<<

Ba¨rbel Schro¨felbauer How do pleiotropic hubs Alexander Hoffmann mediate specific signaling by TNFR superfamily members?

Authors’ address Summary: Tumor necrosis factor receptor (TNFR) superfamily mem- 1 1 Ba¨rbel Schro¨felbauer , Alexander Hoffmann bers mediate the cellular response to a wide variety of biological inputs. 1 Signaling Systems Laboratory, Department of Chemistry and The responses range from death, survival, differentiation, prolifera- Biochemistry, University of California, San Diego, La Jolla, tion, to the regulation of immunity. All these physiological responses are CA, USA. regulated by a limited number of highly pleiotropic . The fact that the same signaling molecules are involved in transducing signals from Correspondence to: TNFR superfamily members that regulate different and even opposing Ba¨prbel Schro¨felbauer processes raises the question of how their specificity is determined. Regu- Signaling Systems Laboratory latory strategies that can contribute to signaling specificity include scaf- Department of Chemistry and Biochemistry folding to control kinase specificity, combinatorial use of several signal University of California, San Diego transducers, and temporal control of signaling. In this review, we discuss NSB 0375 these strategies in the context of TNFR superfamily member signaling. 9500 Gilman Dr La Jolla, CA 92093-0375, USA Keywords: TNFR, MAPK, IKK, NFjB, scaffold, signaling specificity Tel.: +1 858 822 4670 Fax: +1 858 822 4671 e-mail: [email protected] Diverse biological responses to TNFR superfamily members Acknowledgements This work was supported by RO1 grant CA141722. BS is a The tumor necrosis factor receptor (TNFR) superfamily Leukemia and Lymphoma Society fellow. The authors have consists of 29 receptors that mediate cellular responses to 19 no conflicts of interest to declare. ligands. While most ligands bind to a single receptor, some bind to numerous receptors. For example, BAFF can associate with three receptors, and TNF-related apoptosis-inducing This article is part of a series of reviews ligand (TRAIL) can even bind to five receptors (1, 2). All TNFRs covering TNF Receptor Family Members are characterized as type I transmembrane , with an appearing in Volume 244 of Immunological Reviews. extracellular N-terminus and intracellular C-terminus necessary for signaling initiation (3). The TNFR superfamily can be cate- gorized into three overlapping classes: activating receptors, death receptors, and decoy receptors. Activating receptors such as TNFR1 and CD40 mediate activation of nuclear factor-jB (NFjB) and -activated kinase (MAPK) path- ways. Death domain containing receptors (such as TNFR1 and FAS) contain an 80 amino acid death domain in their cytoplas- Immunological Reviews 2011 mic domain. Its deletion abolishes ligand-induced cell death. Vol. 244: 29–43 Printed in Singapore. All rights reserved Through sequestration of the ligand, decoy receptors (e.g. DCR1, OPG) have been shown to inhibit . 2011 John Wiley & Sons A/S Immunological Reviews Most receptors are expressed on a wide variety of cell types. 0105-2896 Receptor engagement by members of the TNF superfamily

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 29 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

can trigger diverse cellular responses, such as apoptosis [for Pleiotropic signal transducers within the TNFR signaling example TNF, lymphotoxin (LT), FAS ligand (FASL)], survival network [receptor activator of NFjB ligand (RANKL) and B-cell activat- The key signal transducers of the TNFR superfamily members ing factor belonging to the TNF family (BAFF)], differentia- are the kinases IKK2, IKK1, JNK, p38, and ERK. Below we tion (such as TNF, RANKL), or proliferation (such as TNF, have summarized some key facts about each kinase, indicating CD40L, OX40L, BAFF). These cellular responses are mediated the wide variety of substrates and biological functions that by the activation of transcription factors NFjB, which com- have been ascribed to each (Fig. 1). prise NFjB1 (p50 and its precursor p105), NFjB2 (p52 and IKK2 its precursor p100), c-Rel, RelA (p65), and RelB, and activator protein-1 (AP-1), composed of Fos (c-Fos, FosB, Fra-1, and IKK2 is a key regulator of NFjB activation induced by Fra-2) and Jun (c-Jun, JunB, and JunD) family members, as inflammatory , pathogens, environmental and meta- well as closely related transcription factors CREB, ATF2, ATF3, bolic stress and some developmental signals. By phosphory- and B-ATF), which are activated by MAPK [p38, c-Jun N-ter- lating the classical IjBs (IjBa,IjBb,IjBe), it triggers their minal kinase (JNK), extracellular signal-regulated kinase proteasomal degradation and release of NFjB to the nucleus (ERK)] and inhibitor of NFjB kinase (IKK) signaling cascades to allow for transcriptional activation. The primary canonical (1, 2). NFjB effectors are RelA, cRel, and p50 in both homodimeric Signaling by TNF superfamily members is essential for a or heterodimeric forms. Induction of their activity by IKK2 large variety of physiological processes including hematopoie- leads, in most cell types, to a general cell activation response, sis, protection from bacterial infection, immune surveillance, that may involve the secretion of inflammatory molecules, and tumor regression. Ligands, including TNF, LTb, and increasing the resistance to cell death-inducing stimuli, and, RANKL, provide crucial signals for the morphogenesis of sec- in lymphocytes particularly, activation of a proliferative pro- ondary lymphoid organs; TNF, FAS, and TRAIL contribute to gram (7, 8). Apart from its role in activating NFjB, IKK2 has the function of cytotoxic effector cells in the recognition and many additional substrates that are not part of the NFjB sig- destruction of virus-infected cells. The expression of FASL on naling system. The IKK2-dependent of the activated T cells induces their cell death, a mechanism to mod- tumor suppressor p53 at S362 and S366 is thought to be a ulate the immune response. Importantly, misregulation of mechanism for regulating its stability (9). Upon TNFa stim- TNFR signaling has been associated with a diverse range of ulation, IKK2 phosphorylates insulin receptor (IR) substrate diseases including autoimmunity, liver disease, tumorigenesis, 1 (IRS-1) at S307, resulting in the termination of metabolic lymphproliferative diseases, diabetes, and even allergic asthma insulin signaling (10). In addition, IKK2 phosphorylates the (1, 2, 4–6). The diverse pathological effects caused by TNFR tumor suppressor tuberous sclerosis 1 (TSC1) at position misregulation reflect the large variety of biological processes S487 and S511, which leads to its suppression and coincides they are involved in and highlight the importance of precise with the activation of the mTOR pathway, enhanced angio- regulation of TNFR superfamily signaling. Interestingly, the genesis and tumor development (11). b-catenin, a key mole- MAPK and IKK cascades are the critical signal transducers for cule in Wnt signaling, and FOXO3a, which acts downstream all TNFRSFs raising the question of how the functional speci- of growth factor signaling (PI3K ⁄ Akt), have also been ficity of these kinases is determined to ensure signaling speci- reported to be specifically phosphorylated by IKK2 (12). ficity. Recently, IKK2 has also been found to play a critical role in

Fig. 1. Kinases involved in TNFRSF-member signaling are highly pleiotropic. The pleiotropic kinases JNK, p38, and ERK1 ⁄ 2 (MAPKs), canonical IKK2, and non-canonical IKK1 are key regulators in in response to a large variety of cellular signals and can trigger highly diverse biological responses.

30 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

AB C

Fig. 2. Activation of signal transducers downstream of TNFR1, CD40, and LTbR engagement. (A) TNF triggers the assembly of a signaling com- plex involving the TRADD, RIPK1, and complexes. TAK1 is recruited and activated by binding of its scaffolds TAB 2 ⁄ 3 to polyubiqu- itin chains. Subsequent binding of NEMO to ubiquitin chains allows for the activation of IKK2. MAP3Ks activate MAPKs through a cascade of phosphorylation events. (B) CD40 engagement triggers the assembly of a complex containing TRAF2, TRAF3, TRAF6, cIAPs, MEKK1, and the LUBAC complex to activate MAPKs, NEMO-IKK2, and non-canonical IKK1. (C) Binding of LTb to the LTbR triggers degradation of TRAF2 and TRAF3 result- ing in NIK stabilization, which activates non-canonical IKK1 (see text for details). starvation-induced autophagy, in an NFjB independent thought to regulate cell proliferation in an NFjB-independent manner (13, 14). The substrates however remain to be manner (16–18). identified. JNK IKK1 JNK is a member of the MAPK family of signaling proteins The IKK1 kinase functions as a homodimer and as a heterodi- (19, 20). It consists of 10 JNK isoforms, which are derived mer with IKK2. As a homodimer it may be activated through through alternative splicing of mRNA transcripts generated the NFjB-inducing kinase (NIK) by developmental signals from the three JNK1, JNK2, and JNK3. It is activated in such as BAFF and LTb (6, 8). IKK1 dependent phosphoryla- response to cellular stress, cytokines, pathogens, and mito- tion of p100 results in p100 processing and induction of tran- gens. JNK activation leads to the phosphorylation of a large scriptional activation. The main mediators of transcription are number of transcription factors, the most prominent of which RelA, RelB, p50, and p52 controlling cell survival and devel- is c-Jun, a component of AP-1, as well as of numerous cellular opmental processes (6, 8). Similar to IKK2, IKK1 has also proteins most of which are associated with apoptosis (such as been reported to have additional NFjB independent sub- Bcl2 and p53). disruption in mice revealed its essential strates. Like IKK2, IKK1 also phosphorylates b-catenin (15). role in TNFa-induced c-Jun phosphorylation and AP-1 tran- Interestingly, while IKK2-induced phosphorylation negatively scription factor activity. JNK is activated through phosphory- regulates b-catenin activity, phosphorylation by IKK1 lation of the Thr-Pro-Tyr motif in its activation loop by MAPK increases b-catenin-dependent transcription, indicating dis- kinases (MAPKKs) MKK4 and MKK7. Activation of these tinct biological roles for these kinases. Many other IKK1 sub- kinases in turn is initiated by a cascade of kinases linked strates localize to the nucleus, including the coactivators through stimulus-dependent association with different scaf- SMRT, SRC3, and CBP, as well as the histone H3, and they are fold proteins (see below). The complex regulation of JNK is

