Entry - *601896 - TNF RECEPTOR-ASSOCIATED FACTOR 3; TRAF3 - OMIM
 
* 601896

TNF RECEPTOR-ASSOCIATED FACTOR 3; TRAF3


Alternative titles; symbols

CD40-BINDING PROTEIN; CD40BP
LMP1-ASSOCIATED PROTEIN 1; LAP1
CD40-ASSOCIATED PROTEIN 1; CAP1
CD40 RECEPTOR-ASSOCIATED FACTOR 1; CRAF1


HGNC Approved Gene Symbol: TRAF3

Cytogenetic location: 14q32.32     Genomic coordinates (GRCh38): 14:102,777,449-102,911,500 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
14q32.32 {?Encephalopathy, acute, infection-induced (herpes-specific), susceptibility to, 5} 614849 AD 3

TEXT

Cloning and Expression

CD40 (109535) is a member of the tumor necrosis factor receptor (TNFR) family. The short cytoplasmic domain of CD40 contains a region with limited homology to the conserved cytosolic death domain of TNFR1 (191190) and FAS (134637). Using a yeast 2-hybrid assay with the cytoplasmic domain of CD40 as bait, Hu et al. (1994) isolated B-cell cDNAs encoding a protein that they called CD40-binding protein (CD40bp). The predicted CD40bp protein contains a RING finger DNA-binding motif, a cys/his-rich region, and a coiled-coil domain. Like TRAF1 (601711) and TRAF2 (601895), both TNFR2 (75-kD TNFR; 191191)-binding proteins, CD40bp contains a C-terminal TRAF domain. In vitro translated CD40bp has an apparent molecular mass of 64 kD. Coimmunoprecipitation studies indicated that CD40bp interacts with CD40 in human B cells. Hu et al. (1994) suggested that CD40bp, along with TRAF1 and TRAF2, comprise a family of proteins that associate with the cytoplasmic faces of the TNFR family and have in common a TRAF domain.

Independently, Sato et al. (1995) and Cheng et al. (1995) identified CD40bp, designating it CAP1 (CD40-associated protein-1) and CRAF1 (CD40 receptor-associated factor 1), respectively. Sato et al. (1995) demonstrated that CAP1 binds specifically to the cytoplasmic domain of CD40, but not to that of TNFR1, TNFR2, or FAS. The C-terminal TRAF domain of CAP1 was sufficient to mediate binding to CD40 and homodimerization. Cheng et al. (1995) isolated mouse and human CRAF1 cDNAs. The predicted 568-amino acid human protein is 96% identical to mouse CRAF1. These authors divided the TRAF domain into 2 regions: TRAF-N, the more N-terminal coiled-coil subdomain, and TRAF-C, which was necessary and sufficient for CRAF1 to interact with CD40. Overexpression of a truncated cDNA encoding the entire TRAF domain of CRAF1 inhibited CD40-mediated upregulation of the CD23 (151445) gene, which suggested to Cheng et al. (1995) that CRAF1 participates in CD40 signaling.


Mapping

Gross (2012) mapped the TRAF3 gene to chromosome 14q32.32 based on an alignment of the TRAF3 sequence (GenBank BC075086) with the genomic sequence (GRCh37).


Gene Function

The cytoplasmic C terminus of the Epstein-Barr virus latent infection membrane protein-1 (LMP1) is essential for B lymphocyte growth transformation. LMP1 is an integral membrane protein that has transforming effects in nonlymphoid cells, and may act by constitutively activating a common cellular growth factor receptor pathway. Mosialos et al. (1995) found that CD40bp, which they called LAP1 (LMP1-associated protein-1), interacted with the LMP1 C-terminal domain. Expression of LMP1 caused LAP1 and TRAF1 (EBI6) to localize to LMP1 clusters in lymphoblast plasma membranes, and LMP1 coimmunoprecipitated with these proteins in cell extracts. LAP1 bound to the cytoplasmic domains of CD40 and LT-beta-R (600979) in vitro, and associated with p80 (TNFR2) in vivo. Northern blot analysis revealed that LAP1 was expressed as a full-length 2.8-kb mRNA and an alternatively spliced 1.8-kb mRNA in all tissues tested. Mosialos et al. (1995) concluded that the interaction of LAP1 with LMP1 and with the cytoplasmic domains of TNFR family members is evidence for a central role of this protein as an effector of cell growth or death signaling pathways.

Dadgostar et al. (2003) determined that the coiled-coil domain of mouse T3jam (608255) interacted with the isoleucine zipper domain of Traf3. T3jam did not associate with other Traf family members. Coexpression of T3jam and Traf3 recruited Traf3 to the detergent-insoluble fraction, and T3jam and Traf3 synergistically activated JNK (601158), but not nuclear factor kappa-B (see 164011).

To dissect biochemically Toll-like receptor signaling, Hacker et al. (2006) established a system for isolating signaling complexes assembled by dimerized adaptors. Using MyD88 (602170) as a prototypic adaptor, they identified TRAF3 as a new component of Toll/interleukin-1 receptor signaling complexes that is recruited along with TRAF6 (602355). Using myeloid cells from Traf3- and Traf6-deficient mice, Hacker et al. (2006) demonstrated that TRAF3 is essential for the induction of type I interferons and the antiinflammatory cytokine interleukin-10 (IL10; 124092), but is dispensable for expression of proinflammatory cytokines. In fact, Traf3-deficient cells overproduced proinflammatory cytokines owing to defective Il10 production. Despite their structural similarity, the functions of TRAF3 and TRAF6 are largely distinct. TRAF3 is also recruited to the adaptor TRIF (607601) and is required for marshalling the protein kinase TBK1 (604834) into Toll/interleukin-1 receptor signaling complexes, thereby explaining its unique role in activation of the interferon response.

