, which is thought to protect IRF5 from degradation by the ubiquitin–proteasome pathway 64. This interaction was mapped to the carboxyl and amino termini of IRF5. Disruption of the CSN–IRF5 interaction was shown to reduce IRF5 transactivation ability, whereas inhibition of IRF5 degradation by the proteasome enhances its transcriptional ability. Therefore, peptide inhibitors that target the CSN–IRF5 binding interaction could destabilize IRF5 and reduce IRF5 activity.

Table 2.

Summary of the interferon regulatory factor 5 (IRF5) interactome

Negative gene regulation Positive gene regulation IRF5 activation
KAP1 RelA
CBP/p300
Histone deacetylase
RIP2
TRAF6
MYD88
IRAK1
IRAK4
CSN
LYN

The recent identity of Lyn from the Src family of tyrosine kinases as an IRF5‐binding partner that negatively regulates IRF5 in the TLR‐MyD88‐IRF5 pathway could be an interesting target to increase IRF5 activity in conditions such as asthma 20, 59. In a kinase‐independent manner, Lyn inhibits IRF5 by directly binding to IRF5 and preventing post‐translational modifications. Lyn deficiency in mice suffering from SLE leads to IRF5 hyperactivation, but reducing IRF5 levels ameliorates the disease development. Like IRF5, Lyn is highly expressed in immune cells (DCs, monocytes, macrophages and B cells), making it an attractive therapeutic target in inflammatory conditions. Since Lyn association with IRF5 is not activation or phosphorylation dependent, available Lyn allosteric activators such as MLR‐1023 91 are unlikely to affect IRF5 function. Instead, peptides could potentially bind and regulate IRF5 activation. The use of Lyn peptide inhibitors have been reported to bind to Lyn and block its association with the Bc receptor 92. In vitro the Lyn peptide inhibitor blocked Lyn‐dependent functions of IL‐5 92, and MD‐2 (Myeloid differentiation) tyrosine phosphorylation 93. Moreover, the peptide inhibitor blocked eosinophil differentiation, survival and airway influx in a murine model of asthma 92, 94. Therefore, peptide inhibitors that block the Lyn–IRF5 interaction could be used in the context of asthma to enhance IRF5 function. The Lyn–IRF5 binding interaction has been mapped to Lyns unique and kinase domains (LYN UD‐IRF5 DBD and Lyn KD‐IRF5 IAD) 59. Based on the amino acid sequence of these domains, peptides could be developed and tested for their ability to increase IRF5 activity.

An alternative approach is the use of ODNs containing the consensus binding site of a transcription factor. Use of decoy ODNs to bind to IRF5 offers another means to inhibit its activity, and has been recently been investigated 95. IRF5 and an ODN termed MS19 share a consensus AAAG repeat‐binding site. In a mouse model of septic peritonitis, MS19 treatment prolonged survival and reduced expression of iNOS, IL‐6 and TNF‐a, suggesting that it might be useful in treating inflammatory conditions. In vitro treatment of LPS‐stimulated RAW264.7 macrophages, also reduced expression of iNOS, IL‐6, TNF‐a and IRF5, with reduced nuclear IRF5 levels. MS19 is thought to bind to IRF5, prevent its nuclear translocation and subsequent induction of target genes. MS19 as a ODN to interfere with IRF5 function could be a potential therapeutic, that warrants further investigation. An efficient means to deliver MS19 to target cells, and its specificity towards IRF5 should be considered in future studies.

Conclusions

There is overwhelming evidence that IRF5 plays a key role in physiology and disease, therefore dampening or enhancing IRF5 expression and activity provides new avenues for the development of therapeutic agents. Among the three strategies for modulation of IRF5 activity and expression discussed in this review, that is, (a) modulating IRF5 expression, (b) interfering with the post‐translational modifications of IRF5, and (c) interfering with IRF5 association with protein partners, the modulation of IRF5 levels using siRNA, CRISPR/Cas9, LNAs or perhaps nanobodies, single‐domain antibody fragments derived from camelid heavy‐chain antibodies that have been successfully utilized to target transcription factors 96, 97, 98, and adenoviruses, may take a long path in the development of new therapies themselves, but provide excellent opportunities for the identification of new points for therapeutic interference.

