Oncogene (1998) 17, 3261 ± 3270 ã 1998 Stockton Press All rights reserved 0950 ± 9232/98 $12.00 http://www.stockton-press.co.uk/onc Modulation of life and death by the TNF receptor superfamily

Stacey J Baker*,1 and E Premkumar Reddy1

1Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, Pennsylvania 19140, USA

The receptor (TNFR) superfamily Cell death is thought to occur by two mechanisms, represents a growing family, with over 20 members necrosis and . While necrosis is believed to having been identi®ed thus far in mammalian cells. These occur in response to physical injury, apoptosis, or share signi®cant homologies in their extracel- programmed cell death, is a genetically encoded and lular ligand binding domains and intracellular e€ector tightly regulated `suicidal' process in which the cell (death) domains. These receptors appear to transmit actively participates in its own destruction. Apoptosis their signals via -protein interactions, which is morphologically characterized by membrane bleb- convey either a death or survival signal. Isolation and bing, cytoplasmic, nuclear and chromatin condensa- characterization of containing proteins tion, and DNA fragmentation. The cell ultimately (TRADD, FADD/MORT-1, RIP), TRAF domain collapses into distinct apoptotic bodies of condensed containing proteins (TRAF1-6) as well as new members chromatin with intact membranes that display speci®c and adaptor proteins such as DAXX have provided new surface markers which direct neighboring phagocytic insights to our understanding of signaling mechanisms cells to engulf the vesicles before they rupture and associated with this family of receptors. While the death subsequently trigger an undesired in¯ammatory im- signals seem to be associated with the activation of both mune response (reviewed in Martin et al., 1994; White, the and JUN kinase pathways, the survival 1995). Some of these stimuli, such as tumor necrosis signals are mediated via the activation of the NF-kB factor (TNF) and some of its relatives, can provide pathway. suicidal signals via proteins containing a `death domain', while others, such as transforming growth Keywords: TNFR; death domain; apoptosis; TRAF; factor beta (TGF-b) are potent and rapid inhibitors of TRADD; FADD; DAXX; ASK; JNK; NF-kB proliferation and inducers of growth arrest and apoptosis (reviewed in Ho€man and Liebermann, 1994; Arch et al., 1998; Ashkenazi and Dixit, 1998). Regulation of apoptosis is genetically controlled by Introduction numerous genes, some of which have remained highly conserved throughout evolution. The continued replenishment of the cellular compo- nents which comprise the hematopoietic system is dependent upon a culmination of signaling events Genetic regulators of apoptosis which instruct uncommitted pluripotent stem cells to proliferate and subsequently commit themselves to Genetic regulation of apoptosis has been best studied di€erentiate along the myeloid, lymphoid or erythroid in the nematode Caenothabditis elegans (C. elegans), lineages. Environmental factors such as the bone where it has been possible to isolate mutants that are marrow microenvironment and cellular responsiveness defective in various aspects of programmed cell death. to interleukins and cytokines were once thought to Among the 1090 cells generated during the develop- govern the establishment and maintenance of each ment of C. elegans, there are 131 cells that undergo hematopoietic lineage. Presumably, by `permitting' programmed cell death and the timing and identities of these soluble factors to dictate whether a cell will these cells are invariant in individual worms. Since the continue to remain in a resting state, proliferate, or body of C. elegans is transparent during embryonic terminally di€erentiate, the individual constituents of development, it has been possible to observe the cells the hematopoietic system are delicately and critically undergoing apoptosis using microscopic techniques balanced in both cell type and number (Wyllie et al., such as Nomarsi optics. Genetic analysis of mutants 1980; Williams et al., 1992; Williams and Smith, 1993). of C. elegans that are defective in programmed cell Disruptions in this physiological balance are clinically death has identi®ed a set of genes which act as positive manifested in several disease states, including cancer, in and negative regulators of this biological process. which the developmental and proliferative constraints Recent studies have shown that several of the genes that are normally placed upon a developing cell are that regulate the apoptotic pathways of C. elegans are ignored. To avoid the onset of such defects in cellular highly conserved throughout evolution and counter- development, cells utilize their capability to respond to parts for these genes can be readily detected in humans additional stimuli which trigger the activation of (Steller, 1995; Yuan, 1996). `suicidal' signal transduction pathways. One of the important genes identi®ed in C. elegans by this type of genetic analysis is Ced-9, in which a dominant gain of function mutation suppresses all normal programmed cell death, while recessive loss-of- function mutation causes apoptosis in many normally *Correspondence: SJ Baker living cells leading to the death of the organism. Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3262 Studies with this gene in C. elegans suggest that the with which the ligands bind to their cognate receptors. product of ced-9 acts as a gatekeeper of apoptosis, In contrast to other protein families with notable where downregulation of this gene is necessary for cells homology, each of these family members displays no to undergo programmed cell death (Yuan, 1996). more than 20 ± 25% homology (which is found mostly Cloning and molecular characterization of ced-9 in the extracellular region) at the protein level. showed that it codes for a protein of 280 amino Although these proteins, for the most part, exist as acids, which is homologous to the mammalian protein trimeric or multimeric membrane bound proteins which bcl-2 (Tsujimoto et al., 1985; reviewed in Yuan, 1996; function to induce receptor aggregation, there are a few Adams and Cory, 1998). members, such as TNF and FasL, which are functional Two other genes which appear to be essential for the in a soluble form. Regardless of conformation, these programmed cell death of C. elegans are ced-3 and ced- ligands, in a mechanism analogous to other cytokines 4 (reviewed in Yuan, 1996; Adams and Cory, 1998). and growth factors, exert their e€ects through receptor- Loss of function mutations in these two genes abolish ligand interactions which induce downstream signal the programmed cell death of all 131 cells during C. transduction events (Gruss and Dower, 1995). elegans hermaphrodite development. The ced-4 gene There are slightly more members of the TNF codes for a 549 amino acid protein which contains two receptor (TNFR) superfamily as there are ligands regions that contains two calcium binding