Skip to main content
Advertisement
  • Loading metrics

Viral manipulation of STAT3: Evade, exploit, and injure

Abstract

Signal transducer and activator of transcription 3 (STAT3) is a key regulator of numerous physiological functions, including the immune response. As pathogens elicit an acute phase response with concerted activation of STAT3, they are confronted with two evolutionary options: either curtail it or employ it. This has important consequences for the host, since abnormal STAT3 function is associated with cancer development and other diseases. This review provides a comprehensive outline of how human viruses cope with STAT3-mediated inflammation and how this affects the host. Finally, we discuss STAT3 as a potential target for antiviral therapy.

Signal transduction through the STAT3 pathway

STAT3 is a transcription factor activated by tyrosine phosphorylation

Signal transducer and activator of transcription 3 (STAT3) was first described in 1994 as a central transcription factor in acute inflammation [1]. Since then, STAT3 has been shown to regulate a wide spectrum of biological programs, including inflammation, tissue regeneration, cell proliferation, cell survival, cellular differentiation, angiogenesis, chemotaxis, and cell adhesion. This functional pleiotropy can be partially explained by the broad number of ligands that lead to STAT3 activation after binding to their respective cytokine receptors [2]. Upon cytokine binding, there is typically recruitment and reciprocal trans-phosphorylation of tyrosine kinases of the Janus kinase (JAK) family comprising JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2) [3,4,5]. They, in turn, recruit and phosphorylate STAT3 (p-STAT3) at the highly conserved tyrosine residue 705 (pY705) [6], resulting in the formation of STAT3 homo- or heterodimers with signal transducer and activator of transcription 1 (STAT1) or signal transducer and activator of transcription 5 (STAT5) [7]. Subsequently, the activated signal transducer and activator of transcription (STAT) dimers translocate to the nucleus and facilitate gene transcription after binding to genomic DNA. Many pathways thus converge in STAT3-mediated gene-expression (Fig 1).

thumbnail
Fig 1. Regulatory circuits of the STAT3 signaling pathway.

STAT3 can be activated by a wide range of ligands binding to cytokine, growth factor, or G-protein-coupled receptors. With the exception of receptor tyrosine kinases, these receptors lack intrinsic kinase activity and thus act by recruiting adaptor kinases (e.g., JAKs, SRC) to propagate downstream signals. As a result, STAT3 is phosphorylated at tyrosine 705 (pY705, pink), forms homodimers or heterodimers, and translocates to the nucleus, where it transcribes regulators of various cellular processes. Additionally, STAT3 can be phosphorylated at serine 727 (pS727, purple) by serine/threonine kinases (e.g., MAPK, mTOR, PKCδ), which enhance STAT3 transcriptional activity in the nucleus or direct STAT3 to mitochondria. Acetylation at lysine 685 (K685, red) by histone acetyltransferases (e.g., CREB binding protein CBP/histone acetyltransferase p300) or methylation at lysine 140 (K140, blue) by histone methyltransferases (e.g., SET9) favor or impair STAT3 transcriptional activity, respectively. Unphosphorylated STAT3 exhibits regulatory functions in the nucleus or can be retained in the cytoplasm, where it associates with microtubules and focal adhesions. The activity of STAT3 is tightly regulated by phosphatases (e.g., PTPRD), SOCS3, PIAS3, and miRNAs that fine-tune the temporal pattern of STAT3 activity and its other pathway components. All miRNAs are degrading the mRNAs of the indicated proteins. A, acetylation; CBP, CREB-binding protein; CT-1R, cardiotrophin 1 receptor; CNTFR, ciliary neurotrophic factor receptor; DUSP2, dual specificity protein phosphatase 2; EGFR, epidermal growth factor receptor; GHR, growth hormone receptor; G-CSFR, granulocyte colony-stimulating factor receptor; GM-CSFR, granulocyte-macrophage colony-stimulating factor receptor; gp130, glycoprotein 130; IFNAR, interferon alpha receptor; IFNGR, interferon gamma receptor; IL, interleukin; JAK, Janus kinase; K140, lysine 140; K685, lysine 685; LIFR, leukemia inhibitory factor receptor; MAPK, mitogen-activated protein kinase; M, methylation; miRNA, microRNA; mTOR, mechanistic target of rapamycin; OSMR, oncostatin-M-specific receptor; P, phosphorylation; p300, histone acetyltransferase p300; PDGFR, platelet-derived growth factor receptor; PIAS3, protein inhibitor of activated STAT protein 3; PKCδ, protein kinase C delta type; pS727, phospho-serine 727; PTPRC, receptor-type tyrosine-protein phosphatase C; PTPRD, receptor-type tyrosine-protein phosphatase D; PTPRT, receptor-type tyrosine-protein phosphatase T; pY705, phospho-tyrosine 705; SET9, histone-lysine N-methyltransferase SET9; SOCS3, suppressor of cytokine signaling 3; SRC, proto-oncogene tyrosine-protein kinase; STAT3, signal transducer and activator of transcription 3; TpoR, thrombopoietin receptor; TRIM28, tripartite motif-containing protein 28.

https://doi.org/10.1371/journal.ppat.1006839.g001

Regulation of STAT3 activation

STAT3 activity is additionally regulated by several post-translational modifications. First, phosphorylation at serine 727 (pS727) by a variety of serine/threonine kinases, such as the mitogen-activated protein (MAP) kinases, mechanistic target of rapamycin (mTOR), and protein kinase C delta type (PKCδ), increases transcriptional activity even further [8]. In mitochondria, pS727 promotes cellular respiration independently from pY705 [9]. Second, STAT3 can be reversible acetylated on K685 by histone acetyltransferase CBP/p300, prolonging transcriptional activity [10]. Contrarily, K140 methylation by histone methyltransferase SET9 impairs transcription [11].

Additional negative feedback regulators include the protein phosphatases receptor-type tyrosine-protein phosphatase C (PTPRC), receptor-type tyrosine-protein phosphatase D (PTPRD), receptor-type tyrosine-protein phosphatase T (PTPRT), and dual specificity protein phosphatase 2 (DUSP2) that hydrolyze p-STAT3 or upstream pathway members [12, 13, 14, 15]. Suppressor of cytokine signaling 3 (SOCS3) prevents STAT3 activation by shielding phospho-tyrosine residues of upstream kinases [16,17], while protein inhibitor of activated STAT protein 3 (PIAS3) prevents binding of STAT3 dimers to DNA [18]. In the nucleus, the phosphorylation and transcriptional activity of STAT3 pS727 is negatively regulated by tripartite motif-containing protein 28 (TRIM28), which binds directly to the central coiled-coil and DNA-binding domains of STAT3 [19]. Furthermore, several microRNAs (miRNAs) directly target STAT3 mRNA, including Let-7a [20], miR-17-5p [21], miR-29b [22], miR-124 [23], and miR-519a [24]. Let-7a also exerts an indirect effect on STAT3 by promoting SOCS3 expression [25]. STAT3-activating miRNAs include miR-24 and miR-629 that impair miR-124 expression via HNF4A mRNA silencing [26]. Similarly, miR-135a-5p and miR-19a enhance pY705 phosphorylation by respectively targeting the mRNA of PTPRD and SOCS3 [27,28].

Although STAT3 phosphorylation is often considered a prerequisite for its transcriptional activity, unphosphorylated STAT3 (u-STAT3) can promote the expression of genes related to cell cycle progression [29,30]. Finally, cytoplasmic STAT3 promotes cell migration by interacting with stathmin, a microtubule destabilizer [31].

Physiological role of STAT3 in inflammation

In mammalian organisms, tissue injuries inflicted by pathogens are met by the release of inflammatory mediators and local infiltration of white blood cells. This eliminates foreign material, removes damaged tissue components, and clears the way for repair. STAT3 plays an essential role in these processes by enabling the expression of a variety of genes in response to specific external signals sensed by cell-surface receptors [32]. Not all cell types and tissues have the same expression patterns of these receptors and their signaling cascade mediators. Therefore, the functional consequence of STAT3 activation is highly context-dependent, which can often lead to conflicting information. As illustrated in the following examples, this is particularly true for the role of STAT3 in inflammation, since it is either able to promote or suppress this process.

IL-6/STAT3 pathway promotes inflammation

Interleukin 6 (IL-6) is a classic proinflammatory cytokine that signals through STAT3 as part of the acute phase response (APR), a nonspecific reaction of the innate immune system to pathogen infection. During acute inflammation, IL-6 is produced in the lesion site to attract neutrophils and increase granulopoiesis [33]. Upon extravasation at the site of injury, neutrophils produce soluble interleukin 6 receptor alpha (sIL-6Rα), which in complex with IL-6 binds to glycoprotein 130 (gp130) at the membrane of resident tissue cells. This process is known as the trans-signaling pathway [34], which subsequently leads to a switch in chemokine expression attracting monocytic and T cells [35,36]. Upon the arrival of monocytic cells in the inflammation site, IL-6 signals govern their transformation into macrophages [37]. Pathogens are thus initially confronted in their initial microenvironment with a potent IL-6 stimulus, which is mounted by the host to combat their very presence.