A more promising strategy at this stage is the use of specific inhibitors to the components of the IRF5‐signalling pathway, for example, kinase inhibitors. An ideal kinase inhibitor should block IRF5 function with limited effects on other signalling pathways. Thus, targeting IKKβ or TAK1 to block IRF5 activity are not ideal approaches, as these kinases target multiple pathways and may produce unwanted side effects. The selectivity of the IRAK4 inhibitor on the IRF5 pathway appears to carry a great potential. Future studies using high‐throughput libraries of kinase inhibitors may help identify novel kinases involved in IRF5 activation.

Another therapeutically promising approach is the use of peptides or small molecules to disrupt interactions with IRF5 cognate partners. The peptide inhibitors or small molecules that block the Lyn–IRF5 interaction could be used in the context of asthma to enhance IRF5 function. In fact, Lyn peptide inhibitors has already been shown to have a beneficial effect in a murine model of asthma 92, 94 and requires further assessment in human setting. Similarly, developing inhibitors or small molecules that could block the IRF5 interactions with RelA or the CSN, would be beneficial for suppressing the unwanted proinflammatory macrophage gene programme in inflammatory diseases.

Although there is still a long way to go, there is reason to believe that eventually some of the strategies discussed here will form the basis of effective therapies in IRF5‐associated conditions.

Acknowledgements

This work was supported by the Arthritis Research UK (PhD studentship number 20966) and Tripartite Immunometabolism Consortium [TrIC] – Novo Nordisk Foundation (grant number NNF15CC0018486). We thank Benedikt Kessler for critically reading the manuscript and his helpful suggestions.

