Extracellular signal-related kinases 1 and 2 (ERK1/2) are the final components of the mitogen-activated protein kinase (MAPK) phosphorylation cascade, an integral module in a diverse array of signalling pathways for shaping cell behaviour and fate. More recently, studies have shown that ERK1/2 plays an essential role downstream of immune receptors to elicit inflammatory gene expression in response to infection and cell or tissue damage. Much of this work has studied ERK1/2 activation in Toll-like receptor (TLR) pathways, providing mechanistic insights into its recruitment, compartmentalisation and activation in cells of the innate immune system. In this review, we summarise the typical activation of ERK1/2 in growth factor receptor pathways before discussing its known roles in immune cell signalling with a focus downstream of TLRs. We examine emerging research uncovering evidence of dysfunctional ERK1/2 signalling in inflammatory diseases and discuss the potential therapeutic benefit of targeting ERK1/2 pathways in inflammation.

Cells utilise a range of receptors on their surface to sense and respond to external cues from the environment. Receptor tyrosine kinases (RTKs) are a widely studied receptor family which recognise growth factors, cytokines and hormones to control cell fate through differentiation, proliferation, survival, metabolism and migration [1]. Intracellular signal transduction from RTKs is propagated through the activation of cytoplasmic kinases including mitogen-activated protein kinases (MAPKs), which function as important signalling mediators to effect downstream responses. MAPKs are a highly conserved group of serine/threonine kinases which are expressed in a wide range of different cell types and play a central role in cell development and function [2]. The three conventional families of MAPKs in mammals are JUN N-terminal kinases (JNK1, JNK2 and JNK3), p38 kinases (p38-α, p38-β, p38-γ and p38-δ) and extracellular signal-regulated kinases (ERK1, ERK2 and ERK5), each of which exist as multiple isoforms and are activated in a cascade comprising orchestrated signal transduction through MAPKKK (MAP3K) phosphorylation of MAPKKs (MAP2Ks), in turn phosphorylating MAPKs for activation of downstream targets. The roles of these MAPKs in a large array of cellular pathways have been extensively reviewed [3].

Amongst MAPK family members, ERK has attracted the greatest characterisation in the literature. The structurally related isoforms ERK1 and ERK2 were the first MAPKs to be identified through early observations of the rapid phosphorylation of two comparably sized proteins in response to growth factor stimulation [4]. Both ERK1 and ERK2 are ubiquitously expressed, with ERK2 generally expressed at slightly higher levels in most mammalian tissues, and they exhibit a level of functional redundancy in many pathways [5]. Whilst mice deficient in ERK1 are viable and show normal development, ERK2-deficient mice display severe developmental defects and embryonic lethality which is rescued by transgenic ERK1 overexpression [6]. Given their similarity, these proteins are hereon collectively referred to using the terms ERK and ERK1/2 interchangeably. In typical ERK1/2 pathways, ligand-induced activation of receptor tyrosine kinases (RTKs) at the plasma membrane activates and recruits the small GTPase RAS in its GTP-bound form which then triggers dimerisation and activation of MAP3K RAF to initiate a downstream phosphorylation cascade for sequential activation of MAP2K MEK and MAPK ERK [7] (Figure 1: Core RAS–ERK pathway). Like ERK1/2, the kinases at each level of the RAS/RAF/MEK/ERK cascade feature distinct isoforms, namely H-RAS/K-RAS/N-RAS, A-RAF/B-RAF/C-RAF and MEK1/MEK2, each encoded by independent genes which often show tissue-specific expression and provide functional diversity [8,9].

Core RAS–ERK pathway downstream of RTKs.

Figure 1.
Core RAS–ERK pathway downstream of RTKs.

Following ligand binding, activated RTKs recruit Grb2 via its SH2 domain. Grb2 associates with the GEF SOS which allows activation of GTP-bound RAS GTPase at the plasma membrane. As part of the core RAS–ERK pathway, RAS activation of RAF then promotes phosphorylation of MEK which phosphorylates and activates ERK. In a negative feedback loop, active ERK phosphorylates SOS to inhibit its interaction with Grb2 and prevent further RAS activation. Cytoplasmic substrates of activated ERK include RSK, MSK and MNK kinase pathways, and nuclear ERK import allows activation of its many nuclear substrates including transcription factors for gene expression in multiple cell fate pathways.

Figure 1.
Core RAS–ERK pathway downstream of RTKs.

Following ligand binding, activated RTKs recruit Grb2 via its SH2 domain. Grb2 associates with the GEF SOS which allows activation of GTP-bound RAS GTPase at the plasma membrane. As part of the core RAS–ERK pathway, RAS activation of RAF then promotes phosphorylation of MEK which phosphorylates and activates ERK. In a negative feedback loop, active ERK phosphorylates SOS to inhibit its interaction with Grb2 and prevent further RAS activation. Cytoplasmic substrates of activated ERK include RSK, MSK and MNK kinase pathways, and nuclear ERK import allows activation of its many nuclear substrates including transcription factors for gene expression in multiple cell fate pathways.

Close modal

ERK1/2 signalling from RTKs

ERK1/2 signalling has been studied extensively downstream of extracellular growth factor receptor (EGFR), an archetypal RTK with a central role in cell proliferation and association with the development of many human cancers [10]. Activated EGFR recruits the adaptor Grb2 and the guanine nucleotide exchange factor (GEF), SOS, to the plasma membrane for activation of membrane-bound RAS GTPase, thereby stimulating the MAPK phosphorylation cascade for ERK1/2 activation (Figure 1). As well as targeting of its downstream substrates, ERK-mediated phosphorylation of SOS causes its disassociation from Grb2 to create a negative feedback mechanism for curtailing RAS activation [11,12]. The recruitment of soluble kinases to specific membrane domains for receptor-mediated signalling has emerged as a critical feature dictating spatiotemporal regulation of signalling cascades. Earlier studies focused only on a general recruitment of RAS to the plasma membrane where it was assumed all receptors were activated to initiate signalling. It has since been recognised that RAS can be partitioned to endomembranes including endosomes, Golgi and endoplasmic reticulum to support RAS–ERK signalling generated by receptors at all of these sites [13]. Thus, mechanisms for the recruitment of specific kinases to subcellular membrane domains or receptors is now a key issue for consideration. To compartmentalise its activity for specific pathways, ERK1/2 is directly recruited to particular membrane domains by scaffolding proteins, which include paxillin, FHL1, GIT1 and PAK1 at focal adhesions [14], CAVIN4, ß-arrestin and MORG1 at G protein-coupled receptors (GPCRs) [15,16] and IQGAPs at the plasma membrane [17]. However, in many cases, the mechanism of ERK1/2 recruitment to these scaffolds or their precise role in ERK signalling remains to be fully established [2].

Upon its activation, ERK1/2 phosphorylates a diverse range of cytoplasmic and nuclear substrates and its repertoire of targets is further expanded through activation of MAPK-activated kinases such as ribosomal protein S6 kinase (RSK), mitogen- and stress-activated protein kinase (MSK) and MAPK-interacting kinase (MNK) pathways [18] (Figure 1). To afford access to its nuclear substrates, activated ERK1/2 is shuttled from the cytoplasm to the nucleus by diffusion through the nuclear pores, a process which is facilitated by an interaction with nucleoporins and phosphorylation of nuclear pore subunits [19,20]. A common class of ERK targets in cell proliferation and growth pathways are immediate early genes such as FOS, JUN, MYC and EGR families of transcription factors which are rapidly transcribed following mitogen stimulation of the cell [21]. In cell metabolism pathways, ERK1/2 activates key metabolic regulatory proteins such as mTOR complex 1 (mTORC1) and transcription factor HIF1α for control of protein synthesis, respiration and glucose metabolism [22,23], and plays a crucial role in cell differentiation and development through regulation of pluripotency transcription factors including OCT4, KLF2 and STAT3 [24–26].

Since its initial discovery, ERK1/2 signalling has attracted a significant weight of literature detailing its role in determining fundamental cellular behaviours [2,27,28]. Further to these diverse pathways, ERK1/2 plays an essential role in immune cells for shaping the inflammatory response to infection and cell damage [29]. In this review, we detail the known roles of ERK1/2 in immune cells with a focus on innate immune signalling downstream of Toll-like receptors (TLRs). We then highlight spatiotemporal aspects of ERK1/2 signalling in these pathways, discuss associations with inflammatory disease and finally discuss ERK1/2 signalling as a therapeutic target in these disease settings.