Apart from the lesion site, the IL-6/STAT3 proinflammatory signaling axis functions in many other cellular and tissue compartments. In secondary lymphoid tissues, where the adaptive immune response takes place, IL-6-mediated STAT3 activation promotes the proliferation and survival of T and B cell populations [38,39]. In addition, together with transforming growth factor beta (TGF-β), the IL-6/STAT3 axis is crucial for differentiating naive CD4+ T cells into Th17 cells [40,41], limiting the generation of regulatory CD4+ T cells (Treg cells) [42]. Moreover, IL-6 promotes the differentiation of follicular helper T cells (TFH cells) via STAT3 [43,44], effectively linking together T and B cell responses [45].

IL-10/STAT3 pathway suppresses inflammation

Interleukin 10 (IL-10) also activates STAT3, but unlike IL-6 the IL-10/STAT3 axis has powerful anti-inflammatory properties. Its function is essential to restrain unwanted immune responses and prevent autoimmune pathologies [46]. IL-10 only exerts an effect on immune cells, as they are the only cells to have the interleukin 10 receptor alpha (IL-10RA). This IL-10 receptor is highly expressed in monocytic cells and macrophages but also to a lesser extent in NK cells, CD4+ and CD8+ T cells, B cells, dendritic cells (DCs), and mast cells [47]. Until recently it was unclear how, in cells responsive to both IL-6 and IL-10, STAT3 orchestrates such opposing functions. In fact, SOCS3 is critical for selecting the transcriptional response. While IL-6 signaling is selectively inhibited by SOCS3 binding to gp130, SOCS3 does not interfere with IL-10R-mediated STAT3 activation [48]. As an effect, STAT3 activation is transient and proinflammatory in response to IL-6, while long lasting and anti-inflammatory in IL-10 [49].

IL-10 exerts its anti-inflammatory effect by suppressing T helper 1 (TH1) cell responses [50] and regulating apoptosis in B cells [51]. In addition, IL-10/STAT3 is necessary for generation of tolerogenic DCs and of induced Tregs out of naïve CD4+ T cells [52].

Interferon activation of STAT3

Upon viral infection, type I and type II interferons (IFNs) initiate a canonical antiviral transcriptional program through STAT1 and STAT2, which results in an inflammatory, proapoptotic, and antiproliferative state [53]. At the same time, IFNs induce STAT3 activation [54,55], which provides a negative feedback by favoring cell proliferation and survival and thus resulting in gene expression with anti-inflammatory properties [56]. In support of this model, silencing of STAT1 or STAT3 expression by RNA interference confirmed the role of STATs as important determinants of IFN-α receptor (IFNAR) function [57] and emphasizes the role of STAT3 to restrain STAT1-mediated proinflammatory signaling [58].

In this context, an initial proinflammatory response to IFNs is mediated by STAT1, which expression is far more abundant, while STAT3-mediated gene induction is prevented by the SIN3 transcription regulator family member A complex (SIN3A). This multimolecular complex, containing histone deacetylases 1 (HDAC1) and 2 (HDAC2), inactivates STAT3 by deacetylation [59]. It has been suggested that only in a second phase is STAT3 activity increased, leading to a sequential counterbalance to the initial flare of apoptosis and decrease in proliferation mediated by IFNs [60].

A potential regulatory layer that remains poorly understood is the role of STAT1 and STAT3 heterodimers induced by IFNs. On one hand, STAT1 and STAT3 heterodimers have been described to bind regulatory elements present in promoters of interferon-stimulated genes (ISGs) such as γ-activated sequence (GAS), supporting a potential antiviral role of STAT1 and STAT3 heterodimers [61]. On the other hand, it has been proposed that STAT1 and STAT3 heterodimers can effectively quench STAT1 and thus provide negative feedback in a later phase of the IFN response [57]. Whatever the effect of STAT1 and STAT3 heterodimers on viral infection, either proviral or antiviral, it provides another layer of potential manipulation for viral gene products that warrants further research.

The suggested temporal dynamics of STAT biology may explain the serious consequences of persistent viral infections, as in the case of hepatitis C virus (HCV) [60]. Here, sustained type I and II IFN signaling may drastically alter the initial STAT dimerization balance, enabling a more pronounced proliferative role of STAT3 and hence increasing oncogenic pressure on hepatocytes.

Role of STAT3 in regeneration and disease

Upon infection, inflammatory cytokines trigger cell signaling in local stem cells or differentiated cells. Among other transcription factors, this eventually leads to the activation of STAT3 that mediates regenerative gene-expression programs. These genes include growth factors, cell-cycle stimulators, cell death inhibitors, and genes promoting dedifferentiation and cell motility and migration [62]. The task of STAT3 in regenerative inflammation is well studied in the liver, a model for organ regeneration as it can easily restore functional capacity after partial resection through compensatory hyperplasia [63,64]. In the liver, the inflammatory response following injury instigates the regenerative process [65]. As part of the APR, liver-residing macrophages (Kupffer cells) release proinflammatory cytokines such as IL-6 and tumor necrosis factor alpha (TNF-α) [66]. These inflammatory cytokines are important components of priming pathways that help sensitize hepatocytes to proliferative signals, such as hepatocyte growth factor (HGF) and epidermal growth factor (EGF) [67]. However, when liver injury persists, as in the case of chronic viral hepatitis, liver inflammation paired with constant STAT3 activity fosters the development of hepatocellular carcinoma (HCC) [27]. A similar oncogenic role of STAT3 has been observed in a wide variety of other malignancies such as colorectal, lung, prostate, gastric, and breast cancers [68].

Given the extensive role of STAT3 in many physiological processes, it is only logical that its perturbation entails a wide variety of pathological consequences. This is exemplified by loss-of-function mutations in the STAT3 gene that lead to the autosomal dominant hyper-immunoglobulin E (IgE) syndrome (AD-HIES) [69]. These patients exhibit an immunodeficiency complex that presents with recurrent episodes of pneumonia and other lung abnormalities, abnormally high levels of IgE, eosinophilia, eczema, and skeletal and connective tissue abnormalities. Inadequate inflammatory capacity due to a broken IL-6/STAT3 axis curtails the APR and leads to "cold" skin abscesses (i.e., without inflammatory signs). As STAT3 is necessary for generating Th17 cells, a defective Th17 response and increased susceptibility for microbial infections are hallmarks of AD-HIES. On the other hand, the defects in the anti-inflammatory IL-10/STAT3 pathway lead to reduced peripheral tolerance, which is clinically translated in atopic dermatitis. Finally, AD-HIES patients exhibit a marked reduction in memory T cells and increased latency of herpesviruses such as varicella-zoster virus (VZV) and Epstein–Barr virus (EBV) [70].

Molecular mechanisms of viral STAT3 manipulation

Viral stimulation of STAT3 function

As STAT3 activation is a pivotal event in the APR elicited by pathogen invasion, many viruses have evolved to thrive in a STAT3-driven microenvironment and have developed strategies to stimulate STAT3 signaling (Fig 2A, Table 1). For example, hepatitis B virus (HBV) promotes the formation of p-STAT3 dimers that bind specifically to an androgen-responsive element site present in the HBV enhancer 1 region and hence stimulates viral gene expression [71]. This is in part mediated by hepatitis B virus X protein (HBx), which induces pY705 phosphorylation via JAK1 [72] and down-regulates miRNA let-7a, a negative regulator of STAT3 mRNA [20]. Additionally, HBV favors STAT3 activation by inducing reactive oxygen species (ROS), which results in epigenetic silencing of SOCS3 mRNA via up-regulation of snail family transcriptional repressor 1 (SNAIL1) [73]. HCV requires STAT3 and therefore promotes STAT3 signaling to maintain infection [74]. HCV stimulates STAT3 directly by interaction with the HCV core protein [75] and indirectly through non-structural protein 5A (NS5A), which activates STAT3 via ROS induction [76]. Furthermore, miR-135a-5p is a negative regulator of STAT3 phosphatase PTPRD and is up-regulated in HCV-infected hepatocytes, leading to an enhanced STAT3 transcriptional activity [27]. Furthermore, HCV-infected hepatocytes secrete miR-19a within exosomes, down-regulating the expression of SOCS3 in hepatic stellate cells (HSCs) and promoting STAT3 phosphorylation [28]. Similarly, Rift Valley fever virus (RVFV) infection induces STAT3 (pY705) phosphorylation by the viral non-structural protein s (NSs) [77]. STAT3 activation is also a frequent feature of the Herpesviridae family. Human cytomegalovirus (HCMV) activates STAT3 through various mechanisms, depending on virus strain and cell type. In U373 MG astrocytes, viral protein US28 of the Titan strain induces IL-6 production, which in turn activates STAT3 in an auto- and paracrine fashion [78]. In hepatoma cells and primary human hepatocytes (PHHs), strains AD169 and HCMV-DB also activate STAT3 via IL-6 in an autocrine and/or paracrine mannerinfluenza strain H5N1 impairs pY705 phosphorylation at I87A [87]. Kaposi’s sarcoma-associated herpesvirus (KSHV) encodes a viral homologue of IL-6 (vIL-6) that signals through the same receptors as cellular IL-6 (IL-6Rα/gp130) but can also activate STAT3 in an IL-6Rα-independent manner in Hep3B liver cells [88]. In human endothelial cells, KSHV increases both pY705 and pS727 phosphorylation [19]. Though pY705 phosphorylation is transient, pS727 persists because the viral protein kaposin B activates the p38/MK2 pathway to suppress TRIM28, which is a negative regulator of pS727 phosphorylation [19]. STAT3 activation in DCs is believed to stem from virions interacting with dendritic cell-specific ICAM-3-grabbing nonintegrin 1 (DC-SIGN) at the cell’s surface, as antibody blockage of DC-SIGN reduces pY705 levels [89]. VZV induces pY705 phosphorylation in epidermal cells and T cells in vivo as well as in fibroblasts in vitro through unknown mechanisms [90]. Resveratrol, an inhibitor of kinases phosphorylating STAT3, hampers VZV infection, suggesting the involvement of host kinases [91]. Similarly, ZIKA virus (ZIKV) infection induces pY705 in primary retinal glial cells [92] and favors the activity of the IL-6/STAT3 pathway in blood mononuclear cells from infected rhesus monkeys, albeit without any known molecular mechanism [93].