References

  • 1. Tamura T, Yanai H, Savitsky D & Taniguchi T (2008) The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26, 535–584. [DOI] [PubMed] [Google Scholar]
  • 2. Mamane Y, Heylbroeck C, Génin P, Algarté M, Servant MJ, LePage C, DeLuca C, Kwon H, Lin R & Hiscott J (1999) Interferon regulatory factors: the next generation. Gene 237, 1–14. [DOI] [PubMed] [Google Scholar]
  • 3. Fujii Y, Shimizu T, Kusumoto M, Kyogoku Y, Taniguchi T & Hakoshima T (1999) Crystal structure of an IRF‐DNA complex reveals novel DNA recognition and cooperative binding to a tandem repeat of core sequences. EMBO J 18, 5028–5041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Taniguchi T, Ogasawara K, Takaoka A & Tanaka N (2001) IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 19, 623–655. [DOI] [PubMed] [Google Scholar]
  • 5. Takaoka A, Yanai H, Kondo S, Duncan G, Negishi H, Mizutani T, Kano S‐I, Honda K, Ohba Y, Mak TW et al (2005) Integral role of IRF‐5 in the gene induction programme activated by Toll‐like receptors. Nature 434, 243–249. [DOI] [PubMed] [Google Scholar]
  • 6. Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, Hussell T, Feldmann M & Udalova IA (2011) IRF5 promotes inflammatory macrophage polarization and TH1‐TH17 responses. Nat Immunol 12, 231–238. [DOI] [PubMed] [Google Scholar]
  • 7. Saliba DG, Heger A, Eames HL, Oikonomopoulos S, Teixeira A, Blazek K, Androulidaki A, Wong D, Goh FG, Weiss M et al (2014) IRF5:RelA interaction targets inflammatory genes in macrophages. Cell Rep 8, 1308–1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Weiss M, Blazek K, Byrne AJ, Perocheau DP & Udalova IA (2013) IRF5 is a specific marker of inflammatory macrophages in vivo. Mediators Inflamm 2013, 245804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Mancl ME, Hu G, Sangster‐Guity N, Olshalsky SL, Hoops K, Fitzgerald‐Bocarsly P, Pitha PM, Pinder K & Barnes BJ (2005) Two discrete promoters regulate the alternatively spliced human interferon regulatory factor‐5 isoforms. Multiple isoforms with distinct cell type‐specific expression, localization, regulation, and function. J Biol Chem 280, 21078–21090. [DOI] [PubMed] [Google Scholar]
  • 10. Graham RR, Kozyrev SV, Baechler EC, Reddy MVPL, Plenge RM, Bauer JW, Ortmann WA, Koeuth T, Escribano MFG, Collaborative Groups the A and S et al (2006) A common haplotype of interferon regulatory factor 5 (IRF5) regulates splicing and expression and is associated with increased risk of systemic lupus erythematosus. Nat Genet 38, 550–555. [DOI] [PubMed] [Google Scholar]
  • 11. Graham RR, Kyogoku C, Sigurdsson S, Vlasova IA, Davies LRL, Baechler EC, Plenge RM, Koeuth T, Ortmann WA, Hom G et al (2007) Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proc Natl Acad Sci USA 104, 6758–6763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hashmueli S, Gleit‐Kielmanowicz M, Meraro D, Azriel A, Melamed D & Levi B‐Z (2003) A truncated IFN‐regulatory factor‐8\IFN consensus sequence‐binding protein acts as dominant‐negative, interferes with endogenous protein±protein interactions and leads to apoptosis of immune cells. Int Immunol 15, 807–815. [DOI] [PubMed] [Google Scholar]
  • 13. Paun A, Reinert JT, Jiang Z, Medin C, Balkhi MY, Fitzgerald KA & Pitha PM (2008) Functional characterization of murine interferon regulatory factor 5 (IRF‐5) and its role in the innate antiviral response. J Biol Chem 283, 14295–14308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Duffau P, Menn‐Josephy H, Cuda CM, Dominguez S, Aprahamian TR, Watkins AA, Yasuda K, Monach P, Lafyatis R, Rice LM et al (2015) Promotion of inflammatory arthritis by interferon regulatory factor 5 in a mouse model. Arthritis Rheumatol 67, 3146–3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Feng D, Yang L, Bi X, Stone RC, Patel P & Barnes BJ (2012) Irf5‐deficient mice are protected from pristane‐induced lupus via increased Th2 cytokines and altered IgG class switching. Eur J Immunol 42, 1477–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Weiss M, Byrne AJ, Blazek K, Saliba DG, Pease JE, Perocheau D, Feldmann M & Udalova IA (2015) IRF5 controls both acute and chronic inflammation. Proc Natl Acad Sci USA 112, 11001–11006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Savitsky DA, Yanai H, Tamura T, Taniguchi T & Honda K (2010) Contribution of IRF5 in B cells to the development of murine SLE‐like disease through its transcriptional control of the IgG2a locus. Proc Natl Acad Sci USA 107, 10154–10159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Bäcklund J, Li C, Jansson E, Carlsen S, Merky P, Nandakumar K‐S, Haag S, Ytterberg J, Zubarev RA & Holmdahl R (2013) C57BL/6 mice need MHC class II Aq to develop collagen‐induced arthritis dependent on autoreactive T cells. Ann Rheum Dis 72, 1225–1232. [DOI] [PubMed] [Google Scholar]