domain with which they interact: TNF-RI (p55), TNF-RII known as the EF-hand. The ced-3 gene, on the other (p75), TNF-RIII (TNF-RP) , OX-40, 4-1BB, CD40, hand, codes for a protein of 503 amino acids which CD30, CD27, the poxvirus gene products PV-T2 and and was originally thought to be homologous to PV-A53R, and p75 NGFR, CAR1, DR3 (also known caspase-1 (Yuan et al., 1993), a cysteine as APO3, WSL-1, TRAMP, LARD), DR4, DR5 (also which processes interleukin-1b (IL-1b) into a mature known as APO2, TRAIL-R2, TRICK 2 or KILLER) and functional form that is required during an (Smith et al., 1993; Armitage, 1994; Gruss and Dower, in¯ammatory response (Salvesen and Dixit, 1997; 1995; Ashkenazi and Dixit, 1998). One of the most Thornberry and Lazebnik, 1998). notable additions to this list is the p75 low anity Apoptotic machinery is normally suppressed or nerve growth factor receptor (NGFR). While this activated by signals from the extracellular environ- protein was actually the ®rst family member to be ment as well as intracellular sensors that monitor DNA identi®ed, its low-anity ligand is structurally un- damage. The absence of external survival signals or related to the TNF ligands and is therefore not irreparable DNA damage are some of the important included as a member of the ligand superfamily. events that appear to trigger apoptosis in lower Almost as notable are the PV-T2 and PV-A53R pox organisms such as C. elegans. While preserving these virus gene products which encode soluble homologs of apoptotic responses, higher organisms such as mam- the TNF receptors. The mammalian TNFR family mals have evolved a distinctive mechanism that enables members are type I membrane proteins and, in spite of the organism to instruct certain cell populations to their low degree of homology (20 ± 25%), are grouped enter apoptotic pathways at di€erent stages of together as such due to the presence of conserved development. This is well exempli®ed in the case of cysteine residues in the extracellular ligand-binding immune system, where it is necessary to eliminate the domain. In general, each receptor contains varying presence of T and B cell populations following their numbers of cysteine rich domains (CRDs), each of expansion in response to external infection (Boise and which is characterized by the presence of approxi- Thompson, 1996; Winoto, 1997). To achieve this, mately six cysteine residues that are interspersed within higher organisms appear to have acquired `death a stretch of 40 amino acids. The presence of the CRDs, receptors' that can transmit apoptic signals in based on available crystallographic data for TNF-RI, response to `death ligands'. These receptors invariably has allowed this protein superfamily to add a di€erent seem to utilize the caspase machinery to achieve their perspective from which these (as opposed to other) goal of triggering cell death. growth factor receptors are studied: a functional TNF superfamily receptor is typically a trimeric or multi- meric complex which is stabilized via intracysteine TNF and TNFR superfamilies disul®de bonds that are formed between the CRDs of individual subunit members (Banner et al., 1993). In 1984, two forms of TNF, TNF-a and LT-a Despite an emphasis on the CRDs and the formation (lymphotoxin, TNF-b), were isolated from activated of membrane bound aggregated complexes, most macrophages and T cells, respectively. Since their receptors also exist in a soluble form. Solubility is, identi®cation, these proteins have become representa- for the most part, achieved by proteolytic cleavage and tives of a unique superfamily of ligands which currently soluble forms of TNF-RI, TNF-RII, CD27, CD30, includes the following family members: TNF, LT-a, CD40, and Fas are generated in this fashion; while 4- (CD95L), OX40L, CD40L, CD27L, 1BB also exists in a soluble form, it is generated via CD30L, 4-1BBL, and LTb, Apo3L/TWEAK, alternative splicing (Gruss and Dower, 1995). The need APO2L/TRAIL and RANKL (Smith et al., 1993; for soluble forms of these receptors, as well as ligands, Armitage, 1994; Gruss and Dower, 1995; Anderson et is currently unknown. al., 1997; Ashkenazi and Dixit, 1998). Several of the Irrespective of ligand and receptor conformation, ligands are type II membrane proteins which are both the TNF related ligands and receptors are characterized by the con®nement of the C-terminus of expressed on (but are not necessarily limited to) the protein within the extracellular space. A 150 amino activated T cells and macrophages and are, in one acid region within the C-terminus is actually the form or another, required during T cell mediated hallmark of the ligand family, as this is the region immune responses. This type of coordinated expression Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3263 and function is thought to ensure that such responses, despite the characterization of a de®ned `death which are largely dependent upon antigen stimulation inducing' region within each of these receptors, the and subsequent cell-cell interactions, are initiated at the intermediates involved in the transmission their signals proper times, in the appropriate places, and involve the were, until recently, completely unknown. As with correct cell types (each of which will express either the other receptors which are devoid of catalytic activity, ligand or the receptor). Frequently the outcome of such TNF-RI and Fas were suspected to utilize cellular cellular responses can be quite pleiotropic, generating a proteins as `downstream messengers of death'. Many broad range of cellular responses in the forms of T-cell charged residues that are well conserved in both activation and stimulation, cellular proliferation and proteins were suspected to be widely dispersed di€erentiation, or cell death in the form of cytotoxicity throughout portions of the death domains which are which proceeds by apoptosis (Armitage, 1994; Gruss oriented to interact with protein components of the and Dower, 1995). cytoplasm (Tartaglia et al., 1993). To date, several death domain-containing proteins which associate with either TNF-RI or Fas have been identi®ed and Death signal transduction by TNF-RI and Fas: the death characterized with respect to their ability to induce domain and induction of apoptosis apoptosis and other downstream signaling events which are activated in immune responses achieved The term `death domain' was originally coined in 1993 through ligand binding to this receptor. by Tartaglia et al. (1993) as a result of deletion mutagenesis studies involving TNF-RI (p55) mediated apoptotic cell death. These studies revealed that an 80 TRADD: a TNF-RI-associated protein amino acid domain, which is localized to the C- terminal portion of the protein's intracellular region, is Although TNF can bind to both TNF-RI and TNF- responsible for the generation of cytotoxic death RII, the former appears to be responsible for the signals, anti-viral responses (Tartaglia et al., 1993), generation of most of the knowledge surrounding