Visual Abstract

As an important effector in response to various intracellular or extracellular stimuli, the NF-κB family extensively participates in a wide spectrum of biological events, and its dysregulation may result in many pathological conditions, such as microbial infection, tumor progression, and neurodegenerative disorders. Previous investigations showed that multiple types of ubiquitination play critical roles in the modulation of the NF-κB signaling pathway, yet the molecular mechanisms are still poorly understood. In the current study, we identified TRIM25, an E3 ubiquitin ligase, as a novel positive regulator in mediating NF-κB activation in human embryonic kidney 293T (HEK293T), HeLa cells, THP-1 cells, and PBMCs. The expression of TRIM25 promoted TNF-α–induced NF-κB signaling, whereas the knockdown had the opposite effect. Furthermore, TRIM25 interacted with TRAF2 and enhanced the K63-linked polyubiquitin chains attached to TRAF2. Moreover, TRIM25 bridged the interaction of TRAF2 and TAK1 or IKKβ. To our knowledge, our study has identified a previously unrecognized role for TRIM25 in the regulation of NF-κB activation by enhancing the K63-linked ubiquitination of TRAF2.

Nuclear factor κB is a pivotal transcription factor that influences cell proliferation, apoptosis, inflammation, and innate immunity. Abnormal regulation of NF-κB signaling has been linked to viral infection, tumorigenesis, autoimmune diseases, and septic shock (14). In unstimulated cells, NF-κB is bound to IκBα (the inhibitory protein of NF-κB) and retains in the cytoplasm. Several stimuli, including TNF (TNF-α), IL-1β, and TLRs, can activate NF-κB signaling by regulating downstream molecules, although the regulatory mechanisms are still not well illuminated.

Ubiquitination is an important modified mode of protein that plays critical roles in the regulation of TNF-α–induced NF-κB signaling (57). A hallmark following TNF-α stimulation is the activation of NF-κB and the transcription of target genes. The binding of TNF-α to TNFR1 leads to the recruitment of TRADD, and subsequently, TRADD recruits TRAF2, TRAF5, cIAP1, cIAP2, plus RIP1 to form a complex. At this time, these molecules may be targeted by multiple polyubiquitin chains, serving as a scaffold to recruit TAK1, TAB1, and TAB2. The active TAK1 further phosphorylates the MAPKs and IKK complex to initiate MAPK and NF-κB cascades. Next, the IKK complex (composed of IKKα, IKKβ, and NEMO) phosphorylates IκB, which results in the degradation of IκB by proteasome pathway, leading to the nuclear translocation of NF-κB (8, 9). Various types of ubiquitination are involved in the signal transduction. For example, TAB2 and TAB3 prefer to bind to K63-linked ubiquitination of RIP1, which results in the activation of TAK1 (10). TAK1 can be activated by tripartite motif (TRIM) protein 8 through K63-linked ubiquitination (11). The K63-linked ubiquitination of NEMO regulates the assembly of the IKK complex (12, 13). The degradation of IκB derives from its K48-linked ubiquitination. All evidence suggests that ubiquitination plays an important role at different levels of TNF-α–induced NF-κB signaling.

TRIM25, which is an E3-ubiquitin ligase containing a RING finger domain, a B box/coiled-coil domain, and a SPRY domain, is involved in various cellular processes, including cell proliferation and innate immunity (14). Previously, TRIM25 was reported to regulate antiviral innate immunity by delivering K63-linked polyubiquitin chains to RIG-I (15). A recent report found that TRIM25 activated RIG-I signaling relying on the isolated 2CARD of RIG-I, whereas RIPLET could activated full-length RIG-I via dual ubiquitin-dependent and -independent manners, which demonstrated the different mechanisms of RIG-I signaling activation (16). TRIM25 promoted type I IFN production by promoting the degradation of MAVS through the ubiquitin-proteasome system (17). It also has been reported that TRIM25 enhanced K63-linked ubiquitination of TRAF6 to regulate NF-κB signaling through the MDA5–MAVS antiviral signaling axis (18). Another study demonstrated that TRIM25 exerted its function through the TGF-β pathway to influence migration and invasion of cancer cells (19). But the role of TRIM25 is not clear in TNF-α–induced NF-κB signaling pathway and inflammation.