  • 19. Inglis JJ, Criado G, Medghalchi M, Andrews M, Sandison A, Feldmann M & Williams RO (2007) Collagen‐induced arthritis in C57BL/6 mice is associated with a robust and sustained T‐cell response to type II collagen. Arthritis Res Ther 9, R113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Byrne AJ, Weiss M, Mathie SA, Walker SA, Eames HL, Saliba D, Lloyd CM & Udalova IA (2017) A critical role for IRF5 in regulating allergic airway inflammation. Mucosal Immunol 10, 716–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Masuda T, Iwamoto S, Yoshinaga R, Tozaki‐Saitoh H, Nishiyama A, Mak TW, Tamura T, Tsuda M & Inoue K (2014) Transcription factor IRF5 drives P2X4R+‐reactive microglia gating neuropathic pain. Nat Commun 5, 3771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Seneviratne AN, Edsfeldt A, Cole JE, Kassiteridi C, Swart M, Park I, Green P, Khoyratty T, Saliba D, Goddard ME et al (2017) Interferon regulatory factor 5 controls necrotic core formation in atherosclerotic lesions by impairing efferocytosis. Circulation 136, 1140–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Weihrauch D, Krolikowski JG, Jones DW, Zaman T, Bamkole O, Struve J, Pillai S, Pagel PS, Lohr NL & Pritchard KA (2016) An IRF5 decoy peptide reduces myocardial inflammation and fibrosis and improves endothelial cell function in tight‐skin mice. PLoS ONE 11, e0151999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Cai H, Yao Z & Li W (2017) IRF‐5 accelerates leukocyte adhesion to endothelial cells in ischemia‐reperfusion injury through regulating the transcription of VCAM‐1. Biochem Biophys Res Commun 492, 192–198. [DOI] [PubMed] [Google Scholar]
  • 25. Dalmas E, Toubal A, Alzaid F, Blazek K, Eames HL, Lebozec K, Pini M, Hainault I, Montastier E, Denis RGP et al (2015) Irf5 deficiency in macrophages promotes beneficial adipose tissue expansion and insulin sensitivity during obesity. Nat Med 21, 610–618. [DOI] [PubMed] [Google Scholar]
  • 26. Alzaid F, Lagadec F, Albuquerque M, Ballaire R, Orliaguet L, Hainault I, Blugeon C, Lemoine S, Lehuen A, Saliba DG et al (2016) IRF5 governs liver macrophage activation that promotes hepatic fibrosis in mice and humans. JCI Insight 1, e88689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Saigusa R, Asano Y, Taniguchi T, Yamashita T, Ichimura Y & Takahashi T (2015) Multifaceted contribution of the TLR4‐activated IRF5 transcription factor in systemic sclerosis. Proc Natl Acad Sci USA 112, 15136–15141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Barnes BJ, Kellum MJ, Pinder KE, Frisancho JA & Pitha PM (2003) Interferon regulatory factor 5, a novel mediator of cell cycle arrest and cell death. Cancer Res 63, 6424–6431. [PubMed] [Google Scholar]
  • 29. Paun A, Bankoti R, Joshi T, Pitha PM & Stäger S (2011) Critical role of IRF‐5 in the development of T helper 1 responses to Leishmania donovani infection. PLoS Pathog 7, e1001246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. del Fresno C, Soulat D, Roth S, Blazek K, Udalova I, Sancho D, Ruland J & Ardavín C (2013) Interferon‐β production via dectin‐1‐Syk‐IRF5 signaling in dendritic cells is crucial for immunity to C. albicans . Immunity 38, 1176–1186. [DOI] [PubMed] [Google Scholar]
  • 31. Hedl M, Yan J & Abraham C (2016) IRF5 and IRF5 disease‐risk variants increase glycolysis and human M1 macrophage polarization by regulating proximal signaling and Akt2 activation. Cell Rep 16, 2442–2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mameli G, Cocco E, Frau J, Marrosu MG & Sechi LA (2016) Epstein Barr virus and Mycobacterium avium subsp. paratuberculosis peptides are recognized in sera and cerebrospinal fluid of MS patients. Sci Rep 6, 22401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Cossu D, Mameli G, Galleri G, Cocco E, Masala S, Frau J, Marrosu MG, Manetti R & Sechi LA (2015) Human interferon regulatory factor 5 homologous epitopes of Epstein‐Barr virus and Mycobacterium avium subsp. paratuberculosis induce a specific humoral and cellular immune response in multiple sclerosis patients. Mult Scler 21, 984–995. [DOI] [PubMed] [Google Scholar]