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 31 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

indicative of its critical role in multiple physiological pro- ERK cesses. Indeed, JNK has been implicated in the regulation of cell survival and apoptosis, inflammation, metabolism, and ERKs are encoded by two genes, ERK1 (MAPK3), and ERK2 development. By phosphorylating several pro-apoptotic mem- (MAPK1), that encode two main proteins, p44 (ERK1), and bers of the Bcl2-related (Bim, Bmf, Bad) and p42 (ERK2). They are preferentially activated by through JNK-dependent activation of AP-1, it can activate the but, similar to JNK and p38, are also activated in response to mitochondrial apoptotic pathway, while its phosphorylation GPCR agonists, cytokines, and environmental stress. Activation ⁄ ⁄ of several anti-apoptotic members (Bcl2, Bcl-xL, Mcl-1) pro- occurs through the Raf MEK ERK signaling cascade through mote cell survival. In addition, JNK has been implicated to different isoforms of the small GTP-binding protein Ras. Acti- play important roles in other types of death including necrosis vated Raf binds to and phosphorylates MEK1 and MEK2, ⁄ ⁄ and autophagic ⁄ lysosomal cell death. JNK is also a potent which in turn phosphorylate ERK1 2. Markedly, ERK1 2 have inducer of inflammatory primarily through been reported to phosphorylate more than 160 substrates, the activation of the transcription factors AP-1 and ATF-2. including numerous important transcriptional regulators such Accordingly, many autoimmune diseases (rheumatoid arthri- as NF-AT, Elk-1, c-Fos, c-, and H3 to control the cell cycle tis, multiple sclerosis, psoriasis) correlate with hyperactivity progression and cell death. Apart from ERK’s critical role in of JNK. The activation of JNK causes insulin resistance at least cell proliferation, it is also critically involved in differentia- in part through the phosphorylation of Insulin receptor sub- tion, migration, and cellular transformation (28, 29). strate-1 (IRS-1) at the inhibitory site Ser-307, thus suppress- ing insulin receptor signaling, indicating its role in Activation of signal transducers downstream of specific TNFRSF members metabolism (19–21). The key kinases are connected to members of the TNFR super- p38 family via complex signaling mechanisms that involve a multitude of protein adapters and enzymatic functions. Their p38 is another member of the MAPK family. It consists of cooperative or sequential regulation control dose response and four isoforms, p38a, p38b, p38c, and p38d, each encoded dynamic behavior of the key kinases. Therefore, knowledge by a distinct gene. They have similar in vitro substrate speci- about the signaling molecules involved in the activation of the ficity but differ in regards to how their activity is regulated key signaling kinases and their regulation is of major impor- or their responsiveness to ligands; the p38 pathway is acti- tance to gain insights into how signaling specificity may be vated by a large number of growth factors, GPCR agonists, achieved. Below we detail what is currently known about these environmental stresses, pathogens, and cytokines. Dependent signaling mechanisms for three TNFRSF members (Fig. 2). on the isotype, they are activated by dual kinases (threonine and tyrosine) MKK3, and the JNK kinases MKK4 and MKK7. TNFR1 Once activated, p38 phosphorylates several cytoplasmic sub- strates (including MBP, HSP27, and MAPKAPK2) and upon Activation of TNFR1 triggers a wide variety of biological nuclear translocation activates the ATF-2 responses from cell death to survival, effects on metabolism, through direct phosphorylation (22–24) as well as CREB differentiation, adhesion, and motility. TNFR1 is particularly (25). p38 can also indirectly regulate transcription through important in inflammatory responses (2). TNFR1 knockout AP-1. One of the main consequences of p38 activation is mice show impaired clearance of bacterial pathogens and increased expression of cytokines and receptors involved in resistance to LPS (30–34). Although lymph nodes develop inflammation and immunity (19, 21, 26). However, it normally, these mice show defects in germinal center forma- remains unclear whether the aforementioned transcription tion and impaired development of Peyer’s patches, indicating factors account for p38’s function in gene expression or a critical role of TNFR1 in acute immune responses (30, 31). another reported p38 function in stimulus-induced mRNA TNFR1 mediates its physiological functions through activation stabilization (27). In addition, p38 is involved in inflamma- of canonical IKK, JNK, MAPK, and ERK. tion, cell death, cell cycle regulation, and cell differentia- Upon binding of TNF to the TNF receptor, the first steps in tion. Inhibition of p38 using specific pharmacological signaling are the recruitment of the adapter molecules TRADD inhibitors results in reduced proliferation and cell cycle and RIPK1 and then of TRAF2 ⁄ 5 to TRADD (35). TRADD is arrest (19, 21, 26). essential for TNF-induced signaling in MEFs as in TRADD-

32 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

deficient MEFs TNFR-induced IjBa phosphorylation and deg- replace TAK1 or whether they act cooperatively is currently radation are completely abolished (36–38). In contrast, in not clear. Activation of the NEMO-IKK complex results in macrophages the TRADD requirement is less complete, sug- phosphorylation and degradation of the canonical IjBs and gesting that in these cells TRAF2 may be recruited in a NFjB activation. TRADD-independent manner (38). The requirement for RIP1 In addition to the activation of IKK, TNFR1 also leads to the varies between cell types; while largely dispensable in MEFs potent activation of JNK, p38, and ERK1 ⁄ 2. Activation of JNK (39–41), it is absolutely required for TNF-induced NFjB acti- depends on TRAF2 and on the formation of ‘non-destructive’ vation in T and B cells, as human T cells lacking RIPK1 and K63 ubiquitin chains (63–70). The involvement of linear pre-B-cell lines derived from RIPK1 knockout mice show chains is still controversial (53, 54, 71). Activation of p38 defects in NFjB activity (42–44). Notably, its kinase activity similarly depends on TRAF2 and RIPK1; however, the contri- is not required (39). TRAF2-deficient mice show only a slight bution of ubiquitin chains is unclear (72, 73). Several reduction in TNFR-induced NFjB activation, while TRAF2 ⁄ 5 MAP3Ks have been implicated in the activation, including double deficiency results in complete impairment, suggesting TAK1 (74), MEKK1 (74), TPL-2 (75), and ASK1 (74, 76, 77). redundant functions for these molecules (39). However, this Genetic deletions of MAP3Ks have not revealed an absolute topic remains controversial. requirement for any kinase, indicating some redundancy in Subsequently, cIAP1 ⁄ 2 are recruited to the complex, which TRAF2-dependent JNK and p38 activation. MAP3Ks then is not only important for NEMO-containing IKK activation phosphorylate and activate MAP2Ks, which are somewhat (45–47) but also for inhibition of the non-canonical IKK1 pathway specific. While MKK3 and MKK6 specifically phos- activity, as these E3 are also involved in constitutive phorylate and activate p38, MKK7 activates JNK. MKK4 is less degradation of NIK (48, 49) (see below). Within this com- specific and can activate both p38 and JNK (reviewed in 78). plex, RIPK1, TRAFs, and NEMO undergo several forms of The main activator of the TNF-induced ERK1 ⁄ 2 cascade is ‘non-destructive’ ubiquitination, primarily conjugation of TPL-2 (79). Macrophages from Tpl2 knockout mice are defec- K63 or head-to tail (linear) polyubiquitin chains. While K63 tive in ERK1 ⁄ 2 activation induced by TNF, while activation of ubiquitination of RIPK1 and NEMO is catalyzed by the E3 lig- other MAPKs and NFjB remain unaffected (79). In contrast, ases cIAP1 ⁄ 2 or TRAF2 together with Uev1A and Ubc13 (44, TNF stimulation of Tpl2-deficient MEFs results in defective 45, 50), the LUBAC complex, consisting of HOIL-1, HOIP, activation of JNK, p38, ERK1 ⁄ 2 as well as NFjB (75), indicat- and SHARPIN, exclusively modifies RIPK1 and NEMO with ing a cell type-specific role for TPL-2. linear polyubiquitin chains (51–54). Conjugation of K63 and linear ubiquitin chains to RIP1 triggers the recruitment of IKK activating kinases. First, the IKK-kinase TAK1 (IKK-K) is CD40 recruited by specific binding of its scaffolds TAB2 ⁄ 3 to the CD40 is expressed on DCs, B cells, and endothelial cells. In ubiquitin chains (39, 50, 55). TAK1 itself is K63 ubiquitinat- DCs, CD40 signaling promotes production, induc- ed, which triggers its autophosphorylation and activation tion of costimulatory molecules, and facilitates cross presenta- (55–57). In vitro unanchored K63 polyubiquitin chains have tion; in B cells, CD40 signaling can promote germinal center been shown to be sufficient for TAK1 activation (58), but formation, isotype switching, somatic hypermutation, and whether conjugation of ubiquitin chains to components of formation of plasma cells and memory B cells. In addition, it the complex may contribute to dose-responsiveness or speci- has been demonstrated to be important for the survival of ficity in vivo remains to be investigated. many cell types, including GC B cells, DCs, and endothelial Binding of NEMO to these ubiquitin chains brings IKK in cells. CD40-deficient mice show defective B-cell development, close proximity to TAK1, allowing for IKK activation by Ig class switching, and GC formation, ultimately causing phosphorylation of the activation loop serines (50–52, 59). immunodeficiency (80–83). In similarity to TNFR1, CD40’s TAK1 appears to be the major kinase responsible for IKK acti- physiological functions are also mediated by the signal trans- vation, as TAK1-deficient cells are largely defective in TNF- ducers IKK1, IKK2, JNK, p38, and ERK. induced NFjB activation (60, 61). However, MEKK3 has also In contrast to TNFR1 signaling, activation of CD40 signal- been reported to act as an IKK kinase. Similar to TAK1, ing does not require the adapter TRADD but instead is initi- MEKK3 binds to RIPK1 and TRAF2 and can phosphorylate ated by binding of ubiquitin ligases TRAF2, TRAF3, and IKK2 (62). Consistently, MEKK3 deficiency impairs IKK and TRAF6 directly to the receptor (84, 85). In the case of CD40 MAPK activation in fibroblasts (62). Whether MEKK3 can activation, the TRAF molecules do not appear to have