In this study, we identified TRIM25 as a positive regulator in TNF-α–induced NF-κB signaling through a luciferase assay screening system. TRIM25 potentiated the activation of NF-κB by targeting TRAF2 to promote its K63-linked ubiquitination through the RING domain and further enhanced the interaction between TRAF2 and TAK1 or IKKβ. Knockdown of TRIM25 inhibited NF-κB activation and, subsequently, resulted in the weak interaction between TRAF2 and TAK1. To our knowledge, our results have identified an unrecognized role for TRIM25 in the positive regulation of NF-κB signaling by promoting K63-linked ubiquitination of TRAF2 that will be helpful to keep gaining insight into many human diseases, including infection, inflammatory diseases, and cancer.

LPS (L4391-1 MG) and IL-1β (SRP3083) were purchased from Sigma-Aldrich. Recombinant Human TNF-α Protein (10602HNAE25) was purchased from Thermo Fisher Scientific.

HRP–anti-Flag (M2) (A8592) and anti–β-actin (A1978) were purchased from Sigma-Aldrich; HRP–anti-hemagglutinin (12013819001) was purchased from Roche Applied Science; anti-c-Myc-HRP (HT101) was purchased from TransGen Biotech; anti–p-IκBα (2859), anti-IκBα (4814), anti-IKKβ (8943), anti–phospho-IKKα/β (2697), and anti-TRIM25 (13773), TAK1 (5206) were purchased from Cell Signaling Technology; anti-rabbit–HRP IgG (sc-2004), anti-mouse IgG–HRP (sc-2005), and TRAF2 (sc-876, sc-136999) were purchased from Santa Cruz Biotechnology.

Empty vector pcDNA3.1 was from Cui Lab at Sun Yat-sen University. TRIM25, TRAF2, TRAF5, TAK1, TAB1, TAB2, IKKβ, and NEMO genes were cloned from A549 cDNA and subcloned into the pcDNA3.1 vector. The plasmids were confirmed by DNA sequencing at Sangon Biotech.

Human embryonic kidney 293T (HEK293T) cells, HeLa cells, TRAF6-knockout (KO) HEK293T, PBMCs, and THP-1 cells were cultured in DMEM (HyClone) or RPMI 1640 (Life Technologies) with 10% FBS (Genestar), and 1% l-glutamine (Life Technologies) at 37°C in 5% CO2. HEK293T, HeLa, and THP-1 cells were provided from Dr. X.-F. Qin’s laboratory (Sun Yat-sen University). TRAF6-KO HEK293T cells were kindly provided by Dr. J. Cui (Sun Yat-sen University). Expression plasmids were transfected with Lipofectamine 2000 (Invitrogen), according to the manufacturer’s instructions.

HEK293T cells were plated in 24-well plates at a density of 6 × 104 cells per well. After overnight incubation, the cells were cotransfected with plasmids encoding NF-κB luciferase reporter (30 ng per well) and the internal control Renilla luciferase (10 ng per well) together with 10–25 ng of empty vector as the control or plasmids encoding HA-TRIM25, Flag-TRAF2, Flag-TRAF5, Flag-TAK1, HA-TAB1, Flag-IKKβ, Flag-P65, and other plasmids with indicated target genes. After 24 h of transfection, the cells were treated with TNF-α for 6 h, and these cells were harvested in passive lysis buffer (Promega). The cell lysates were measured using the Dual-Luciferase Assay Kit according to the manufacturer’s protocol (Promega). All the assays were performed in triplicate and repeated a minimum of three times.