  • 34. Bo M, Erre GL, Niegowska M, Piras M, Taras L, Longu MG, Passiu G & Sechi LA (2018) Interferon regulatory factor 5 is a potential target of autoimmune response triggered by Epstein‐Barr virus and Mycobacterium avium subsp. paratuberculosis in rheumatoid arthritis: investigating a mechanism of molecular mimicry. Clin Exp Rheumatol 36, 376–381. [PubMed] [Google Scholar]
  • 35. Dieguez‐Gonzalez R, Calaza M, Perez‐Pampin E, de la Serna AR, Fernandez‐Gutierrez B, Castañeda S, Largo R, Joven B, Narvaez J, Navarro F et al (2008) Association of interferon regulatory factor 5 haplotypes, similar to that found in systemic lupus erythematosus, in a large subgroup of patients with rheumatoid arthritis. Arthritis Rheum 58, 1264–1274. [DOI] [PubMed] [Google Scholar]
  • 36. Dideberg V, Kristjansdottir G, Milani L, Libioulle C, Sigurdsson S, Louis E, Wiman A‐C, Vermeire S, Rutgeerts P, Belaiche J et al (2007) An insertion‐deletion polymorphism in the interferon regulatory factor 5 (IRF5) gene confers risk of inflammatory bowel diseases. Hum Mol Genet 16, 3008–3016. [DOI] [PubMed] [Google Scholar]
  • 37. Eames HL, Corbin AL & Udalova IA (2016) Interferon regulatory factor 5 in human autoimmunity and murine models of autoimmune disease. Transl Res 167, 167–182. [DOI] [PubMed] [Google Scholar]
  • 38. Miceli‐Richard C, Comets E, Loiseau P, Puechal X, Hachulla E & Mariette X (2007) Association of an IRF5 gene functional polymorphism with Sjögren's syndrome. Arthritis Rheum 56, 3989–3994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Kristjansdottir G, Sandling JK, Bonetti A, Roos IM, Milani L, Wang C, Gustafsdottir SM, Sigurdsson S, Lundmark A, Tienari PJ et al (2008) Interferon regulatory factor 5 (IRF5) gene variants are associated with multiple sclerosis in three distinct populations. J Med Genet 45, 362–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Courties G, Heidt T, Sebas M, Iwamoto Y, Jeon D, Truelove J, Tricot B, Wojtkiewicz G, Dutta P, Sager HB et al (2014) In vivo silencing of the transcription factor IRF5 reprograms the macrophage phenotype and improves infarct healing. J Am Coll Cardiol 63, 1556–1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kanasty R, Dorkin JR, Vegas A & Anderson D (2013) Delivery materials for siRNA therapeutics. Nat Mater 12, 967–977. [DOI] [PubMed] [Google Scholar]
  • 42. Fitzgerald K, Frank‐Kamenetsky M, Shulga‐Morskaya S, Liebow A, Bettencourt BR, Sutherland JE, Hutabarat RM, Clausen VA, Karsten V, Cehelsky J et al (2014) Effect of an RNA interference drug on the synthesis of proprotein convertase subtilisin/kexin type 9 (PCSK9) and the concentration of serum LDL cholesterol in healthy volunteers: a randomised, single‐blind, placebo‐controlled, phase 1 trial. Lancet 383, 60–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, Perez J, Chiesa J, Warrington S, Tranter E et al (2013) Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med 369, 819–829. [DOI] [PubMed] [Google Scholar]
  • 44. Bobbin ML & Rossi JJ (2016) RNA interference (RNAi)‐based therapeutics: delivering on the promise? Annu Rev Pharmacol Toxicol 56, 103–122. [DOI] [PubMed] [Google Scholar]
  • 45. Jeong W, Rapisarda A, Park SR, Kinders RJ, Chen A, Melillo G, Turkbey B, Steinberg SM, Choyke P, Doroshow JH et al (2014) Pilot trial of EZN‐2968, an antisense oligonucleotide inhibitor of hypoxia‐inducible factor‐1 alpha (HIF‐1α), in patients with refractory solid tumors. Cancer Chemother Pharmacol 73, 343–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. van Poelgeest EP, Hodges MR, Moerland M, Tessier Y, Levin AA, Persson R, Lindholm MW, Dumong Erichsen K, Ørum H, Cohen AF et al (2015) Antisense‐mediated reduction of proprotein convertase subtilisin/kexin type 9 (PCSK9): a first‐in‐human randomized, placebo‐controlled trial. Br J Clin Pharmacol 80, 1350–1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. van der Ree MH, van der Meer AJ, de Bruijne J, Maan R, van Vliet A, Welzel TM, Zeuzem S, Lawitz EJ, Rodriguez‐Torres M, Kupcova V et al (2014) Long‐term safety and efficacy of microRNA‐targeted therapy in chronic hepatitis C patients. Antiviral Res 111, 53–59. [DOI] [PubMed] [Google Scholar]