and the activation of acid sphingomyelinase (Wieg- TNF-mediated cellular responses. TRADD, a novel mann et al., 1994); it is also partially responsible for, in gene product having little or no homology with other conjunction with residues in the N-terminal portion on known proteins in the database, was one of the ®rst of the intracellular region, the induction of nitric oxide several TNF-RI-associated proteins identi®ed (Hsu et (NO) synthase activity (Tartaglia et al., 1993). al., 1995). Although levels of TRADD expression in Homology searches have revealed that the TNF-RI most tissues examined are relatively low, the 1.5 kb death domain is approximately 65% similar (28% TRADD message, as well as that of TNF-RI, are identical) to a 65 amino acid region within the ubiquitously expressed. Deletion mutagenesis of this intracellular domain of the Fas antigen; mutagenesis oligomeric 34.2 kDa protein has suggested that studies have con®rmed that these 65 amino acids are interaction with TNF-RI via residues of the TNF-RI required for the induction of cell death following death domain is mediated by a C-terminal 111 amino treatment with an anti-Fas antibody in conjunction acid TRADD death domain. The TRADD death with actinomycin D (Itoh and Nagata, 1993). domain also appears to modulate TRADD self- Supporting evidence for a functional overlap between association, NF-kB activation and induction of the domains of these two receptors was achieved apoptosis. This region (residues 195 ± 305) is 23% through the generation of a `death signal delivering' identical to that of TNFR-I, but shares only 13% chimeric receptor which replaced TNF-RI amino acid identity with that of Fas. Despite the fact that a residues 324 ± 426 with the corresponding amino acids chimeric TNF-RI protein which contains a Fas death of the Fas antigen (Tartaglia et al., 1993). domain can initiate an apoptotic response, the death The death domain, aside from being the only domain of Fas is unable to associate with TRADD. homologous intracellular domain that is shared by These results, in conjunction with others which have members of the TNFR superfamily, generates a suggested a divergence in TNF and Fas mediated cytotoxic signal irrespective of its position with respect apoptotic pathways, further demonstrate the unique- to the extracellular domain (Tartaglia et al., 1993). In ness and individuality of each receptor despite the addition, this domain appears to mediate self-associa- presence of a death domain. Overexpression of tion of both TNF-RI and Fas, thereby mimicking the TRADD induces a similar CrmA-inhibitable apopto- aggregation events which are induced by ligand binding tic phenotype as TNF-RI overexpression, including an to each of these receptors (Boldin et al., 1995a). These alteration in cell morphology and DNA fragmentation results, which demonstrate that the death domain is an (Hsu et al., 1995). independent domain at both the structural and functional levels, were recently con®rmed by the identi®cation and subsequent characterization of three FADD /MORT-1 and RIP death domain-containing proteins, each of which can generate an apoptotic signal when overexpressed in cells. FADD (designated MORT-1 by Boldin et al. (1995b), who simultaneously isolated the identical cDNA) is another addition to the rapidly growing list of death Transducers of death: novel mediators of TNF-RI signal domain-containing proteins; the protein is appropri- transduction ately named given that it shares many characteristics with TRADD. First, the 1.6 kb FADD/MORT-1 The rapid induction of cell death via the death domain message is ubiquitously expressed (as is that of the is thus far unique to TNFR superfamily; however, Fas antigen). Like TRADD, FADD, with a molecular Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3264 weight of 23.3 kDa, appears to be highly selective in its those of FADD (Thome et al., 1997; Shu et al., 1997; interactions in that it strongly associates with Fas via Hu et al., 1997). death domain-death domain interactions. In addition, FADD is also able to associate with TNF-RI (Chinnaiyan et al., 1995), and therefore explains some Daxx and JNK pathway of the similarities between TNF-RI and Fas-mediated apoptosis. In addition to the activation of caspase-8, Fas-ligand Database searches have revealed that the FADD interactions appear to activate apoptosis by a second death domain shares 25 ± 30% identity with those of mechanism which involves activation of the Jun kinase TNF-RI and Fas; more importantly, amino acid cascade, culminating in the phosphorylation and residues 111 ± 170 within the death domain of human activation of factors such as c-Jun. This FADD/MORT-1 are identical to amino acid residues second pathway appears to be mediated by a novel 233 ± 292 of rat Fas (rFas) which localize to the death protein termed Daxx, which binds to the death domain domain of the protein. Since it has been previously of Fas via its C-terminal end (Yang et al., 1997). stated that Fas and TNF-RI can self-associate, and Overexpression of Daxx was found to enhance Fas- that this association is via the death domain, it may be mediated apoptosis and activation of the JNK path- that the death domain may actually be a domain which way. Recent analysis of the mechanisms associated is required for protein-protein interactions (Boldin et with Daxx-mediated apoptosis revealed that this al., 1995b; Chinnaiyan et al., 1995), thus facilitating the protein interacts with the N-terminal 648 amino acids signal transduction pathways that lead to apoptosis. In of Ask1 (a MAP3K). Based on their deletional fact, Chinnaiyan et al. (1995) and others (Feinstein et mutagenesis studies, Chang et al. (1998) propose that al., 1995) have remarked that a death domain has been ASK1 normally exists in an inactive state because of found in various other proteins such as , a the intra-molecular interaction between the N- and C- protein involved in cell adhesion and cell-cell interac- terminal ends of this protein. Recruitment of Daxx to tions. Analogous to TRADD, overexpression of the complex facilitates its interaction with FADD/MORT-1 can induce apoptosis which is Ask1, resulting in the `opening up' of the ASK1 kinase inhibitable by CrmA. However, what appears to be into an active conformation. Activated ASK1 appears particularly unique about FADD/MORT-1 is that the to initiate a phosphorylation cascade that ultimately e€ector domain which actually induces apoptosis results in the phosphorylation of c-Jun N-terminal resides at the N-terminus of the protein (the death kinase, JNK. c-Jun, the JNK substrate, has been domain with which Fas interacts is located at the C- shown to activate apoptotic pathways by a mechanism terminus). that is yet to be determined. FADD couples the TNF-RI-TRADD and Fas- FADD complexes to activation of Caspase-8, by recruiting pro-caspase-8 to the receptor complex. Survival signals transduced by TNF-RI FADD appears to accomplish this important task via a `death e€ector