HEK293T cells were plated in 24-well plates at a density of 6 × 104 cells per well. Cells were transfected with target plasmids using Lipofectamine 2000 according to the manufacturer’s instructions and extracted in 120 μl of low-salt lysis buffer (50 mM HEPES [pH 7.5], 150 mM NaCl, 1 mM EDTA, 1.5 mM MgCl2, 10% glycerol, 1% Triton X-100) supplemented with 5 mg/ml protease inhibitor (Thermo Fischer Scientific) and Phosphatase Inhibitor Cocktail (Roche), then samples of 20-μl total proteins were subjected to SDS-PAGE.

For immunoprecipitation (IP) experiments, whole cell extracts were prepared after transfection or stimulation with ligands as the method of immunoblot assay, followed by incubation with the anti-Flag agarose gels (Sigma-Aldrich) overnight at 4°C. The beads were washed five times with low-salt lysis buffer, resuspended with 2× SDS Loading Buffer (FD Biotechnology), and boiled for 5 min. The released proteins were subjected to SDS-PAGE and transferred onto PVDF membranes with subsequent blocking using 5% skim milk. Membranes were incubated with specific Abs and detected using chemiluminescence (Millipore).

Cells were treated with target plasmids or small interfering RNAs (siRNAs) as indicated, then stimulated by ligands in 24-well plates. The total cellular RNA was isolated by TRIzol Reagent (Invitrogen) according to the manufacturer’s protocol, and the first-strand cDNA was synthesized from total RNA using a PrimeScript 1st strand cDNA Synthesis Kit (Takara Bio). Real-time PCR (RT-PCR) was performed with SYBR Green qPCR Master Mix (Takara Bio). IL-1β, IL-6, and TNF-α induced by NF-κB were analyzed by RT-PCR at 94°C for 5 min, followed by 40 cycles at 94°C for 20 s, at 55°C for 20 s, at 72°C for 20 s, and finally, at 72°C for 10 min. The following specific primers were used for RT-PCR: hGAPDH forward primer, 5′-ACAACTTTGGTATCGTGGAAGG-3′ and hGAPDH reverse primer, 5′-GCCATCACGCCACAGTTTC-3′; hIL-1β forward primer, 5′-ATGATGGCTTATTACAGTGGCAA-3′ and hIL-1β reverse primer, 5′-GTCGGAGATTCGTAGCTGGA-3′; hIL-6 forward primer, 5′-AGAGGCACTGGCAGAAAACAAC-3′ and hIL-6 reverse primer, 5′-AGGCAAGTCTCCTCATTGAATCC-3′; hTNF-α forward primer, 5′-CCAGACCAAGGTCAACCTCC-3′ and hTNF-α reverse primer, 5′-CAGACTCGGCAAAGTCGAGA-3′; TRIM25 forward primer, 5′-AGCAGCTACAACAAGAATACACG-3′ and TRIM25 reverse primer, 5′-GGCTCTGTTCAATCTCCTCCT-3′.

LipoRNAiMax (Invitrogen) was used according to the manufacturer’s protocols for transfection of siRNAs into HEK293T cells, HeLa cells, THP-1 cells, or PBMCs. The sequences of TRIM25 siRNAs are as follows: human TRIM25-specific siRNA 1) sense sequencing (SenseSeq): 5′-AGGUCCACCUGAUGUAUAATT-3′ and antisense sequencing (AntiSeq): 5′-UUAUACAUCAGGUGGACCUTT-3′; 2) SenseSeq: 5′-CCCUGAGGCACAAACUAACTT-3′ and AntiSeq: 5′-GUUAGUUUGUGCCUCAGGGTG-3′; and 3) SenseSeq: 5′-GCAAAUGUUCCCAGCACAATT-3′ and AntiSeq: 5′-UUGUGCUGGGAACAUUUGCTT-3′.

Data were compared between the different test groups using a Student t test and GraphPad Prism 5.0 Software. All the experiments were repeated at least three times independently, and the differences between groups were considered significant when *p < 0.05, **p < 0.01, and ***p < 0.001.