thumbnail
Fig 2. Viral manipulation of the STAT3 signaling pathway.

(A) Viruses activating STAT3 function and the mechanisms involved. Viral proteins such as HBx, NS5A, core, NSs, EBNA2, LMP1, US28, and IE1 induce STAT3 activation either directly or by favoring the action of upstream positive regulators. Viruses like HCMV and KSHV code for homologues of human interleukins such as IL-10 and IL-6. Alternatively, virus-induced activation of STAT3 can be achieved by the inhibition of negative regulators such as SOCS3, PTPRD, TRIM28, and Let-7a. In the case of some viruses, STAT3 activation (VZV and ZIKV) or STAT3-mediated effects (IAV) have been described, but the mechanisms involved have not been fully elucidated. All miRNAs are degrading the mRNAs of the indicated proteins. (B) Viruses suppressing STAT3 function and the mechanisms involved. Virus-mediated inactivation of STAT3 can be attained by decreasing its phosphorylation (KSHV, IAV, and hMPV), inducing STAT3 protein degradation (MuV), hampering its transcriptional activity (MeV), or altering its subcellular localization (HCMV, RABV, HEV, and hMPV). EBNA2, Epstein–Barr virus nuclear antigen 2; EBV, Epstein–Barr virus; HBV, hepatitis B virus; HBx, hepatitis B virus X protein; HCMV, human cytomegalovirus; HCV, hepatitis C virus; HEV, hepatitis E virus; hMPV, human metapneumovirus; IAV, influenza A virus; IE1, intermediate-early protein 1; IL-6, interleukin 6; IL-10, interleukin 10; IRAK1, interleukin 1 receptor-associated kinase 1; JAK1, Janus kinase 1; KSHV, Kaposi’s sarcoma-associated herpesvirus; LMP1, latent membrane protein 1; miRNA, microRNA; MeV, measles virus; MK2, mitogen-activated protein kinase 2; MuV, mumps virus; NS5A, non-structural protein 5A; NSs, non-structural proteins; P, phosphorylation; PKCδ, protein kinase C delta type; PTPRD, receptor-type tyrosine-protein phosphatase D; RABV, rabies virus; ROS, reactive oxygen species; RVFV, Rift Valley fever virus; SOCS3, suppressor of cytokine signaling 3; STAT3, signal transducer and activator of transcription 3; TRIM28, tripartite motif-containing protein 28; u-STAT3, unphosphorylated STAT3; vIL-10, viral IL-10; vIL-6, viral IL-6; VZV, varicella-zoster virus; ZIKV, Zika virus.

https://doi.org/10.1371/journal.ppat.1006839.g002

thumbnail
Table 1. Virus/STAT3 interactions: Summary of observations and employed methods.

https://doi.org/10.1371/journal.ppat.1006839.t001

Viral suppression of STAT3 function

In the acute phase, viral suppression of STAT3 reduces the host cell's ability to respond to inflammatory cytokines. On the other hand, inhibiting STAT3 also removes negative feedback on the antiviral response. To understand the beneficial effect of blocking STAT3 for viruses, it thus requires a temporal dissection of each individual virus/STAT3 interaction. Most viruses that suppress STAT3, however, do this to avoid the antiviral pressure exerted by STAT3 responsive genes in the acute phase of infection (Fig 2B, Table 1). Mumps virus (MuV) viral protein V (MuV V) induces STAT3 degradation by promoting STAT3-directed ubiquitin E3 ligase complexes [94]. Similarly, measles virus (MeV) viral protein V (MeV V) reduces STAT3-mediated transcription but through an unknown mechanism that is, however, independent of ubiquitin ligase subunits [95]. Influenza A virus (IAV) infection induces STAT3 activation in the early phase of the inflammatory response. As the infection progresses, STAT3 activity is suppressed to a degree that inversely correlates with the pathogenicity of each IAV strain. For instance, the highly pathogenic avian influenza strain H5N1 impairs pY705 phosphorylation, but in the case of the low pathogenic seasonal H1N1 strain this decrease is even more pronounced [96]. This inhibition could be partly mediated by viral protein NS1, which increases SOCS3 expression [97]. Other viruses have developed alternative strategies to impair STAT3 function, such as manipulating its subcellular localization during infection. Hepatitis E virus (HEV) ORF3 protein blocks the nuclear translocation of p-STAT3 [98]. Likewise, in rabies virus (RABV) infections, viral protein P associates with p-STAT3 in the cytoplasm, impeding its nuclear translocation. In addition, P protein interferes with gp130 receptor signaling [99]. Human metapneumovirus (hMPV) infection prevents the nuclear translocation of STAT3 in a cytokine-specific manner, as this was only observed following stimulation with IL-6 and not in case of interleukin 22 (IL-22) [100]. Contrary to the occasions where HCMV induces STAT3 phosphorylation [78,79], HCMV can also rapidly disrupt IL-6/STAT3 signaling in U-373 cells by sequestering u-STAT3 to the nucleus via viral protein IE1 [101]. Apart from inducing STAT3 activation, KSHV can also target and inhibit STAT3 or its activators in vitro through a panel of virally encoded miRNAs. KSHV miR-K6-5, miR-K8, and miR-K9* reduce STAT3 levels, while upon IL-6 treatment, miR-K6-5 and miR-K9 decrease PKCδ and interleukin 1 receptor-associated kinase 1 (IRAK1) expression, respectively, which is accompanied by reduced p-STAT3 levels [102]. Whether in the end KSHV-induced STAT3 activation or the negative regulation of STAT3 by viral miRNAs act predominantly in endothelial cells remains unclear. But it is conceivable that both opposing mechanisms are required in a time-dependent manner to regulate the transition from the latent to the lytic stage of the viral life cycle.

Consequences of viral perturbations in STAT3 activity

Recalibration of apoptosis dynamics

Apoptosis is perhaps the most primordial response of a host cell to infection, designed to thwart the virus spread. Generally, viruses need to prevent host cell apoptosis to maintain a compartment of infected cells [104]. However, there are also examples where viruses induce apoptosis to spark the release of virions and galvanize viral spread [105]. STAT3 is mainly considered a negative regulator of apoptosis by up-regulating the expression of several antiapoptotic factors [106] (Fig 3A). IAV H5N1 causes higher pY705 levels than seasonal H1N1. Therefore, apoptosis is delayed during H5N1 infection, allocating additional time to infected cells for progeny virus production. Ultimately, this leads to an accumulation of apoptotic cells at later stages [96]. Similarly, VZV prevents apoptosis by increasing STAT3 phosphorylation, which up-regulates baculoviral IAP repeat-containing protein 5 (BIRC5) expression, a VZV host factor belonging to the family of inhibitors of apoptosis (IAP) [90]. During EBV infection, virus-induced STAT3 activation up-regulates poly(rC)-binding protein 2 (PCBP2) expression, limiting susceptibility of latently infected cells to lytic signals and fostering persistence [107]. This goes as well for KSHV, in which STAT3 restrains the exit from latency into the lytic cycle by repressing the expression of the viral protein R transactivator (RTA) [108]. MuV is yet another example in which the cytopathic effects of infection are associated with the induction of apoptosis, partly via V protein-mediated STAT3 degradation [94]. Finally, RVFV reins in apoptosis by enhancing the nuclear translocation of phosphorylated STAT3 and impairs the expression of proapoptotic genes such as proto-oncogene c-Fos (FOS), proto-oncogene c-Jun (JUN), and nuclear receptor subfamily 4 group A member 2 (NR4A2) [77].

thumbnail
Fig 3. Viral replicative advantages and pathological consequences related to STAT3-altered function.