Oganesyan et al. (2006) demonstrated that cells lacking TRAF3 are defective in type I interferon responses activated by several different Toll-like receptors. Furthermore, they showed that TRAF3 associates with the Toll-like receptor adaptors TRIF and IRAK1 (300283), as well as downstream IRF3/7 kinases TBK1 and IKK-epsilon (IKKE, or IKBKE; 605048), suggesting that TRAF3 serves as a critical link between Toll-like receptor adaptors and downstream regulatory kinases important for IRF activation. In addition to TLR stimulation, Oganesyan et al. (2006) showed that TRAF3-deficient fibroblasts are defective in their type I interferon response to direct infection with vesicular stomatitis virus, indicating that TRAF3 is also an important component of TLR-independent viral recognition pathways. Oganesyan et al. (2006) concluded that TRAF3 is a major regulator of type I interferon production and the innate antiviral response.

Production of type I interferon is a critical host defense triggered by pattern-recognition receptors (PRRs) of the innate immune system. Kayagaki et al. (2007) demonstrated that reduction of DUBA (300713) augmented the PRR-induced type I interferon response in transfected HEK293 cells, whereas ectopic expression of DUBA had the converse effect. DUBA bound TRAF3, an adaptor protein essential for type I interferon response. TRAF3 is an E3 ubiquitin ligase that preferentially assembled lys63-linked polyubiquitin chains in cotransfection assays. DUBA selectively cleaved the lys63-linked polyubiquitin chains on TRAF3, resulting in its dissociation from the downstream signaling complex containing TBK1. A discrete ubiquitin interaction motif within DUBA was required for efficient deubiquitination of TRAF3 and optimal suppression of type I interferon. Kayagaki et al. (2007) concluded that their data identified DUBA as a negative regulator of innate immune responses.

Cytokine signaling is thought to require assembly of multicomponent signaling complexes at cytoplasmic segments of membrane-embedded receptors, in which receptor-proximal protein kinases are activated. Matsuzawa et al. (2008) reported that, upon ligation, CD40 formed a complex containing adaptor molecules TRAF2 and TRAF3, ubiquitin-conjugating enzyme UBC13 (UBE2N; 603679), cellular inhibitor of apoptosis protein-1 (CIAP1, or BIRC2; 601712) and -2 (CIAP2, or BIRC3; 601721), IKK-gamma (IKBKG; 300248), and MEKK1 (MAP3K1; 600982). TRAF2, UBC13, and IKK-gamma were required for complex assembly and activation of MEKK1 and MAP kinase cascades. However, the kinases were not activated unless the complex was translocated from the membrane to the cytosol upon CIAP1/CIAP2-induced degradation of TRAF3. Matsuzawa et al. (2008) proposed that this 2-stage signaling mechanism may apply to other innate immune receptors and may account for spatial and temporal separation of MAPK and IKK signaling.

Hu et al. (2013) identified the deubiquitinase OTUD7B (611748) as a pivotal regulator of the noncanonical NF-kappa-B pathway. OTUD7B deficiency in mice has no appreciable effect on canonical NF-kappa-B activation but causes hyperactivation of noncanonical NF-kappa-B. In response to noncanonical NF-kappa-B stimuli, OTUD7B binds and deubiquitinates TRAF3, thereby inhibiting TRAF3 proteolysis and preventing aberrant noncanonical NF-kappa-B activation. Consequently, the OTUD7B deficiency results in B-cell hyperresponsiveness to antigens, lymphoid follicular hyperplasia in the intestinal mucosa, and elevated host-defense ability against an intestinal bacterial pathogen, Citrobacter rodentium. Hu et al. (2013) concluded that their findings established OTUD7B as a crucial regulator of signal-induced noncanonical NF-kappa-B activation, and indicated a mechanism of immune regulation that involves OTUD7B-mediated deubiquitination and stabilization of TRAF3.

The herpes simplex virus-1 (HSV-1) tegument protein UL36 contains an N-terminal deubiquitinase (DUB) motif called UL36 ubiquitin-specific protease (UL36USP). By expressing UL36USP in human embryonic kidney cells, Wang et al. (2013) identified host pathways affected by HSV-1 infection that resulted in inhibition of IFNB (147640) expression. UL36USP inhibited Sendai virus (SeV)-induced IRF3 (603734) dimerization and activation and transcription of IFNB. Mutation analysis confirmed that the DUB activity of UL36USP1 was required to block IFNB production. UL36USP also inhibited IFNB promoter activity induced by overexpression of the RIGI (DDX58; 609631) N terminus or MAVS (609676), but not TBK1, IKKE, or the active form of IRF3. UL36USP deubuitinated TRAF3 and prevented recruitment of TBK1. Cells infected with recombinant HSV-1 lacking UL36USP DUB activity produced more IFNB than cells infected with wildtype HSV-1. Wang et al. (2013) concluded that HSV-1 UL36USP removes polyubiquitin chains on TRAF3 and counteracts the IFNB pathway.

Using transfected HEK293T cells, Chen et al. (2015) showed that overexpression of RNF166 (617178) enhanced activation of the IFNB promoter after infection with SeV. Knockdown of RNF166 in HEK293T cells inhibited IFNB promoter activation, IFNB transcription, and IFNB secretion in response to SeV infection. Similar results were observed with knockdown of RNF166 in HeLa cells. RNF166 interacted with TRAF3 and TRAF6, and knockdown of RNF166 suppressed SeV-induced ubiquitination of TRAF3 and TRAF6. Chen et al. (2015) proposed that RNF166 positively regulates RNA virus-triggered IFNB production by enhancing ubiquitination of TRAF3 and TRAF6.