domain' that binds to an analogous One of the earlier intriguing observations concerns the domain in pro-caspase-8 (also called a FLICE or activation of NF-kB by TRADD, a phenomenon MACH) (Boldin et al., 1996; Muzio et al., 1996). This which is also associated with TNF-R1 engagement interaction domain, also known as CARD (caspase and is involved in TNF-mediated responses (reviewed recruitment domain), is found in a large number of in Heller and Kronke, 1994; Ashkenazi and Dixit, including Caspase-2, 8, 9 and 10 (Ho€mann et 1998). Unlike the TRADD-induced apoptotic response, al., 1997). The recruitment of pro-caspase-8 appears to NF-kB activation is not inhibited by CrmA (Hsu et al., result in its oligomerization, activation of its auto- 1995). It has been known for some time that TNF-R1 proteolytic activity and ultimately the generation of rarely triggers apoptosis unless protein synthesis is enzymatically active caspase-8 (Yeh et al., 1998; Zhang blocked (Itoh and Nagata, 1993), suggesting the et al., 1998). Activated caspase-8 is known to cleave existence of signaling pathways that can suppress other pro-caspases, such as caspase-9 (the functional apoptotic stimuli generated by this receptor-ligand homolog of CED3), thereby initiating apoptosis. interactions. Recent studies have determined that Besides FADD, TNF-RI can engage an adapter called indeed activation of NF-kB is a survival signal that is RAIDD or CARDD. RAIDD associates with the transmitted by some of the death receptors. death domain of RIP, and through a CARD motif, TNF-RI-mediated induction of cell survival appears associates with caspase-2, thereby inducing apoptosis. to involve a second receptor-associated protein, Studies with FADD null mutant mice demonstrate designated RIP (receptor interaction protein) (Stanger that FADD is essential for normal development as et al., 1995). Although its pattern of expression is also FADD7/7 mice are embryonic lethal (Yeh et al., constitutive and increases in conA activated T cells, the 1998; Zhang et al., 1998). While the T cells derived from structure of the 74 kDa RIP protein is somewhat these mice are resistant to Fas-mediated apoptosis, they unique from the others which have been discussed here. seem to exhibit defects in antigen induced proliferation Despite the presence of a death domain which is most suggesting that FADD is associated with other critical closely related to that of TNF-RI (30% identical; 59% functions besides apoptosis. similar) at the C-terminus of the protein, the N- Recently, a family of viral proteins called v-FLIPs terminus of the protein displays the characteristics of a and their cellular homologs, termed c-FLIPs have been kinase domain, which shares key residues in common described, whose function is at present unclear. Since with the serine-threonine family kinases, suggesting these proteins contain a death e€ector domain, it is that the encoded protein might function as a serine/ possible that they perform functions analogous to threonine kinase. Accumulating evidence suggests that Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3265 ligand-induced trimerization of the TNF-RI results in activation (Boldin et al., 1995b; Chinnaiyan et al., the recruitment of the death domain adaptor protein 1995; Stanger, et al., 1995). TRADD, which in turn recruits RIP. RIP interacts with TRADD via its death domain, and this interaction appears to be TNF-dependent (Hsu et al., TRAF proteins: the ®rst isolated TNFR superfamily 1996; Kelliher et al, 1998). Over-expression of RIP associated proteins induces NF-kB (Hsu et al, 1996; Ting et al., 1996) as well as JNK activation. In addition, dominant negative Despite the recent attention received by the receptor forms of RIP have been shown to speci®cally inhibit associated proteins of TNF-RI and Fas, the receptor TNFa-induced NF-kB and JNK activation. While the associated proteins of the less commonly studied TNF- role of JNK activation in this process is unclear, RII (p75) protein were actually the ®rst TNFR activation of NF-kB has recently been shown to be superfamily associated proteins to be identi®ed. It associated with cell survival (Beg and Baltimore, 1996; was known early on that activation of TNF-RII results Liu et al., 1996; Van Antwerp et al., 1996; Wang et al., in the activation of NF-kB. Deletional mutagenesis has 1996); however, the mechanisms, by which RIP determined that a C-terminal 78 amino acid region is activates this transcription factor is not entirely clear. responsible for the generation of signals which are NF-kB proteins are a family of cytoplasmic hetero- sucient to induce cellular proliferation and NF-kB dimeric transcription factors that are inactive due to activation. Since the entire intracellular region of TNF- their association with an inhibitory protein, termed IkB RII, like those of TNF-RI and Fas, does not possess (reviewed in Gilmore et al., 1996). Upon phosphoryla- any domains with intrinsic catalytic activity, receptor tion by IKKa and IKKb,IkB becomes ubiquitinated associated proteins were thought to function as the and degraded by proteosome complexes (Zandi et al., actual signal transducers during TNF-RII generated 1997). Once free, unbound NF-kB dimerizes and signals. Experiments aimed at the identi®cation and becomes an active transcription factor that translo- characterization of these proteins resulted in the cates to the nucleus and activates transcription of NF- isolation of two novel proteins, termed TRAF-1 kB-responsive genes. IKKs are phosphorylated by NIK (TNF receptor-associated factor-1) and TRAF-2 (NF-kB-inducing kinase), a MAP kinase that is part of (Rothe et al., 1994). Since then, six distinct TRAF the TNF-RI receptor complex (reviewed in Gilmore et proteins (termed TRAF-1 through 6) have been al., 1996; Malinin et al., 1997). According to this identi®ed in mammalian cells (reviewed in Arch et model, TNF-RI activation may stimulate RIP kinase al., 1998). activity, which then phosphorylates NIK, which in All TRAF proteins share several structural motifs. turn, activates IKKa and IKKb. This could lead to Ik- TRAFs range in size from 409 amino acids (TRAF-1) B phsophorylation and degradation, resulting in the to 567 amino acids (TRAF-3) and contain distinct activation of NF-kB. This model is supported by the domains (reviewed in Arch et al., 1998 and references analysis of RIP7/7 cells, which are defective in TNF- therein). The C-termini of the proteins, which has been induced NF-kB activation. Interestingly, these cells designated by Rothe et al. (1994) as the TRAF retain TNF-mediated JNK activation, suggesting that domain, is further divided into two sub-domains. The RIP may not be directly involved in the activation of C-terminal portion of the TRAF domain is highly JNK (Kelliher et al., 1998). This observation poses a conserved between TRAF proteins and appears to be problem in explaining earlier results, where Hsu et al. unique to TRAF domain containing proteins. The N- (1996) have shown