To investigate the roles of TRIM family proteins in NF-κB signaling induced by TNF-α, we screened the function of a panel of TRIM proteins using luciferase assays. We transfected HEK293T cells with an NF-κB luciferase reporter and an internal control Renilla thymidine kinase luciferase, as well as one of the candidate genes encoding TRIM, and stimulated them with TNF-α. Notably, TRIM8, TRIM13, and TRIM38, which were previously reported to positively or negatively modulate NF-κB signaling by targeting TAK1, NEMO, or TAB2/3, were identified as hits in the screening (Fig. 1A), thus validating this experimental approach (11, 20, 21). These data also led to the identification of TRIM25 as a potent activator of NF-κB signaling. To further determine whether TRIM25 functions in TLR- and/or cytokine-mediated NF-κB activation, we transfected HEK293T or HEK293T/TLR4 cells with an NF-κB luciferase reporter and the internal control Renilla luciferase, as well as an increasing amount of TRIM25 plasmid, and then stimulated the cells for 6 h with TNF-α, IL-1β, or LPS. As shown in Fig. 1B–D, TRIM25 potently activated NF-κB activation induced by TNF-α, IL-1β, or LPS in a dose-dependent manner. It is reported that TRIM25 can interact TRAF6 and enhance its K63-linked ubiquitination, so we speculated that TRIM25 might regulate IL-1β– or LPS-mediated NF-κB signaling through TRAF6 (18). Next, we used TNF-α to treat TRAF6 KO cells and found TRIM25 also enhances TNF-α–induced NF-κB signaling (Fig. 1E), indicating that TRIM25 activated TNF-α–induced NF-κB in a TRAF6-independent manner.

FIGURE 1.

TRIM25 is a potent activator of NF-κB signaling. (A) HEK293T cells were transfected with an empty vector (Ctrl) or plasmids of 15 TRIMs along with NF-κB luciferase reporter and the internal control Renilla luciferase, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The cells were analyzed for NF-κB activity by luciferase reporter assay. (BE) HEK293T or HEK293T/TLR4 or HEK293T/TRAF6-KO cells were transfected with an NF-κB luciferase reporter and the internal control Renilla luciferase, as well as an empty vector or an increasing amount (wedge) of TRIM25 plasmid, and mock stimulated or stimulated the cells for 6 h with IL-1β (B) (10 ng/ml), TNF-α (C and E) (5 ng/ml) or LPS (D) (100 ng/ml). The cells were analyzed for NF-κB activity by luciferase reporter assay. (F) HEK293T cells were transfected with NF-κB luciferase reporter and the internal control Renilla luciferase along with no-tagged or differently tagged TRIM25 expression plasmids, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The cells were analyzed for NF-κB activity by a reporter gene assay. (G) HEK293T cells were transfected with an empty vector or HA-TRIM25 and treated by TNF-α (2 ng/ml) for 0, 15, 30, or 60 min. IκBα turnover was detected by Western blotting with indicated Abs. (H) HeLa cells were transfected with an empty vector or HA-TRIM25, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 8 h. The total mRNA was harvested, and IL-1β, IL-6, and TNF-α mRNA abundance were analyzed by quantitative real-time PCR. The empty vector (pcDNA3.1) used to clone TRIM25 was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, Student t test.

FIGURE 1.