Molecular Genetics

Acute Infection-Induced (Herpes-Specific) Encephalopathy, Susceptibility to

Perez de Diego et al. (2010) investigated an 18-year-old French female who had suffered from herpes simplex encephalitis (HSE) (IIAE5; 614849) at age 4 years and who lacked mutations in either the UNC93B1 (608204) or TLR3 (603029) genes. They identified a heterozygous C-to-T substitution at position 352 in exon 4 of the TRAF3 gene, resulting in a nonconservative missense change, arg118 to trp (R118W; 601896.0001). The mutation was not found in her parents or brothers, and none had a history of HSE. RT-PCR analysis of the patient's cells detected normal levels of TRAF3 mRNA, but Western blot analysis showed severely reduced levels of TRAF3 protein. Responsiveness to TLR3 agonists was impaired, as indicated by deficient NFKB activation and poor production of IFNB, IFNL (IL29; 607403), and IL6 (147620). Expression of wildtype and mutant alleles in cell lines showed that the mutant was dominant-negative. TRAF3-deficient fibroblasts had impaired type I and type III IFN-dependent control of viruses and deficient responses through the TNFR (e.g., TNFRSF5; 109535) pathways. Perez de Diego et al. (2010) concluded that, whereas complete Traf3 deficiency is neonatal lethal in mice, decreases in TRAF3 production and function result in predisposition to HSE, a condition that is usually fatal if untreated.

Associations Pending Confirmation

Braggio et al. (2009) identified biallelic inactivation of TRAF3 in 3 (5.3%) of 57 Waldenstrom macroglobulinemia (WM; see 153600) samples. TRAF3 inactivation was associated with transcriptional activation of NF-kappa-B (NFKB1; 164011). In addition, 1 of 24 patients with a 6q deletion had an inactivating somatic mutation in TNFAIP3 (191163), another negative regulator of NF-kappa-B. Monoallelic deletions of chromosome 6q23, including the TNFAIP3 gene, were identified in 38% of patients, suggesting that haploinsufficiency can predispose to the development of WM. The results indicated that mutational activation of the NF-kappa-B pathway plays a role in the pathogenesis of WM.


Animal Model

Mikula et al. (2001) reported that mice lacking Craf1 died at midgestation with placenta and liver anomalies. The livers were hypocellular, but hepatoblast proliferation was not impaired. Fibroblast and hemopoietic cell proliferation was poor due to increased apoptosis with greater sensitivity to apoptotic stimuli, such as Fasl (TNFSF6; 134638)/Fas activation. Mikula et al. (2001) concluded that the essential function of CRAF1 is to counteract apoptosis using effectors distinct from the MEK/ERK (see 601795) cascade.

By generating mice deficient in Traf3 specifically in regulatory T (Treg) cells, Chang et al. (2014) showed that Traf3 was dispensable for homeostasis of Treg cells but was critical for regulation of follicular Treg (TFR) cell generation and germinal center reactions. Mice lacking Traf3 in Treg cells had severe inhibition of antigen-stimulated activation of TFR cells, coupled with deregulated activation of T follicular helper (TFH) cells and heightened germinal center reactions, with elevated production of high-affinity antibodies of the IgG subtypes. Chang et al. (2014) concluded that TRAF3 is a signaling factor that mediates the effector functions of Treg cells, particularly in the control of humoral immune responses, and that TRAF3 is involved in induction of ICOS (604558) as a result of impaired ERK activation.


ALLELIC VARIANTS ( 1 Selected Example):

.0001 ENCEPHALOPATHY, ACUTE, INFECTION-INDUCED (HERPES-SPECIFIC), SUSCEPTIBILITY TO, 5 (1 patient)

TRAF3, ARG118TRP
  
RCV000030822...

Perez de Diego et al. (2010) reported a female French patient with herpes simplex encephalitis (IIAE5; 614849) at age 4 years who was healthy without prophylaxis and showed normal resistance to other infectious diseases, including other herpesvirus family members, at age 18 years. They identified a de novo heterozygous 352C-T transition in exon 4 of the TRAF3 gene, resulting in an arg118-to-trp (R118W) substitution, in this patient, whose HSE was successfully treated with acyclovir for 3 weeks.


REFERENCES

  1. Braggio, E., Keats, J. J., Leleu, X., Van Wier, S., Jimenez-Zepeda, V. H., Valdez, R., Schop, R. F. J., Price-Troska, T., Henderson, K., Sacco, A., Azab, F., Greipp, P., and 11 others. Identification of copy number abnormalities and inactivating mutations in two negative regulators of nuclear factor-kappa-B signaling pathways in Waldenstrom's macroglobulinemia. Cancer Res. 69: 3579-3588, 2009. [PubMed: 19351844, images, related citations] [Full Text]

  2. Chang, J.-H., Hu, H., Jin, J., Puebla-Osorio, N., Xiao, Y., Gilbert, B. E., Brink, R., Ullrich, S. E., Sun, S.-C. TRAF3 regulates the effector function of regulatory T cells and humoral immune responses. J. Exp. Med. 211: 137-151, 2014. [PubMed: 24378539, images, related citations] [Full Text]

  3. Chen, H.-W., Yang, Y.-K., Xu, H., Yang, W.-W., Zhai, Z.-H., Chen, D.-Y. Ring finger protein 166 potentiates RNA virus-induced interferon-beta production via enhancing the ubiquitination of TRAF3 and TRAF6. Sci. Rep. 5: 14770, 2015. Note: Electronic Article. [PubMed: 26456228, images, related citations] [Full Text]

  4. Cheng, G., Cleary, A. M., Ye, Z., Hong, D. I., Lederman, S., Baltimore, D. Involvement of CRAF1, a relative of TRAF, in CD40 signaling. Science 267: 1494-1498, 1995. [PubMed: 7533327, related citations] [Full Text]

  5. Dadgostar, H., Doyle, S. E., Shahangian, A., Garcia, D. E., Cheng, G. T3JAM, a novel protein that specifically interacts with TRAF3 and promotes the activation of JNK. FEBS Lett. 553: 403-407, 2003. [PubMed: 14572659, related citations] [Full Text]