Innate immune cells provide the first line of defence against infection through the activation of germline-encoded transmembrane pattern recognition receptors (PRRs). PRRs detect molecular signatures from foreign pathogens, termed pathogen-associated molecular patterns (PAMPs), as well as endogenous damage-associated molecular patterns (DAMPs) associated with tissue damage and inflammation [30]. Activation of PRRs induces the production of pathogen-targeting molecules such as antimicrobial peptides and inducible nitric oxide synthase as well as inflammatory cytokines and chemokines for the recruitment of additional immune cells and induction of the adaptive immune response [31]. Innate immune PRRs include TLRs, NOD-like receptors (NLRs), RIG-I-like receptors (RLRs) and C-type lectin receptors (CLRs), and the activation of p38, JNK and ERK1/2 MAPK pathways by these PRRs, as part of the inflammatory response, is well described [32,33].

ERK1/2 signalling from TLRs

MAPK activation in innate immune cells such as macrophages and dendritic cells (DCs) has largely been studied downstream of TLRs, a highly conserved family of transmembrane receptors which are poised to recognise a diverse range of specific DAMPs or PAMPs both from the cell surface and within the cell from endosomal membranes [34]. Due to their exposed position, cell surface TLRs typically recognise extracellular microbial signatures, whereas endosomal TLRs detect nucleic acids from internalised pathogens as well as self-derived nucleic acids from damaged cells or tissue [35]. Stimulated TLRs recruit specific sets of cytoplasmic adaptors through their homologous toll–interleukin 1 receptor (TIR) domains which orchestrate the initiation, duration and spatial organisation of downstream signalling pathways for inflammatory gene expression, and include classical signalling adaptors MYD88, MAL, TRIF and TRAM [36]. Activation of MYD88 downstream of all TLRs, with the exception of TLR3 which instead signals solely through TRIF [37], rapidly induces the recruitment of kinases IRAK1, IRAK2 and IRAK4 to form the multiprotein ‘myddosome’ complex which triggers a signal transduction cascade to activate the inflammatory transcription factor nuclear factor κB (NF-κB) [36,38] (Figure 2A). NF-κB is a master regulator of inflammation, promoting the expression of a wide range of pro-inflammatory cytokines including IL-1ß, IL-6, IL-12p40 and TNF in response to activation of both MYD88-dependent and MYD88-independent signalling arms [39]. The E3 ubiquitin ligase TRAF6 plays a critical role in this canonical signalling cascade by activating the kinase TAK1 through adaptors TAB2/3 which then allows IκB kinase (IKK) to promote proteasomal degradation of the cytoplasmic inhibitor of NF-κB, IκB, thereby releasing NF-κB to translocate into the nucleus for modulation of gene transcription [40]. Importantly, TRAF6-activated TAK1 also induces MAPK signalling for activation of the activator protein 1 (AP-1) transcription factor complex [41] (Figure 2A). The gene expression regulatory complex AP-1 is activated in response to a broad range of external stimuli where it regulates cellular processes such as cell survival, differentiation and proliferation, and it is a key promoter of pro-inflammatory gene expression in immune cells [42]. AP-1 is formed through homodimer or heterodimer assembly of c-Fos, c-Jun and activating transcription factor (ATF) monomers and its transcriptional activity is tightly regulated though subunit abundance, composition and phosphorylation [43].

ERK activation downstream of TLR4.

Figure 2.
ERK activation downstream of TLR4.

(A) LPS binding to TLR4 on the cell surface stimulates the recruitment of MYD88 through a homologous TIR domain interaction (shown in purple). Subsequent MYD88-IRAK oligomer formation activates TRAF6 and TAK1 to promote IKK-mediated degradation of IκB to allow the nuclear translocation of NF-κB for inflammatory cytokine expression. IKK also promotes proteolysis of TPL2 inhibitory protein p105 to activate the TPL2-ERK pathway for ERK phosphorylation and activation of nuclear AP-1 component c-Fos. (B) LPS-activated TLR4 directly interacts with transmembrane adaptor SCIMP through a TIR-non-TIR interaction. SCIMP scaffolds Lyn kinase through its proline-rich domain (PRD; shown in green) and recruits a subset of ERK to TLR4 on the cell surface and macropinosomes for phosphorylation of c-Fos which translocates to the nucleus for pro-inflammatory cytokine expression. Dashed blue arrow between pathways shows possible TPL2 activation of SCIMP-scaffolded ERK.

Figure 2.
ERK activation downstream of TLR4.

(A) LPS binding to TLR4 on the cell surface stimulates the recruitment of MYD88 through a homologous TIR domain interaction (shown in purple). Subsequent MYD88-IRAK oligomer formation activates TRAF6 and TAK1 to promote IKK-mediated degradation of IκB to allow the nuclear translocation of NF-κB for inflammatory cytokine expression. IKK also promotes proteolysis of TPL2 inhibitory protein p105 to activate the TPL2-ERK pathway for ERK phosphorylation and activation of nuclear AP-1 component c-Fos. (B) LPS-activated TLR4 directly interacts with transmembrane adaptor SCIMP through a TIR-non-TIR interaction. SCIMP scaffolds Lyn kinase through its proline-rich domain (PRD; shown in green) and recruits a subset of ERK to TLR4 on the cell surface and macropinosomes for phosphorylation of c-Fos which translocates to the nucleus for pro-inflammatory cytokine expression. Dashed blue arrow between pathways shows possible TPL2 activation of SCIMP-scaffolded ERK.

Close modal

As is the case in cell growth pathways, MAPKs are intricately associated with transcription factor phosphorylation downstream of TLRs. ERK1/2 primarily phosphorylates c-Fos for inflammatory cytokine production [44,45]. c-Jun is primarily phosphorylated by JNK [46,47] which also promotes the nuclear translocation of ATF4 to form heterodimers with c-Jun for AP-1 activation [48]. p38 phosphorylates members of the myocyte-enhancer factor 2 (MEF2) family of transcription factors, MEF2A and MEF2C, downstream of activated TLRs to control anti-inflammatory IL-10 production [49] as well as stimulating expression of c-Jun, thereby influencing AP-1-mediated cytokine expression [50–52].

Whilst early studies indicated that ERK1/2 also induces NF-κB activity through direct IKK and p65 phosphorylation in TLR and IL-1 pathways [53,54], this concept has now been superseded by the premise that ERK1/2 activity instead drives gene expression changes and controls other regulators that support NF-κB activation. For instance, ERK2-mediated phosphorylation of PARP1 has been shown to control NF-κB transcriptional activation in response to TNF stimulation [55], indicating how ERK1/2 can play a role in NF-κB regulation in response to other inflammatory mediators. Ternary complex factors (TCFs) are a major direct downstream target of ERK1/2 in RAS–ERK1/2 pathways [56], and a recent study shows that ERK1/2 also controls c-Fos transcription through activation of the TCF, ELK1, to further modulate c-Fos-dependent cytokine expression following TLR stimulation [57]. As an NF-κB target gene, ELK1 regulation by IKK, therefore, provides a level of cross-talk between transcription factor signalling arms downstream of activated TLRs.

In contrast with RTK pathways which signal through RAF, immune receptors including TLRs, tumour necrosis factor receptor 1 (TNFR1) and interleukin-1 receptor (IL-1R), activate tumour progression locus 2 (TPL2), a MAP3K expressed highly in myeloid cells and fibroblasts which is involved in inflammatory responses and angiogenesis [58]. Downstream of these immune receptors and their adaptors, IKK-dependent proteolysis of p105 couples activation of NF-κB pathways to MAPK signalling by releasing TPL2 from p105 inhibition. After liberation from p105, TPL2 directly phosphorylates MEK and thereby activates downstream ERK MAPK signalling [58,59] (Figure 2A). TPL2 is also directly regulated by IKK-mediated phosphorylation which recruits the scaffold 14-3-3 to its C-terminal tail to stimulate TPL2 kinase activity [60]. In addition to controlling MEK/ERK signalling, TPL2 can also regulate the activation of p38 in macrophages [61] and neutrophils [62] as well as ERK1/2 and JNK in mouse embryonic fibroblasts [63], positioning it as an important regulator of multiple MAPKs in several cell types. Many recent studies have demonstrated that TPL2-mediated activation of ERK1/2 in immune cells can generate diverse cytokine responses, including pro-inflammatory TNF, IL-1ß, IL-6 and IL-12p40 [64–67] as well as anti-inflammatory IL-10 and IFN-ß [68,69], highlighting the many genetic targets of ERK and its substrates in inflammatory pathways.