(A) Virus-induced perturbation of STAT3 as regulator of apoptosis. In the context of viral infections, apoptosis can be restrained via STAT3, since it favors the expression of antiapoptotic factors (e.g., PCBP2 and BIRC5) or prevents proapoptotic ones (e.g., RTA, FOS, JUN, and NR4A2). In contrast, inhibition of STAT3 by viruses such as IAV and MuV has been associated with the induction of the apoptotic process. (B) Viral manipulation of STAT3 and its effect on immune responses. Viral inhibition of STAT3 can induce a decrease of ISG and APR gene expression and favor immune evasion, as in the case of KSHV and HEV. Virus-mediated STAT3 activation can also have immunosuppressive actions such as impairing DC function (KSHV and HCMV) and favoring the expansion of MDSCs (HCV). In other cases, the proinflammatory actions of STAT3 have been associated with the development of host pathologies such as cancer (KSHV). (C) Virus-induced alteration of STAT3 and its impact on cell and tissue organization. STAT3 activation during HCV infection has been associated with alterations of the MT network. This represents a potential advantage for HCV by favoring virus trafficking along MTs. At the tissue and organ level, STAT3 activation has been associated with the development of fibrosis (HCV), the disruption of endothelial vascular junctions (IAV), and enhanced cell invasion, which favors cancer development (EBV). ANGPTL4, angiopoietin-like protein 4; APR, acute phase response; BIRC5, baculoviral IAP repeat-containing protein 5; CCL5, C-C motif chemokine ligand 5; DCs, dendritic cells; DC-SIGN, dendritic cell-specific ICAM-3-grabbing non-integrin 1; EBV, Epstein–Barr virus; FOS, proto-oncogene c-Fos; HCC, hepatocellular carcinoma; HCMV, human cytomegalovirus; HCV, hepatitis C virus; HEV, hepatitis E virus; HSCs, hepatic stellate cells; IAV, influenza A virus; IDO1, indoleamine 2,3-dioxygenase 1; IL-10, interleukin 10; ISG, interferon-stimulated gene; JUN, proto-oncogene c-Jun; KSHV, Kaposi’s sarcoma-associated herpesvirus; MCL1, induced myeloid leukemia cell differentiation protein Mcl-1; MDSCs, myeloid-derived suppressor cells; MT, microtubule; MUC1, mucin 1 cell surface associated; MuV, mumps virus; NPC, nasopharyngeal carcinoma; NR4A2, nuclear receptor subfamily 4 group A member 2; PCBP2, poly(rC)-binding protein 2; PD-L1, programmed cell death 1 ligand 1; RTA, R transactivator; RVFV, Rift Valley fever virus; STAT3, signal transducer and activator of transcription 3; TGF-β, transforming growth factor beta; Treg, regulatory CD4+ T cell; VZV, varicella-zoster virus.

https://doi.org/10.1371/journal.ppat.1006839.g003

Perturbing the immune response

The benefit for a virus to dampen STAT3 signaling lies in controlling antiviral innate immunity responses such as the APR (Fig 3B). Many of the APR genes are modulators of inflammation. C-reactive protein (CRP) for example is a target gene of STAT3 and has several biological functions related to nonspecific host defense [109]. Increased plasma levels of metal-binding APRs (e.g., haptoglobin and hemopexin) help protect host cells from iron loss during infection and the associated injury. Moreover, they act as scavengers for potentially damaging free oxygen radicals. Protease inhibitors among APR genes (e.g., alpha-1-antitrypsin) neutralize lysosomal proteases. These inhibiting factors are released in response to tissue infiltration of activated neutrophils and macrophages, modulating the activity of proinflammatory enzyme cascades. HEV impairs the expression of these APR genes by inhibiting STAT3, attenuating inflammatory responses and creating a favorable environment for viral replication and survival [98].

In contrast to HEV, the KSHV-mediated activation of STAT3 is associated with increased expression of C-C motif chemokine ligand 5 (CCL5) [19], a potent chemoattractant for monocytic cells, eosinophils, NKs, and DCs [110]. Many of these cell types have been shown to be present in Kaposi’s sarcoma lesions, suggesting that STAT3 contributes to the chronic inflammatory state observed in this pathology [19]. Moreover, KSHV-induced STAT3 activation correlates with up-regulated induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) expression levels, which can be reverted by inhibiting STAT3 [89]. MCL1 inhibits Beclin-1, a positive regulator of autophagosome formation, to interfere with antigen processing and presentation by DCs to avoid recognition and clearance [89]. KSHV also inhibits STAT3 via the action of viral miRNAs, and by doing so it hinders the expression of ISGs such as CXCL10, ISG15, IFITM1, IRF1, OAS2, and MX1 [102]. The vIL-10 coded by HCMV up-regulates expression of its receptor DC-SIGN in DCs, their target cells [81,111]. vIL-10 stimulation of DCs also prevents the expression of costimulatory molecules (i.e., CD40, CD80, and CD86), inhibiting maturation of DCs, enhancing their susceptibility to infection, and hampering the immune response [81]. Chronic HCV infection has been associated with the presence of myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid cells that suppress the function of NK, CD4+, and CD8+ T cells [112]. Analysis of myeloid and lymphoid cells from chronically HCV-infected patients has shown that activation of STAT3 up-regulates the expression of suppressive genes (i.e., IL-10, programmed cell death 1 ligand 1 [PD-L1], indoleamine 2,3-dioxygenase 1 [IDO1]) in monocytic cells. They acquire MDSC-like characteristics and favor the expansion of Treg cells [113,114]. MDSCs have been linked to an increased tumor burden and a higher metastasis rate in patients with HCC and in liver cancer mouse models [115]. Thus, by the STAT3-mediated induction of MDSCs, HCV can establish a microenvironment that supports viral immune evasion and accelerates HCC development.

Altering cell architecture and tissue organization

STAT3 also plays a role in cell morphology, which viruses exploit to promote viral persistence, with grave consequences for host cell physiology (Fig 3C). HCV-induced p-STAT3 directly controls microtubule (MT) dynamics through contact inhibition with stathmin [74]. Both HCV core and NS5A are transported along MTs [116]. Moreover, HCV core integrates into the MT lattice by a direct binding to tubulin [117]. Viral attenuation of stathmin enhances intracellular trafficking of the virus and increases replication [74]. In addition, regenerative STAT3 activation in HSCs precipitates fibrotic gene expression (i.e., TGF-β1, TIMP-1) [28], eventually leading to cirrhosis, which constitutes the procarcinogenic field on which most HCCs grow [118]. IAV triggers a STAT3-mediated up-regulation of angiopoietin-like protein 4 (ANGPTL4), a protein that compromises the integrity of endothelial vascular junctions. This leads to enhanced tissue leakiness and exacerbation of inflammatory lung damage in infected mice [103]. EBV is the most distinct etiological agent for the development of nasopharyngeal carcinoma (NPC), a type of cancer in which STAT3 activation or overexpression is associated with more than 75% of tumors in regions where EBV is endemic [119]. EBV-mediated activation of STAT3 spurs cell invasiveness in vitro, and constitutive expression of STAT3 in NPC cell lines results in an increase of mesenchymal markers such as fibronectin and N-cadherin [120]. In accordance, STAT3 activation via LMP1 induces the expression of mucin 1 cell surface-associated (MUC1), a glycoprotein involved in the early steps of cancer cell detachment [121].

Disruption of STAT3 function as antiviral therapy

In the cases where STAT3 activity has a proviral or pathogenic effect, blocking STAT3 represents an interesting therapeutic strategy. Unfortunately, no molecule directly targeting STAT3 has received Food and Drug Administration (FDA) approval for any pathology so far [122], and candidate compounds targeting viral disease have not advanced beyond preclinical evaluation (Table 2). Small-molecule inhibitors targeting STAT3 phosphorylation (e.g., Cpd188, IB-32, Stattic) or dimerization (e.g., STA-21, S3I-201) have been evaluated as antivirals in vitro or in animal models. For instance, HCV replication but not entry is inhibited by STA-21, S3I-201, Cpd188, and IB-32 in Huh7 hepatoma cells or derivatives thereof [58,74,123]. Similarly, S3I-201 and Stattic reduce HCMV replication in cell culture [101], while S3I-201 limits VZV infection both in vitro and in animal models [90]. Oligodeoxynucleotide decoys (ODNs) are DNA-binding domain inhibitors that compete for binding of transcription factors with endogenous promoter sequences in their target genes. STAT3-targeting ODNs significantly decrease HBV RNA expression and DNA replication in hepatoma cell lines [124].

thumbnail
Table 2. STAT3 signaling inhibitors, their mechanisms and in vitro antiviral applications.

https://doi.org/10.1371/journal.ppat.1006839.t002

In addition, several natural products such as resveratrol or curcumin have been described to exhibit STAT3 inhibitory properties [125]. Resveratrol impairs EBV and VZV infection. For EBV, at least, it has been demonstrated that resveratrol suppresses STAT3 phosphorylation [126,127], while the antiviral mechanism by which resveratrol inhibits VZV is not yet understood [91]. Curcumin hinders HCMV replication in U373 cells by reducing nuclear accumulation of STAT3 [101], and while it exerts antiviral properties for IAV [128] and HCV [129], a mechanistic link to STAT3 has not been demonstrated yet.