  6. Gross, M. B. Personal Communication. Baltimore, Md. 10/5/2012.

  7. Hacker, H., Redecke, V., Blagoev, B., Kratchmarova, I., Hsu, L.-C., Wang, G. G., Kamps, M. P., Raz, E., Wagner, H., Hacker, G., Mann, M., Karin, M. Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature 439: 204-207, 2006. [PubMed: 16306937, related citations] [Full Text]

  8. Hu, H., Brittain, G. C., Chang, J.-H., Puebla-Osorio, N., Jin, J., Zal, Z., Xiao, Y., Cheng, X., Chang, M., Fu, Y.-X., Zal, T., Zhu, C., Sun, S.-C. OTUD7B controls noncanonical NF-kappa-B activation through deubiquitination of TRAF3. Nature 494: 371-374, 2013. [PubMed: 23334419, images, related citations] [Full Text]

  9. Hu, H. M., O'Rourke, K., Boguski, M. S., Dixit, V. M. A novel RING finger protein interacts with the cytoplasmic domain of CD40. J. Biol. Chem. 269: 30069-30072, 1994. [PubMed: 7527023, related citations]

  10. Kayagaki, N., Phung, Q., Chan, S., Chaudhari, R., Quan, C., O'Rourke, K. M., Eby, M., Pietras, E., Cheng, G., Bazan, J. F., Zhang, Z., Arnott, D., Dixit, V. M. DUBA: a deubiquitinase that regulates type I interferon production. Science 318: 1628-1632, 2007. [PubMed: 17991829, related citations] [Full Text]

  11. Matsuzawa, A., Tseng, P.-H., Vallabhapurapu, S., Luo, J.-L., Zhang, W., Wang, H., Vignali, D. A. A., Gallagher, E., Karin, M. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science 321: 663-668, 2008. Note: Erratum: Science 322: 375 only, 2008. [PubMed: 18635759, images, related citations] [Full Text]

  12. Mikula, M., Schreiber, M., Husak, Z., Kucerova, L., Ruth, J., Wieser, R., Zatloukal, K., Beug, H., Wagner, E. F., Baccarini, M. Embryonic lethality and fetal liver apoptosis in mice lacking the c-raf-1 gene. EMBO J. 20: 1952-1962, 2001. [PubMed: 11296228, images, related citations] [Full Text]

  13. Mosialos, G., Birkenbach, M., Yalamanchili, R., VanArsdale, T., Ware, C., Kieff, E. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell 80: 389-399, 1995. [PubMed: 7859281, related citations] [Full Text]

  14. Oganesyan, G., Saha, S. K., Guo, B., He, J. Q., Shahangian, A., Zarnegar, B., Perry, A., Cheng, G. Critical role of TRAF3 in the Toll-like receptor-dependent and -independent antiviral response. Nature 439: 208-211, 2006. [PubMed: 16306936, related citations] [Full Text]

  15. Perez de Diego, R., Sancho-Shimizu, V., Lorenzo, L., Puel, A., Plancoulaine, S., Picard, C., Herman, M., Cardon, A., Durandy, A., Bustamante, J., Vallabhapurapu, S., Bravo, J., and 12 others. Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis. Immunity 33: 400-411, 2010. [PubMed: 20832341, images, related citations] [Full Text]

  16. Sato, T., Irie, S., Reed, J. C. A novel member of the TRAF family of putative signal transducing proteins binds to the cytoplasmic domain of CD40. FEBS Lett. 358: 113-118, 1995. [PubMed: 7530216, related citations] [Full Text]

  17. Wang, S., Wang, K., Li, J., Zheng, C. Herpes simplex virus 1 ubiquitin-specific protease UL36 inhibits beta interferon production by deubiquitinating TRAF3. J. Virol. 87: 11851-11860, 2013. [PubMed: 23986588, images, related citations] [Full Text]


Paul J. Converse - updated : 10/31/2016
Paul J. Converse - updated : 10/8/2015
Paul J. Converse - updated : 6/10/2014
Ada Hamosh - updated : 3/21/2013
Matthew B. Gross - updated : 10/5/2012
Paul J. Converse - updated : 10/4/2012
Cassandra L. Kniffin - updated : 1/15/2010
Paul J. Converse - updated : 8/28/2008
Ada Hamosh - updated : 5/7/2008
Ada Hamosh - updated : 5/1/2006
Patricia A. Hartz - updated : 11/13/2003
Rebekah S. Rasooly - updated : 6/11/1999
Creation Date:
Lori M. Kelman : 3/20/1997
carol : 03/14/2018
mgross : 10/31/2016
mgross : 10/08/2015
mgross : 10/8/2015
carol : 8/31/2015
mgross : 6/10/2014
mcolton : 5/29/2014
alopez : 4/2/2013
terry : 3/21/2013
joanna : 12/7/2012
mgross : 10/5/2012
mgross : 10/5/2012
mgross : 10/5/2012
terry : 10/4/2012
wwang : 1/15/2010
ckniffin : 12/22/2009
alopez : 11/18/2008
mgross : 8/28/2008
terry : 8/28/2008
alopez : 5/7/2008
wwang : 4/23/2008
alopez : 5/3/2006
terry : 5/1/2006
mgross : 11/13/2003
mgross : 11/13/2003
alopez : 6/11/1999
alopez : 6/11/1999
carol : 6/18/1998
mark : 9/9/1997
alopez : 8/1/1997
alopez : 7/23/1997

* 601896

TNF RECEPTOR-ASSOCIATED FACTOR 3; TRAF3


Alternative titles; symbols

CD40-BINDING PROTEIN; CD40BP
LMP1-ASSOCIATED PROTEIN 1; LAP1
CD40-ASSOCIATED PROTEIN 1; CAP1
CD40 RECEPTOR-ASSOCIATED FACTOR 1; CRAF1