TRIM25 is a potent activator of NF-κB signaling. (A) HEK293T cells were transfected with an empty vector (Ctrl) or plasmids of 15 TRIMs along with NF-κB luciferase reporter and the internal control Renilla luciferase, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The cells were analyzed for NF-κB activity by luciferase reporter assay. (BE) HEK293T or HEK293T/TLR4 or HEK293T/TRAF6-KO cells were transfected with an NF-κB luciferase reporter and the internal control Renilla luciferase, as well as an empty vector or an increasing amount (wedge) of TRIM25 plasmid, and mock stimulated or stimulated the cells for 6 h with IL-1β (B) (10 ng/ml), TNF-α (C and E) (5 ng/ml) or LPS (D) (100 ng/ml). The cells were analyzed for NF-κB activity by luciferase reporter assay. (F) HEK293T cells were transfected with NF-κB luciferase reporter and the internal control Renilla luciferase along with no-tagged or differently tagged TRIM25 expression plasmids, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The cells were analyzed for NF-κB activity by a reporter gene assay. (G) HEK293T cells were transfected with an empty vector or HA-TRIM25 and treated by TNF-α (2 ng/ml) for 0, 15, 30, or 60 min. IκBα turnover was detected by Western blotting with indicated Abs. (H) HeLa cells were transfected with an empty vector or HA-TRIM25, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 8 h. The total mRNA was harvested, and IL-1β, IL-6, and TNF-α mRNA abundance were analyzed by quantitative real-time PCR. The empty vector (pcDNA3.1) used to clone TRIM25 was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, Student t test.

Close modal

Next, we used no-tagged or differently tagged TRIM25 to determine the function of TRIM25, and the luciferase assay showed that they had the same function in activating TNF-α–mediated NF-κB–luc activation (Fig. 1F). More importantly, we found that TRIM25 significantly enhanced the degradation of endogenous IκBα protein in the presence of TNF-α (Fig. 1G). In addition, we found that TNF-α treatment resulted in higher expression levels of TNF-α, IL-6, or IL-1β mRNA in HeLa cells with TRIM25-overexpression than in those transfected with empty vector (Fig. 1H). Taken together, these results suggest that TRIM25 activates TNF-α–induced NF-κB activation.

We next detected whether endogenous TRIM25 was involved in the regulation of TNF-α–mediated NF-κB signaling. We generated three pairs of siRNAs specific for TRIM25, all of which efficiently inhibited the expression of endogenous TRIM25 in protein and mRNA levels (Fig. 2A, 2B). As expected, we found that treatment with TNF-α resulted in much lower p-IKK and p-IκBα in HEK293T cells transfected with TRIM25-specific siRNA than in those transfected with control siRNA (Fig. 2C). In RT-PCR assays, we found that TNF-α treatment resulted in lower expression levels of TNF-α, IL-6, or IL-1β mRNA in TRIM25-deficient HeLa cells or PBMCs (Fig. 2D, 2E). Hence, these data suggest knockdown of TRIM25 inhibits NF-κB activity.

FIGURE 2.

Knockdown of TRIM25 inhibits NF-κB activation. (A) The knockdown efficiency of TRIM25-specific siRNAs in protein level. Immunoblot analysis of the knockdown of endogenous TRIM25 in HEK293T cells treated with three pairs of TRIM25-specific siRNAs or control (Ctrl) siRNA (40 nM). β-actin serves as a loading control. (B) The knockdown efficiency of TRIM25-specific siRNAs in mRNA level. The quantitative real-time PCR analysis of endogenous TRIM25 in HEK293T cells treated with TRIM25-specific siRNAs or control siRNA (40 nM). (C) HEK293T cells were transfected with control siRNA or TRIM25-specific siRNA and treated with TNF-α (2 ng/ml) for the indicated time points. TNF-α–induced IKK activation was measured by immunoblotting with indicated Abs. (D and E) HeLa cells (D) or PBMCs (E) were transfected with control siRNA or TRIM25-specific siRNAs for 36 h, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 8 h. The total mRNA was harvested, and IL-1β, IL-6, and TNF-α mRNAs abundance was analyzed by quantitative real-time PCR. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, ***p < 0.001, Student t test.

FIGURE 2.