that dominant negative mutants of terminal portion of the TRAF domain is somewhat less RIP block activation of both NF-kB and JNK. This conserved, and appears to conform to the coiled-coil a model also does not explain how a kinase-defective helix motif (Rothe et al., 1994). While it is possible that RIP mutant restores the NF-kB inducibility in a RIP- the coiled-coil may account for the oligomeric qualities de®cient JURKAT clone (Ting et al., 1996). Additional of TRAFs, additional experiments have implicated the studies with RIP7/7 cells are required to resolve more conserved C-terminal TRAF domain sequences these discrepancies. in protein oligomerization in addition to their function Clinical relevance of FADD/MORT-1 and RIP in mediating TRAF protein-receptor interactions interactions with the Fas death domain are evident (Cheng et al., 1995; Hu et al., 1995; Mosialos et al., when viewed in the context of Fas alleles which contain 1995; Rothe et al., 1995; Sato et al., 1995). the lymphoproliferation (lpr) and lprcg mutations, both All of TRAFs, with the exception of TRAF-1, share of which abolish the ability of Fas to transduce a few characteristics with some more commonly known apoptotic signals. Mice which inherit these alleles transcription factors. The N-terminus of these proteins display phenotypic characteristics which resemble the contains a RING ®nger motif which is believed to autoimmune disease state of systemic lupus erythema- mediate both DNA-protein as well as protein-protein tosus (SLE), autoantibody production, and increased interactions via the formation of two zinc ®nger levels of immunoglobulins (Nagata and Goldstein, structures. TRAF-2,3,4,5 and 6 also contain a zinc- 1995). Neither FADD/MORT-1 nor RIP is able to ®nger domain consisting of ®ve zinc ®nger structures. interact with the lpr and lprcg alleles respectively, The zinc ®nger domain of TRAFs is most closely further con®rming that these mice (and perhaps those related to that of TFIIIA and the presence of one or patients su€ering from disease states which are both of these domains may implicate TRAFs in signal mimicked by the lpr mice (Fisher et al., 1995)) are transduction events occurring in the cytoplasm as well unable to mediate the downstream signal transduction as the nucleus. Interestingly, TRAF-3 and TRAF-5 events which are required for the proper elimination of contain an isoleucine zipper domain between the last certain lymphocytes which, under normal circum- zinc ®nger and the TRAF-N domain. With the stances, are removed in a timely fashion following exception of TRAF-4, all of the TRAFs have been Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3266 found to be localized in the soluble fraction of the designated as TRAF-2A, which di€ers from TRAF-2 cytosol. TRAF-4, on the other hand appears to be a in that it contains a seven amino acid insertion in its nuclear protein, and interestingly has not been shown ring ®nger domain. Interestingly, over-expression of to associate with any of the TNF family of receptors. TRAF-2A failed to activate the NF-kB pathway, while Both a heteromeric complex composed of TRAF-1 activating the JNK pathway (Dadgostar and Cheng, and TRAF-2 as well as TRAF-2 homodimers can 1998). These results implicate the existence of pathway- associate with the C-terminal signal transducing restricted TRAFs and demonstrate that these two component of TNF-RII; although TRAF-1 is also signal transduction pathways could be di€erentially capable of forming homodimers, association with regulated by minor alterations in the structure of TNF-RII appears to be mediated by TRAF-2. TRAFs. Additional use of the yeast two hybrid system has JNK, which activates the transcription factor c-Jun determined that although TRAF-2 can bind strongly to by phosphorylation at its amino terminal end, is itself TNF-RII and TRAF-1, interaction between TRAF-1 activated by phosphorylation which is mediated by a and the receptor is relatively weak. These, as well as MAP kinase (MAPKK/SEK/MEK). MEK, in turn is other deletion experiments suggest that the interaction activated by phosphorylation by a MAPKK, termed between TRAF-1 and TNF-RII, is likely to be MEKK1 (reviewed in Dhanasekaran and Reddy, dependent upon its heterodimerization with TRAF-2. 1998). MEKK1 is activated through phosphorylation More importantly, the TRAF domain, like the death by GCKR protein (Germinal Center Kinase Related domain, appears to constitute an yet another unique Protein) (Shi and Kehrl, 1995). Dominant negative protein dimerization motif which functions to mediate mutants of MEKK1 and GCKR block the TRAF- the protein aggregation events that are required to mediated activation of JNK suggesting that TRAF-2 initiate downstream signal transduction pathways might mediate activation of this pathway at a very (Rothe et al., 1994, 1995; Cheng et al., 1995; Hu et proximal step. None of these kinases were shown to al., 1995; Mosialos et al., 1995; Sato et al., 1995). associate with TRAF-2. However, recently Nishitoh et Interestingly, Northern blot analysis reveals the al. (1998) showed that ASK1 interacts with members of expression of the 2.2 kb TRAF-2 transcript in most the TRAF family and is activated by over-expression tissues, with the highest levels being observed in the of TRAF-2, 5 and 6. A dominant negative mutant of spleen. The 2.1 kb TRAF-1 transcript, on the other TRAF-2, which inhibits JNK activation by TNF, also hand, is only detected in the spleen, testis, and lung. In was found to block TNF-induced ASK1 activation. In those tissues where multiple TRAF proteins are addition, a kinase negative mutant of ASK1 was found expressed, they may heterodimerize and form a to act as dominant negative inhibitor of TNF and functional signal transducing component; however, in TRAF-2-induced activation of JNK. Since ASK1 those tissues where only TRAF-2 is expressed alone, it rapidly associates with TRAF-2 in a TNF-dependent may form a homodimer which can function as a sole manner, this kinase acts as an important intermediary signal transducing component of TNF-RII (Rothe et kinase associated with the activation of both the JNK al., 1994). and NF-kB activation pathways. As can be expected, TRAF-2-de®cient mice fail to activate JNK in response to TNF (Yeh et al., 1997). Role of TRAFs in TNFR-mediated signal transduction Surprisingly, however, these cells seem to activate the NF-kB pathway in the absence of TRAF-2. These During the past few years, several groups have results suggest that TRAF-2 is not essential for NF-kB examined the role of TRAFs in signal transduction activation. It is possible that other TRAFs compensate by examining the nature of intracellular kinases that for the loss of TRAF-2 in these cells; alternatively are activated as a consequence of over-expression of other pathways, such as those associated with RIP TRAFs. These studies have shown that overexpression might also by compensatory. Dominant negative forms of TRAF-2, 5 and 6 activate the JNK and NF-kB