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 33 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

redundant functions. TRAF2 has been shown to be the pri- In similarity to TNFR1 signaling, both TAK1 and MEKK3 mary mediator of JNK and p38 activation (67, 68, 86). are implicated as the activating kinases for canonical IKK by Engagement of TRAF2 to CD40 results in the recruitment of CD40 ligand engagement (57, 106, 107). Several kinases have MEKK1 (87), which drives the phosphorylation of JNK and been suggested to mediate activation of MAPK pathways. As p38. TRAF2-deficient fibroblasts are defective in CD40L- mentioned above, MEKK1 is recruited to the CD40 complex induced JNK and p38 activation, with only little defect in through its interaction with TRAF2 to induce JNK and p38 NFjB activation (67, 68, 86). Attenuation of MAPK activation activation (87). Accordingly, in MEKK1-deficient B cells no was also observed in TRAF2-deficient B cells (88). In addi- JNK and p38 activation can be detected, while there are con- tion, TRAF2 deficiency caused constitutive p100 processing flicting results on the effects on NFjB activation (108–111). and elevated c-Rel activity (88). This reflects the fact that in Several other MAP3Ks have been suggested to be important contrast to TNFR1, CD40 engagement also triggers the activa- for JNK and p38 activation, including TAK1, MEKK3, and tion of non-canonical IKK through NIK (89). In unstimulated TPL-2. Although TAK1 is critical for TNF, BCR, and TLR cells, NIK expression is very low due to its constitutive degra- ligand induced activation of MAPKs, it appears to only play a dation by cIAPs, TRAF2, and TRAF3 (48, 49). CD40 engage- minor role in CD40-induced signaling, as CD40 engagement ment triggers self-degradation of TRAF2 and cIAP-dependent in TAK1-deficient B cells shows only modest defects in JNK degradation of TRAF3, resulting in stabilization and accumu- and p38 activation (61). In overexpression experiments lation of NIK (90–93). Indeed, germ line inactivation of MEKK3 has been demonstrated to be able to induce JNK and either TRAF2 or TRAF3 leads to NIK accumulation and con- p38 activation (112), due to embryonic lethality the physio- stitutive p100 processing, indicating that NIK accumulation is logical relevance for CD40-induced MAPK activation however sufficient for its activation of non-canonical NFjB pathway remains unclear (113). Interestingly, the MAP3K TPL-2 does (94–96). It subsequently activates IKK1 through phosphoryla- not appear to be involved in JNK, p38, or NFjB activation in tion of activation loop serines. NIK and IKKa phosphorylate CD40 stimulated B cells (79) but instead plays a critical role in p100 resulting in its processing and induction of NFjB tran- the activation of the ERK pathway (79). Tpl2-deficient mice scription (97–99). show a partial activation defect of ERK in response to CD40 Binding of TRAF6 to CD40 is important for activation of and TLR activation resulting in partial inhibition of IgE pro- JNK and p38 activation as well as NFjB (100). Its deficiency duction (79, 114). TPL-2 has also been implicated to be results in reduction or abrogation in the activation of canoni- involved in processing of NFjB1 p105 (115); however, pro- cal NFjB, JNK, and p38 (101, 102). Similar to TNFR signal- cessing appeared to be normal in Tpl2 knockout mice (79). ing, the cIAP1 ⁄ 2 and LUBAC ubiquitin ligase complexes have also been shown to be recruited to the CD40 receptor (103– 105). Whereas TNFR1 signaling involves conjugation of K63 LTbR and linear Ub-chains to recruit RIPK1 and NEMO and mediate LTbR signaling primarily controls the development of second- activation of TAK1 and downstream MAPK and canonical IKKs ary lymphoid organs (lymph nodes and Peyer’s patches). (44, 45, 50–54), CD40 signaling requires ubiquitination of LTbR-deficient mice lack all lymph nodes, Peyer’s patches, TRAF2 and TRAF6 for these downstream signaling events. and display a disturbed splenic architecture (116–118). In Interestingly, K63 ubiquitination appears to be critical for JNK adults, LTbR also controls the maturation and maintenance of and p38 activation and less important for CD40-induced the microarchitecture of lymphoid organs through expression ) ) NFjB activation, as B cells and macrophages from ubc13 ⁄ of specific chemokines (such as CXCL13 and CCL19), which mice, which are defective in catalyzing K63 chains, show induce stromal cell differentiation (117, 119). strong defects in JNK and p38 activation, while NFjB activa- In contrast to CD40 signaling, which leads to the activation tion is largely intact (105). In contrast, interfering with com- of MAPKs, the canonical NEMO-IKK2 complex and non- ponents of the LUBAC complex results in defective NFjB and canonical-NIK-IKK1, LTb, and BAFFR only induce NEMO- JNK activation (52, 54), indicating differential requirements independent, non-canonical IKK1. In similarity to CD40, the of Ub chains for MAPK and NFjB activation. A more detailed cytoplasmic domain of LTbR associates with TRAF3 and understanding of the nature of Ub-chains important for sig- TRAF2 (120, 121). Ligation of LTab or LIGHT to the LTb naling, the proteins they are conjugated to (if any) and the receptor triggers the degradation of TRAF2 and TRAF3 result- kinetics of Ub-chain formation will be important for a better ing in accumulation of NIK and subsequent activation of IKK1 understanding of signaling specificity. (see CD40 signaling). Through IKK1-induced degradation

34 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

and processing of p100 RelA:p50 and RelB:p52 dimers are interactions with substrates, binding can trigger allosteric con- released into the nucleus to activate transcription (122–124). formational changes that can in turn affect strength and dura- NFjB activation induced by developmental signals such as tion of MAPK signaling (133). However, not all known LTb is considerably slower and weaker as compared to inflam- substrates have docking sites, suggesting that additional mech- matory signals. LTbR engagement also weakly induces JNK anisms are likely to exist in order to achieve substrate selectiv- activation in a TRAF2-dependent manner (125, 126); the ity to ensure efficient and specific signaling (127). exact mechanism however remains elusive. IKK1 and IKK2 are also known to be pleiotropic kinases that can phosphorylate a wide range of substrates influencing Mechanisms that ensure signaling specificity diverse cellular responses (134, 135). Interestingly, in in vitro assays IKKs do not show a high degree of substrate specificity, As described above, TNFRSF signal transduction pathways and in contrast to MAPKs, IKKs do not appear to have docking involve a small number of signaling , yet their bio- sites raising the question of how their activity is regulated to logical responses are highly diverse. Over the past few years, ensure signaling specificity. To understand the molecular basis many studies using a variety of genetic tools have revealed that for signaling specificity in cellular pathways, it will be of the functional requirements of specific signaling proteins in major importance to further elucidate mechanisms that con- signal transduction are cell type specific. Cell type-specific trol substrate specificity in vivo. genetic requirements may be the result of differential expres- sion of parallel, potentially compensating pathway compo- nents. However, it may also be the result of cell-type specific Signaling specificity via scaffolding expression of proteins that modulate the function or kinetics When analyzing signaling responses to diverse biological of the key signaling enzymes that thereby control signaling inputs, it becomes clear that many signal transducers are specificity. shared, and, as described above, their substrate specificities Broadly, the specificity of signaling can be controlled at the appear limited. Yet, the biological outputs are highly specific. level of substrate specificity of the key kinases, differential Signaling specificity can be achieved by organizing discrete wiring of signaling inputs through scaffolding, the kinetics of subsets of proteins in space and time. One way to achieve this the kinase activities, as well as through combinatorial and is by sequestering functionally interacting proteins into temporal control mechanisms. Below we discuss some of specific subcellular compartments, such as organelles or the these mechanisms of regulation and how they may contribute plasma membrane. Another prevalent strategy is the assembly to signaling specificity. of functionally interacting proteins into specific complexes through protein scaffolds. Scaffolds bind to two or more com- Regulation of the enzymatic substrate specificity of kinases ponents of a cascade, bringing them in close proximity, MAPKs and IKKs are highly pleiotropic kinases, involved in thereby not only facilitating efficient propagation of the signal numerous distinct biological responses by phosphorylating a but also mediating its insulation from other signals. In addi- diverse array of substrates. MAPKs have been estimated to have tion to organizing signaling molecules into signalosomes, as many as 200–300 substrates each (127–129). One mecha- scaffolds can also have allosteric effects on the kinases thus nism by which specificity may be ensured in vivo may involve regulating kinase activity itself. co-factors that alter the intrinsic enzymatic specificity. How- The most prominent signaling cascades that are regulated by ever, unlike other enzymes such as prokaryotic RNA polyme- scaffolds are the MAPK pathways. As mentioned above, initia- rases whose DNA binding specificity is regulated by sigma tion of JNK and p38 pathways is triggered by many MAP3Ks factors, surprisingly little information has emerged about including TAK1, ASK1, MEKKs, and TPL-2 (74–77), which, specificity factors for the pleiotropic kinases in the TNFR sig- among others, can phosphorylate MKK4 and MKK7 to activate naling network. JNK, and MKK3, MKK4, and MKK6 to activate p38 (78). The MAPKs are known to have substrate binding sites, usually usage of alternative kinases at each step of the cascade might referred to as docking domains (130–132). These domains allow for the precise stimulus-dependent control of MAPKs. are distinct from the serine ⁄ threonine phospho-acceptor sites Indeed, the stimulus-specific organization of different kinases and consist of positively and negatively charged residues. into cascades by scaffolds can create functional signaling mod- Docking interactions themselves can be regulated by post- ules to control specificity of signal transduction (Fig. 3). For translational modifications (130–132). Apart from regulating example, filamin has been identified as a scaffold that may

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 35 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