  • 48. Yeung ATY, Hale C, Lee AH, Gill EE, Bushell W, Parry‐Smith D, Goulding D, Pickard D, Roumeliotis T, Choudhary J et al (2017) Exploiting induced pluripotent stem cell‐derived macrophages to unravel host factors influencing Chlamydia trachomatis pathogenesis. Nat Commun 8, 15013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Cyranoski D (2016) CRISPR gene‐editing tested in a person for the first time. Nature 539, 479. [DOI] [PubMed] [Google Scholar]
  • 50. Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjöstrand M, Eloranta M‐L, Ní Gabhann J, Winqvist O et al (2009) Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL‐23‐Th17 pathway. J Exp Med 206, 1661–1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Lazzari E, Korczeniewska J, Ní Gabhann J, Smith S, Barnes BJ & Jefferies CA (2014) TRIpartite motif 21 (TRIM21) differentially regulates the stability of interferon regulatory factor 5 (IRF5) isoforms. PLoS ONE 9, e103609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Stone RC, Du P, Feng D, Dhawan K, Rönnblom L, Eloranta M‐L, Donnelly R & Barnes BJ (2013) RNA‐seq for enrichment and analysis of IRF5 transcript expression in SLE. PLoS ONE 8, e54487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Yamamoto Y, Nagasato M, Yoshida T & Aoki K (2017) Recent advances in genetic modification of adenovirus vectors for cancer treatment. Cancer Sci 108, 831–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Beatty MS & Curiel DT (2012) Chapter two–adenovirus strategies for tissue‐specific targeting. Adv Cancer Res 115, 39–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Ryzhakov G, Eames HL & Udalova IA (2015) Activation and function of interferon regulatory factor 5. J Interf Cytok Res 35, 71–78. [DOI] [PubMed] [Google Scholar]
  • 56. Schoenemeyer A, Barnes BJ, Mancl ME, Latz E, Goutagny N, Pitha PM, Fitzgerald KA & Golenbock DT (2005) The interferon regulatory factor, IRF5, is a central mediator of toll‐like receptor 7 signaling. J Biol Chem 280, 17005–17012. [DOI] [PubMed] [Google Scholar]
  • 57. Balkhi MY, Fitzgerald KA & Pitha PM (2008) Functional regulation of MyD88‐activated interferon regulatory factor 5 by K63‐linked polyubiquitination. Mol Cell Biol 28, 7296–7308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Cushing L, Winkler A, Jelinsky SA, Lee K, Korver W, Hawtin R, Rao VR, Fleming M & Lin L‐L (2017) IRAK4 activity controls TLR inflammation through IRF5 in monocytes. IRAK4 kinase activity controls Toll‐like receptor induced inflammation through the transcription factor IRF5 in primary human monocytes. J Biol Chem 292, 18689–18698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ban T, Sato GR, Nishiyama A, Akiyama A, Takasuna M, Umehara M, Suzuki S, Ichino M, Matsunaga S, Kimura A et al (2016) Lyn kinase suppresses the transcriptional activity of IRF5 in the TLR‐MyD88 pathway to restrain the development of autoimmunity. Immunity 45, 319–332. [DOI] [PubMed] [Google Scholar]
  • 60. Barnes BJ, Kellum MJ, Field AE & Pitha PM (2002) Multiple regulatory domains of IRF‐5 control activation, cellular localization, and induction of chemokines that mediate recruitment of T lymphocytes. Mol Cell Biol 22, 5721–5740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Barnes BJ, Moore PA & Pitha PM (2001) Virus‐specific activation of a novel interferon regulatory factor, IRF‐5, results in the induction of distinct interferon alpha genes. J Biol Chem 276, 23382–23390. [DOI] [PubMed] [Google Scholar]
  • 62. Yanai H, Chen H‐M, Inuzuka T, Kondo S, Mak TW, Takaoka A, Honda K & Taniguchi T (2007) Role of IFN regulatory factor 5 transcription factor in antiviral immunity and tumor suppression. Proc Natl Acad Sci USA 104, 3402–3407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hu G, Mancl ME & Barnes BJ (2005) Signaling through IFN regulatory factor‐5 sensitizes p53‐deficient tumors to DNA damage‐induced apoptosis and cell death. Cancer Res 65, 7403–7412. [DOI] [PubMed] [Google Scholar]