The multikinase inhibitor sorafenib exhibits an antiviral effect against various HCMV strains by inhibiting the expression of immediate early genes of HCMV at clinically relevant concentrations [130]. However, sorafenib is not selective for STAT3; therefore, it is likely that a combination of unspecific effects may account for the observed antiviral effect of sorafenib on HCMV.

Outlook

STAT3 is a key regulator in inflammation and tissue regeneration triggered by almost every pathogenic infection. Therefore, viruses must deal with STAT3 activity by either curtailing it or employing it. STAT3 dependencies of viruses put a spotlight on the diverse role of signal transduction during viral infections and represent a target for potential antiviral strategies. Deregulated STAT3 signaling is an oncogenic driver and is associated with virus-induced complications, including cancers. However, targeting STAT3 during viral infection and cancer is currently an untapped reservoir, and the question still remains as to why it has not yet resulted in a broad range of clinical applications.

Currently, unspecific tyrosine kinase inhibitors (e.g., sorafenib) and monoclonal antibodies (e.g., tocilizumab) that block upstream components in the STAT3 pathway are readily administered to patients as cancer chemotherapeutics [131,132]. Similarly, other indirect STAT3-targeting strategies, including the modulation of STAT3 regulators, are promising. These include the use of histone deacetylase or proteasome inhibitors that promote expression of the endogenous STAT3 inhibitors SOCS3 and PIAS3, respectively [133]. While the use of approved indirect STAT3 modulators in clinical practice allows an indirect safety evaluation for STAT3-targeting strategies, their use does not allow conclusions on the specific clinical tolerance and efficacy of a STAT3-based antiviral approach.

Several natural products targeting STAT3 are currently being explored and seem promising; however, many (including curcumin and resveratrol) have been described as pan-assay interference compounds (PAINs). In other words, it currently cannot be ruled out that the observed effects of these natural compounds are due to an interference with the experimental readout rather than an interaction with their specific targets [134].

Due to multiple and redundant pathways that converge in STAT3 activation, direct STAT3-targeting agents would be a gold standard to assess the potential benefit of this approach. One reason why we have not observed a breakthrough in STAT3-targeting drugs so far may be that transcription factors are notoriously difficult to target and that many of the STAT3 inhibitors evaluated to date have shown to be problematic regarding their potency, bioavailability, and specificity [122]. Nevertheless, as we have explored in this review, there is strong scientific rationale to continue the development of novel STAT3-targeting therapies. Recently emerged agents that appear encouraging include AZD9150, an antisense oligonucleotide targeting STAT3 mRNA that is in early phase I and II studies for advanced solid and hematological cancers [135137], and napabucasin, a small-molecule inhibitor that has advanced to phase III clinical trials [138]. The evaluation of these and similar compounds for the treatment of cancers is expected to result in a broad range of clinical applications and holds great promise for future antiviral strategies as well.

Acknowledgments

We would like to thank the investigators whose work is cited here and apologize to those whose work could not be cited due to space restrictions. The authors thank Philippe Georgel, Université de Strasbourg, for helpful discussions.