ABC

Fig. 3. Scaffolding to achieve signaling specificity. Scaffolds can organize pleiotropic kinases into stimulus-specific signaling modules to direct them to one specific pathway. (A) Filamin interacts with TRAF2, MEKK4, and JNK to allow for JNK activation upon TNFR1 engagement. ERK1 ⁄ 2 and p38 might similarly be organized into stimulus-specific modules through yet unidentified scaffolding proteins. (B) Binding of NEMO to IKK2 allows for its activation but is also required for proper signaling propagation to IjBa in response to inflammatory signals. (C) NIK is not only the activating kinase of IKK1 but also channels IKK1 activity to p100. coordinate the specific activation of JNK upon stimulation with 50–52, 55, 59). Thus, NEMO links upstream inflammatory TNF (136). The actin binding protein interacts with the signals to IKK2 through binding to ubiquitin chains. How is TNFR1 adapter TRAF2, the MAP2K MKK4, and JNK, thereby active IKK2 able to phosphorylate IjBs so efficiently and rap- coordinating a module that channels the upstream TNFR1 sig- idly while phosphorylation of alternative substrates is much nal to JNK (137). Accordingly, filamin deficiency causes a weaker and occurs with much slower kinetics? Our unpub- selective loss of TNF-induced JNK activation (136). The dual lished data suggest that IKK2-dependent phosphorylation of specificity SKRP1 has also been reported IjB is also mediated by NEMO (BS and AH, unpublished to be involved in TNF-induced JNK activation through a scaf- data). Binding of NEMO to IjBs specifically channels IKK2 folding function facilitating the interaction of the MAP3K kinase activity towards inflammatory responses. Thus, scaf- ASK1, MAP2K MKK7, and JNK (138–141). Similar examples folding of NEMO ensures downstream signaling specificity by for a stimulus-specific organization of MAPK cascades by scaf- directing the pleiotropic IKK2 kinase to the canonical NFjB folds can be found for the activation of p38. The osmo-sensing pathway. The complex between NEMO-IKK2 and IjBa scaffold for MEKK3 (OSM) for example associates with Rac, appears to be constitutive, which might not only ensure MEKK3, and MKK3 to regulate p38 activation specifically in specific phosphorylation of IjBs but also enables the rapid response to osmotic shock (142). Other p38-specific scaffolds response, necessary for efficient inflammatory signal transduc- include JIP4 (143) and JLP1, which is implicated in p38- tion. In analogy to stimulus-specific MAPK kinase modules, dependent regulation of differentiation (144–146). Interest- ingly, the MAP3K MEKK1 not only acts as a kinase to activate JNK upon CD40 ligation, but itself possesses a scaffolding function by interacting with components of the JNK module (108, 147–151). It also constitutively interacts with Raf-1, MEK1, and ERK2 to facilitate stimulus-induced activation of ERK1 ⁄ 2 (147, 152, 153). TAK1, which is involved in trans- ducing signals from diverse upstream receptors (e.g. TNFRs, TLRs, NLRs) to activate downstream JNK, p38, as well as NFjB pathways, associates with the scaffolds TAB1, TAB2, and TAB3. Apart from mediating TAK1 activation (see below), through binding to poly-ubiquitin chains, TAB2 ⁄ 3 also recruit TAK1 to NEMO-IKK2 to mediate NFjB activation. A well-known scaffold necessary for the activation of the Fig. 4. Combinatorial control of kinases facilitates stimulus specific- canonical IKKs is NEMO (154–156). Binding of NEMO to ity. Engagement of TNFRSFs activates pathways leading to the activation stimulus-induced polyubiquitin chains brings IKKs in close of ERK, JNK, p38, IKK2, and IKK1. Phosphorylation of downstream proximity to each other (allowing for trans-autophosphoryla- effectors induces a transcriptional program. Some genes require a syner- gistic function of several transcription factors and may thus be transcribed tion) and to the IKK-kinase TAK1, which then activates IKK2 in a stimulus-specific manner. Specificity can also be achieved post- through phosphorylation of its activation loop serines (39, transcriptionally through p38-dependent regulation of mRNA stability.

36 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

NEMO assembles an inflammation-specific IKK module by results in defective JNK activation and shifts the balance of linking upstream IKK activators with the downstream effector signaling towards non-canonical IKK, despite a defect in IjBa. The specificity of IKK1 for p100 also seems to be regu- TRAF3 degradation (86, 88). This observation highlights the lated through scaffolding. NIK forms a complex with IKK1 importance of TRAF2 in negatively regulating the activation and p100, thereby not only activating IKK1 but also directing of non-canonical IKKs. In B cells, TRAF3 deficiency similarly IKK1s kinase activity towards p100. Thus, active IKK1 will results in enhanced activation of NIK-IKK but also in enhanced only induce p100 processing when NIK is stabilized by spe- canonical IKK and JNK activity (48, 49, 159, 160). Thus, cific developmental signals (e.g. LTb, CD40) (99). although both TRAF2 and TRAF3 are important negative regu- In summary, pathway-specific scaffolds can direct a pleio- lators of non-canonical IKK, they have different functions in tropic kinase to one specific pathway and at the same time regulating JNK activity. TRAF6 is dispensable for TNFR1 and prevent the activation of pathways irrelevant to that particular LTb signaling but is essential for CD40-induced JNK, p38, stimulus. Thus, scaffolds ensure stimulus-specific functions of ERK1 ⁄ 2, and canonical IKK activation (84, 85). kinases. The identification of scaffolds that are specific for each For activation of downstream kinases, TRAFs do not just act TNFR superfamily member will be critical to gain a better as adapters or scaffolds, but their E3 ubiquitin ligase activity is understanding about the regulation of signaling specificity. required. In conjunction with Uev1A and Ubc13, TRAFs mediate the conjugation of K63-linked ubiquitin chains onto other TRAFs, RIPK1, and NEMO, which has been shown to be Combinatorial coding critical for activation of NEMO-IKK2 and JNK by TNF as well Signaling specificity may also be mediated through the combi- as CD40L (44, 50, 70, 103–105). In fact, non-destructive natorial use of several pleiotropic signal transducers (Fig. 4). ubiquitin chains (such as K63 and linear chains) might add an In this scenario, stimulation of a single receptor triggers the additional level of regulation to achieve signaling specificity. formation of a signalosome that allows for the activation of a The type of ubiquitin chain as well as the molecules they are subset of signal transducers. Downstream effectors (e.g. the conjugated onto are likely to be stimulus specific. Interest- promoters of potential target genes) may integrate the combi- ingly, TNF and IL1b signaling appear to have differential nations of signal transducers (e.g. transcription factors) to requirements for ubiquitin chain types (161). While for determine the stimulus-specific activity level (e.g. of target IL-1b-induced IKK activation the Uev1A ⁄ Ubc13 ⁄ TRAF2 E3 genes). ligase complex that is restricted to K63 chain linkage is neces- Because they lack intrinsic enzymatic activities, TNFRs asso- sary for IKK activation, TNF-induced signaling does not ciate with TRAF proteins to initiate intracellular signaling. require catalysis of K63 chains (161). Recently, mass spec TRAF protein recruitment forms the basis for the encoding of analysis revealed that RIPK1 and NEMO are heavily ubiquiti- a combinatorial code by which a receptor-associated complex nated with K63, linear and K11-linked chains upon stimula- leads to the activation of several pleiotropic kinases. The fam- tion with TNF, indicating the high level of diversity in ily of TRAF proteins consists of 6 members, TRAF1-6, that ubiquitin modifications that occur during signaling (54). have both overlapping as well as distinct roles. The TRAF pro- More work is required to get a better understanding of the teins with which the receptor associates at least in part deter- stimulus specific role of ubiquitin chains. The use of ubiqu- mines the pathways that are activated. TRAFs are not only itin-chains as scaffolds allows for highly flexible and dynamic critically involved in signaling of TNFR superfamily members assembly of diverse signaling complexes but also for the but also play important roles in TLR ⁄ IL-1R signaling, where recruitment of specific activators and inactivators. Binding of TRAF3 is indispensable for IRF3 activation upon TLR3 engage- the scaffolds TAB 2 ⁄ 3 to polyubiquitin chains for example ment (157), or TRAF6 is necessary for activation of NFjBby recruits the MAP3K TAK1 to the TNFR1 complex to allow for LPS (101, 158). TRAFs have a RING-finger domain (RING) IKK2 activation, which in turn also depends on the ability of with an associated E3 ligase activity, a Zinc-finger (ZF) motif, its scaffold NEMO to bind to ubiquitin chains (39, 50–52, 55, and a highly conserved C-terminal domain that mediates 59). In addition to coordinating a signaling cascade, one can homo- and heterodimerization of TRAFs as well as association imagine that ubiquitin chains may branch one upstream signal with cell surface receptors (85, 159). to multiple downstream kinases through association of dis- The association of TRAF2 to CD40 is important for activa- tinct ubiquitin binding proteins to different types of poly- tion of MAPKs and to a lesser extent for the activation of ubiquitin chains, thereby playing a key role in encoding a canonical IKKs (67, 68, 86). Accordingly, TRAF2 deficiency combinatorial signaling code.