  • 64. Korczeniewska J & Barnes BJ (2018) Corrected and republished from: the COP9 signalosome interacts with and regulates interferon regulatory factor 5 protein stability. Mol Cell Biol 38, e00493‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Chang Foreman H‐C, Van Scoy S, Cheng T‐F & Reich NC (2012) Activation of interferon regulatory factor 5 by site specific phosphorylation. PLoS ONE 7, e33098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Chen W, Lam SS, Srinath H, Jiang Z, Correia JJ, Schiffer CA, Fitzgerald KA, Lin K & Royer WE (2008) Insights into interferon regulatory factor activation from the crystal structure of dimeric IRF5. Nat Struct Mol Biol 15, 1213–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Cheng T‐F, Brzostek S, Ando O, Van Scoy S, Kumar KP & Reich NC (2006) Differential activation of IFN regulatory factor (IRF)‐3 and IRF‐5 transcription factors during viral infection. J Immunol 176, 7462–7470. [DOI] [PubMed] [Google Scholar]
  • 68. Balkhi MY, Fitzgerald KA & Pitha PM (2010) IKKα negatively regulates IRF‐5 function in a MyD88–TRAF6 pathway. Cell Signal 22, 117–127. [DOI] [PubMed] [Google Scholar]
  • 69. Lin R, Yang L, Arguello M, Penafuerte C & Hiscott J (2005) A CRM1‐dependent nuclear export pathway is involved in the regulation of IRF‐5 subcellular localization. J Biol Chem 280, 3088–3095. [DOI] [PubMed] [Google Scholar]
  • 70. Marchlik E, Thakker P, Carlson T, Jiang Z, Ryan M, Marusic S, Goutagny N, Kuang W, Askew GR, Roberts V et al (2010) Mice lacking Tbk1 activity exhibit immune cell infiltrates in multiple tissues and increased susceptibility to LPS‐induced lethality. J Leukoc Biol 88, 1171–1180. [DOI] [PubMed] [Google Scholar]
  • 71. Pandey AK, Yang Y, Jiang Z, Fortune SM, Coulombe F, Behr MA, Fitzgerald KA, Sassetti CM & Kelliher MA (2009) NOD2, RIP2 and IRF5 play a critical role in the Type I interferon response to Mycobacterium tuberculosis . PLoS Pathog 5, e1000500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Lopez‐Pelaez M, Lamont DJ, Peggie M, Shpiro N, Gray NS & Cohen P (2014) Protein kinase IKKβ‐catalyzed phosphorylation of IRF5 at Ser462 induces its dimerization and nuclear translocation in myeloid cells. Proc Natl Acad Sci USA 111, 17432–17437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ren J, Chen X & Chen ZJ (2014) IKKβ is an IRF5 kinase that instigates inflammation. Proc Natl Acad Sci USA 111, 17438–17443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hu MC‐T, Lee D‐F, Xia W, Golfman LS, Ou‐Yang F, Yang J‐Y, Zou Y, Bao S, Hanada N, Saso H et al (2004) IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117, 225–237. [DOI] [PubMed] [Google Scholar]
  • 75. Gringhuis SI, García‐Vallejo JJ, van Het Hof B & van Dijk W (2005) Convergent actions of I kappa B kinase beta and protein kinase C delta modulate mRNA stability through phosphorylation of 14‐3‐3 beta complexed with tristetraprolin. Mol Cell Biol 25, 6454–6463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Gao Z, Hwang D, Bataille F, Lefevre M, York D, Quon MJ & Ye J (2002) Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. J Biol Chem 277, 48115–48121. [DOI] [PubMed] [Google Scholar]
  • 77. Lee S, Andrieu C, Saltel F, Destaing O, Auclair J, Pouchkine V, Michelon J, Salaun B, Kobayashi R, Jurdic P et al (2004) IkappaB kinase beta phosphorylates Dok1 serines in response to TNF, IL‐1, or gamma radiation. Proc Natl Acad Sci USA 101, 17416–17421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Bergstrøm B, Aune MH, Awuh JA, Kojen JF, Blix KJ, Ryan L, Flo TH, Mollnes TE, Espevik T & Stenvik J (2015) TLR8 senses Staphylococcus aureus RNA in human primary monocytes and macrophages and induces IFN‐β production via a TAK1‐IKKβ‐IRF5 signaling pathway. J Immunol 195, 1100–1111. [DOI] [PubMed] [Google Scholar]
  • 79. Shim J‐H, Xiao C, Paschal AE, Bailey ST, Rao P, Hayden MS, Lee K‐Y, Bussey C, Steckel M, Tanaka N et al (2005) TAK1, but not TAB 1 or TAB 2, plays an essential role in multiple signaling pathways in vivo. Genes Dev 19, 2668–2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Koziczak‐Holbro M, Littlewood‐Evans A, Pöllinger B, Kovarik J, Dawson J, Zenke G, Burkhart C, Müller M & Gram H (2009) The critical role of kinase activity of interleukin‐1 receptor‐associated kinase 4 in animal models of joint inflammation. Arthritis Rheum 60, 1661–1671. [DOI] [PubMed] [Google Scholar]
  • 81. Yeh JE, Toniolo PA & Frank DA (2013) Targeting transcription factors. Curr Opin Oncol 25, 652–658. [DOI] [PubMed] [Google Scholar]