Whilst scaffolds for targeted ERK1/2 recruitment in other receptor signalling pathways and other cells have largely been identified (see ‘ERK signalling from RTKs’), the mechanism by which ERK1/2 activity is compartmentalised for TLR signalling has so far remained unresolved. We previously described the atypical TLR adaptor SCIMP, a non-TIR transmembrane protein which directly interacts with TLRs and scaffolds effectors and kinases to potentiate TLR phosphorylation, pro-inflammatory signalling and cytokine output [70–72]. As an immune-specific palmitoylated transmembrane adaptor protein (pTRAP), SCIMP clusters with TLRs in lipid raft domains and tetraspanin-enriched microdomains (TEMs) on the cell surface or on endomembranes where it directly recruits the Src-family kinase Lyn for phosphorylation of both activated TLRs, and itself, to drive transient downstream MAPK signalling responses including phosphorylation of ERK1/2 [70,72]. Recently, we showed that SCIMP acts as a transmembrane scaffold for ERK1/2 in TLR pathways, rapidly recruiting ERK2 to SCIMP at TLR4 signalling sites on membrane domains, including on cell surface ruffles and macropinosomes in macrophages [73]. Using advanced live imaging techniques, it was possible to discern the recruitment and enrichment of cytoplasmic ERK2 to these dynamic membrane domains with high-spatiotemporal resolution, for the first time providing evidence for spatiotemporal partitioning of ERK during signalling in TLR pathways (Figure 2B). Biochemical analyses showed that ERK1/2 is recruited to SCIMP that is in a complex with TLR4, and this ERK1/2 primarily activates c-Fos for increased production of pro-inflammatory cytokines TNF, IL-1ß, IL-6 and IL-12p40 but not anti-inflammatory cytokines [73], in contrast with canonical TLR-activated ERK1/2 (Figure 2A) which directs a broader cytokine profile [74]. In this study and previously, SCIMP has been shown to also activate NF-κB in response to TLR stimulation [70,72,73], which may be through ERK1/2 or rather other signalling kinases, for example, MAPKs JNK and p38 also regulated by SCIMP.

Whilst it is unlikely to act in a mutually exclusive manner to TPL2-activated ERK1/2 (Figure 2B), this implies that, as a transmembrane adaptor, SCIMP scaffolds a subset of ERK1/2 to stimulated TLRs to selectively drive rapid, pro-inflammatory responses, perhaps to give spatial or temporal emphasis or indeed to control thresholding (see ‘Homeostatic control of ERK1/2 in immune signalling’), for pro-inflammatory ERK activation. Driving ERK1/2 signalling in TLR pathways aligns with SCIMP's promotion of ERK1/2 activation downstream of other immune receptors in DCs and B cells. As a transmembrane adaptor clustered alongside receptors within signalling microdomains, SCIMP has unique capacity to act as an immune-specific scaffold for membrane recruitment of ERK1/2 and spatiotemporal compartmentalisation of ERK1/2 signalling. With SCIMP setting a precedent as a spatiotemporal, pro-inflammatory ERK adaptor, it will be important to seek other immune adaptors, perhaps other pTRAP family members, and beyond, which can also influence innate immune activation of this critical inflammatory kinase.

ERK1/2 in adaptive immunity

Following the innate immune signalling that issues acute inflammatory responses, ERK1/2 signalling directs the differentiation and activation of cells of the adaptive immune system, providing longer-term protection against re-infection with invading pathogens. In B cells, Raf-mediated ERK1/2 activation by B cell receptors (BCRs) involves adaptor proteins Grb2 and GRAP [75] and controls a transcription factor network to drive B cell development [76]. Using tissue-specific inactivation of ERK2 in ERK1-deficient mice to avoid embryonic lethality, ERK1/2 has been shown to be important for multiple stages of thymic development during T cell differentiation and maturation [77], and ERK2 co-ordinates B cell and CD8 T cell survival through regulation of the transcription and stability of the apoptotic BCL-2 family member, Bim [77,78]. Recently, proteomic analysis of TCR-activated CD8 T cells has shown that ERK1/2 controls many of the key transcriptional regulators of T cell differentiation and activation [79]. ERK1/2, therefore, guides multiple signalling pathways in cells of both the innate and adaptive immune systems and is emerging as a central regulator of immune cell function.

Homeostatic control of ERK1/2 in immune signalling

The threshold of ERK1/2 activation plays an important role in determining the level and type of cellular responses. In RTK proliferation pathways, ERK1/2 signalling exhibits clear threshold effects, whereby too little ERK1/2 activation prevents proliferation, yet too much ERK1/2 activation can drive cell cycle arrest and death [80]. Similarly, ERK1/2 activation thresholds are apparent in the context of immune signalling. BCL3, an IκB protein which regulates NF-κB, also promotes TPL2 degradation to control ERK1/2 activity following TLR stimulation [81]. The stability of TPL2, therefore, determines the activity threshold of ERK1/2 required to initiate the production of inflammatory cytokines in response to TLR stimulation. The identification of SCIMP, as a pro-inflammatory ERK1/2 scaffold for TLRs [73], may also provide a level of thresholding to direct the activity of SCIMP-recruited ERK1/2 towards a pro-inflammatory output.

Another way of controlling the activation threshold of ERK1/2 signalling is through feedback inhibition. In much the same way as ERK1/2 phosphorylates upstream components in a negative feedback mechanism in RAS–ERK pathways, it is possible that ERK1/2 demonstrates a level of feedback inhibition in inflammatory pathways to constrain excessive responses. In B cells, chronic ERK1/2 activation downstream of BCR inhibits TLR-induced B cell differentiation, introducing tolerance of B cells to self-antigens and thereby providing resistance against autoimmunity [82]. However, to date, there have been no reports of direct ERK1/2 phosphorylation of upstream TLR signalling components such as TPL2, and so it remains to be seen if ERK1/2-mediated feedback inhibition or cross-talk plays a role in the homeostatic control of TLR pathways.

The significance of ERK1/2 in immune cell signalling is emphasised through the many ways in which internalised pathogens have evolved mechanisms to modulate ERK signalling in inflammatory pathways as a means of immune evasion. For example, Mycobacterium tuberculosis secretion of tyrosine phosphatase MptpB decreases ERK1/2 phosphorylation in the cytoplasm of macrophages to block the production of IL-6 and activate Akt pathways to enhance cell survival [83]. Salmonella enterica and Shigella flexneri secrete phospho-threonine lyases which target the Thr–X–Tyr activation motif of ERK1/2 and irreversibly inhibit its kinase activities through phosphate cleavage [84,85]. As well as direct targeting of ERK1/2, other bacteria such as Yersinia spp. and Vibrio parahaemolyticus secrete acetyltransferase enzymes to acetylate upstream MEK1/2, preventing its kinase activity and inhibiting ERK1/2 phosphorylation [86,87].

Given the fundamental role of MAPK signalling in the regulation of diverse cellular processes, MAPK signalling dysfunction is well established in many diseases [88]. The RAS–RAF–MEK–ERK pathway is dysregulated in approximately one-third of all human cancers through its central role in cell growth, proliferation and survival pathways [89]. In addition, a recent genome sequencing study found that activating TPL2 mutations are present in 33% of childhood spitzoid melanomas [90]. Although no cancer-associated mutations in ERK itself have been identified to date, K-RAS and B-RAF are widely recognised oncogenes in which constitutively active mutations such as K-RASG12D and B-RAFV600E lead to colorectal cancer, breast cancer, leukaemia and melanoma through persistent activation of RAF–MEK–ERK and PI3K–Akt/mTOR signalling pathways [91,92]. As such, B-RAF and MEK inhibitors have shown promise as cancer therapeutics, however, the development of resistance to inhibitors through gain-of-function mutations may compromise their use in the clinic [93]. ERK1/2-specific inhibitors have been proposed as a way of overcoming resistance to these upstream targets in cancer therapy [94].