HGNC Approved Gene Symbol: TRAF3

Cytogenetic location: 14q32.32     Genomic coordinates (GRCh38): 14:102,777,449-102,911,500 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
14q32.32 {?Encephalopathy, acute, infection-induced (herpes-specific), susceptibility to, 5} 614849 Autosomal dominant 3

TEXT

Cloning and Expression

CD40 (109535) is a member of the tumor necrosis factor receptor (TNFR) family. The short cytoplasmic domain of CD40 contains a region with limited homology to the conserved cytosolic death domain of TNFR1 (191190) and FAS (134637). Using a yeast 2-hybrid assay with the cytoplasmic domain of CD40 as bait, Hu et al. (1994) isolated B-cell cDNAs encoding a protein that they called CD40-binding protein (CD40bp). The predicted CD40bp protein contains a RING finger DNA-binding motif, a cys/his-rich region, and a coiled-coil domain. Like TRAF1 (601711) and TRAF2 (601895), both TNFR2 (75-kD TNFR; 191191)-binding proteins, CD40bp contains a C-terminal TRAF domain. In vitro translated CD40bp has an apparent molecular mass of 64 kD. Coimmunoprecipitation studies indicated that CD40bp interacts with CD40 in human B cells. Hu et al. (1994) suggested that CD40bp, along with TRAF1 and TRAF2, comprise a family of proteins that associate with the cytoplasmic faces of the TNFR family and have in common a TRAF domain.

Independently, Sato et al. (1995) and Cheng et al. (1995) identified CD40bp, designating it CAP1 (CD40-associated protein-1) and CRAF1 (CD40 receptor-associated factor 1), respectively. Sato et al. (1995) demonstrated that CAP1 binds specifically to the cytoplasmic domain of CD40, but not to that of TNFR1, TNFR2, or FAS. The C-terminal TRAF domain of CAP1 was sufficient to mediate binding to CD40 and homodimerization. Cheng et al. (1995) isolated mouse and human CRAF1 cDNAs. The predicted 568-amino acid human protein is 96% identical to mouse CRAF1. These authors divided the TRAF domain into 2 regions: TRAF-N, the more N-terminal coiled-coil subdomain, and TRAF-C, which was necessary and sufficient for CRAF1 to interact with CD40. Overexpression of a truncated cDNA encoding the entire TRAF domain of CRAF1 inhibited CD40-mediated upregulation of the CD23 (151445) gene, which suggested to Cheng et al. (1995) that CRAF1 participates in CD40 signaling.


Mapping

Gross (2012) mapped the TRAF3 gene to chromosome 14q32.32 based on an alignment of the TRAF3 sequence (GenBank BC075086) with the genomic sequence (GRCh37).


Gene Function

The cytoplasmic C terminus of the Epstein-Barr virus latent infection membrane protein-1 (LMP1) is essential for B lymphocyte growth transformation. LMP1 is an integral membrane protein that has transforming effects in nonlymphoid cells, and may act by constitutively activating a common cellular growth factor receptor pathway. Mosialos et al. (1995) found that CD40bp, which they called LAP1 (LMP1-associated protein-1), interacted with the LMP1 C-terminal domain. Expression of LMP1 caused LAP1 and TRAF1 (EBI6) to localize to LMP1 clusters in lymphoblast plasma membranes, and LMP1 coimmunoprecipitated with these proteins in cell extracts. LAP1 bound to the cytoplasmic domains of CD40 and LT-beta-R (600979) in vitro, and associated with p80 (TNFR2) in vivo. Northern blot analysis revealed that LAP1 was expressed as a full-length 2.8-kb mRNA and an alternatively spliced 1.8-kb mRNA in all tissues tested. Mosialos et al. (1995) concluded that the interaction of LAP1 with LMP1 and with the cytoplasmic domains of TNFR family members is evidence for a central role of this protein as an effector of cell growth or death signaling pathways.

Dadgostar et al. (2003) determined that the coiled-coil domain of mouse T3jam (608255) interacted with the isoleucine zipper domain of Traf3. T3jam did not associate with other Traf family members. Coexpression of T3jam and Traf3 recruited Traf3 to the detergent-insoluble fraction, and T3jam and Traf3 synergistically activated JNK (601158), but not nuclear factor kappa-B (see 164011).

To dissect biochemically Toll-like receptor signaling, Hacker et al. (2006) established a system for isolating signaling complexes assembled by dimerized adaptors. Using MyD88 (602170) as a prototypic adaptor, they identified TRAF3 as a new component of Toll/interleukin-1 receptor signaling complexes that is recruited along with TRAF6 (602355). Using myeloid cells from Traf3- and Traf6-deficient mice, Hacker et al. (2006) demonstrated that TRAF3 is essential for the induction of type I interferons and the antiinflammatory cytokine interleukin-10 (IL10; 124092), but is dispensable for expression of proinflammatory cytokines. In fact, Traf3-deficient cells overproduced proinflammatory cytokines owing to defective Il10 production. Despite their structural similarity, the functions of TRAF3 and TRAF6 are largely distinct. TRAF3 is also recruited to the adaptor TRIF (607601) and is required for marshalling the protein kinase TBK1 (604834) into Toll/interleukin-1 receptor signaling complexes, thereby explaining its unique role in activation of the interferon response.

Oganesyan et al. (2006) demonstrated that cells lacking TRAF3 are defective in type I interferon responses activated by several different Toll-like receptors. Furthermore, they showed that TRAF3 associates with the Toll-like receptor adaptors TRIF and IRAK1 (300283), as well as downstream IRF3/7 kinases TBK1 and IKK-epsilon (IKKE, or IKBKE; 605048), suggesting that TRAF3 serves as a critical link between Toll-like receptor adaptors and downstream regulatory kinases important for IRF activation. In addition to TLR stimulation, Oganesyan et al. (2006) showed that TRAF3-deficient fibroblasts are defective in their type I interferon response to direct infection with vesicular stomatitis virus, indicating that TRAF3 is also an important component of TLR-independent viral recognition pathways. Oganesyan et al. (2006) concluded that TRAF3 is a major regulator of type I interferon production and the innate antiviral response.