of TRAF-2 were also found to block TNF-mediated pathways (reviewed in Arch et al., 1998). Neither of activation of NF-kB. The mechanisms by which these pathways appear to be activated by over- TRAFs activate NF-kB remains to be solved. expression of TRAFs 1, 3 and 4 suggesting that Recently, TRAF-2 was found to interact with NIK, di€erent TRAFs, in spite of their structural homology, suggesting that TRAFs could directly activate this might perform very di€erent functions. In a recent kinase. study, Dadgostar and Cheng (1998) examined the molecular basis for the di€erent biochemical activities observed between TRAF-3 and TRAF-5. Taking TRAF-3 and CD40 interactions advantage of the high degree of structural similarity between TRAF-5, which activates both NF-kBand Characterization of TRAF-3 (CRAF-1, CAP-1, JNK, and TRAF-3, which activates neither, they CD40 bp, LAP-1) has provided additional informa- constructed a number of chimeric molecules by tion regarding the mechanisms by which TRAFs domain swapping between the two cDNAs. Their dimerize and transduce signals (Cheng et al., 1995; result demonstrate that the structural basis for the Hu et al., 1995; Mosialos et al., 1995; Sato et al., 1995). di€erences in signaling by TRAF-3 and TRAF-5 can The 62 kDa TRAF-3 protein, like TRAF-2, is be overcome by replacing the ®rst zinc ®nger and ®rst expressed in virtually all tissues examined to date. 10 amino acids of the second zinc ®nger of TRAF-3 Despite the fact that receptor-protein interaction with those of TRAF-5. occurs via a CD40 sub-domain which shares a low In addition to the six TRAFs, the TRAF-2 gene was degree of homology with regions within TNF-RI, Fas, found to code for an alternatively spliced form and the p75 subunit of NGFR, an overall anity for Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3267 the 62 amino acid intracellular region of CD40 transduce speci®c signals for NF-kB activation presumably accounts for a more speci®c involvement (Grammer et al., 1998). It has also been hypothesized in CD40L (Cheng et al., 1995; Hu et al., 1995; that the RING ®nger domain of this protein is also Mosialos et al., 1995; Rothe et al., 1995; Sato et al., able to modulate downstream events, as an N-terminal 1995), and possibly LTb (Mosialos et al., 1995; Force deletion mutant of TRAF-3 has been observed to et al., 1997) and OX-40 ligand- induced signal suppress CD40 dependent induction of CD23 (Cheng transduction pathways (Arch and Thompson, 1998; et al., 1995; Rothe et al, 1995). Dominant negative Kawamata et al., 1998). TRAF-3 mutants have also been shown to block LT-b- The majority of CD40 mediated signal transduction induced apoptosis (Force et al., 1997). knowledge originates from studies involving CD40 At the protein level, TRAF-3 is most closely related which is expressed on the surface of B lymphocytes. to TRAF-2; however, the most notable di€erence is the CD40 is activated upon binding to the CD40L (gp39) presence of an isoleucine zipper which is located protein which is present on the surface of immune between the N-terminal RING and zinc ®nger system constituents such as helper T cells. Although domains and the C-terminal TRAF domain in CD40L mediated engagement of CD40 protects B TRAF-3 (Cheng et al., 1995). The presence of an lymphocytes from Fas mediated apoptosis, it also isoleucine zipper, either alone or in conjunction with induces B cell proliferation and di€erentiation, the other domains which have been previously expression of several cluster of di€erentiation antigens identi®ed in transcription factors, is again indicative such as CD23 and CD80 (BB-1 or B7), generation of of a protein directly linking cytoplasmic generated memory B cells, and isotype switching. The pleiotropic signals to those occurring in the nucleus. CD40:TRAF- nature of this interaction is further complicated by the 3 and TRAF-3: TRAF-3 interactions occur via the C- fact that CD40, which is expressed at all stages of B terminal portion of the TRAF domain (Cheng et al., cell di€erentiation with the exception of plasma cells, 1995; Hu et al., 1995; Mosialos et al., 1995; Rothe et exerts varying e€ects at di€erent stages of the B al., 1995; Sato et al., 1995). Although these results lymphocyte maturation process. Clinical evidence clearly indicate that the residues within the TRAF which supports these ®ndings as well as stresses the domain (which in contrast to the N-terminal TRAF importance of CD40 is found in individuals with domain residues are unique to, and are well conserved mutations in the CD40L gene. Genetic lesions within among members of the family) constitute a protein CD40L, like Fas and FasL, wreak havoc on cells of the dimerization motif, they do not completely account for immune system. Disruption of CD40L:CD40 interac- the speci®city of interaction among family members. tions results in an X-linked immunode®ciency syn- While both TNF-RII and CD40 utilize TRAF-2, each drome (X-linked hyper-IgM syndrome) which is protein appears to preferentially recruit other TRAFs characterized by the absence of lymphoid follicles as in order to generate signals which are partially distinct well as increased levels of IgM and the absence of IgG, and non-overlapping. Exactly what accounts for the IgE, and IgA due to an inability to undergo isotype speci®city of interactions between closely related switching (reviewed in Conley, 1994; Gruss and Dower, members of a protein superfamily remains to be 1995; Cheng et al., 1995). elucidated. Because TRAF-3 is expressed during all stages of B Although the mechanisms of protein dimerization lymphocyte di€erentiation (Cheng et al., 1995), the among these proteins is not clear, the identi®cation of consequences of its interaction with CD40 are diverse. TRAF proteins, in addition to enriching our under- It will be of equal interest to determine the roles of standing of TNFR superfamily signal transduction TRAFs 1, 2 and 6 in CD40 generated immune pathways, has also allowed us to gain insight into the responses, as these proteins have also been shown to mechanisms which are used by the Epstein Barr Virus interact with CD40 (Pullen et al., 1998), and can (EBV) (the causative agent of infectious mononucleo- transduce di€erent signals form other TNFR family sis) to immortalize B lymphocytes. During life cycle of than TRAF-3. Residues which comprise the N-terminal the virus, expression of the latent infection membrane RING ®nger domain are of paramount importance protein (LMP1) and other viral gene products are with respect to NF-kB activation (which is also crucial for B cell transformation. LMP1, which also observed upon overexpression of the TRAF-2 pro- transforms non-lymphoid cells, induces the majority of tein). In a manner analogous to death domain molecular characteristics observed in EBV positive cells mediated receptor aggregation, it is thought that including NF-kB activation, altered surface marker overexpression of TNFR superfamily associated expression, and possibly the abrogation of