Whilst ERK1/2 signalling dysregulation in cancer has been extensively detailed in the literature, the role of ERK1/2 in inflammatory pathways underlies an emerging association with several immune-linked diseases characterised by chronic states of inflammation. Elevated ERK1/2 activity has been found in inflamed joints of patients with the autoimmune disease rheumatoid arthritis (RA) [95] and ERK1/2 inhibitors show therapeutic promise by reducing levels of RA-associated inflammatory cytokines including TNF, IL-1 and IL-6 [96]. ERK1/2 activity has also been linked with chronic lung inflammation through sustaining a cellular state of excessive NF-κB activation in bronchial epithelial cells from patients with a homozygous for the Z mutation in alpha1-antitrypsin, a genetic driver of early onset emphysema [97]. In rodent models of chronic alcohol-induced liver inflammation, ERK1/2 activation downstream of TLR4 promotes altered TNF expression in liver macrophages [98]. In experimental autoimmune encephalomyelitis, an animal model of human multiple sclerosis, inhibition of ERK activation using the upstream MEK inhibitor U0126 blocked IL-23 and IL-1β production in DCs to suppress Th17 and Th1 cell auto-antigen responses and reduce the severity of disease [99]. As in cancer, no inflammation-associated ERK1/2 genetic variants have been identified to date. However, genetic associations between ERK1/2 pathway components and inflammatory disease strengthens the pathogenic role of ERK1/2 signalling in these conditions. For example, a single nucleotide polymorphism (SNP) in TPL2 (rs1042058) confers increased risk of inflammatory bowel disease including Crohn's disease and ulcerative colitis [100], corresponding with enhanced ERK phosphorylation and IL-1β and IL-18 secretion in human monocyte-derived macrophages in vitro[101], and reduced levels of anti-inflammatory IL-10 in patient intestinal biopsies [102].

ERK signalling has also been linked to neurodegenerative conditions such as Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS) which are characterised by aberrant protein aggregation in the central nervous system (CNS), and in which neuroinflammation is commonly a hallmark of disease [103]. ERK1/2 is emerging as a key player in the activation of microglia, the resident macrophages of the CNS, in these diseases. For example, recent in vitro and in vivo studies show that ERK1/2 promotes TLR-mediated pro-inflammatory microglial activation and disease-related gene expression in AD [104,105]. In PD models, ERK1/2 promotes leucine-rich repeat kinase 2 (LRRK2)-mediated microglia activation leading to inflammation and apoptosis as well as mounting in vivo inflammatory responses [106,107]. In non-immune pathways, ERK1/2 activity is associated with defects in mitochondrial morphology and function in AD [108] as well as mitochondrial biogenesis defects and dopaminergic neuronal degeneration in PD [109,110]. ERK1/2 signalling in motor neurons has been also linked to glutamate excitotoxicity and axonal transport defects associated with ALS [111,112]. Crucially, many of these pathogenic defects can be attenuated through pharmacological ERK inhibition in disease models, reflecting the broad functions of ERK signalling in neurodegenerative pathways and highlighting ERK as a potential therapeutic target in these diseases.

Further to ERK1/2 functions in immune cell signalling, its central role in many fundamental non-immune cell pathways must be considered, possibly giving rise to unintended consequences for the use of systemic MEK/ERK inhibition as anti-inflammatory therapeutics. One promising approach to avoid this global blockade of ERK pathways is through inhibition of TPL2 upstream of ERK1/2 in inflammatory pathways, thereby having a more targeted effect on ERK1/2-mediated inflammation. TPL2 itself has been linked to autoimmune inflammation in experimental autoimmune encephalomyelitis mouse models [113,114] and a genetic polymorphism in TPL2 resulting in increased expression is associated with inflammatory bowel disease [101]. Indeed, TPL2 kinase inhibitors demonstrate therapeutic potential in multiple in vivo models of inflammatory diseases including RA, MS, immune thrombocytopenic purpura and inflammatory bowel disease [59]. A TPL2 inhibitor (GS-4875; tilpisertib) showed promise in suppressing inflammation in primary human monocytes [115] and was taken forward to clinical trial for the treatment of ulcerative colitis, and has now been superseded by an updated molecule with greater target coverage. It is, therefore, becoming increasingly clear that targeting ERK pathways either through direct inhibition of ERK1/2 or inhibition of its upstream regulators shows therapeutic potential for the treatment of many inflammatory and autoimmune diseases. Targeting the spatiotemporal membrane recruitment of ERK1/2 through immune-specific adaptors such as SCIMP may offer another disruptive strategy. Future studies will be of great importance to progress the widespread feasibility of ERK1/2 inhibitors and interventions in the clinic.

  • ERK1/2 signalling plays a central role in many fundamental cell behaviours including survival, proliferation, differentiation, migration and critically shapes many aspects of the inflammatory response and fate of immune cells.

  • In innate immune cells, compartmentalisation of ERK1/2 downstream of TLRs allows for spatiotemporal control of signalling and thresholding of inflammatory responses.

  • ERK1/2 signalling dysregulation is linked to many human pathological conditions and emerging studies continue to reveal its impact in chronic or uncontrolled inflammation. Future studies will play an important role in advancing the therapeutic targeting of ERK1/2 pathways in inflammatory disease.

The authors declare that there are no competing interests associated with the manuscript.

The authors are supported by fellowships and grant funding from the Australian Research Council (L.L.: DE180100524) and the National Health and Medical Research Council of Australia (J.L.S.: APP1176209, J.L.S.: APP1138723; L.L.: APP1159106).

Open access for this article was enabled by the participation of the University of Queensland in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with CAUL.

Figures were created with BioRender.com.