Production of type I interferon is a critical host defense triggered by pattern-recognition receptors (PRRs) of the innate immune system. Kayagaki et al. (2007) demonstrated that reduction of DUBA (300713) augmented the PRR-induced type I interferon response in transfected HEK293 cells, whereas ectopic expression of DUBA had the converse effect. DUBA bound TRAF3, an adaptor protein essential for type I interferon response. TRAF3 is an E3 ubiquitin ligase that preferentially assembled lys63-linked polyubiquitin chains in cotransfection assays. DUBA selectively cleaved the lys63-linked polyubiquitin chains on TRAF3, resulting in its dissociation from the downstream signaling complex containing TBK1. A discrete ubiquitin interaction motif within DUBA was required for efficient deubiquitination of TRAF3 and optimal suppression of type I interferon. Kayagaki et al. (2007) concluded that their data identified DUBA as a negative regulator of innate immune responses.

Cytokine signaling is thought to require assembly of multicomponent signaling complexes at cytoplasmic segments of membrane-embedded receptors, in which receptor-proximal protein kinases are activated. Matsuzawa et al. (2008) reported that, upon ligation, CD40 formed a complex containing adaptor molecules TRAF2 and TRAF3, ubiquitin-conjugating enzyme UBC13 (UBE2N; 603679), cellular inhibitor of apoptosis protein-1 (CIAP1, or BIRC2; 601712) and -2 (CIAP2, or BIRC3; 601721), IKK-gamma (IKBKG; 300248), and MEKK1 (MAP3K1; 600982). TRAF2, UBC13, and IKK-gamma were required for complex assembly and activation of MEKK1 and MAP kinase cascades. However, the kinases were not activated unless the complex was translocated from the membrane to the cytosol upon CIAP1/CIAP2-induced degradation of TRAF3. Matsuzawa et al. (2008) proposed that this 2-stage signaling mechanism may apply to other innate immune receptors and may account for spatial and temporal separation of MAPK and IKK signaling.

Hu et al. (2013) identified the deubiquitinase OTUD7B (611748) as a pivotal regulator of the noncanonical NF-kappa-B pathway. OTUD7B deficiency in mice has no appreciable effect on canonical NF-kappa-B activation but causes hyperactivation of noncanonical NF-kappa-B. In response to noncanonical NF-kappa-B stimuli, OTUD7B binds and deubiquitinates TRAF3, thereby inhibiting TRAF3 proteolysis and preventing aberrant noncanonical NF-kappa-B activation. Consequently, the OTUD7B deficiency results in B-cell hyperresponsiveness to antigens, lymphoid follicular hyperplasia in the intestinal mucosa, and elevated host-defense ability against an intestinal bacterial pathogen, Citrobacter rodentium. Hu et al. (2013) concluded that their findings established OTUD7B as a crucial regulator of signal-induced noncanonical NF-kappa-B activation, and indicated a mechanism of immune regulation that involves OTUD7B-mediated deubiquitination and stabilization of TRAF3.

The herpes simplex virus-1 (HSV-1) tegument protein UL36 contains an N-terminal deubiquitinase (DUB) motif called UL36 ubiquitin-specific protease (UL36USP). By expressing UL36USP in human embryonic kidney cells, Wang et al. (2013) identified host pathways affected by HSV-1 infection that resulted in inhibition of IFNB (147640) expression. UL36USP inhibited Sendai virus (SeV)-induced IRF3 (603734) dimerization and activation and transcription of IFNB. Mutation analysis confirmed that the DUB activity of UL36USP1 was required to block IFNB production. UL36USP also inhibited IFNB promoter activity induced by overexpression of the RIGI (DDX58; 609631) N terminus or MAVS (609676), but not TBK1, IKKE, or the active form of IRF3. UL36USP deubuitinated TRAF3 and prevented recruitment of TBK1. Cells infected with recombinant HSV-1 lacking UL36USP DUB activity produced more IFNB than cells infected with wildtype HSV-1. Wang et al. (2013) concluded that HSV-1 UL36USP removes polyubiquitin chains on TRAF3 and counteracts the IFNB pathway.

Using transfected HEK293T cells, Chen et al. (2015) showed that overexpression of RNF166 (617178) enhanced activation of the IFNB promoter after infection with SeV. Knockdown of RNF166 in HEK293T cells inhibited IFNB promoter activation, IFNB transcription, and IFNB secretion in response to SeV infection. Similar results were observed with knockdown of RNF166 in HeLa cells. RNF166 interacted with TRAF3 and TRAF6, and knockdown of RNF166 suppressed SeV-induced ubiquitination of TRAF3 and TRAF6. Chen et al. (2015) proposed that RNF166 positively regulates RNA virus-triggered IFNB production by enhancing ubiquitination of TRAF3 and TRAF6.