Knockdown of TRIM25 inhibits NF-κB activation. (A) The knockdown efficiency of TRIM25-specific siRNAs in protein level. Immunoblot analysis of the knockdown of endogenous TRIM25 in HEK293T cells treated with three pairs of TRIM25-specific siRNAs or control (Ctrl) siRNA (40 nM). β-actin serves as a loading control. (B) The knockdown efficiency of TRIM25-specific siRNAs in mRNA level. The quantitative real-time PCR analysis of endogenous TRIM25 in HEK293T cells treated with TRIM25-specific siRNAs or control siRNA (40 nM). (C) HEK293T cells were transfected with control siRNA or TRIM25-specific siRNA and treated with TNF-α (2 ng/ml) for the indicated time points. TNF-α–induced IKK activation was measured by immunoblotting with indicated Abs. (D and E) HeLa cells (D) or PBMCs (E) were transfected with control siRNA or TRIM25-specific siRNAs for 36 h, then mock stimulated or stimulated with TNF-α (5 ng/ml) for 8 h. The total mRNA was harvested, and IL-1β, IL-6, and TNF-α mRNAs abundance was analyzed by quantitative real-time PCR. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, ***p < 0.001, Student t test.

Close modal

To determine the molecular mechanisms by which TRIM25 potentiates TNF-α–induced NF-κB signaling, we transfected HEK293T cells with TRAF2, TRAF5, TAK1-TAB1, IKKβ, or p65 subunit together with TRIM25 plus the NF-κB luciferase reporter. We found that the activation of NF-κB by TRAF2 and TRAF5 was markedly enhanced by TRIM25 (Fig. 3A). In contrast, TRIM25 did not increase TAK1-TAB1–, IKKβ-, or p65-mediated NF-κB activation (Fig. 3A), suggesting that TRIM25 potentiates the NF-κB pathway upstream of TAK1, most likely targeting the TRAF complex. Consistent with these results, we found that knockdown of TRIM25 inhibited NF-κB luciferase activity induced by TRAF2 and TRAF5 but not TAK1-TAB1, IKKβ, or p65 (Fig. 3B). These results suggest that TRIM25 enhances NF-κB signaling upstream of TAK1, at the level of the TRAF complex.

FIGURE 3.

TRIM25 enhances NF-κB signaling at the level of the TRAF2–TRAF5 complex. (A) TRIM25 promoted NF-κB activation induced by NF-κB pathway downstream signaling molecules. HEK293T cells transfected with an NF-κB luciferase reporter, together with an empty vector, TRAF2, TRAF5, TAK1-TAB1, IKKβ, or P65, along with an empty vector or plasmid of TRIM25. (B) Knockdown of endogenous TRIM25 inhibited TRAF2- or TRAF5-mediated NF-κB activation. Luciferase activity in HEK293T cells transfected with TRIM25-specific siRNA or control siRNA (40 nM) and transfected with an empty vector, TRAF2, TRAF5, TAK1-TAB1, IKKβ, or P65 together with an NF-κB luciferase reporter. The empty vector (pcDNA3.1) used to clone TRIM25 was transfected as the “–.” All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. ***p < 0.001, Student t test.

FIGURE 3.

TRIM25 enhances NF-κB signaling at the level of the TRAF2–TRAF5 complex. (A) TRIM25 promoted NF-κB activation induced by NF-κB pathway downstream signaling molecules. HEK293T cells transfected with an NF-κB luciferase reporter, together with an empty vector, TRAF2, TRAF5, TAK1-TAB1, IKKβ, or P65, along with an empty vector or plasmid of TRIM25. (B) Knockdown of endogenous TRIM25 inhibited TRAF2- or TRAF5-mediated NF-κB activation. Luciferase activity in HEK293T cells transfected with TRIM25-specific siRNA or control siRNA (40 nM) and transfected with an empty vector, TRAF2, TRAF5, TAK1-TAB1, IKKβ, or P65 together with an NF-κB luciferase reporter. The empty vector (pcDNA3.1) used to clone TRIM25 was transfected as the “–.” All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. ***p < 0.001, Student t test.