References

  1. 1. Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, et al. (1994) Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell 77: 63–71. pmid:7512451
  2. 2. Mackey-Lawrence NM, Petri WA Jr. (2012) Leptin and mucosal immunity. Mucosal immunology 5: 472–479. pmid:22692456
  3. 3. Wilks AF, Harpur AG, Kurban RR, Ralph SJ, Zurcher G, et al. (1991) Two novel protein-tyrosine kinases, each with a second phosphotransferase-related catalytic domain, define a new class of protein kinase. Molecular and cellular biology 11: 2057–2065. pmid:1848670
  4. 4. Rane SG, Reddy EP (1994) JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene 9: 2415–2423. pmid:7518579
  5. 5. Firmbach-Kraft I, Byers M, Shows T, Dalla-Favera R, Krolewski JJ (1990) tyk2, prototype of a novel class of non-receptor tyrosine kinase genes. Oncogene 5: 1329–1336. pmid:2216457
  6. 6. Kaptein A, Paillard V, Saunders M (1996) Dominant negative stat3 mutant inhibits interleukin-6-induced Jak-STAT signal transduction. The Journal of biological chemistry 271: 5961–5964. pmid:8626374
  7. 7. Delgoffe GM, Vignali DA (2013) STAT heterodimers in immunity: A mixed message or a unique signal? JAK-STAT 2: e23060. pmid:24058793
  8. 8. Wen Z, Zhong Z, Darnell JE Jr. (1995) Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82: 241–250. pmid:7543024
  9. 9. Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, et al. (2009) Function of mitochondrial Stat3 in cellular respiration. Science 323: 793–797. pmid:19131594
  10. 10. Yuan ZL, Guan YJ, Chatterjee D, Chin YE (2005) Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science 307: 269–273. pmid:15653507
  11. 11. Yang J, Huang J, Dasgupta M, Sears N, Miyagi M, et al. (2010) Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proceedings of the National Academy of Sciences of the United States of America 107: 21499–21504. pmid:21098664
  12. 12. Kumar V, Cheng P, Condamine T, Mony S, Languino LR, et al. (2016) CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation. Immunity 44: 303–315. pmid:26885857
  13. 13. Veeriah S, Brennan C, Meng S, Singh B, Fagin JA, et al. (2009) The tyrosine phosphatase PTPRD is a tumor suppressor that is frequently inactivated and mutated in glioblastoma and other human cancers. Proceedings of the National Academy of Sciences of the United States of America 106: 9435–9440. pmid:19478061
  14. 14. Zhang X, Guo A, Yu J, Possemato A, Chen Y, et al. (2007) Identification of STAT3 as a substrate of receptor protein tyrosine phosphatase T. Proceedings of the National Academy of Sciences of the United States of America 104: 4060–4064. pmid:17360477
  15. 15. Lu D, Liu L, Ji X, Gao Y, Chen X, et al. (2015) The phosphatase DUSP2 controls the activity of the transcription activator STAT3 and regulates TH17 differentiation. Nature immunology 16: 1263–1273. pmid:26479789
  16. 16. Nicholson SE, De Souza D, Fabri LJ, Corbin J, Willson TA, et al. (2000) Suppressor of cytokine signaling-3 preferentially binds to the SHP-2-binding site on the shared cytokine receptor subunit gp130. Proceedings of the National Academy of Sciences of the United States of America 97: 6493–6498. pmid:10829066
  17. 17. Sasaki A, Yasukawa H, Suzuki A, Kamizono S, Syoda T, et al. (1999) Cytokine-inducible SH2 protein-3 (CIS3/SOCS3) inhibits Janus tyrosine kinase by binding through the N-terminal kinase inhibitory region as well as SH2 domain. Genes to cells: devoted to molecular & cellular mechanisms 4: 339–351.
  18. 18. Chung CD, Liao J, Liu B, Rao X, Jay P, et al. (1997) Specific inhibition of Stat3 signal transduction by PIAS3. Science 278: 1803–1805. pmid:9388184
  19. 19. King CA (2013) Kaposi's sarcoma-associated herpesvirus kaposin B induces unique monophosphorylation of STAT3 at serine 727 and MK2-mediated inactivation of the STAT3 transcriptional repressor TRIM28. J Virol 87: 8779–8791. pmid:23740979
  20. 20. Wang Y, Lu Y, Toh ST, Sung WK, Tan P, et al. (2010) Lethal-7 is down-regulated by the hepatitis B virus x protein and targets signal transducer and activator of transcription 3. Journal of hepatology 53: 57–66. pmid:20447714
  21. 21. Liao XH, Xiang Y, Yu CX, Li JP, Li H, et al. (2017) STAT3 is required for MiR-17-5p-mediated sensitization to chemotherapy-induced apoptosis in breast cancer cells. Oncotarget 8: 15763–15774. pmid:28178652
  22. 22. Qin L, Li R, Zhang J, Li A, Luo R (2015) Special suppressive role of miR-29b in HER2-positive breast cancer cells by targeting Stat3. American journal of translational research 7: 878–890. pmid:26175849
  23. 23. Cheng Y, Li Y, Nian Y, Liu D, Dai F, et al. (2015) STAT3 is involved in miR-124-mediated suppressive effects on esophageal cancer cells. BMC cancer 15: 306. pmid:25928665
  24. 24. Hong L, Ya-Wei L, Hai W, Qiang Z, Jun-Jie L, et al. (2016) MiR-519a functions as a tumor suppressor in glioma by targeting the oncogenic STAT3 pathway. Journal of neuro-oncology 128: 35–45. pmid:26970980
  25. 25. Patel K, Kollory A, Takashima A, Sarkar S, Faller DV, et al. (2014) MicroRNA let-7 downregulates STAT3 phosphorylation in pancreatic cancer cells by increasing SOCS3 expression. Cancer letters 347: 54–64. pmid:24491408
  26. 26. Hatziapostolou M, Polytarchou C, Aggelidou E, Drakaki A, Poultsides GA, et al. (2011) An HNF4alpha-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell 147: 1233–1247. pmid:22153071
  27. 27. Van Renne N, Roca Suarez AA, Duong FH, Gondeau C, Calabrese D, et al. (2017) miR-135a-5p-mediated downregulation of protein tyrosine phosphatase receptor delta is a candidate driver of HCV-associated hepatocarcinogenesis. Gut pii: gutjnl-2016-312270.
  28. 28. Devhare PB, Sasaki R, Shrivastava S, Di Bisceglie AM, Ray R, et al. (2017) Exosome-Mediated Intercellular Communication between Hepatitis C Virus-Infected Hepatocytes and Hepatic Stellate Cells. J Virol 91.
  29. 29. Yang J, Chatterjee-Kishore M, Staugaitis SM, Nguyen H, Schlessinger K, et al. (2005) Novel roles of unphosphorylated STAT3 in oncogenesis and transcriptional regulation. Cancer research 65: 939–947. pmid:15705894
  30. 30. Yang J, Liao X, Agarwal MK, Barnes L, Auron PE, et al. (2007) Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes & development 21: 1396–1408.
  31. 31. Ng DC, Lin BH, Lim CP, Huang G, Zhang T, et al. (2006) Stat3 regulates microtubules by antagonizing the depolymerization activity of stathmin. The Journal of cell biology 172: 245–257. pmid:16401721
  32. 32. Sehgal PB (2008) Paradigm shifts in the cell biology of STAT signaling. Seminars in cell & developmental biology 19: 329–340.
  33. 33. Liu F, Poursine-Laurent J, Wu HY, Link DC (1997) Interleukin-6 and the granulocyte colony-stimulating factor receptor are major independent regulators of granulopoiesis in vivo but are not required for lineage commitment or terminal differentiation. Blood 90: 2583–2590. pmid:9326224
  34. 34. Hurst SM, Wilkinson TS, McLoughlin RM, Jones S, Horiuchi S, et al. (2001) Il-6 and its soluble receptor orchestrate a temporal switch in the pattern of leukocyte recruitment seen during acute inflammation. Immunity 14: 705–714. pmid:11420041
  35. 35. McLoughlin RM, Jenkins BJ, Grail D, Williams AS, Fielding CA, et al. (2005) IL-6 trans-signaling via STAT3 directs T cell infiltration in acute inflammation. Proceedings of the National Academy of Sciences of the United States of America 102: 9589–9594. pmid:15976028
  36. 36. Romano M, Sironi M, Toniatti C, Polentarutti N, Fruscella P, et al. (1997) Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment. Immunity 6: 315–325. pmid:9075932
  37. 37. Chomarat P, Banchereau J, Davoust J, Palucka AK (2000) IL-6 switches the differentiation of monocytes from dendritic cells to macrophages. Nat Immunol 1: 510–514. pmid:11101873
  38. 38. Rochman I, Paul WE, Ben-Sasson SZ (2005) IL-6 increases primed cell expansion and survival. Journal of immunology 174: 4761–4767.
  39. 39. Yu CR, Dambuza IM, Lee YJ, Frank GM, Egwuagu CE (2013) STAT3 regulates proliferation and survival of CD8+ T cells: enhances effector responses to HSV-1 infection, and inhibits IL-10+ regulatory CD8+ T cells in autoimmune uveitis. Mediators of inflammation 2013: 359674. pmid:24204098
  40. 40. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, et al. (2006) The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126: 1121–1133. pmid:16990136
  41. 41. Nowell MA, Williams AS, Carty SA, Scheller J, Hayes AJ, et al. (2009) Therapeutic targeting of IL-6 trans signaling counteracts STAT3 control of experimental inflammatory arthritis. Journal of immunology 182: 613–622.
  42. 42. Korn T, Mitsdoerffer M, Croxford AL, Awasthi A, Dardalhon VA, et al. (2008) IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells. Proceedings of the National Academy of Sciences of the United States of America 105: 18460–18465. pmid:19015529
  43. 43. Eto D, Lao C, DiToro D, Barnett B, Escobar TC, et al. (2011) IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS ONE 6: e17739. pmid:21423809
  44. 44. Ma CS, Avery DT, Chan A, Batten M, Bustamante J, et al. (2012) Functional STAT3 deficiency compromises the generation of human T follicular helper cells. Blood 119: 3997–4008. pmid:22403255