AD

Alzheimer's disease

ALS

amyotrophic lateral sclerosis

AP-1

activator protein 1

ATF

activating transcription factor

BCR

B cell receptor

CLR

C-type lectin receptor

CNS

central nervous system

DAMP

damage-associated molecular pattern

EGFR

extracellular growth factor receptor

ERK

extracellular-related kinase

GEF

guanine exchange factor

GPCR

G protein-coupled receptor

IKK

IκB kinase

IL-1R

interleukin 1 receptor

IκB

inhibitor of NF-κB

JNK

c-Jun N-terminal kinase

MAL

MYD88 adaptor-like

MAP2K

MAPK kinase

MAP3K

MAPK kinase kinase

MAPK

mitogen-activated protein kinase

MEF

myocyte-enhancer factor

MNK

MAPK-interacting protein kinase

MSK

mitogen and stress-activated protein kinase

MYD88

myeloid differentiation primary response 88

NLR

NOD-like receptor

PAMP

pathogen-associated molecular pattern

PD

Parkinson's disease

PRR

pattern recognition receptor

pTRAP

palmitoylated transmembrane adaptor protein

RA

rheumatoid arthritis

RLR

RIG-I-like receptor

RSK

ribosomal S6 kinase

RTK

receptor tyrosine kinase

TEM

tetraspanin-enriched microdomain

TIR

Toll/interleukin-1 receptor

TLR

Toll-like receptor

TNF

tumor necrosis factor

TNFR

TNF receptor

TRAM

TRIF-related adaptor molecule

TRIF

TIR-domain-containing adapter-inducing interferon-β

1
Lemmon
,
M.A.
and
Schlessinger
,
J.
(
2010
)
Cell signaling by receptor-tyrosine kinases
.
Cell
141
,
1117
1134
2
Lavoie
,
H.
,
Gagnon
,
J.
and
Therrien
,
M.
(
2020
)
ERK signalling: a master regulator of cell behaviour, life and fate
.
Nat. Rev. Mol. Cell Biol.
21
,
607
632
3
Johnson
,
G.L.
and
Lapadat
,
R.
(
2002
)
Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases
.
Science
298
,
1911
1912
4
Cooper
,
J.A.
,
Sefton
,
B.M.
and
Hunter
,
T.
(
1984
)
Diverse mitogenic agents induce the phosphorylation of two related 42,000-dalton proteins on tyrosine in quiescent chick cells
.
Mol. Cell. Biol.
4
,
30
37
5
Buscà
,
R.
,
Pouysségur
,
J.
and
Lenormand
,
P.
(
2016
)
ERK1 and ERK2 map kinases: specific roles or functional redundancy?
Front. Cell Dev. Biol.
4
53
6
Frémin
,
C.
,
Saba-El-Leil
,
M.K.
,
Lévesque
,
K.
,
Ang
,
S.-L.
and
Meloche
,
S.
(
2015
)
Functional redundancy of ERK1 and ERK2 MAP kinases during development
.
Cell Rep.
12
,
913
921
7
Lavoie
,
H.
and
Therrien
,
M.
(
2015
)
Regulation of RAF protein kinases in ERK signalling
.
Nat. Rev. Mol. Cell Biol.
16
,
281
298
8
Di Benedetto
,
B.
,
Hitz
,
C.
,
Hölter
,
S.M.
,
Kühn
,
R.
,
Vogt Weisenhorn
,
D.M.
and
Wurst
,
W.
(
2007
)
Differential mRNA distribution of components of the ERK/MAPK signalling cascade in the adult mouse brain
.
J. Comp. Neurol.
500
,
542
556
9
Desideri
,
E.
,
Cavallo
,
A.L.
and
Baccarini
,
M.
(
2015
)
Alike but different: RAF paralogs and their signaling outputs
.
Cell
161
,
967
970
10
Sigismund
,
S.
,
Avanzato
,
D.
and
Lanzetti
,
L.
(
2018
)
Emerging functions of the EGFR in cancer
.
Mol. Oncol.
12
,
3
20
11
Dong
,
C.
,
Waters
,
S.B.
,
Holt
,
K.H.
and
Pessin
,
J.E.
(
1996
)
SOS phosphorylation and disassociation of the Grb2-SOS complex by the ERK and JNK signaling pathways
.
J. Biol. Chem.
271
,
6328
6332
12
Corbalan-Garcia
,
S.
,
Yang
,
S.S.
,
Degenhardt
,
K.R.
and
Bar-Sagi
,
D.
(
1996
)
Identification of the mitogen-activated protein kinase phosphorylation sites on human Sos1 that regulate interaction with Grb2
.
Mol. Cell. Biol.
16
,
5674
5682
13
Prior
,
I.A.
and
Hancock
,
J.F.
(
2012
)
Ras trafficking, localization and compartmentalized signalling
.
Semin. Cell Dev. Biol.
23
,
145
153
14
Dhanasekaran
,
D.N.
,
Kashef
,
K.
,
Lee
,
C.M.
,
Xu
,
H.
and
Reddy
,
E.P.
(
2007
)
Scaffold proteins of MAP-kinase modules
.
Oncogene
26
,
3185
3202
15
Ogata
,
T.
,
Naito
,
D.
,
Nakanishi
,
N.
,
Hayashi
,
Y.K.
,
Taniguchi
,
T.
,
Miyagawa
,
K.
et al (
2014
)
MURC/Cavin-4 facilitates recruitment of ERK to caveolae and concentric cardiac hypertrophy induced by α1-adrenergic receptors
.
Proc. Natl Acad. Sci. U.S.A.
111
,
3811
3816
16
Vomastek
,
T.
,
Schaeffer
,
H.-J.
,
Tarcsafalvi
,
A.
,
Smolkin
,
M.E.
,
Bissonette
,
E.A.
and
Weber
,
M.J.
(
2004
)
Modular construction of a signaling scaffold: MORG1 interacts with components of the ERK cascade and links ERK signaling to specific agonists
.
Proc. Natl Acad. Sci. U.S.A.
101
,
6981
6986
17
Roy
,
M.
,
Li
,
Z.
and
Sacks
,
D.B.
(
2005
)
IQGAP1 is a scaffold for mitogen-activated protein kinase signaling
.
Mol. Cell. Biol.
25
,
7940
7952
18
Cargnello
,
M.
and
Roux
,
P.P.
(
2011
)
Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases
.
Microbiol. Mol. Biol. Rev.
75
,
50
83
19
Ranganathan
,
A.
,
Yazicioglu
,
M.N.
and
Cobb
,
M.H.
(
2006
)
The nuclear localization of ERK2 occurs by mechanisms both independent of and dependent on energy
.
J. Biol. Chem.
281
,
15645
15652
20
Kosako
,
H.
,
Yamaguchi
,
N.
,
Aranami
,
C.
,
Ushiyama
,
M.
,
Kose
,
S.
,
Imamoto
,
N.
et al (
2009
)
Phosphoproteomics reveals new ERK MAP kinase targets and links ERK to nucleoporin-mediated nuclear transport
.
Nat. Struct. Mol. Biol.
16
,
1026
1035
21
Fowler
,
T.
,
Sen
,
R.
and
Roy
,
A.L.
(
2011
)
Regulation of primary response genes
.
Mol. Cell
44
,
348
360
22
Mylonis
,
I.
,
Chachami
,
G.
,
Samiotaki
,
M.
,
Panayotou
,
G.
,
Paraskeva
,
E.
,
Kalousi
,
A.
et al (
2006
)
Identification of MAPK phosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1alpha
.
J. Biol. Chem.
281
,
33095
33106
23
Carrière
,
A.
,
Cargnello
,
M.
,
Julien
,
L.-A.
,
Gao
,
H.
,
Bonneil
,
E.
,
Thibault
,
P.
et al (
2008
)
Oncogenic MAPK signaling stimulates mTORC1 activity by promoting RSK-mediated raptor phosphorylation
.
Curr. Biol
18
,
1269
1277
24
Spelat
,
R.
,
Ferro
,
F.
and
Curcio
,
F.
(
2012
)
Serine 111 phosphorylation regulates OCT4A protein subcellular distribution and degradation
.
J. Biol. Chem.
287
,
38279
38288
25
Yeo
,
J.-C.
,
Jiang
,
J.
,
Tan
,
Z.-Y.
,
Yim
,
G.-R.
,
Ng
,
J.-H.
,
Göke
,
J.
et al (
2014
)
Klf2 is an essential factor that sustains ground state pluripotency
.
Cell Stem Cell
14
,
864
872
26
Chung
,
J.
,
Uchida
,
E.
,
Grammer
,
T.C.
and
Blenis