Close modal

Next, we sought to determine whether TRIM25 could directly interact with the TRAF complex or other signaling proteins within the NF-κB signaling pathway. Coimmunoprecipitation experiments revealed that TRIM25 specifically interacted with TRAF2 rather than the TRAF5 or TAK1 complex (Fig. 4A, 4B). To determine the interaction, we treated HeLa cells with TNF-α and then collected the cell lysates. IP and immunoblot analysis revealed that TRIM25 only weakly interacted with TRAF2 in unstimulated cells. However, TRIM25 strongly interacted with TRAF2 after TNF-α treatment (Fig. 4C). We also found that endogenous TRIM25 interacted with TRAF2 in unstimulated THP-1 cells and PBMCs, and the treatment of TNF-α could enhance their interaction (Fig. 4D, 4E). To identify the domain of TRIM25 responsible for its interaction with TRAF2, we generated three TRIM25 deletion constructs (Fig. 4F), and we found that only ∆RING domain (KD) interacted with TRAF2 (Fig. 4G). It suggests the SPRY domain of TRIM25 is critical for binding to TRAF2. In addition, we found NF-κB activation, as assessed by a luciferase assay, was increased in the cells transfected with TRIM25 (wild type [WT]) but not its deletion constructs (Fig. 4H). It should be noted that both the RING and SPRY domain were critical for TRIM25-mediated enhancement of TNF-α–induced NF-κB signaling.

FIGURE 4.

TRIM25 interacts with TRAF2. (A and B) TRIM25 interacted with TRAF2. Coimmunoprecipitation and immunoblot analysis of HEK293T cells transfected with various combinations of plasmids, including an empty vector, Flag-TRAF2, Flag-TRAF5 (A), Flag-TAK1, Flag-TAB1, Flag-TAB2 (B), and HA-TRIM25. Flag-tagged proteins were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab and whole cell lysates (WCL). (C) TRIM25 interacted with TRAF2. HeLa cells were transfected with an empty vector or Flag-TRIM25 and mock treated or treated by TNF-α (5 ng/ml) for 2 h. Flag-tagged TRIM25 was immunoprecipitated with anti-Flag beads and blotted with TRAF2. (D and E) THP-1 cells (D) or PBMCs (E) were treated by TNF-α (5 ng/ml) for 2 h. They were immunoprecipitated with endogenous TRAF2 and blotted with TRIM25 in cell lysates. (F) The domain structure of TRIM25. Numbers indicated the amino acid position in construct. (G) The interaction between HA-TRAF2 and Flag-TRIM25 domains. HEK293T cells transfected with HA-TRAF2 and an empty vector, Flag-TRIM25, or Flag-TRIM25 domains. Whole cell extracts were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab. (H) The activation of TRIM25 in NF-κB signaling depended on RING and SPRY domains. Luciferase activity of HEK293T cells transfected with plasmids of NF-κB luciferase reporter and an empty vector, Flag-TRIM25, Flag-TRIM25 B Box/CCD-SPRY (ΔRING), Flag-TRIM25 RING-B Box/CCD (ΔSPRY), or Flag-TRIM25 B Box/CCD (BBOX), then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The B Box/CCD was the B box/coiled-coil domain of TRIM25. The empty vector (pcDNA3.1) used to subclone was transfected as the “–” in all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, Student t test.

FIGURE 4.