Molecular Genetics

Acute Infection-Induced (Herpes-Specific) Encephalopathy, Susceptibility to

Perez de Diego et al. (2010) investigated an 18-year-old French female who had suffered from herpes simplex encephalitis (HSE) (IIAE5; 614849) at age 4 years and who lacked mutations in either the UNC93B1 (608204) or TLR3 (603029) genes. They identified a heterozygous C-to-T substitution at position 352 in exon 4 of the TRAF3 gene, resulting in a nonconservative missense change, arg118 to trp (R118W; 601896.0001). The mutation was not found in her parents or brothers, and none had a history of HSE. RT-PCR analysis of the patient's cells detected normal levels of TRAF3 mRNA, but Western blot analysis showed severely reduced levels of TRAF3 protein. Responsiveness to TLR3 agonists was impaired, as indicated by deficient NFKB activation and poor production of IFNB, IFNL (IL29; 607403), and IL6 (147620). Expression of wildtype and mutant alleles in cell lines showed that the mutant was dominant-negative. TRAF3-deficient fibroblasts had impaired type I and type III IFN-dependent control of viruses and deficient responses through the TNFR (e.g., TNFRSF5; 109535) pathways. Perez de Diego et al. (2010) concluded that, whereas complete Traf3 deficiency is neonatal lethal in mice, decreases in TRAF3 production and function result in predisposition to HSE, a condition that is usually fatal if untreated.

Associations Pending Confirmation

Braggio et al. (2009) identified biallelic inactivation of TRAF3 in 3 (5.3%) of 57 Waldenstrom macroglobulinemia (WM; see 153600) samples. TRAF3 inactivation was associated with transcriptional activation of NF-kappa-B (NFKB1; 164011). In addition, 1 of 24 patients with a 6q deletion had an inactivating somatic mutation in TNFAIP3 (191163), another negative regulator of NF-kappa-B. Monoallelic deletions of chromosome 6q23, including the TNFAIP3 gene, were identified in 38% of patients, suggesting that haploinsufficiency can predispose to the development of WM. The results indicated that mutational activation of the NF-kappa-B pathway plays a role in the pathogenesis of WM.


Animal Model

Mikula et al. (2001) reported that mice lacking Craf1 died at midgestation with placenta and liver anomalies. The livers were hypocellular, but hepatoblast proliferation was not impaired. Fibroblast and hemopoietic cell proliferation was poor due to increased apoptosis with greater sensitivity to apoptotic stimuli, such as Fasl (TNFSF6; 134638)/Fas activation. Mikula et al. (2001) concluded that the essential function of CRAF1 is to counteract apoptosis using effectors distinct from the MEK/ERK (see 601795) cascade.

By generating mice deficient in Traf3 specifically in regulatory T (Treg) cells, Chang et al. (2014) showed that Traf3 was dispensable for homeostasis of Treg cells but was critical for regulation of follicular Treg (TFR) cell generation and germinal center reactions. Mice lacking Traf3 in Treg cells had severe inhibition of antigen-stimulated activation of TFR cells, coupled with deregulated activation of T follicular helper (TFH) cells and heightened germinal center reactions, with elevated production of high-affinity antibodies of the IgG subtypes. Chang et al. (2014) concluded that TRAF3 is a signaling factor that mediates the effector functions of Treg cells, particularly in the control of humoral immune responses, and that TRAF3 is involved in induction of ICOS (604558) as a result of impaired ERK activation.


ALLELIC VARIANTS 1 Selected Example):

.0001   ENCEPHALOPATHY, ACUTE, INFECTION-INDUCED (HERPES-SPECIFIC), SUSCEPTIBILITY TO, 5 (1 patient)

TRAF3, ARG118TRP
SNP: rs143813189, gnomAD: rs143813189, ClinVar: RCV000030822, RCV003407376, RCV003952376

Perez de Diego et al. (2010) reported a female French patient with herpes simplex encephalitis (IIAE5; 614849) at age 4 years who was healthy without prophylaxis and showed normal resistance to other infectious diseases, including other herpesvirus family members, at age 18 years. They identified a de novo heterozygous 352C-T transition in exon 4 of the TRAF3 gene, resulting in an arg118-to-trp (R118W) substitution, in this patient, whose HSE was successfully treated with acyclovir for 3 weeks.


REFERENCES

  1. Braggio, E., Keats, J. J., Leleu, X., Van Wier, S., Jimenez-Zepeda, V. H., Valdez, R., Schop, R. F. J., Price-Troska, T., Henderson, K., Sacco, A., Azab, F., Greipp, P., and 11 others. Identification of copy number abnormalities and inactivating mutations in two negative regulators of nuclear factor-kappa-B signaling pathways in Waldenstrom's macroglobulinemia. Cancer Res. 69: 3579-3588, 2009. [PubMed: 19351844] [Full Text: https://doi.org/10.1158/0008-5472.CAN-08-3701]

  2. Chang, J.-H., Hu, H., Jin, J., Puebla-Osorio, N., Xiao, Y., Gilbert, B. E., Brink, R., Ullrich, S. E., Sun, S.-C. TRAF3 regulates the effector function of regulatory T cells and humoral immune responses. J. Exp. Med. 211: 137-151, 2014. [PubMed: 24378539] [Full Text: https://doi.org/10.1084/jem.20131019]

  3. Chen, H.-W., Yang, Y.-K., Xu, H., Yang, W.-W., Zhai, Z.-H., Chen, D.-Y. Ring finger protein 166 potentiates RNA virus-induced interferon-beta production via enhancing the ubiquitination of TRAF3 and TRAF6. Sci. Rep. 5: 14770, 2015. Note: Electronic Article. [PubMed: 26456228] [Full Text: https://doi.org/10.1038/srep14770]

  4. Cheng, G., Cleary, A. M., Ye, Z., Hong, D. I., Lederman, S., Baltimore, D. Involvement of CRAF1, a relative of TRAF, in CD40 signaling. Science 267: 1494-1498, 1995. [PubMed: 7533327] [Full Text: https://doi.org/10.1126/science.7533327]

  5. Dadgostar, H., Doyle, S. E., Shahangian, A., Garcia, D. E., Cheng, G. T3JAM, a novel protein that specifically interacts with TRAF3 and promotes the activation of JNK. FEBS Lett. 553: 403-407, 2003. [PubMed: 14572659] [Full Text: https://doi.org/10.1016/s0014-5793(03)01072-x]