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 37 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

The decoding of combinatorially coded signals has been Evidence for the temporal coding model for generating studied primarily at the level of gene expression. Gene pro- stimulus-specific signaling can be found in JNK, p38, and ERK moters or enhancers often contain binding sites for several signaling (170, 172). A prominent example demonstrating signal-responsive transcription factors that are the down- the importance of temporal control in MAPK signaling is the stream effectors of pleiotropic kinases. Synergistic function neuronal cell line PC12, in which ERK signaling can induce by several transcription factors has been documented in syn- proliferation (in response to EGF), as well as differentiation thetic experimental systems (162). The multivalent interac- (in response to NGF) depending on the duration of the signal tions by the ubiquitous co-activators CBP ⁄ p300 suggest a (173). While EGF induced MAPK signaling is transient, MAPK possible decoding mechanism in vivo. For some specific activity induced by NGF is sustained. Extensive evidence for genes, a combinatorial requirement has been established and the importance of temporal coding also comes from canonical examined at a mechanistic level. The classic example is the NFjB signaling. Quantitative measurements of the canonical control of the IFNb enhancer, which requires the activity of IKK activity revealed that its induced activity profile is stimu- AP-1, IFN response factor 3 (IRF3), and NFjB. A nucleo- lus-specific (171). Whereas TNF activates a transient IKK some located over the transcription start site was identified activity, LPS leads to prolonged IKK activity. In the former as the block for transcription initiation, and only the assem- case, the negative feedback regulator A20 limits late IKK activ- bly of an ‘enhanceosome’ (163) allowed for the recruitment ity, whereas in the latter, cytokine feedback via TNF ensures of chromatin remodeling factors that in turn allow for pre- an elevated late phase. Within the TNFRSF network, temporal initiation complex assembly (164). Interestingly, although control of effectors is mediated by the kinetics of key reaction AP-1 and NFjB are separated by four IRFs on the enhancer, mechanisms, such as receptor internalization, recycling, and protein-protein interactions between them were found to be replenishment, ubiquitin chain formation rates and their deg- critical (165). These are facilitated by HMG proteins capable radation through deubiquitin enzymes (DUSPs) such as A20, of inducing DNA bends (166). Similarly, expression of the nuclear translocation rates, and negative feedback mechanism chemokine MCP1 ⁄ CCL2 requires communication between a impacting IKK or NFjB(Fig. 5). proximally bound SP-1 and a distally bound NFjB that How may target genes distinguish between different tem- suggests looping (167). A different mechanism poral profiles of NFjB activity? We may imagine a variety of of combinatorial control has been proposed for the TNF gene. Expression of TNF requires NFjB binding to its promoter but also ERK and p38 signals to control mRNA nuclear-cytoplasmic transport, mRNA stability, and transla- tion (114, 168, 169). These examples indicate different mechanisms in the gene regulatory network may be targeted by the combinatorial control of TNFRSF-induced intracellu- lar signals.

Temporal coding Biochemical cell population timecourse studies and single cell studies with in vivo reporters have revealed that the activity profile of TNFRS-induced transcription factors is dynamic and that the observed temporal profiles are stimulus-specific (170, 171). These observations led to the proposal that stimulus- Fig. 5. Temporal control can facilitate stimulus specificity. The tem- specific temporal control of pleiotropic transducers may allow poral profile of key signal transducers, whether pleiotropic kinases or for stimulus-specific signaling (7). The temporal code may be transcription factors, may determine signaling specificity. Temporal pro- files are encoded and transduced through kinetic mechanisms that control encoded via receptor-associated mechanisms, transmitted via receptor internalization and recycling, half-life control of signaling medi- stimulus-specific temporal control of a pleiotropic transducer, ators, including ubiquitin chain second messengers or scaffolds, as well and decoded by mechanisms associated with the effector, for as negative and positive feedback. Signaling dynamics may be decoded to determine the activity of effectors, such as downstream genes, by mRNA example the gene regulatory network controlling the expres- half-life control, with other transcription factors or chroma- sion of a target gene. tin regulatory mechanisms.

38 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

mechanisms by which the temporal code may be decoded by begin to explain the mechanisms by which signaling specific- gene regulatory networks (GRNs). At its simplest, short versus ity is achieved. The most prominent of these may be the utili- long mRNA half-life may sensitize a gene to transient versus zation of scaffolding proteins, which may recruit kinases to prolonged NFjB activity. Coupled to non-linear or threshol- upstream activators or to downstream substrates. Indeed when ded dose response curves of promoters, the GRN may decode such scaffolds are multivalent, the same protein may deter- complex temporal profiles of NFjB in specific ways. In more mine both upstream activators and downstream targets, thus sophisticated GRNs, NFjB may need to coincident with or placing the kinase within a cascade and effectively insulating it phase shifted relative to other transcription factors to allow for from other signaling pathways. Within the MAPK field, prom- gene activation. On others yet, NFjB may be required to be inent examples of such scaffolds are the yeast Ste5 ⁄ Pbs2 scaf- coincident with a transcription factor induced by the early fold modules, and in mammals, the MAPK kinase cascades NFjB activity, forming a feed forward control loop. that are organized by various stimulus specific scaffolds. In NFjB signaling, NEMO appears to play a similar role (Schro¨- Conclusion felbauer et al. unpublished data). However, we expect that The signaling network innervated by members of the TNFR ongoing efforts to understand specificity of signaling within superfamily consists of a number of kinases with highly pleio- the TNFRSF network will lead to the identification of other tropic functions. Generally, these kinases show too little such scaffolding proteins and the characterization of their intrinsic specificity in vitro to account for their apparent signal- roles in signaling insulation and as mediators of signaling ing specificity in cells, but several models have emerged that crosstalk.

References

1. Aggarwal BB. Signaling pathways of the kinase complex. J Biol Chem 2002; 19. Huang G, Shi LZ, Chi H. Regulation of JNK TNF superfamily: a double-edged sword. 277:48115–48121. and p38 MAPK in the : sig- Nat Rev Immunol 2003;3:745–756. 11. Lee DF, et al. IKK beta suppression of TSC1 nal integration, propagation and termina- 2. Locksley RM, Killeen N, Lenardo MJ. The links inflammation and tumor angiogenesis tion. Cytokine 2009;48:161–169. TNF and TNF receptor superfamilies: inte- via the mTOR pathway. Cell 2007; 20. Manning AM, Davis RJ. Targeting JNK for grating mammalian biology. Cell 130:440–455. therapeutic benefit: from junk to gold? Nat 2001;104:487–501. 12. Lamberti C, et al. Regulation of beta-catenin Rev Drug Discov 2003;2:554–565. 3. Bodmer JL, Schneider P, Tschopp J. The function by the IkappaB kinases. J Biol 21. Rincon M, Davis RJ. Regulation of the molecular architecture of the TNF super- Chem 2001;276:42276–42286. immune response by stress-activated protein family. Trends Biochem Sci 2002;27:19– 13. Comb WC, Cogswell P, Sitcheran R, Bald- kinases. Immunol Rev 2009;228:212–224. 26. win AS. IKK-dependent, NF-kappaB-inde- 22. Gupta S, Campbell D, Derijard B, Davis RJ. 4. Vandenabeele P, Declercq W, Van Herrewe- pendent control of autophagic gene Transcription factor ATF2 regulation by the ghe F, Vanden Berghe T. The role of the expression. Oncogene 2011;30:1727– JNK signal transduction pathway. Science kinases RIP1 and RIP3 in TNF-induced 1732. 1995;267:389–393. necrosis. Sci Signal 2010;3:re4. 14. Criollo A, et al. The IKK complex contrib- 23. Livingstone C, Patel G, Jones N. ATF-2 con- 5. Vandenabeele P, Galluzzi L, Vanden Berghe utes to the induction of autophagy. EMBO J tains a phosphorylation-dependent tran- T, Kroemer G. Molecular mechanisms of 2010;29:619–631. scriptional activation domain. EMBO J necroptosis: an ordered cellular explosion. 15. Carayol N, Wang CY. IKKalpha stabilizes 1995;14:1785–1797. Nat Rev Mol Cell Biol 2010;11:700–714. cytosolic beta-catenin by inhibiting both 24. van Dam H, Wilhelm D, Herr I, Steffen A, 6. Mackay F, Schneider P. Cracking the BAFF canonical and non-canonical degradation Herrlich P, Angel P. ATF-2 is preferentially code. Nat Rev Immunol 2009;9:491–502. pathways. Cell Signal 2006;18:1941–1946. activated by stress-activated protein kinases 7. Hoffmann A, Baltimore D. Circuitry of 16. Hoberg JE, Yeung F, Mayo MW. SMRT to mediate c-jun induction in response to nuclear factor kappaB signaling. Immunol derepression by the IkappaB kinase alpha: a genotoxic agents. EMBO J 1995;14:1798– Rev 2006;210:171–186. prerequisite to NF-kappaB transcription and 1811. 8. Shih VF, Tsui R, Caldwell A, Hoffmann A. A survival. Mol Cell 2004;16:245–255. 25. Chrivia JC, Kwok RP, Lamb N, Hagiwara M, single NFkappaB system for both canonical 17. Park KJ, Krishnan V, O’Malley BW, Yamam- Montminy MR, Goodman RH. Phosphory- and non-canonical signaling. Cell Res oto Y, Gaynor RB. Formation of an IKKal- lated CREB binds specifically to the nuclear 2011;21:86–102. pha-dependent transcription complex is protein CBP. Nature 1993;365:855–859. 9. Xia Y, Padre RC, De Mendoza TH, Bottero required for estrogen receptor-mediated 26. Ashwell JD. The many paths to p38 mito- V, Tergaonkar VB, Verma IM. Phosphoryla- gene activation. Mol Cell 2005;18:71–82. gen-activated activation in tion of p53 by IkappaB kinase 2 promotes 18. Huang WC, Ju TK, Hung MC, Chen CC. the immune system. Nat Rev Immunol its degradation by beta-TrCP. Proc Natl Acad Phosphorylation of CBP by IKKalpha pro- 2006;6:532–540. Sci USA 2009;106:2629–2634. motes cell growth by switching the binding 27. Winzen R, et al. The p38 MAP kinase path- 10. Gao Z, et al. Serine phosphorylation of insu- preference of CBP from p53 to NF-kappaB. way signals for cytokine-induced mRNA lin receptor substrate 1 by inhibitor kappa B Mol Cell 2007;26:75–87. stabilization via MAP kinase-activated