,
J.
(
1997
)
STAT3 serine phosphorylation by ERK-dependent and -independent pathways negatively modulates its tyrosine phosphorylation
.
Mol. Cell. Biol.
17
,
6508
6516
27
Meloche
,
S.
and
Pouysségur
,
J.
(
2007
)
The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1- to S-phase transition
.
Oncogene
26
,
3227
3239
28
Balmanno
,
K.
and
Cook
,
S.J.
(
2009
)
Tumour cell survival signalling by the ERK1/2 pathway
.
Cell Death Differ.
16
,
368
377
29
Dong
,
C.
,
Davis
,
R.J.
and
Flavell
,
R.A.
(
2002
)
MAP kinases in the immune response
.
Annu. Rev. Immunol.
20
,
55
72
30
Kawai
,
T.
and
Akira
,
S.
(
2010
)
The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors
.
Nat. Immunol.
11
,
373
384
31
Iwasaki
,
A.
and
Medzhitov
,
R.
(
2010
)
Regulation of adaptive immunity by the innate immune system
.
Science
327
,
291
295
32
O'Neill
,
L.A.J.
,
Golenbock
,
D.
and
Bowie
,
A.G.
(
2013
)
The history of Toll-like receptors — redefining innate immunity
.
Nat. Rev. Immunol.
13
,
453
460
33
Brubaker
,
S.W.
,
Bonham
,
K.S.
,
Zanoni
,
I.
and
Kagan
,
J.C.
(
2015
)
Innate immune pattern recognition: a cell biological perspective
.
Annu. Rev. Immunol.
33
,
257
290
34
Akira
,
S.
,
Uematsu
,
S.
and
Takeuchi
,
O.
(
2006
)
Pathogen recognition and innate immunity
.
Cell
124
,
783
801
35
Kawasaki
,
T.
and
Kawai
,
T.
(
2014
)
Toll-like receptor signaling pathways
.
Front. Immunol.
5
,
461
36
Luo
,
L.
,
Lucas
,
R.M.
,
Liu
,
L.
and
Stow
,
J.L.
(
2019
)
Signalling, sorting and scaffolding adaptors for Toll-like receptors
.
J. Cell Sci.
133
.
37
Yamamoto
,
M.
,
Sato
,
S.
,
Hemmi
,
H.
,
Hoshino
,
K.
,
Kaisho
,
T.
,
Sanjo
,
H.
et al (
2003
)
Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway
.
Science
301
,
640
643
38
Gay
,
N.J.
,
Gangloff
,
M.
and
O'Neill
,
L.A.J.
(
2011
)
What the Myddosome structure tells us about the initiation of innate immunity
.
Trends Immunol.
32
,
104
109
39
Kawai
,
T.
and
Akira
,
S.
(
2007
)
Signaling to NF-κB by Toll-like receptors
.
Trends Mol. Med.
13
,
460
469
40
Walsh
,
M.C.
,
Lee
,
J.
and
Choi
,
Y.
(
2015
)
Tumor necrosis factor receptor associated factor 6 (TRAF6) regulation of development, function, and homeostasis of the immune system
.
Immunol. Rev.
266
,
72
92
41
Karin
,
M.
(
1995
)
The regulation of AP-1 activity by mitogen-activated protein kinases
.
J. Biol. Chem.
270
,
16483
16486
42
Shaulian
,
E.
and
Karin
,
M.
(
2002
)
AP-1 as a regulator of cell life and death
.
Nat. Cell Biol.
4
,
E131
E136
43
Hess
,
J.
,
Angel
,
P.
and
Schorpp-Kistner
,
M.
(
2004
)
AP-1 subunits: quarrel and harmony among siblings
.
J. Cell Sci.
117
,
5965
5973
44
Chalmers
,
C.J.
,
Gilley
,
R.
,
March
,
H.N.
,
Balmanno
,
K.
and
Cook
,
S.J.
(
2007
)
The duration of ERK1/2 activity determines the activation of c-Fos and Fra-1 and the composition and quantitative transcriptional output of AP-1
.
Cell Signal.
19
,
695
704
45
Gilley
,
R.
,
March
,
H.N.
and
Cook
,
S.J.
(
2009
)
ERK1/2, but not ERK5, is necessary and sufficient for phosphorylation and activation of c-Fos
.
Cell Signal.
21
,
969
977
46
Ip
,
Y.T.
and
Davis
,
R.J.
(
1998
)
Signal transduction by the c-Jun N-terminal kinase (JNK) — from inflammation to development
.
Curr. Opin. Cell Biol.
10
,
205
219
47
Cui
,
J.
,
Zhang
,
M.
,
Zhang
,
Y.
and
Xu
,
Z.
(
2007
)
JNK pathway: diseases and therapeutic potential
.
Acta Pharm. Sin.
28
,
601
608
48
Erlanson
,
D.A.
,
Chytil
,
M.
and
Verdine
,
G.L.
(
1996
)
The leucine zipper domain controls the orientation of AP-1 in the NFAT.AP-1.DNA complex
.
Chem. Biol.
3
,
981
991
49
Pattison
,
M.J.
,
Naik
,
R.J.
,
Reyskens
,
K.M.S.E.
and
Arthur
,
J.S.C.
(
2020
)
Loss of Mef2D function enhances TLR induced IL-10 production in macrophages
.
Biosci. Rep.
40
,
BSR20201859
50
Han
,
J.
,
Jiang
,
Y.
,
Li
,
Z.
,
Kravchenko
,
V.V.
and
Ulevitch
,
R.J.
(
1997
)
Activation of the transcription factor MEF2C by the MAP kinase p38 in inflammation
.
Nature
386
,
296
299
51
Yang
,
S.H.
,
Galanis
,
A.
and
Sharrocks
,
A.D.
(
1999
)
Targeting of p38 mitogen-activated protein kinases to MEF2 transcription factors
.
Mol. Cell. Biol.
19
,
4028
4038
52
Zhao
,
M.
,
New
,
L.
,
Kravchenko
,
V.V.
,
Kato
,
Y.
,
Gram
,
H.
,
di Padova
,
F.
et al (
1999
)
Regulation of the MEF2 family of transcription factors by p38
.
Mol. Cell. Biol.
19
,
21
30
53
Chen
,
B.-C.
and
Lin
,
W.-W.
(
2001
)
PKC- and ERK-dependent activation of IκB kinase by lipopolysaccharide in macrophages: enhancement by P2Y receptor-mediated CaMK activation
.
Br. J. Pharmacol.
134
,
1055
1065
54
Jefferies
,
C.A.
and
O'Neill
,
L.A.J.
(
2000
)
Rac1 regulates interleukin 1-induced nuclear factor κB activation in an inhibitory protein κBα-independent manner by enhancing the ability of the p65 subunit to transactivate gene expression
.
J. Biol. Chem.
275
,
3114
3120
55
Vuong
,
B.
,
Hogan-Cann
,
A.D.J.
,
Alano
,
C.C.
,
Stevenson
,
M.
,
Chan
,
W.Y.
,
Anderson
,
C.M.
et al (
2015
)
NF-κB transcriptional activation by TNFα requires phospholipase C, extracellular signal-regulated kinase 2 and poly(ADP-ribose) polymerase-1
.
J. Neuroinflammation
12
,
229
56
Gualdrini
,
F.
,
Esnault
,
C.
,
Horswell
,
S.
,
Stewart
,
A.
,
Matthews
,
N.
and
Treisman
,
R.
(
2016
)
SRF Co-factors control the balance between cell proliferation and contractility
.
Mol. Cell
64
,
1048
1061
57
Blair
,
L.
,
Pattison
,
M.J.
,
Chakravarty
,
P.
,
Papoutsopoulou
,
S.
,
Bakiri
,
L.
,
Wagner
,
E.F.
et al (
2022
)
TPL-2 inhibits IFN-β expression via an ERK1/2-TCF-FOS axis in TLR4-stimulated macrophages
.
J. Immunol.
208
,
941
954
58
Gantke
,
T.
,
Sriskantharajah
,
S.
and
Ley
,
S.C.
(
2011
)
Regulation and function of TPL-2, an IκB kinase-regulated MAP kinase kinase kinase
.
Cell Res.
21
,
131
145
59
Xu
,
D.
,
Matsumoto
,
M.L.
,
McKenzie
,
B.S.
and
Zarrin
,
A.A.
(
2018
)
TPL2 kinase action and control of inflammation
.
Pharmacol. Res.
129
,
188
193
60
Ben-Addi
,
A.
,
Mambole-Dema
,
A.
,
Brender
,
C.
,
Martin
,
S.R.
,
Janzen
,
J.
,
Kjaer
,
S.
et al (
2014
)
Iκb kinase-induced interaction of TPL-2 kinase with 14-3-3 is essential for Toll-like receptor activation of ERK-1 and -2 MAP kinases