  45. 45. Ma CS, Deenick EK, Batten M, Tangye SG (2012) The origins, function, and regulation of T follicular helper cells. The Journal of experimental medicine 209: 1241–1253. pmid:22753927
  46. 46. Ouyang W, Rutz S, Crellin NK, Valdez PA, Hymowitz SG (2011) Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annual review of immunology 29: 71–109. pmid:21166540
  47. 47. Wu C, Orozco C, Boyer J, Leglise M, Goodale J, et al. (2009) BioGPS: an extensible and customizable portal for querying and organizing gene annotation resources. Genome Biol 10: R130. pmid:19919682
  48. 48. Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, et al. (2003) IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nature immunology 4: 551–556. pmid:12754507
  49. 49. Yoshimura A, Naka T, Kubo M (2007) SOCS proteins, cytokine signalling and immune regulation. Nature reviews Immunology 7: 454–465. pmid:17525754
  50. 50. Cavani A, Nasorri F, Prezzi C, Sebastiani S, Albanesi C, et al. (2000) Human CD4+ T lymphocytes with remarkable regulatory functions on dendritic cells and nickel-specific Th1 immune responses. J Invest Dermatol 114: 295–302. pmid:10651989
  51. 51. Itoh K, Hirohata S (1995) The role of IL-10 in human B cell activation, proliferation, and differentiation. J Immunol 154: 4341–4350. pmid:7722292
  52. 52. Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, et al. (2011) Defective IL-10 signaling in hyper-IgE syndrome results in impaired generation of tolerogenic dendritic cells and induced regulatory T cells. J Exp Med 208: 235–249. pmid:21300911
  53. 53. Raftery N, Stevenson NJ (2017) Advances in anti-viral immune defence: revealing the importance of the IFN JAK/STAT pathway. Cell Mol Life Sci 74: 2525–2535. pmid:28432378
  54. 54. Darnell JE Jr., Kerr IM, Stark GR (1994) Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264: 1415–1421. pmid:8197455
  55. 55. Velichko S, Wagner TC, Turkson J, Jove R, Croze E (2002) STAT3 activation by type I interferons is dependent on specific tyrosines located in the cytoplasmic domain of interferon receptor chain 2c. Activation of multiple STATS proceeds through the redundant usage of two tyrosine residues. The Journal of biological chemistry 277: 35635–35641. pmid:12105218
  56. 56. Wang WB, Levy DE, Lee CK (2011) STAT3 negatively regulates type I IFN-mediated antiviral response. Journal of immunology 187: 2578–2585.
  57. 57. Ho HH, Ivashkiv LB (2006) Role of STAT3 in type I interferon responses. Negative regulation of STAT1-dependent inflammatory gene activation. The Journal of biological chemistry 281: 14111–14118. pmid:16571725
  58. 58. Lupberger J, Duong FH, Fofana I, Zona L, Xiao F, et al. (2013) Epidermal growth factor receptor signaling impairs the antiviral activity of interferon-alpha. Hepatology 58: 1225–1235. pmid:23519785
  59. 59. Icardi L, Mori R, Gesellchen V, Eyckerman S, De Cauwer L, et al. (2012) The Sin3a repressor complex is a master regulator of STAT transcriptional activity. Proceedings of the National Academy of Sciences of the United States of America 109: 12058–12063. pmid:22783022
  60. 60. Ivashkiv LB, Donlin LT (2014) Regulation of type I interferon responses. Nat Rev Immunol 14: 36–49. pmid:24362405
  61. 61. Heim MH (2015) Interferon signaling. In: Jean-François Dufour P-AC, editor. Signaling pathways in liver diseases. 3rd ed: Wiley Blackwell. pp. 214–225.
  62. 62. Karin M, Clevers H (2016) Reparative inflammation takes charge of tissue regeneration. Nature 529: 307–315. pmid:26791721
  63. 63. Michalopoulos GK (2013) Principles of liver regeneration and growth homeostasis. Comprehensive Physiology 3: 485–513. pmid:23720294
  64. 64. Michalopoulos GK (2014) Advances in liver regeneration. Expert review of gastroenterology & hepatology 8: 897–907.
  65. 65. Robinson MW, Harmon C, O'Farrelly C (2016) Liver immunology and its role in inflammation and homeostasis. Cellular & molecular immunology 13: 267–276.
  66. 66. Selzner N, Selzner M, Odermatt B, Tian Y, Van Rooijen N, et al. (2003) ICAM-1 triggers liver regeneration through leukocyte recruitment and Kupffer cell-dependent release of TNF-alpha/IL-6 in mice. Gastroenterology 124: 692–700. pmid:12612908
  67. 67. Michalopoulos GK (2007) Liver regeneration. Journal of cellular physiology 213: 286–300. pmid:17559071
  68. 68. Yu H, Lee H, Herrmann A, Buettner R, Jove R (2014) Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nature reviews Cancer 14: 736–746. pmid:25342631
  69. 69. Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, et al. (2007) STAT3 mutations in the hyper-IgE syndrome. The New England journal of medicine 357: 1608–1619. pmid:17881745
  70. 70. Siegel AM, Heimall J, Freeman AF, Hsu AP, Brittain E, et al. (2011) A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity 35: 806–818. pmid:22118528
  71. 71. Waris G, Siddiqui A (2002) Interaction between STAT-3 and HNF-3 leads to the activation of liver-specific hepatitis B virus enhancer 1 function. J Virol 76: 2721–2729. pmid:11861839
  72. 72. Lee YH, Yun Y (1998) HBx protein of hepatitis B virus activates Jak1-STAT signaling. J Biol Chem 273: 25510–25515. pmid:9738022
  73. 73. Yuan K, Lei Y, Chen HN, Chen Y, Zhang T, et al. (2016) HBV-induced ROS accumulation promotes hepatocarcinogenesis through Snail-mediated epigenetic silencing of SOCS3. Cell Death Differ 23: 616–627. pmid:26794444
  74. 74. McCartney EM, Helbig KJ, Narayana SK, Eyre NS, Aloia AL, et al. (2013) Signal transducer and activator of transcription 3 is a proviral host factor for hepatitis C virus. Hepatology 58: 1558–1568. pmid:23703790
  75. 75. Yoshida T, Hanada T, Tokuhisa T, Kosai K, Sata M, et al. (2002) Activation of STAT3 by the hepatitis C virus core protein leads to cellular transformation. The Journal of experimental medicine 196: 641–653. pmid:12208879
  76. 76. Gong G, Waris G, Tanveer R, Siddiqui A (2001) Human hepatitis C virus NS5A protein alters intracellular calcium levels, induces oxidative stress, and activates STAT-3 and NF-kappa B. Proc Natl Acad Sci U S A 98: 9599–9604. pmid:11481452
  77. 77. Pinkham C, An S, Lundberg L, Bansal N, Benedict A, et al. (2016) The role of signal transducer and activator of transcription 3 in Rift Valley fever virus infection. Virology 496: 175–185. pmid:27318793
  78. 78. Slinger E, Maussang D, Schreiber A, Siderius M, Rahbar A, et al. (2010) HCMV-encoded chemokine receptor US28 mediates proliferative signaling through the IL-6-STAT3 axis. Sci Signal 3: ra58. pmid:20682912
  79. 79. Lepiller Q, Abbas W, Kumar A, Tripathy MK, Herbein G (2013) HCMV activates the IL-6-JAK-STAT3 axis in HepG2 cells and primary human hepatocytes. PLoS ONE 8: e59591. pmid:23555719
  80. 80. Kotenko SV, Saccani S, Izotova LS, Mirochnitchenko OV, Pestka S (2000) Human cytomegalovirus harbors its own unique IL-10 homolog (cmvIL-10). Proc Natl Acad Sci U S A 97: 1695–1700. pmid:10677520
  81. 81. Raftery MJ, Wieland D, Gronewald S, Kraus AA, Giese T, et al. (2004) Shaping phenotype, function, and survival of dendritic cells by cytomegalovirus-encoded IL-10. J Immunol 173: 3383–3391. pmid:15322202
  82. 82. Spencer JV (2007) The cytomegalovirus homolog of interleukin-10 requires phosphatidylinositol 3-kinase activity for inhibition of cytokine synthesis in monocytes. J Virol 81: 2083–2086. pmid:17121792
  83. 83. Chen H, Hutt-Fletcher L, Cao L, Hayward SD (2003) A positive autoregulatory loop of LMP1 expression and STAT activation in epithelial cells latently infected with Epstein-Barr virus. J Virol 77: 4139–4148. pmid:12634372
  84. 84. Kung CP, Meckes DG Jr., Raab-Traub N (2011) Epstein-Barr virus LMP1 activates EGFR, STAT3, and ERK through effects on PKCdelta. J Virol 85: 4399–4408. pmid:21307189
  85. 85. Muromoto R, Ikeda O, Okabe K, Togi S, Kamitani S, et al. (2009) Epstein-Barr virus-derived EBNA2 regulates STAT3 activation. Biochem Biophys Res Commun 378: 439–443. pmid:19032945
  86. 86. Moore KW, Vieira P, Fiorentino DF, Trounstine ML, Khan TA, et al. (1990) Homology of cytokine synthesis inhibitory factor (IL-10) to the Epstein-Barr virus gene BCRFI. Science 248: 1230–1234. pmid:2161559
  87. 87. Ding Y, Qin L, Kotenko SV, Pestka S, Bromberg JS (2000) A single amino acid determines the immunostimulatory activity of interleukin 10. J Exp Med 191: 213–224. pmid:10637267
  88. 88. Wan X, Wang H, Nicholas J (1999) Human herpesvirus 8 interleukin-6 (vIL-6) signals through gp130 but has structural and receptor-binding properties distinct from those of human IL-6. J Virol 73: 8268–8278. pmid:10482577
  89. 89. Santarelli R, Gonnella R, Di Giovenale G, Cuomo L, Capobianchi A, et al. (2014) STAT3 activation by KSHV correlates with IL-10, IL-6 and IL-23 release and an autophagic block in dendritic cells. Sci Rep 4: 4241. pmid:24577500
  90. 90. Sen N, Che X, Rajamani J, Zerboni L, Sung P, et al. (2012) Signal transducer and activator of transcription 3 (STAT3) and survivin induction by varicella-zoster virus promote replication and skin pathogenesis. Proc Natl Acad Sci U S A 109: 600–605. pmid:22190485