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 39 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

protein kinase 2 and an AU-rich region-tar- mediates IKK activation. Immunity 54. Gerlach B, et al. Linear ubiquitination pre- geted mechanism. EMBO J 1999;18:4969– 2000;12:419–429. vents inflammation and regulates immune 4980. 41. Wong WW, Gentle IE, Nachbur U, Ander- signalling. Nature 2011;471:591–596. 28. Roux PP, Blenis J. ERK and p38 MAPK-acti- ton H, Vaux DL, Silke J. RIPK1 is not essen- 55. Kanayama A, et al. TAB 2 and TAB 3 activate vated protein kinases: a family of protein tial for TNFR1-induced activation of NF- the NF-kappaB pathway through binding to kinases with diverse biological functions. kappaB. Cell Death Differ 2010;17:482– polyubiquitin chains. Mol Cell 2004;15: Microbiol Mol Biol Rev 2004;68:320–344. 487. 535–548. 29. Cagnol S, Chambard JC. ERK and cell death: 42. Ting AT, Pimentel-Muinos FX, Seed B. RIP 56. Scholz R, Sidler CL, Thali RF, Winssinger N, mechanisms of ERK-induced cell death – mediates tumor necrosis factor receptor 1 Cheung PC, Neumann D. Autoactivation of apoptosis, autophagy and senescence. FEBS J activation of NF-kappaB but not Fas ⁄ APO- transforming growth factor beta-activated 2010;277:2–21. 1-initiated apoptosis. EMBO J 1996; kinase 1 is a sequential bimolecular process. 30. Pfeffer K, et al. Mice deficient for the 55 kd 15:6189–6196. J Biol Chem 2010;285:25753–25766. tumor necrosis factor receptor are resistant 43. Kelliher MA, Grimm S, Ishida Y, Kuo F, 57. Ishitani T, Takaesu G, Ninomiya-Tsuji J, to endotoxic shock, yet succumb to L. mono- Stanger BZ, Leder P. The death domain Shibuya H, Gaynor RB, Matsumoto K. Role cytogenes infection. Cell 1993;73:457–467. kinase RIP mediates the TNF-induced NF- of the TAB 2-related protein TAB 3 in IL-1 31. Rothe J, et al. Mice lacking the tumour kappaB signal. Immunity 1998;8:297–303. and TNF signaling. EMBO J 2003;22:6277– necrosis factor receptor 1 are resistant to 44. Li H, Kobayashi M, Blonska M, You Y, Lin 6288. TNF-mediated toxicity but highly suscepti- X. Ubiquitination of RIP is required for 58. Xia ZP, et al. Direct activation of protein ble to infection by Listeria monocytogenes. Nat- tumor necrosis factor alpha-induced NF- kinases by unanchored polyubiquitin ure 1993;364:798–802. kappaB activation. J Biol Chem 2006; chains. Nature 2009;461:114–119. 32. Peschon JJ, et al. TNF receptor-deficient 281:13636–13643. 59. Wu CJ, Conze DB, Li T, Srinivasula SM, Ash- mice reveal divergent roles for p55 and p75 45. Bertrand MJ, et al. cIAP1 and cIAP2 facilitate well JD. Sensing of Lys 63-linked polyubiq- in several models of inflammation. J Immu- cancer cell survival by functioning as E3 lig- uitination by NEMO is a key event in NF- nol 1998;160:943–952. ases that promote RIP1 ubiquitination. Mol kappaB activation [corrected]. Nat Cell Biol 33. O’Brien DP, Briles DE, Szalai AJ, Tu AH, Cell 2008;30:689–700. 2006;8:398–406. Sanz I, Nahm MH. Tumor necrosis factor 46. Mahoney DJ, et al. Both cIAP1 and cIAP2 60. Fan Y, et al. Lysine 63-linked polyubiquiti- alpha receptor I is important for survival regulate TNFalpha-mediated NF-kappaB nation of TAK1 at lysine 158 is required for from Streptococcus pneumoniae infections. Infect activation. Proc Natl Acad Sci USA tumor necrosis factor alpha- and interleu- Immun 1999;67:595–601. 2008;105:11778–11783. kin-1beta-induced IKK ⁄ NF-kappaB and 34. Deckert-Schluter M, Bluethmann H, Rang A, 47. Varfolomeev E, et al. c-IAP1 and c-IAP2 are JNK ⁄ AP-1 activation. J Biol Chem Hof H, Schluter D. Crucial role of TNF critical mediators of tumor necrosis factor 2010;285:5347–5360. receptor type 1 (p55), but not of TNF alpha (TNFalpha)-induced NF-kappaB acti- 61. Sato S, et al. Essential function for the kinase receptor type 2 (p75), in murine toxoplas- vation. J Biol Chem 2008;283:24295– TAK1 in innate and adaptive immune mosis. J Immunol 1998;160:3427–3436. 24299. responses. Nat Immunol 2005;6:1087– 35. Micheau O, Tschopp J. Induction of TNF 48. Vallabhapurapu S, et al. Non-redundant and 1095. receptor I-mediated apoptosis via two complementary functions of TRAF2 and 62. Yang J, et al. The essential role of MEKK3 in sequential signaling complexes. Cell 2003; TRAF3 in a ubiquitination cascade that acti- TNF-induced NF-kappaB activation. Nat 114:181–190. vates NIK-dependent alternative NF-kappaB Immunol 2001;2:620–624. 36. Chen NJ, et al. Beyond tumor necrosis fac- signaling. Nat Immunol 2008;9:1364– 63. Reinhard C, Shamoon B, Shyamala V, tor receptor: TRADD signaling in Toll-like 1370. Williams LT. Tumor necrosis factor alpha- receptors. Proc Natl Acad Sci USA 49. Zarnegar BJ, et al. Non-canonical NF-kap- induced activation of c-jun N-terminal 2008;105:12429–12434. paB activation requires coordinated assem- kinase is mediated by TRAF2. EMBO J 37. Ermolaeva MA, et al. Function of TRADD in bly of a regulatory complex of the adapters 1997;16:1080–1092. tumor necrosis factor receptor 1 signaling cIAP1, cIAP2, TRAF2 and TRAF3 and the 64. Natoli G, et al. Activation of SAPK ⁄ JNK by and in TRIF-dependent inflammatory kinase NIK. Nat Immunol 2008;9:1371– TNF receptor 1 through a non-cytotoxic responses. Nat Immunol 2008;9:1037– 1378. TRAF2-dependent pathway. Science 1046. 50. Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ. 1997;275:200–203. 38. Pobezinskaya YL, et al. The function of Activation of IKK by TNFalpha requires site- 65. Liu ZG, Hsu H, Goeddel DV, Karin M. Dis- TRADD in signaling through tumor necrosis specific ubiquitination of RIP1 and poly- section of TNF receptor 1 effector functions: factor receptor 1 and TRIF-dependent Toll- ubiquitin binding by NEMO. Mol Cell JNK activation is not linked to apoptosis like receptors. Nat Immunol 2008;9:1047– 2006;22:245–257. while NF-kappaB activation prevents cell 1054. 51. Tokunaga F, et al. Involvement of linear death. Cell 1996;87:565–576. 39. Lee TH, Shank J, Cusson N, Kelliher MA. polyubiquitylation of NEMO in NF-kappaB 66. Song HY, Regnier CH, Kirschning CJ, The kinase activity of Rip1 is not required activation. Nat Cell Biol 2009;11:123–132. Goeddel DV, Rothe M. Tumor necrosis for tumor necrosis factor-alpha-induced 52. Haas TL, et al. Recruitment of the linear factor (TNF)-mediated kinase cascades: IkappaB kinase or p38 MAP kinase activa- ubiquitin chain assembly complex stabilizes bifurcation of nuclear factor-kappaB and tion or for the ubiquitination of Rip1 by the TNF-R1 signaling complex and is c-jun N-terminal kinase (JNK ⁄ SAPK) path- Traf2. J Biol Chem 2004;279:33185– required for TNF-mediated gene induction. ways at TNF receptor-associated factor 2. 33191. Mol Cell 2009;36:831–844. Proc Natl Acad Sci USA 1997;94:9792– 40. Devin A, Cook A, Lin Y, Rodriguez Y, Kelli- 53. Ikeda F, et al. SHARPIN forms a linear 9796. her M, Liu Z. The distinct roles of TRAF2 ubiquitin ligase complex regulating 67. Lee SY, Reichlin A, Santana A, Sokol KA, and RIP in IKK activation by TNF-R1: NF-kappaB activity and apoptosis. Nature Nussenzweig MC, Choi Y. TRAF2 is essen- TRAF2 recruits IKK to TNF-R1 while RIP 2011;471:637–641. tial for JNK but not NF-kappaB activation