.
Proc. Natl Acad. Sci. U.S.A.
111
,
E2394
E2403
61
Pattison
,
M.J.
,
Mitchell
,
O.
,
Flynn
,
H.R.
,
Chen
,
C.-S.
,
Yang
,
H.-T.
,
Ben-Addi
,
H.
et al (
2016
)
TLR and TNF-R1 activation of the MKK3/MKK6-p38α axis in macrophages is mediated by TPL-2 kinase
.
Biochem. J.
473
,
2845
2861
62
Senger
,
K.
,
Pham
,
V.C.
,
Varfolomeev
,
E.
,
Hackney
,
J.A.
,
Corzo
,
C.A.
,
Collier
,
J.
et al (
2017
)
The kinase TPL2 activates ERK and p38 signaling to promote neutrophilic inflammation
.
Sci. Signal.
10
,
eaah4273
63
Das
,
S.
,
Cho
,
J.
,
Lambertz
,
I.
,
Kelliher
,
M.A.
,
Eliopoulos
,
A.G.
,
Du
,
K.
et al (
2005
)
Tpl2/Cot signals activate ERK, JNK, and NF-κB in a cell-type and stimulus-specific manner
.
J. Biol. Chem.
280
,
23748
23757
64
Rousseau
,
S.
,
Papoutsopoulou
,
M.
,
Symons
,
A.
,
Cook
,
D.
,
Lucocq
,
J.M.
,
Prescott
,
A.R.
et al (
2008
)
TPL2-mediated activation of ERK1 and ERK2 regulates the processing of pre-TNFα in LPS-stimulated macrophages
.
J. Cell Sci.
121
,
149
154
65
Flynn
,
C.M.
,
Garbers
,
Y.
,
Lokau
,
J.
,
Wesch
,
D.
,
Schulte
,
D.M.
,
Laudes
,
M.
et al (
2019
)
Activation of Toll-like receptor 2 (TLR2) induces interleukin-6 trans-signaling
.
Sci. Rep.
9
,
7306
66
Singh
,
A.
,
Singh
,
V.
,
Tiwari
,
R.L.
,
Chandra
,
T.
,
Kumar
,
A.
,
Dikshit
,
M.
et al (
2016
)
The IRAK-ERK-p67phox-Nox-2 axis mediates TLR4, 2-induced ROS production for IL-1 β transcription and processing in monocytes
.
Cell. Mol. Immunol.
13
,
745
763
67
Jin
,
X.
,
Gong
,
P.
,
Zhang
,
X.
,
Li
,
G.
,
Zhu
,
T.
,
Zhang
,
M.
et al (
2017
)
Activation of ERK signaling via TLR11 induces IL-12p40 production in peritoneal macrophages challenged by Neospora caninum
.
Front. Microbiol.
8
,
1393
68
Sanin
,
D.E.
,
Prendergast
,
C.T.
and
Mountford
,
A.P.
(
2015
)
IL-10 production in macrophages is regulated by a TLR-driven CREB-mediated mechanism that is linked to genes involved in cell metabolism
.
J. Immunol.
195
,
1218
1232
69
Kaiser
,
F.
,
Cook
,
D.
,
Papoutsopoulou
,
S.
,
Rajsbaum
,
R.
,
Wu
,
X.
,
Yang
,
H.-T.
et al (
2009
)
TPL-2 negatively regulates interferon-β production in macrophages and myeloid dendritic cells
.
J. Exp. Med.
206
,
1863
1871
70
Luo
,
L.
,
Bokil
,
N.J.
,
Wall
,
A.A.
,
Kapetanovic
,
R.
,
Lansdaal
,
N.M.
,
Marceline
,
F.
et al (
2017
)
SCIMP is a transmembrane non-TIR TLR adaptor that promotes proinflammatory cytokine production from macrophages
.
Nat. Commun.
8
,
1
14
71
Curson
,
J.E.B.
,
Luo
,
L.
,
Sweet
,
M.J.
and
Stow
,
J.L.
(
2018
)
pTRAPs: transmembrane adaptors in innate immune signaling
.
J. Leukoc. Biol.
72
Luo
,
L.
,
Curson
,
J.E.B.
,
Liu
,
L.
,
Wall
,
A.A.
,
Tuladhar
,
N.
,
Lucas
,
R.M.
et al (
2019
)
SCIMP is a universal Toll-like receptor adaptor in macrophages
.
J. Leukoc. Biol.
73
Lucas
,
R.M.
,
Liu
,
L.
,
Curson
,
J.E.B.
,
Koh
,
Y.W.H.
,
Tuladhar
,
N.
,
Condon
,
N.D.
et al (
2021
)
SCIMP is a spatiotemporal transmembrane scaffold for Erk1/2 to direct pro-inflammatory signaling in TLR-activated macrophages
.
Cell Rep.
36
,
109662
74
Banerjee
,
A.
,
Gugasyan
,
R.
,
McMahon
,
M.
and
Gerondakis
,
S.
(
2006
)
Diverse Toll-like receptors utilize Tpl2 to activate extracellular signal-regulated kinase (ERK) in hemopoietic cells
.
Proc. Natl Acad. Sci. U.S.A.
103
,
3274
3279
75
Vanshylla
,
K.
,
Bartsch
,
C.
,
Hitzing
,
C.
,
Krümpelmann
,
L.
,
Wienands
,
J.
and
Engels
,
N.
(
2018
)
Grb2 and GRAP connect the B cell antigen receptor to Erk MAP kinase activation in human B cells
.
Sci. Rep.
8
,
4244
76
Gold
,
M.R.
(
2008
)
B cell development: important work for ERK
.
Immunity
28
,
488
490
77
Fischer
,
A.M.
,
Katayama
,
C.D.
,
Pagès
,
G.
,
Pouysségur
,
J.
and
Hedrick
,
S.M.
(
2005
)
The role of Erk1 and Erk2 in multiple stages of T cell development
.
Immunity
23
,
431
443
78
Adem
,
J.
,
Hämäläinen
,
A.
,
Ropponen
,
A.
,
Eeva
,
J.
,
Eray
,
M.
,
Nuutinen
,
U.
et al (
2015
)
ERK1/2 has an essential role in B cell receptor- and CD40-induced signaling in an in vitro model of germinal center B cell selection
.
Mol. Immunol.
67
,
240
247
79
Damasio
,
M.P.
,
Marchingo
,
J.M.
,
Spinelli
,
L.
,
Hukelmann
,
J.L.
,
Cantrell
,
D.A.
and
Howden
,
A.J.M.
(
2021
)
Extracellular signal-regulated kinase (ERK) pathway control of CD8+ T cell differentiation
.
Biochem. J.
478
,
79
98
80
Sale
,
M.J.
,
Minihane
,
E.
,
Monks
,
N.R.
,
Gilley
,
R.
,
Richards
,
F.M.
,
Schifferli
,
K.P.
et al (
2019
)
Targeting melanoma's MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors
.
Nat. Commun.
10
,
5167
81
Collins
,
P.E.
,
Somma
,
D.
,
Kerrigan
,
D.
,
Herrington
,
F.
,
Keeshan
,
K.
,
Nibbs
,
R.J.B.
et al (
2019
)
The IκB-protein BCL-3 controls Toll-like receptor-induced MAPK activity by promoting TPL-2 degradation in the nucleus
.
Proc. Natl Acad. Sci. U.S.A.
116
,
25828
25838
82
Rui
,
L.
,
Vinuesa
,
C.G.
,
Blasioli
,
J.
and
Goodnow
,
C.C.
(
2003
)
Resistance to CpG DNA-induced autoimmunity through tolerogenic B cell antigen receptor ERK signaling
.
Nat. Immunol.
4
,
594
600
83
Zhou
,
B.
,
He
,
Y.
,
Zhang
,
X.
,
Xu
,
J.
,
Luo
,
Y.
,
Wang
,
Y.
et al (
2010
)
Targeting mycobacterium protein tyrosine phosphatase B for antituberculosis agents
.
Proc. Natl Acad. Sci. U.S.A.
107
,
4573
4578
84
Li
,
H.
,
Xu
,
H.
,
Zhou
,
Y.
,
Zhang
,
J.
,
Long
,
C.
,
Li
,
S.
et al (
2007
)
The phosphothreonine lyase activity of a bacterial type III effector family
.
Science
315
,
1000
1003
85
Arthur
,
J.S.C.
and
Ley
,
S.C.
(
2013
)
Mitogen-activated protein kinases in innate immunity
.
Nat. Rev. Immunol.
13
,
679
692
86
Trosky
,
J.E.
,
Li
,
Y.
,
Mukherjee
,
S.
,
Keitany
,
G.
,
Ball
,
H.
and
Orth
,
K.
(
2007
)
Vopa inhibits ATP binding by acetylating the catalytic loop of MAPK kinases
.
J. Biol. Chem.
282
,
34299
34305
87
Mukherjee
,
S.
,
Keitany
,
G.
,
Li
,
Y.
,
Wang
,
Y.
,
Ball
,
H.L.
,
Goldsmith
,
E.J.
et al (
2006
)
Yersinia YopJ acetylates and inhibits kinase activation by blocking phosphorylation
.
Science
312
,
1211
1214
88
Kim
,
E.K.
and
Choi
,
E.-J.
(
2015
)
Compromised MAPK signaling in human diseases: an update