require- proteins mirrors the phenomenon of ligand-induced ments for certain growth factors (Mosialos et al., receptor aggregation (Rothe et al., 1995). Unlike the 1995). LMP1 is an integral membrane protein which activation e€ects induced by TRAF-2 in both TNF-RII contains six hydrpohobic transmembrane domains that and CD40 generated signals, TRAF-3 appears to are required for protein aggregation at the plasma negatively regulate protein activation and expression membrane. TRAF-1, 2, 3 and 5 are expressed in EBV since overexpression of TRAF-3 suppressed TNF-RII positive B lymphoblasts (Devergne et al., 1998). and CD40-dependent activation of NF-kB dependent TRAF-3 interacts with the ®rst 44 amino acids of the reporter genes in 293 cells. This has been further LMP1 C-terminus through residues within the TRAF supported by studies in which dominant negative domain. (The C-terminus of the protein is responsible mutants of TRAF-3 did not e€ect NF-kB activation for induction of the transformed state). As with CD40, and only weakly e€ected JNK activation (Grammer et altered expression of TRAF-3 mutants do not e€ect al., 1998). Because these mutants have been shown to NF-kB activation by LMP1 (Miller et al., 1997), and in inhibit p38 activation and immunoglobulin production, fact, LMP1-induced NF-kB activation and TRAF- CD40 may utilize other TRAFs to which it binds to mediated signaling are presumed to travel via Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3268

Figure 1 Schematic representation of the proposed interactions between selected members of the TNFR superfamily and their associated proteins

independent pathways (Miller et al., 1997) through I-TRAF and A20. By associating with members of the di€erent LMP1 motifs (Brodeur et al., 1997). Because TRAF family, these proteins have been shown to block it is presumed that LMP1 sequesters TRAF proteins at TRAF-mediated NF-kB activation and/or apoptosis, the plasma membrane, its expression may mimic the depending on the cell type in which they are expressed e€ects induced by members of the TNF superfamily and the signals by which they are induced. The precise such as CD40 (which, in conjunction with IL-4 mechanisms of activation of these proteins is ate treatment of primary B cells, induces the character- present unclear. istics of EBV transformed cells) and LTb (Mosialos et al., 1995). In addition to members of the TNFR superfamily New members of the TNFR superfamily: DR3, DR4, and other death domain-containing proteins, TRAF DR5 and the identi®cation of decoy receptors proteins are also able to associate with other regulatory `modifying' proteins that can subvert TRAF-mediated The newest members of the TNFR superfamily, DR3, responses (reviewed in Arch et al., 1998). Proteins that DR4, DR5 and RANK were only isolated in the past fall into this class include c-IAP1 and 2, TRIP, TANK/ two years (reviewed in Ashkenazi and Dixit (1998) and Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3269 references therein). DR3, which shares the most procaspase-8, whose dimerization activates its proteo- homology with TNF-RI, can induce apoptosis as well lytic activity and other participants in the caspase as NF-kB activation using TNF-RI-analogous path- cascade. ways in response to ligand (Apo3L/TWEAK) binding. Another variation is seen in the activation of the DR4, which shares the most homology with Fas, JNK pathway, which is often associated with stress- induces apoptosis along the caspase-8-mediated path- induced apoptosis. TNFR family receptors appear to way when bound to its ligand, Apo2/TRAIL. While activate this pathway by multiple mechanisms which this pathway is analogous to Fas-mediated apoptosis, it are mediated by di€erent cellular proteins that are is at present unclear as to whether FADD is required recruited to the receptors following their oligomeriza- in DR4-mediated signaling. Apo2L/TRAIL also bind tion. This is exempli®ed by the DAXX-ASK and to DR5, and it is presumed that ligand activation of TRAF-2-ASK connection, both of which appear to this receptor triggers signaling pathways that are activate the JUN kinase pathway. Interestingly closely related to that of DR4. Interestingly, DR4 TRAFs, which exhibit extensive structural homology and DR5-mediated signaling can be modulated by amongst themselves, seem to function primarily as Apo2L/TRAIL interactions with decoy receptors that transducers of survival signals via activation of the compete with DR4 and 5 for ligand binding. There are NF-kB pathway. The TRAF proteins were originally currently two decoy receptors, DcR1 and DcR2. DcRI identi®ed and characterized with respect to their ability (LIT/TRID/TRAIL-R3) is structurally similar to both to bind to TNF-RII (as well as to each other) via a DR4 and 5 is GPI-linked to the outer surface of the novel domain, the TRAF domain. While the resulting plasma membrane. Unlike DR4 and 5, DcR1 lacks an phenotype associated with the overexpression of these intracellular domain and therefore is incapable of proteins is rather pleiotropic, they appear to modulate transducing DR4 or DR5-like signals. DcR2 the expression of genes which often lead to the (TRUNDD/TRAIL-R4) also shares structural related- induction of an immune response and the maturation ness to DR4 and 5, but has a smaller death domain of those cells which contribute to these responses. that lacks the regions that are responsible for the While the exact mechanism by which TRAF proteins induction of apoptosis and NF-kB activation. activate NIK or ASK1 is at present unclear, further study of these proteins at the biochemical and molecular levels should aid in the understanding of Conclusions immune system development and the nature of the signaling pathways that induce immune system Delineation of the signal transduction pathways constituent proliferation, di€erentiation, and removal associated with the signals mediated by the TNFR following activation. It is intriguing that TRAFs often superfamily have demonstrated the nature of pleio- mediate the activation of both the JNK and NF-kB tropic responses generated by these receptors through pathways. It appears that when these receptors activate their interaction with multiple components of cellular the JNK pathway alone, the phenotypic result appears machinery. As depicted in Figure 1, all of these to be cell death. In contrast, the activation of both the receptors appear to activate apoptotic pathways via NF-kB and JNK pathways appear to result in cell the activation of the caspase cascade. These studies, survival, suggesting that the target genes of c-Jun- which focused on the ability of the TNF-RI and Fas associated complexes could be quite di€erent in the death domains to induce apoptosis, resulted in the presence or absence of activated NF-kB. Additional identi®cation of additional death domain-containing studies aimed at understanding the target genes of proteins (TRADD, FADD/MORT-1, RIP) which are these two important transcription factors that mediate capable of inducing apoptosis when overexpressed in survival and death could provide us with important cells. Not only have these discoveries allowed clues regarding cell proliferation, di€erentiation, and investigators to further dissect the signal transduction death following activation of the death receptors. pathways which are mediated by these receptors, they further con®rm preliminary results which suggested that the death domain may actually be a dimerization motif. Presumably protein multimerization via the death domain allows for the aggregation of signal Acknowledgements transducers and the subsequent engagement of down- This work was supported by a grant # CA 65779 from stream targets, the most important of which is NIH.