  91. 91. Docherty JJ, Sweet TJ, Bailey E, Faith SA, Booth T (2006) Resveratrol inhibition of varicella-zoster virus replication In vitro. Antiviral Res 72: 171–177. pmid:16899306
  92. 92. Zhu S, Luo H, Liu H, Ha Y, Mays ER, et al. (2017) p38MAPK plays a critical role in induction of a pro-inflammatory phenotype of retinal Muller cells following Zika virus infection. Antiviral Res 145: 70–81. pmid:28739278
  93. 93. Aid M, Abbink P, Larocca RA, Boyd M, Nityanandam R, et al. (2017) Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys. Cell 169: 610–620 e614. pmid:28457610
  94. 94. Ulane CM, Rodriguez JJ, Parisien JP, Horvath CM (2003) STAT3 ubiquitylation and degradation by mumps virus suppress cytokine and oncogene signaling. J Virol 77: 6385–6393. pmid:12743296
  95. 95. Palosaari H, Parisien JP, Rodriguez JJ, Ulane CM, Horvath CM (2003) STAT protein interference and suppression of cytokine signal transduction by measles virus V protein. J Virol 77: 7635–7644. pmid:12805463
  96. 96. Hui KP, Li HS, Cheung MC, Chan RW, Yuen KM, et al. (2016) Highly pathogenic avian influenza H5N1 virus delays apoptotic responses via activation of STAT3. Sci Rep 6: 28593. pmid:27344974
  97. 97. Jia D, Rahbar R, Chan RW, Lee SM, Chan MC, et al. (2010) Influenza virus non-structural protein 1 (NS1) disrupts interferon signaling. PLoS ONE 5: e13927. pmid:21085662
  98. 98. Chandra V, Kar-Roy A, Kumari S, Mayor S, Jameel S (2008) The hepatitis E virus ORF3 protein modulates epidermal growth factor receptor trafficking, STAT3 translocation, and the acute-phase response. J Virol 82: 7100–7110. pmid:18448545
  99. 99. Lieu KG, Brice A, Wiltzer L, Hirst B, Jans DA, et al. (2013) The rabies virus interferon antagonist P protein interacts with activated STAT3 and inhibits Gp130 receptor signaling. J Virol 87: 8261–8265. pmid:23698294
  100. 100. Mitzel DN, Jaramillo RJ, Stout-Delgado H, Senft AP, Harrod KS (2014) Human metapneumovirus inhibits the IL-6-induced JAK/STAT3 signalling cascade in airway epithelium. J Gen Virol 95: 26–37. pmid:24114793
  101. 101. Reitsma JM, Sato H, Nevels M, Terhune SS, Paulus C (2013) Human cytomegalovirus IE1 protein disrupts interleukin-6 signaling by sequestering STAT3 in the nucleus. J Virol 87: 10763–10776. pmid:23903834
  102. 102. Ramalingam D, Ziegelbauer JM (2017) Viral microRNAs Target a Gene Network, Inhibit STAT Activation, and Suppress Interferon Responses. Sci Rep 7: 40813. pmid:28102325
  103. 103. Li L, Chong HC, Ng SY, Kwok KW, Teo Z, et al. (2015) Angiopoietin-like 4 Increases Pulmonary Tissue Leakiness and Damage during Influenza Pneumonia. Cell Rep.
  104. 104. Kane M, Golovkina T (2010) Common threads in persistent viral infections. J Virol 84: 4116–4123. pmid:19955304
  105. 105. Danthi P (2016) Viruses and the Diversity of Cell Death. Annu Rev Virol 3: 533–553. pmid:27501259
  106. 106. Yu H, Kortylewski M, Pardoll D (2007) Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol 7: 41–51. pmid:17186030
  107. 107. Koganti S, Clark C, Zhi J, Li X, Chen EI, et al. (2015) Cellular STAT3 functions via PCBP2 to restrain Epstein-Barr Virus lytic activation in B lymphocytes. J Virol 89: 5002–5011. pmid:25717101
  108. 108. King CA, Li X, Barbachano-Guerrero A, Bhaduri-McIntosh S (2015) STAT3 Regulates Lytic Activation of Kaposi's Sarcoma-Associated Herpesvirus. J Virol 89: 11347–11355. pmid:26339061
  109. 109. Zhang D, Sun M, Samols D, Kushner I (1996) STAT3 participates in transcriptional activation of the C-reactive protein gene by interleukin-6. J Biol Chem 271: 9503–9509. pmid:8621622
  110. 110. Lv D, Zhang Y, Kim HJ, Zhang L, Ma X (2013) CCL5 as a potential immunotherapeutic target in triple-negative breast cancer. Cell Mol Immunol 10: 303–310. pmid:23376885
  111. 111. Halary F, Amara A, Lortat-Jacob H, Messerle M, Delaunay T, et al. (2002) Human cytomegalovirus binding to DC-SIGN is required for dendritic cell infection and target cell trans-infection. Immunity 17: 653–664. pmid:12433371
  112. 112. Shin EC, Sung PS, Park SH (2016) Immune responses and immunopathology in acute and chronic viral hepatitis. Nat Rev Immunol 16: 509–523. pmid:27374637
  113. 113. Ren JP, Wang L, Zhao J, Ning SB, El Gazzar M, et al. (2017) Decline of miR-124 in myeloid cells promotes regulatory T-cell development in hepatitis C virus infection. Immunology 150: 213–220. pmid:27753084
  114. 114. Zhai N, Li H, Song H, Yang Y, Cui A, et al. (2017) Hepatitis C Virus Induces MDSCs-Like Monocytes through TLR2/PI3K/AKT/STAT3 Signaling. PLoS ONE 12: e0170516. pmid:28114346
  115. 115. Wan S, Kuo N, Kryczek I, Zou W, Welling TH (2015) Myeloid cells in hepatocellular carcinoma. Hepatology 62: 1304–1312. pmid:25914264
  116. 116. Wolk B, Buchele B, Moradpour D, Rice CM (2008) A dynamic view of hepatitis C virus replication complexes. J Virol 82: 10519–10531. pmid:18715913
  117. 117. Roohvand F, Maillard P, Lavergne JP, Boulant S, Walic M, et al. (2009) Initiation of hepatitis C virus infection requires the dynamic microtubule network: role of the viral nucleocapsid protein. J Biol Chem 284: 13778–13791. pmid:19269968
  118. 118. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, et al. (2016) Hepatocellular carcinoma. Nat Rev Dis Primers 2: 16018. pmid:27158749
  119. 119. Ho Y, Tsao SW, Zeng M, Lui VW (2013) STAT3 as a therapeutic target for Epstein-Barr virus (EBV): associated nasopharyngeal carcinoma. Cancer Lett 330: 141–149. pmid:23220625
  120. 120. Lui VW, Wong EY, Ho Y, Hong B, Wong SC, et al. (2009) STAT3 activation contributes directly to Epstein-Barr virus-mediated invasiveness of nasopharyngeal cancer cells In vitro. Int J Cancer 125: 1884–1893. pmid:19588483
  121. 121. Kondo S, Yoshizaki T, Wakisaka N, Horikawa T, Murono S, et al. (2007) MUC1 induced by Epstein-Barr virus latent membrane protein 1 causes dissociation of the cell-matrix interaction and cellular invasiveness via STAT signaling. J Virol 81: 1554–1562. pmid:17151127
  122. 122. Wong ALA, Hirpara JL, Pervaiz S, Eu JQ, Sethi G, et al. (2017) Do STAT3 inhibitors have potential in the future for cancer therapy? Expert Opin Investig Drugs 26: 883–887. pmid:28714740
  123. 123. Niu Y, Si Y, Li Y, Chi X, Li X, et al. (2015) A novel small-molecule inhibitor of hepatitis C virus replication acts by suppressing signal transducer and activator of transcription 3. J Antimicrob Chemother 70: 2013–2023. pmid:25858355
  124. 124. Yang Y, Zheng B, Han Q, Zhang C, Tian Z, et al. (2016) Targeting blockage of STAT3 inhibits hepatitis B virus-related hepatocellular carcinoma. Cancer Biol Ther 17: 449–456. pmid:26934469
  125. 125. Miklossy G, Hilliard TS, Turkson J (2013) Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov 12: 611–629. pmid:23903221
  126. 126. Espinoza JL, Takami A, Trung LQ, Kato S, Nakao S (2012) Resveratrol prevents EBV transformation and inhibits the outgrowth of EBV-immortalized human B cells. PLoS ONE 7: e51306. pmid:23251493
  127. 127. De Leo A, Arena G, Lacanna E, Oliviero G, Colavita F, et al. (2012) Resveratrol inhibits Epstein Barr Virus lytic cycle in Burkitt's lymphoma cells by affecting multiple molecular targets. Antiviral Res 96: 196–202. pmid:22985630
  128. 128. Liu CL, Hung HC, Lo SC, Chiang CH, Chen IJ, et al. (2016) Using mutagenesis to explore conserved residues in the RNA-binding groove of influenza A virus nucleoprotein for antiviral drug development. Sci Rep 6: 21662. pmid:26916998
  129. 129. Kim K, Kim KH, Kim HY, Cho HK, Sakamoto N, et al. (2010) Curcumin inhibits hepatitis C virus replication via suppressing the Akt-SREBP-1 pathway. FEBS Lett 584: 707–712. pmid:20026048
  130. 130. Michaelis M, Paulus C, Loschmann N, Dauth S, Stange E, et al. (2011) The multi-targeted kinase inhibitor sorafenib inhibits human cytomegalovirus replication. Cell Mol Life Sci 68: 1079–1090. pmid:20803231
  131. 131. Furtek SL, Backos DS, Matheson CJ, Reigan P (2016) Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem Biol 11: 308–318. pmid:26730496
  132. 132. Gharwan H, Groninger H (2016) Kinase inhibitors and monoclonal antibodies in oncology: clinical implications. Nat Rev Clin Oncol 13: 209–227. pmid:26718105
  133. 133. Heppler LN, Frank DA (2017) Targeting Oncogenic Transcription Factors: Therapeutic Implications of Endogenous STAT Inhibitors. Trends Cancer 3: 816–827. pmid:29198438
  134. 134. Baell J, Walters MA (2014) Chemistry: Chemical con artists foil drug discovery. Nature 513: 481–483. pmid:25254460
  135. 135. Clinicaltrials.gov (2017) AZD9150 With MEDI4736 in Patients With Advanced Pancreatic, Non-Small Lung and Colorectal Cancer.
  136. 136. Clinicaltrials.gov (2017) MEDI4736 Alone and in Combination With Tremelimumab or AZD9150 in Adult Subjects With Relapsed/Refractory DLBCL.
  137. 137. Clinicaltrials.gov (2017) Study to Assess MEDI4736 With Either AZD9150 or AZD5069 in Advanced Solid Tumors & Relapsed Metastatic Squamous Cell Carcinoma of Head & Neck.
  138. 138. Hubbard JM, Grothey A (2017) Napabucasin: An Update on the First-in-Class Cancer Stemness Inhibitor. Drugs 77: 1091–1103. pmid:28573435