40 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

and regulates lymphocyte proliferation and 81. Quezada SA, Jarvinen LZ, Lind EF, Noelle homeostasis in secondary lymphoid organs. survival. Immunity 1997;7:703–713. RJ. CD40 ⁄ CD154 interactions at the inter- Immunity 2007;27:253–267. 68. Yeh WC, et al. Early lethality, functional face of tolerance and immunity. Annu Rev 96. Gardam S, Sierro F, Basten A, Mackay F, NF-kappaB activation, and increased sensi- Immunol 2004;22:307–328. Brink R. TRAF2 and TRAF3 signal adapters tivity to TNF-induced cell death in TRAF2- 82. Danese S, Sans M, Fiocchi C. The act cooperatively to control the maturation deficient mice. Immunity 1997;7:715–725. CD40 ⁄ CD40L costimulatory pathway in and survival signals delivered to B cells by 69. Habelhah H, Takahashi S, Cho SG, Kadoya inflammatory bowel disease. Gut the BAFF receptor. Immunity 2008;28: T, Watanabe T, Ronai Z. Ubiquitination and 2004;53:1035–1043. 391–401. translocation of TRAF2 is required for acti- 83. Elgueta R, Benson MJ, de Vries VC, Wasiuk 97. Xiao G, Harhaj EW, Sun SC. NF-kappaB- vation of JNK but not of p38 or NF-kappaB. A, Guo Y, Noelle RJ. Molecular mechanism inducing kinase regulates the processing of EMBO J 2004;23:322–332. and function of CD40 ⁄ CD40L engagement NF-kappaB2 p100. Mol Cell 2001;7:401– 70. Fukushima T, et al. Ubiquitin-conjugating in the immune system. Immunol Rev 409. Ubc13 is a critical component of 2009;229:152–172. 98. Senftleben U, et al. Activation by IKKalpha of TNF receptor-associated factor (TRAF)- 84. Ha H, Han D, Choi Y. TRAF-mediated a second, evolutionary conserved, NF-kappa mediated inflammatory responses. Proc Natl TNFR-family signaling. Curr Protoc Immu- B signaling pathway. Science 2001; Acad Sci USA 2007;104:6371–6376. nol 2009;87:11.9D.1–11.9D.19. 293:1495–1499. 71. Tokunaga F, et al. SHARPIN is a component 85. Bishop GA, Moore CR, Xie P, Stunz LL, 99. Xiao G, Fong A, Sun SC. Induction of p100 of the NF-kappaB-activating linear ubiquitin Kraus ZJ. TRAF proteins in CD40 signaling. processing by NF-kappaB-inducing kinase chain assembly complex. Nature Adv Exp Med Biol 2007;597:131–151. involves docking IkappaB kinase alpha (IKK- 2011;471:633–636. 86. Hostager BS, Haxhinasto SA, Rowland SL, alpha) to p100 and IKKalpha-mediated 72. Yuasa T, Ohno S, Kehrl JH, Kyriakis JM. Bishop GA. Tumor necrosis factor receptor- phosphorylation. J Biol Chem 2004; Tumor necrosis factor signaling to stress- associated factor 2 (TRAF2)-deficient B lym- 279:30099–30105. activated protein kinase (SAPK) ⁄ Jun NH2- phocytes reveal novel roles for TRAF2 in 100. Rowland SL, Tremblay MM, Ellison JM, terminal kinase (JNK) and p38. Germinal CD40 signaling. J Biol Chem 2003;278: Stunz LL, Bishop GA, Hostager BS. A novel center kinase couples TRAF2 to mitogen- 45382–45390. mechanism for TNFR-associated factor activated protein kinase ⁄ ERK kinase kinase 87. Gallagher E, et al. Kinase MEKK1 is required 6-dependent CD40 signaling. J Immunol 1 and SAPK while receptor interacting pro- for CD40-dependent activation of the kinas- 2007;179:4645–4653. tein associates with a mitogen-activated es Jnk and p38, germinal center formation, 101. Lomaga MA, et al. TRAF6 deficiency results protein kinase kinase kinase upstream of B cell proliferation and produc- in osteopetrosis and defective interleukin-1, MKK6 and p38. J Biol Chem 1998; tion. Nat Immunol 2007;8:57–63. CD40, and LPS signaling. Genes Dev 273:22681–22692. 88. Grech AP, Amesbury M, Chan T, Gardam S, 1999;13:1015–1024. 73. Lee TH, et al. The death domain kinase RIP1 Basten A, Brink R. TRAF2 differentially reg- 102. Matsuzawa A, et al. Essential cytoplasmic is essential for tumor necrosis factor alpha ulates the canonical and non-canonical path- translocation of a cytokine receptor-assem- signaling to p38 mitogen-activated protein ways of NF-kappaB activation in mature B bled signaling complex. Science 2008; kinase. Mol Cell Biol 2003;23:8377–8385. cells. Immunity 2004;21:629–642. 321:663–668. 74. Takaesu G, et al. TAB 2, a novel adapter pro- 89. Karin M, Gallagher E. TNFR signaling: 103. Deng L, et al. Activation of the IkappaB tein, mediates activation of TAK1 MAPKKK ubiquitin-conjugated TRAFfic signals con- kinase complex by TRAF6 requires a by linking TAK1 to TRAF6 in the IL-1 signal trol stop-and-go for MAPK signaling dimeric ubiquitin-conjugating enzyme transduction pathway. Mol Cell complexes. Immunol Rev 2009;228:225– complex and a unique polyubiquitin chain. 2000;5:649–658. 240. Cell 2000;103:351–361. 75. Das S, et al. Tpl2 ⁄ cot signals activate ERK, 90. Brown KD, Hostager BS, Bishop GA. Regula- 104. Hofmann RM, Pickart CM. In vitro assembly JNK, and NF-kappaB in a cell-type and stim- tion of TRAF2 signaling by self-induced and recognition of Lys-63 polyubiquitin ulus-specific manner. J Biol Chem 2005; degradation. J Biol Chem 2002;277:19433– chains. J Biol Chem 2001;276:27936– 280:23748–23757. 19438. 27943. 76. Ichijo H, et al. Induction of apoptosis by 91. Liao G, Zhang M, Harhaj EW, Sun SC. Regu- 105. Yamamoto M, et al. Key function for the ASK1, a mammalian MAPKKK that activates lation of the NF-kappaB-inducing kinase by Ubc13 E2 ubiquitin-conjugating enzyme in SAPK ⁄ JNK and p38 signaling pathways. Sci- tumor necrosis factor receptor-associated immune receptor signaling. Nat Immunol ence 1997;275:90–94. factor 3-induced degradation. J Biol Chem 2006;7:962–970. 77. Nishitoh H, et al. ASK1 is essential for 2004;279:26243–26250. 106. Blonska M, You Y, Geleziunas R, Lin X. Res- JNK ⁄ SAPK activation by TRAF2. Mol Cell 92. Moore CR, Bishop GA. Differential regula- toration of NF-kappaB activation by tumor 1998;2:389–395. tion of CD40-mediated TNF receptor-asso- necrosis factor alpha receptor complex-tar- 78. Wang X, Destrument A, Tournier C. Physio- ciated factor degradation in B lymphocytes. geted MEKK3 in receptor-interacting pro- logical roles of MKK4 and MKK7: insights J Immunol 2005;175:3780–3789. tein-deficient cells. Mol Cell Biol 2004; from animal models. Biochim Biophys Acta 93. Varfolomeev E, et al. IAP antagonists induce 24:10757–10765. 2007;1773:1349–1357. autoubiquitination of c-IAPs, NF-kappaB 107. Wang C, Deng L, Hong M, Akkaraju GR, 79. Eliopoulos AG, Wang CC, Dumitru CD, activation, and TNFalpha-dependent apop- Inoue J, Chen ZJ. TAK1 is a ubiquitin- Tsichlis PN. Tpl2 transduces CD40 and TNF tosis. Cell 2007;131:669–681. dependent kinase of MKK and IKK. Nature signals that activate ERK and regulates IgE 94. He JQ, et al. Rescue of TRAF3-null mice by 2001;412:346–351. induction by CD40. EMBO J 2003;22: p100 NF-kappa B deficiency. J Exp Med 108. Yujiri T, et al. MEK kinase 1 gene disruption 3855–3864. 2006;203:2413–2418. alters cell migration and c-Jun NH2-terminal 80. Grewal IS, Flavell RA. CD40 and CD154 in 95. Xie P, Stunz LL, Larison KD, Yang B, Bishop kinase regulation but does not cause a mea- cell-mediated immunity. Annu Rev Immu- GA. Tumor necrosis factor receptor-associ- surable defect in NF-kappa B activation. Proc nol 1998;16:111–135. ated factor 3 is a critical regulator of B cell Natl Acad Sci USA 2000;97:7272–7277.

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 41 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