References

Adams JM and Cory S. (1998). Science, 281, 1322 ± 1326. Banner DW, D'Arcy A, Janes W, Gentz R, Schoenfeld HJ, Anderson DM, Maraskovsky E, Billingsley WL, Dougall Broger C, Loetscher H and Lesslauer W. (1993). Cell, 73, WC, Tometsko ME, Roux ER, Teepe MC, DuBose RF, 431 ± 445. Cosman D and Galibert L. (1997). Nature, 390, 175 ± 179. Beg AA and Baltimore D. (1996). Science, 274, 782 ± 784. Arch RH and Thompson CB. (1998). Mol. Cell. Biol., 18, Boise LH and Thompson CB. (1996). Science, 274, 67 ± 68. 558 ± 565. Boldin MP, Goncharov TM, Golstev YV and Wallach D. Arch RH, Gedrich RW and Thompson CB. (1998). Genes. (1996). Cell, 85, 803 ± 815. Dev., 12, 2821 ± 2830. Boldin MP, Mett IL, Varfolomeev EE, Shemer-Avni Y, Ashkenazi A and Dixit VM. (1998). Science, 281, 1305 ± Camonis JH and Wallach D. (1995a). J. Biol. Chem., 270, 1308. 387 ± 391. Armitage RJ. (1994). Curr. Opin. Immunol., 6, 407 ± 413. Signal transduction by TNFR superfamily SJ Baker and EP Reddy 3270 Boldin MP, Varfolomeev EE, Pancer Z, Mett IL, Camonis Nishitoh H, Saitoh M, Mochida Y, Takeda K, Nakano H, JH and Wallach D. (1995b). J. Biol. Chem., 270, 7795 ± Rothe M, Miyazono K and Ichijo H. (1998). Molecular 7798. Cell, 2, 389 ± 395. Brodeur SR, Cheng G, Baltimore D and Throley-Dawson Pullen SS, Miller HG, Everdeen DS, Dang TTA, Crute JJ, DA. (1997). J. Biol. Chem., 272, 19777 ± 19784. and Kehry MR. (1998). Biochemistry, 37, 11836 ± 11845. Chang HY, Nishitoh H, Yang X, Ichijo H and Baltimore D. Rothe M, Sarma V, Dixit VM and Goeddel DV. (1995). (1998). Science, 281, 1860 ± 1863. Science, 269, 1424 ± 1427. Cheng G, Cleary AM, Ye Z, Hong DI, Lederman S and Rothe M., Wong SC, Henzel WJ and Goeddel DV. (1994). Baltimore D. (1995). Science, 267, 1494 ± 1497. Cell, 78, 681 ± 692. Chinnaiyan AM, O'Rourke K, Tewari M and Dixit VM. Salvesen GS and Dixit VM. (1997). Cell, 91, 443 ± 446. (1995). Cell, 81, 505 ± 512. Sato T, Irie S and Reed JC. (1995). FEBS Letters, 358, 113 ± Conley ME. (1994). Curr. Opin. Genes. Dev., 4, 401 ± 406 118. Dadogstar H and Cheng G. (1998). J. Biol. Chem., 273, Shi C-S and Kehrl JH. (1997). J. Biol. Chem., 272, 32102 ± 24775 ± 24780. 32107. Devergne O, McFraland EC, Mosialos G, Izumi KM, Ware Shu HB, Halpin DR, Goeddel and DV. (1997). Immunity, 6, CF and Kie€ E. (1998). J. Virol., 72, 7900 ± 7908. 751 ± 763. Dhanasekaran N and Reddy EP. (1998). Oncogene, 17, Smith CA, Farrah T and Goodwin RG. (1993). Cell, 76, 1447 ± 1455. 959 ± 962. Feinstein E, Kimchi A, Wallach D, Boldin M and Stanger BZ, Leder P, Lee T, Kim E and Seed B. (1995). Cell, Varfolomeev E. (1995). TIBS, 20, 342 ± 344. 81, 513 ± 523. Force WR, Cheung TC and Ware CF. (1997). J. Biol. Chem., Steller H. (1995). Science, 267, 1445 ± 1449. 272, 30835 ± 30840. Tartaglia LA and Goeddel DV. (1992). Immunology Today, Fisher GH, Rosenberg FJ, Straus SE, Dale JK, Middelton 13, 151 ± 153. SE, Lin AY, Strober W, Lenardo MJ and Puck JM. Tartaglia LA, Ayres TM, Wong GH and Goeddel DV. (1995). Cell, 81, 935 ± 946. (1993). Cell, 74, 845 ± 853. Gilmore TD, Koedood M, Pi€at KA and White DW. (1996). Thome M, Schneider P, Ho€mann K, Fickenscher H, Meinl Oncogene, 13, 1367 ± 1378. E,NeiplF,MattmannC,BurnsK,BodmerJL,Schroter Grammer HC, Swantek JL, McFarland RD, Miura Y, M, Scadi C, Krammer PH, Peter ME and Tschopp J. Geppert T and Lipsky PE. (1998). J. Immunol., 161, (1997). Nature, 386, 517 ± 521. 1183 ± 1193. Thornberry NA and Lazebnik Y. (1998). Science, 281, Gruss H and Dower SK. (1995). Blood, 85, 3378 ± 3404. 1312 ± 1316. Heller RA and Kronke M. (1994). J. Cell Biol., 126, 5±9. Ting AT, Pimentel-Muinos FX and Seed B. (1996). EMBO Ho€man B and Liebermann DA. (1994). Oncogene, 9, 1807 ± J., 15, 6189 ± 6196. 1812. Tsujimoto Y, Gorham J, Cossman J, Ja€e E and Croce CM. Ho€mann K, Bucher P and Tschopp J. (1997). Trends (1985). Science, 229, 1390 ± 1393. Biochem. Sci., 22, 155 ± 156. VanAntwerpDJ,MartinSJ,KafriT,GreenDRandVerma Hsu H, Xiong J and Goeddel D. (1995). Cell, 81, 495 ± 504. IM. (1996). Science, 274, 787 ± 789. Hsu H, Huang J, Shu H-B, Baichwal V and Goeddel DV. Wang C-Y, Mayo MW and Baldwin AS. (1996). Science, (1996). Immunity, 4, 387 ± 396. 274, 784 ± 787. Hu HM, O'Rourke K, Boguski MS and Dixit VM. (1995). J. White E. (1995). Genes. Dev., 10, 1±15. Biol. Chem., 269, 30069 ± 30072. Wiegmann K, Schutze S, Machleidt T, Witte D and Kronke Hu S, Vincenz C, Ni J, Gentz R and Dixit VM. (1997). J. M. (1994). Cell, 78, 1005 ± 1015. Biol. Chem., 272, 17255 ± 17257. Williams GT and Smith CA. (1993). Cell, 74, 777 ± 778. Itoh N and Nagata S. (1993). J. Biol. Chem., 268, 10932 ± Williams GT, Smith CA, McCarthy NJ, and Grimes EA. 10937. (1992). Trends Cell Biol., 2, 263 ± 265. Kawamata S, Hori T, Imura A, Takaori-Kondo A and Winoto A. (1997). Curr. Opin. Immunol., 9, 365 ± 370. Uchiyama T. (1998). J. Biol. Chem., 273, 5808 ± 5814. Wyllie AH, Kerr JFR and Currie AR. (1980). Int. Rev. Kelliher MA, Grimm S, Ishida Y, Kuo F, Stanger BZ and Cytol., 68, 251 ± 255. Leder P. (1998). Cell, 8, 297 ± 303. Yang X, Khosrsravi-Far R, Chang HY and Baltimore D. Liu Z-G, Hsu H, Goeddel DV and Karin M. (1996). Cell, 87, (1997). Cell, 89, 1067 ± 1076. 565 ± 576. Yeh WC, Pompa JL, McCurrach ME, Shu HB, Elia AJ, Malinin NL, Boldin MP, Kovalenko AV and Wallach D. ShahinianA,NgM.,WakehamA,KhooW,MitchellK, (1997). Nature, 385, 540 ± 544. El-Deiry WS, Lowe SW, Goeddel DV and Mak TW. Martin SJ, Green DR and Cotter TG. (1994). TIBS, 19, 26 ± (1998). Science, 279, 1954 ± 1958. 30. Yeh WC, Shahinian A, Speiser D, Kraunus J, Bilia F, Miller WE, Mosialos G, Kie€ E and Raab-Traub N. (1997). Wakeham A, de la Pompa JL, et al. (1997). Immunity, 7, J. Virol., 71, 586 ± 594. 715 ± 725. Mosialos G, Birkenbach M, Yalamanchili R, VanArsdale T, Yuan J. (1996). J. Cell. Biochem., 60, 4±11. Ware C and Kie€ E. (1995). Cell, 80, 389 ± 399. Yuan J, Shaham S, Ledoux S, Ellis HM and Horvitz HR. Muzio M, Chinnayan AM, Kischkel FC, O'Rourke K, (1993). Cell, 75, 641 ± 652. Shevchenko A, Scadi C, Brentz JD, Zhang M, Gentz R, Zandi E, Rothwarf DM, Delhase M, Hayakawa M and Mannmkrammer PH, Peter ME and Dixit VM. (1996). Karin M. (1997). Cell, 91, 243 ± 252. Cell, 85, 817 ± 827. Zhang J, Cado D, Chen A, Kabra NH and Winoto A. (1998). Nagata S and Goldstein P. (1995). Science, 267, 1449 ± 1456. Nature, 392, 296 ± 300.