  6. Gross, M. B. Personal Communication. Baltimore, Md. 10/5/2012.

  7. Hacker, H., Redecke, V., Blagoev, B., Kratchmarova, I., Hsu, L.-C., Wang, G. G., Kamps, M. P., Raz, E., Wagner, H., Hacker, G., Mann, M., Karin, M. Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature 439: 204-207, 2006. [PubMed: 16306937] [Full Text: https://doi.org/10.1038/nature04369]

  8. Hu, H., Brittain, G. C., Chang, J.-H., Puebla-Osorio, N., Jin, J., Zal, Z., Xiao, Y., Cheng, X., Chang, M., Fu, Y.-X., Zal, T., Zhu, C., Sun, S.-C. OTUD7B controls noncanonical NF-kappa-B activation through deubiquitination of TRAF3. Nature 494: 371-374, 2013. [PubMed: 23334419] [Full Text: https://doi.org/10.1038/nature11831]

  9. Hu, H. M., O'Rourke, K., Boguski, M. S., Dixit, V. M. A novel RING finger protein interacts with the cytoplasmic domain of CD40. J. Biol. Chem. 269: 30069-30072, 1994. [PubMed: 7527023]

  10. Kayagaki, N., Phung, Q., Chan, S., Chaudhari, R., Quan, C., O'Rourke, K. M., Eby, M., Pietras, E., Cheng, G., Bazan, J. F., Zhang, Z., Arnott, D., Dixit, V. M. DUBA: a deubiquitinase that regulates type I interferon production. Science 318: 1628-1632, 2007. [PubMed: 17991829] [Full Text: https://doi.org/10.1126/science.1145918]

  11. Matsuzawa, A., Tseng, P.-H., Vallabhapurapu, S., Luo, J.-L., Zhang, W., Wang, H., Vignali, D. A. A., Gallagher, E., Karin, M. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science 321: 663-668, 2008. Note: Erratum: Science 322: 375 only, 2008. [PubMed: 18635759] [Full Text: https://doi.org/10.1126/science.1157340]

  12. Mikula, M., Schreiber, M., Husak, Z., Kucerova, L., Ruth, J., Wieser, R., Zatloukal, K., Beug, H., Wagner, E. F., Baccarini, M. Embryonic lethality and fetal liver apoptosis in mice lacking the c-raf-1 gene. EMBO J. 20: 1952-1962, 2001. [PubMed: 11296228] [Full Text: https://doi.org/10.1093/emboj/20.8.1952]

  13. Mosialos, G., Birkenbach, M., Yalamanchili, R., VanArsdale, T., Ware, C., Kieff, E. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell 80: 389-399, 1995. [PubMed: 7859281] [Full Text: https://doi.org/10.1016/0092-8674(95)90489-1]

  14. Oganesyan, G., Saha, S. K., Guo, B., He, J. Q., Shahangian, A., Zarnegar, B., Perry, A., Cheng, G. Critical role of TRAF3 in the Toll-like receptor-dependent and -independent antiviral response. Nature 439: 208-211, 2006. [PubMed: 16306936] [Full Text: https://doi.org/10.1038/nature04374]

  15. Perez de Diego, R., Sancho-Shimizu, V., Lorenzo, L., Puel, A., Plancoulaine, S., Picard, C., Herman, M., Cardon, A., Durandy, A., Bustamante, J., Vallabhapurapu, S., Bravo, J., and 12 others. Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis. Immunity 33: 400-411, 2010. [PubMed: 20832341] [Full Text: https://doi.org/10.1016/j.immuni.2010.08.014]

  16. Sato, T., Irie, S., Reed, J. C. A novel member of the TRAF family of putative signal transducing proteins binds to the cytoplasmic domain of CD40. FEBS Lett. 358: 113-118, 1995. [PubMed: 7530216] [Full Text: https://doi.org/10.1016/0014-5793(94)01406-q]

  17. Wang, S., Wang, K., Li, J., Zheng, C. Herpes simplex virus 1 ubiquitin-specific protease UL36 inhibits beta interferon production by deubiquitinating TRAF3. J. Virol. 87: 11851-11860, 2013. [PubMed: 23986588] [Full Text: https://doi.org/10.1128/JVI.01211-13]


Contributors:
Paul J. Converse - updated : 10/31/2016
Paul J. Converse - updated : 10/8/2015
Paul J. Converse - updated : 6/10/2014
Ada Hamosh - updated : 3/21/2013
Matthew B. Gross - updated : 10/5/2012
Paul J. Converse - updated : 10/4/2012
Cassandra L. Kniffin - updated : 1/15/2010
Paul J. Converse - updated : 8/28/2008
Ada Hamosh - updated : 5/7/2008
Ada Hamosh - updated : 5/1/2006
Patricia A. Hartz - updated : 11/13/2003
Rebekah S. Rasooly - updated : 6/11/1999

Creation Date:
Lori M. Kelman : 3/20/1997

Edit History:
carol : 03/14/2018
mgross : 10/31/2016
mgross : 10/08/2015
mgross : 10/8/2015
carol : 8/31/2015
mgross : 6/10/2014
mcolton : 5/29/2014
alopez : 4/2/2013
terry : 3/21/2013
joanna : 12/7/2012
mgross : 10/5/2012
mgross : 10/5/2012
mgross : 10/5/2012
terry : 10/4/2012
wwang : 1/15/2010
ckniffin : 12/22/2009
alopez : 11/18/2008
mgross : 8/28/2008
terry : 8/28/2008
alopez : 5/7/2008
wwang : 4/23/2008
alopez : 5/3/2006
terry : 5/1/2006
mgross : 11/13/2003
mgross : 11/13/2003
alopez : 6/11/1999
alopez : 6/11/1999
carol : 6/18/1998
mark : 9/9/1997
alopez : 8/1/1997
alopez : 7/23/1997