109. Xia Y, et al. MEK kinase 1 is critically 123. Mordmuller B, Krappmann D, Esen M, factor receptor-associated factor 2. J Biol required for c-Jun N-terminal kinase activa- Wegener E, Scheidereit C. Lymphotoxin and Chem 2000;275:271–278. tion by proinflammatory stimuli and lipopolysaccharide induce NF-kappaB-p52 138. Chen AJ, et al. The dual specificity JKAP spe- growth factor-induced cell migration. Proc generation by a co-translational mechanism. cifically activates the c-Jun N-terminal Natl Acad Sci USA 2000;97:5243–5248. EMBO Rep 2003;4:82–87. kinase pathway. J Biol Chem 2002;277: 110. Lee FS, Peters RT, Dang LC, Maniatis T. 124. Derudder E, Dejardin E, Pritchard LL, Green 36592–36601. MEKK1 activates both IkappaB kinase alpha DR, Korner M, Baud V. RelB ⁄ p50 dimers 139. Zama T, Aoki R, Kamimoto T, Inoue K, Ike- and IkappaB kinase beta. Proc Natl Acad Sci are differentially regulated by tumor necro- da Y, Hagiwara M. A novel dual specificity USA 1998;95:9319–9324. sis factor-alpha and lymphotoxin-beta phosphatase SKRP1 interacts with the MAPK 111. Lee FS, Hagler J, Chen ZJ, Maniatis T. Acti- receptor activation: critical roles for p100. J kinase MKK7 and inactivates the JNK MAPK vation of the IkappaB alpha kinase complex Biol Chem 2003;278:23278–23284. pathway. Implication for the precise regula- by MEKK1, a kinase of the JNK pathway. 125. Kim YS, Nedospasov SA, Liu ZG. TRAF2 tion of the particular MAPK pathway. J Biol Cell 1997;88:213–222. plays a key, non-redundant role in LIGHT- Chem 2002;277:23909–23918. 112. Blank JL, Gerwins P, Elliott EM, Sather S, lymphotoxin beta receptor signaling. Mol 140. Zama T, Aoki R, Kamimoto T, Inoue K, Johnson GL. Molecular cloning of mitogen- Cell Biol 2005;25:2130–2137. Ikeda Y, Hagiwara M. Scaffold role of a activated protein ⁄ ERK kinase kinases 126. Chang YH, Hsieh SL, Chen MC, Lin WW. mitogen-activated protein kinase phospha- (MEKK) 2 and 3. Regulation of sequential Lymphotoxin beta receptor induces inter- tase, SKRP1, for the JNK signaling pathway. phosphorylation pathways involving mito- leukin 8 gene expression via NF-kappaB and J Biol Chem 2002;277:23919–23926. gen-activated protein kinase and c-Jun AP-1 activation. Exp Cell Res 2002; 141. Morrison DK, Davis RJ. Regulation of MAP kinase. J Biol Chem 1996;271:5361–5368. 278:166–174. kinase signaling modules by scaffold pro- 113. Yang J, et al. Mekk3 is essential for early 127. Cuadrado A, Nebreda AR. Mechanisms and teins in mammals. Annu Rev Cell Dev Biol embryonic cardiovascular development. Nat functions of p38 MAPK signalling. Biochem 2003;19:91–118. Genet 2000;24:309–313. J 2010;429:403–417. 142. Uhlik MT, et al. Rac-MEKK3-MKK3 scaf- 114. Dumitru CD, et al. TNF-alpha induction by 128. Chuderland D, Seger R. Protein-protein folding for p38 MAPK activation during LPS is regulated post-transcriptionally via a interactions in the regulation of the extracel- hyperosmotic shock. Nat Cell Biol Tpl2 ⁄ ERK-dependent pathway. Cell lular signal-regulated kinase. Mol Biotech- 2003;5:1104–1110. 2000;103:1071–1083. nol 2005;29:57–74. 143. Kelkar N, Standen CL, Davis RJ. Role of the 115. Belich MP, Salmeron A, Johnston LH, Ley 129. Perkins ND. Integrating cell-signalling path- JIP4 scaffold protein in the regulation of SC. TPL-2 kinase regulates the proteolysis of ways with NF-kappaB and IKK function. Nat mitogen-activated protein kinase signaling the NF-kappaB-inhibitory protein NF-kap- Rev Mol Cell Biol 2007;8:49–62. pathways. Mol Cell Biol 2005;25:2733– paB1 p105. Nature 1999;397:363–368. 130. Tanoue T, Adachi M, Moriguchi T, Nishida 2743. 116. Drayton DL, Liao S, Mounzer RH, Ruddle E. A conserved docking motif in MAP kinas- 144. Takaesu G, et al. Activation of p38alpha ⁄ NH. Lymphoid organ development: from es common to substrates, activators and reg- beta MAPK in myogenesis via binding of the ontogeny to neogenesis. Nat Immunol ulators. Nat Cell Biol 2000;2:110–116. scaffold protein JLP to the cell surface pro- 2006;7:344–353. 131. Enslen H, Davis RJ. Regulation of MAP tein Cdo. J Cell Biol 2006;175:383–388. 117. Fu YX, Chaplin DD. Development and matu- kinases by docking domains. Biol Cell 145. Kang JS, et al. A Cdo-Bnip-2-Cdc42 signal- ration of secondary lymphoid tissues. Annu 2001;93:5–14. ing pathway regulates p38alpha ⁄ beta MAPK Rev Immunol 1999;17:399–433. 132. Bardwell L, Shah K. Analysis of mitogen- activity and myogenic differentiation. J Cell 118. Gommerman JL, Browning JL. Lymphotox- activated protein kinase activation and inter- Biol 2008;182:497–507. in ⁄ light, lymphoid microenvironments and actions with regulators and substrates. 146. Oh JE, et al. Cdo promotes neuronal differ- autoimmune disease. Nat Rev Immunol Methods 2006;40:213–223. entiation via activation of the p38 mitogen- 2003;3:642–655. 133. Good M, Tang G, Singleton J, Remenyi A, activated protein kinase pathway. FASEB J 119. McCarthy DD, Summers-Deluca L, Vu F, Lim WA. The Ste5 scaffold directs mating 2009;23:2088–2099. Chiu S, Gao Y, Gommerman JL. The lym- signaling by catalytically unlocking the Fus3 147. Minden A, et al. Differential activation of photoxin pathway: beyond lymph node MAP kinase for activation. Cell 2009;136: ERK and JNK mitogen-activated protein kin- development. Immunol Res 2006;35:41– 1085–1097. ases by Raf-1 and MEKK. Science 1994; 54. 134. Scheidereit C. IkappaB kinase complexes: 266:1719–1723. 120. Force WR, Glass AA, Benedict CA, Cheung gateways to NF-kappaB activation and 148. Su YC, Han J, Xu S, Cobb M, Skolnik EY. TC, Lama J, Ware CF. Discrete signaling transcription. Oncogene 2006;25: NIK is a new Ste20-related kinase that binds regions in the lymphotoxin-beta receptor 6685–6705. NCK and MEKK1 and activates the for tumor necrosis factor receptor-associ- 135. Chariot A. The NF-kappaB-independent SAPK ⁄ JNK cascade via a conserved regula- ated factor binding, subcellular localization, functions of IKK subunits in immunity and tory domain. EMBO J 1997;16:1279–1290. and activation of cell death and NF-kappaB cancer. Trends Cell Biol 2009;19:404–413. 149. Xia Y, Wu Z, Su B, Murray B, Karin M. pathways. J Biol Chem 2000;275:11121– 136. Marti A, et al. Actin-binding protein-280 JNKK1 organizes a MAP kinase module 11129. binds the stress-activated protein kinase through specific and sequential interactions 121. Li C, et al. Structurally distinct recognition (SAPK) activator SEK-1 and is required for with upstream and downstream compo- motifs in lymphotoxin-beta receptor and tumor necrosis factor-alpha activation of nents mediated by its amino-terminal CD40 for tumor necrosis factor receptor- SAPK in melanoma cells. J Biol Chem extension. Genes Dev 1998;12:3369– associated factor (TRAF)-mediated signal- 1997;272:2620–2628. 3381. ing. J Biol Chem 2003;278:50523–50529. 137. Leonardi A, Ellinger-Ziegelbauer H, Franz- 150. Xu S, Cobb MH. MEKK1 binds directly to 122. Basak S, et al. A fourth IkappaB protein oso G, Brown K, Siebenlist U. Physical and the c-Jun N-terminal kinases ⁄ stress-acti- within the NF-kappaB signaling module. functional interaction of filamin (actin- vated protein kinases. J Biol Chem 1997; Cell 2007;128:369–381. binding protein-280) and tumor necrosis 272:32056–32060.

42 2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 Schro¨felbauer & Hoffmann Æ Signaling specificity of TNFRSFs

151. Yan M, et al. Activation of stress-activated node organogenesis in TRAF6-deficient human IFN-beta gene. Cell 1992;71:777– protein kinase by MEKK1 phosphorylation mice. Genes Cells 1999;4:353–362. 789. of its activator SEK1. Nature 1994;372: 159. He JQ, Oganesyan G, Saha SK, Zarnegar B, 167. Ping D, Boekhoudt GH, Rogers EM, Boss 798–800. Cheng G. TRAF3 and its biological function. JM. Nuclear factor-kappa B p65 mediates 152. Xu S, Robbins DJ, Christerson LB, English Adv Exp Med Biol 2007;597:48–59. the assembly and activation of the TNF- JM, Vanderbilt CA, Cobb MH. Cloning of rat 160. Xie P, Hostager BS, Bishop GA. Requirement responsive element of the murine monocyte MEK kinase 1 cDNA reveals an endogenous for TRAF3 in signaling by LMP1 but not chemoattractant-1 gene. J Immunol membrane-associated 195-kDa protein with CD40 in B lymphocytes. J Exp Med 1999;162:727–734. a large regulatory domain. Proc Natl Acad 2004;199:661–671. 168. Kontoyiannis D, Pasparakis M, Pizarro TT, Sci USA 1996;93:5291–5295. 161. Xu M, Skaug B, Zeng W, Chen ZJ. A ubiqu- Cominelli F, Kollias G. Impaired on ⁄ off reg- 153. Yujiri T, Sather S, Fanger GR, Johnson GL. itin replacement strategy in human cells ulation of TNF biosynthesis in mice lacking Role of MEKK1 in cell survival and activa- reveals distinct mechanisms of IKK activa- TNF AU-rich elements: implications for tion of JNK and ERK pathways defined by tion by TNFalpha and IL-1beta. Mol Cell joint and gut-associated immunopatholo- targeted gene disruption. Science 1998; 2009;36:302–314. gies. Immunity 1999;10:387–398. 282:1911–1914. 162. Carey M, Lin YS, Green MR, Ptashne M. A 169. Carballo E, Lai WS, Blackshear PJ. Feedback 154. Rothwarf DM, Zandi E, Natoli G, Karin M. mechanism for synergistic activation of a inhibition of macrophage tumor necrosis IKK-gamma is an essential regulatory sub- mammalian gene by GAL4 derivatives. Nat- factor-alpha production by tristetraprolin. unit of the IkappaB kinase complex. Nature ure 1990;345:361–364. Science 1998;281:1001–1005. 1998;395:297–300. 163. Thanos D, Maniatis T. Virus induction of 170. Ventura JJ, Hubner A, Zhang C, Flavell RA, 155. Yamaoka S, et al. Complementation cloning human IFN beta gene expression requires Shokat KM, Davis RJ. Chemical genetic anal- of NEMO, a component of the IkappaB the assembly of an enhanceosome. Cell ysis of the time course of signal transduc- kinase complex essential for NF-kappaB acti- 1995;83:1091–1100. tion by JNK. Mol Cell 2006;21:701–710. vation. Cell 1998;93:1231–1240. 164. Lomvardas S, Thanos D. Modifying gene 171. Werner SL, Barken D, Hoffmann A. Stimulus 156. Mercurio F, et al. IkappaB kinase (IKK)- expression programs by altering core pro- specificity of gene expression programs associated protein 1, a common component moter chromatin architecture. Cell determined by temporal control of IKK of the heterogeneous IKK complex. Mol Cell 2002;110:261–271. activity. Science 2005;309:1857–1861. Biol 1999;19:1526–1538. 165. Kim TK, Maniatis T. The mechanism of 172. Murphy LO, Blenis J. MAPK signal specific- 157. Oganesyan G, et al. Critical role of TRAF3 in transcriptional synergy of an in vitro assem- ity: the right place at the right time. Trends the Toll-like receptor-dependent and -inde- bled interferon-beta enhanceosome. Mol Biochem Sci 2006;31:268–275. pendent antiviral response. Nature 2006; Cell 1997;1:119–129. 173. Marshall CJ. Specificity of receptor tyrosine 439:208–211. 166. Thanos D, Maniatis T. The high mobility kinase signaling: transient versus sustained 158. Naito A, et al. Severe osteopetrosis, defec- group protein HMG I(Y) is required for NF- extracellular signal-regulated kinase activa- tive interleukin-1 signalling and lymph kappa B-dependent virus induction of the tion. Cell 1995;80:179–185.

2011 John Wiley & Sons A/S • Immunological Reviews 244/2011 43