  • 82. Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, Kong N, Kammlott U, Lukacs C, Klein C et al (2004) In vivo activation of the p53 pathway by small‐molecule antagonists of MDM2. Science 303, 844–848. [DOI] [PubMed] [Google Scholar]
  • 83. Moellering RE, Cornejo M, Davis TN, Del Bianco C, Aster JC, Blacklow SC, Kung AL, Gilliland DG, Verdine GL & Bradner JE (2009) Direct inhibition of the NOTCH transcription factor complex. Nature 462, 182–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Higueruelo AP, Jubb H & Blundell TL (2013) Protein‐protein interactions as druggable targets: recent technological advances. Curr Opin Pharmacol 13, 791–796. [DOI] [PubMed] [Google Scholar]
  • 85. Wójcik P & Berlicki Ł (2016) Peptide‐based inhibitors of protein–protein interactions. Bioorg Med Chem Lett 26, 707–713. [DOI] [PubMed] [Google Scholar]
  • 86. Xu H, Krolikowski JG, Jones DW, Ge Z‐D, Pagel PS, Pritchard KA & Weihrauch D (2012) 4F Decreases IRF5 expression and activation in hearts of tight skin mice. PLoS ONE 7, e52046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Krausgruber T, Saliba D, Ryzhakov G, Lanfrancotti A, Blazek K & Udalova IA (2010) IRF5 is required for late‐phase TNF secretion by human dendritic cells. Blood 115, 4421–4430. [DOI] [PubMed] [Google Scholar]
  • 88. Eames H (2014) The interferon regulatory factor 5 (IRF5) interactome: investigating the role of co‐factors in regulation of inflammation. PhD Thesis, Imperial College London, London, UK. [Google Scholar]
  • 89. Feng D, Sangster‐Guity N, Stone R, Korczeniewska J, Mancl ME, Fitzgerald‐Bocarsly P & Barnes BJ (2010) Differential requirement of histone acetylase and deacetylase activities for IRF5‐mediated proinflammatory cytokine expression. J Immunol 185, 6003–6012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Eames HL, Saliba DG, Krausgruber T, Lanfrancotti A, Ryzhakov G & Udalova IA (2012) KAP1/TRIM28: an inhibitor of IRF5 function in inflammatory macrophages. Immunobiology 217, 1315–1324. [DOI] [PubMed] [Google Scholar]
  • 91. Saporito MS, Ochman AR, Lipinski CA, Handler JA & Reaume AG (2012) MLR‐1023 is a potent and selective allosteric activator of Lyn kinase in vitro that improves glucose tolerance in vivo. J Pharmacol Exp Ther 342, 15–22. [DOI] [PubMed] [Google Scholar]
  • 92. Adachi T, Pazdrak K, Stafford S & Alam R (1999) The mapping of the Lyn kinase binding site of the common β subunit of IL‐3/granulocyte‐macrophage colony‐ stimulating factor/IL‐5 receptor. J Immunol 162, 1496–1501. [PubMed] [Google Scholar]
  • 93. Gray P, Dagvadorj J, Michelsen KS, Brikos C, Rentsendorj A, Town T, Crother TR & Arditi M (2011) Myeloid differentiation factor‐2 interacts with Lyn kinase and is tyrosine phosphorylated following lipopolysaccharide‐induced activation of the TLR4 signaling pathway. J Immunol 187, 4331–4337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Adachi T, Stafford S, Sur S & Alam R (1999) A novel Lyn‐binding peptide inhibitor blocks eosinophil differentiation, survival, and airway eosinophilic inflammation1, 2. J Immunol 163, 939–946. [PubMed] [Google Scholar]
  • 95. Gao S, Li X, Nie S, Yang L, Tu L, Dong B, Zhao P, Wang Y, Yu Y, Wang L et al (2017) An AAAG‐rich oligodeoxynucleotide rescues mice from bacterial septic peritonitis by interfering interferon regulatory factor 5. Int J Mol Sci 18, 1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Bethuyne J, De Gieter S, Zwaenepoel O, Garcia‐Pino A, Durinck K, Verhelle A, Hassanzadeh‐Ghassabeh G, Speleman F, Loris R & Gettemans J (2014) A nanobody modulates the p53 transcriptional program without perturbing its functional architecture. Nucleic Acids Res 42, 12928–12938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Möller A, Pion E, Narayan V & Ball KL (2010) Intracellular activation of interferon regulatory factor‐1 by nanobodies to the multifunctional (Mf1) domain. J Biol Chem 285, 38348–38361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Li M, Fan X, Liu J, Hu Y & Huang H (2015) Selection by phage display of nanobodies directed against hypoxia inducible factor‐1α (HIF‐1α). Biotechnol Appl Biochem 62, 738–745. [DOI] [PubMed] [Google Scholar]

Articles from The Febs Journal are provided here courtesy of Wiley

RESOURCES