.
Arch. Toxicol.
89
,
867
882
89
Dhillon
,
A.S.
,
Hagan
,
S.
,
Rath
,
O.
and
Kolch
,
W.
(
2007
)
MAP kinase signalling pathways in cancer
.
Oncogene
26
,
3279
3290
90
Newman
,
S.
,
Fan
,
L.
,
Pribnow
,
A.
,
Silkov
,
A.
,
Rice
,
S.V.
,
Lee
,
S.
et al (
2019
)
Clinical genome sequencing uncovers potentially targetable truncations and fusions of MAP3K8 in spitzoid and other melanomas
.
Nat. Med.
25
,
597
602
91
Diaz-Flores
,
E.
,
Goldschmidt
,
H.
,
Depeille
,
P.
,
Ng
,
V.
,
Akutagawa
,
J.
,
Krisman
,
K.
et al (
2013
)
PLC-γ and PI3K link cytokines to ERK activation in hematopoietic cells with Normal and oncogenic Kras
.
Sci. Signal.
6
,
ra105
ra105
92
Holderfield
,
M.
,
Deuker
,
M.M.
,
McCormick
,
F.
and
McMahon
,
M.
(
2014
)
Targeting RAF kinases for cancer therapy: BRAF mutated melanoma and beyond
.
Nat. Rev. Cancer
14
,
455
467
93
Nikolaev
,
S.I.
,
Rimoldi
,
D.
,
Iseli
,
C.
,
Valsesia
,
A.
,
Robyr
,
D.
,
Gehrig
,
C.
et al (
2012
)
Exome sequencing identifies recurrent somatic MAP2K1 and MAP2K2 mutations in melanoma
.
Nat. Genet.
44
,
133
139
94
Liu
,
F.
,
Yang
,
X.
,
Geng
,
M.
and
Huang
,
M.
(
2018
)
Targeting ERK, an Achilles’ Heel of the MAPK pathway, in cancer therapy
.
Acta Pharm. Sin. B
8
,
552
562
95
de Launay
,
D.
,
van de Sande
,
M.G.H.
,
van de Sande
,
G.P.M.
,
Wijbrandts
,
C.A.
,
Tak
,
P.P.
and
Reedquist
,
K.A.
(
2010
)
Selective involvement of ERK and JNK MAP kinases in the synovial tissue of patients with early arthritis
.
Ann. Rheum. Dis.
69
,
A16
96
Thalhamer
,
T.
,
McGrath
,
M.A.
and
Harnett
,
M.M.
(
2008
)
MAPKs and their relevance to arthritis and inflammation
.
Rheumatology
47
,
409
414
97
van ‘t Wout
,
E.F.A.
,
Dickens
,
J.A.
,
van Schadewijk
,
A.
,
Haq
,
I.
,
Kwok
,
H.F.
,
Ordóñez
,
A.
et al (
2014
)
Increased ERK signalling promotes inflammatory signalling in primary airway epithelial cells expressing Z α1-antitrypsin
.
Hum. Mol. Genet.
23
,
929
941
98
Mandrekar
,
P.
and
Szabo
,
G.
(
2009
)
Signalling pathways in alcohol-induced liver inflammation
.
J. Hepatol.
50
,
1258
1266
99
Brereton
,
C.F.
,
Sutton
,
C.E.
,
Lalor
,
S.J.
,
Lavelle
,
E.C.
and
Mills
,
K.H.G.
(
2009
)
Inhibition of ERK MAPK suppresses IL-23- and IL-1-driven IL-17 production and attenuates autoimmune disease
.
J. Immunol.
183
,
1715
1723
100
Jostins
,
L.
,
Ripke
,
S.
,
Weersma
,
R.K.
,
Duerr
,
R.H.
,
McGovern
,
D.P.
,
Hui
,
K.Y.
et al (
2012
)
Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease
.
Nature
491
,
119
124
101
Hedl
,
M.
and
Abraham
,
C.
(
2016
)
A TPL2 (MAP3K8) disease-risk polymorphism increases TPL2 expression thereby leading to increased pattern recognition receptor-initiated caspase-1 and caspase-8 activation, signalling and cytokine secretion
.
Gut
65
,
1799
1811
102
Morsy
,
Y.
,
Brillant
,
N.
,
Franc
,
Y.
,
Scharl
,
M.
,
Wawrzyniak
,
M.
and
On behalf of the Swiss IBD Cohort Study Group
. (
2021
)
Unravelling the impact of the genetic variant rs1042058 within the TPL2 risk gene locus on molecular and clinical disease course patients with inflammatory bowel disease
.
Cells
10
,
3589
. .
103
Chitnis
,
T.
and
Weiner
,
H.L.
(
2017
)
CNS inflammation and neurodegeneration
.
J. Clin. Invest.
127
,
3577
3587
104
Chen
,
M.J.
,
Ramesha
,
S.
,
Weinstock
,
L.D.
,
Gao
,
T.
,
Ping
,
L.
,
Xiao
,
H.
et al (
2021
)
Extracellular signal-regulated kinase regulates microglial immune responses in Alzheimer's disease
.
J. Neurosci. Res.
99
,
1704
1721
105
Long
,
H.
,
Zhong
,
G.
,
Wang
,
C.
,
Zhang
,
J.
,
Zhang
,
Y.
,
Luo
,
J.
et al (
2019
)
TREM2 attenuates Aβ1-42-mediated neuroinflammation in BV-2 cells by downregulating TLR signaling
.
Neurochem. Res.
44
,
1830
1839
106
Kim
,
J.
,
Pajarillo
,
E.
,
Rizor
,
A.
,
Son
,
D.-S.
,
Lee
,
J.
,
Aschner
,
M.
et al (
2019
)
LRRK2 kinase plays a critical role in manganese-induced inflammation and apoptosis in microglia
.
PLoS ONE
14
,
e0210248
107
Lv
,
Y.
,
Zhang
,
Z.
,
Hou
,
L.
,
Zhang
,
L.
,
Zhang
,
J.
,
Wang
,
Y.
et al (
2015
)
Phytic acid attenuates inflammatory responses and the levels of NF-κB and p-ERK in MPTP-induced Parkinson's disease model of mice
.
Neurosci. Lett.
597
,
132
136
108
Du
,
H.
,
Guo
,
L.
and
Yan
,
S.S.
(
2012
)
Synaptic mitochondrial pathology in Alzheimer's disease
.
Antioxid. Redox Signal.
16
,
1467
1475
109
Reinhardt
,
P.
,
Schmid
,
B.
,
Burbulla
,
L.F.
,
Schöndorf
,
D.C.
,
Wagner
,
L.
,
Glatza
,
M.
et al (
2013
)
Genetic correction of a LRRK2 mutation in human iPSCs links parkinsonian neurodegeneration to ERK-dependent changes in gene expression
.
Cell Stem Cell
12
,
354
367
110
Wang
,
K.Z.Q.
,
Zhu
,
J.
,
Dagda
,
R.K.
,
Uechi
,
G.
,
Cherra
,
S.J.
,
Gusdon
,
A.M.
et al (
2014
)
ERK-mediated phosphorylation of TFAM downregulates mitochondrial transcription: implications for Parkinson's disease
.
Mitochondrion
17
,
132
140
111
Perlson
,
E.
,
Jeong
,
G.-B.
,
Ross
,
J.L.
,
Dixit
,
R.
,
Wallace
,
K.E.
,
Kalb
,
R.G.
et al (
2009
)
A switch in retrograde signaling from survival to stress in rapid-onset neurodegeneration
.
J. Neurosci.
29
,
9903
9917
112
Lim
,
E.
,
Lee
,
S.
,
Li
,
E.
,
Kim
,
Y.
and
Park
,
S.
(
2011
)
Ghrelin protects spinal cord motoneurons against chronic glutamate-induced excitotoxicity via ERK1/2 and phosphatidylinositol-3-kinase/Akt/glycogen synthase kinase-3β pathways
.
Exp. Neurol.
230
,
114
122
113
Xiao
,
Y.
,
Jin
,
J.
,
Chang
,
M.
,
Nakaya
,
M.
,
Hu
,
H.
,
Zou
,
Q.
et al (
2014
)
TPL2 mediates autoimmune inflammation through activation of the TAK1 axis of IL-17 signaling
.
J. Exp. Med.
211
,
1689
1702
114
Sriskantharajah
,
S.
,
Gückel
,
E.
,
Tsakiri
,
N.
,
Kierdorf
,
K.
,
Brender
,
C.
,
Ben-Addi
,
A.
et al (
2014
)
Regulation of experimental autoimmune encephalomyelitis by TPL-2 kinase
.
J. Immunol.
192
,
3518
3529
115
Zarrin
,
A.A.
,
Bao
,
K.
,
Lupardus
,
P.
and
Vucic
,
D.
(
2021
)
Kinase inhibition in autoimmunity and inflammation
.
Nat. Rev. Drug Discov.
20
,
39
63
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of the University of Queensland in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with CAUL.