TRIM25 interacts with TRAF2. (A and B) TRIM25 interacted with TRAF2. Coimmunoprecipitation and immunoblot analysis of HEK293T cells transfected with various combinations of plasmids, including an empty vector, Flag-TRAF2, Flag-TRAF5 (A), Flag-TAK1, Flag-TAB1, Flag-TAB2 (B), and HA-TRIM25. Flag-tagged proteins were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab and whole cell lysates (WCL). (C) TRIM25 interacted with TRAF2. HeLa cells were transfected with an empty vector or Flag-TRIM25 and mock treated or treated by TNF-α (5 ng/ml) for 2 h. Flag-tagged TRIM25 was immunoprecipitated with anti-Flag beads and blotted with TRAF2. (D and E) THP-1 cells (D) or PBMCs (E) were treated by TNF-α (5 ng/ml) for 2 h. They were immunoprecipitated with endogenous TRAF2 and blotted with TRIM25 in cell lysates. (F) The domain structure of TRIM25. Numbers indicated the amino acid position in construct. (G) The interaction between HA-TRAF2 and Flag-TRIM25 domains. HEK293T cells transfected with HA-TRAF2 and an empty vector, Flag-TRIM25, or Flag-TRIM25 domains. Whole cell extracts were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab. (H) The activation of TRIM25 in NF-κB signaling depended on RING and SPRY domains. Luciferase activity of HEK293T cells transfected with plasmids of NF-κB luciferase reporter and an empty vector, Flag-TRIM25, Flag-TRIM25 B Box/CCD-SPRY (ΔRING), Flag-TRIM25 RING-B Box/CCD (ΔSPRY), or Flag-TRIM25 B Box/CCD (BBOX), then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. The B Box/CCD was the B box/coiled-coil domain of TRIM25. The empty vector (pcDNA3.1) used to subclone was transfected as the “–” in all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, Student t test.

Close modal

Because the RING domain of TRIM25 is critical for its function (Fig. 4H), we examined whether the E3 ligase activity of TRIM25 was essential for its function. An enzymatically inactive mutant, TRIM25-CS (C53S), could not facilitate NF-κB reporter activation (Fig. 5A), suggesting that TRIM25 may affect the NF-κB pathways through its E3 ligase activity. Next, we tested whether TRIM25 affected the ubiquitination of TRAF2. We observed that TRIM25 remarkably enhanced the ubiquitination of TRAF2 rather than TRIM25-CS mutant or TRIM25 ΔRING domain (Fig. 5B), and knockdown of TRIM25 limited the ubiquitination of TRAF2 (Fig. 5C). Next, we want to know which kinds of TRAF2 ubiquitination can be affected by TRIM25. Our findings revealed that TRIM25 only enhanced K63-ubiquitination of TRAF2 but not others (Fig. 5D). This finding was further confirmed in the knockdown system (Fig. 5E). Collectively, these results suggest that TRIM25 enhances the K63-linked ubiquitination of TRAF2 using its E3 ligase activity.

FIGURE 5.

TRIM25 enhances the K63-linked ubiquitination of TRAF2. (A) TRIM25 enhanced NF-κB activation depending on its E3 ubiquitin ligase activity. HEK293T cells were transfected with an NF-κB luciferase reporter, along with empty vector or plasmids for Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S), then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. (B) TRIM25 but not its inactive mutants enhanced the ubiquitination of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged ubiquitin and empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S). They have proceeded to IP and immunoblot analysis with anti-Flag beads and were analyzed with anti-HA Ab by immunoblot. (C) Knockdown of TRIM25 reduced the ubiquitination of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged ubiquitin and TRIM25-specific siRNA, control siRNA, or Myc-TRIM25 (WT). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. (D) TRIM25 enhanced the K63-linked polyubiquitin on TRAF2. HEK293T cells were transfected with plasmids of Flag-TRAF2 and empty vector or Myc-TRIM25, together with HA-tagged ubiquitin, HA-tagged K6, K11, K27, K29, K33, K48, or K63 ubiquitin mutants. Whole cell extracts were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab. (E) Knockdown of TRIM25 reduced the K63-linked ubiquitin chains of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged K63 ubiquitin, along with TRIM25-specific siRNA or control siRNA (40 nM). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. The empty vector (pcDNA3.1) used to subclone was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. ***p < 0.001, Student t test.

FIGURE 5.

TRIM25 enhances the K63-linked ubiquitination of TRAF2. (A) TRIM25 enhanced NF-κB activation depending on its E3 ubiquitin ligase activity. HEK293T cells were transfected with an NF-κB luciferase reporter, along with empty vector or plasmids for Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S), then mock stimulated or stimulated with TNF-α (5 ng/ml) for 6 h. (B) TRIM25 but not its inactive mutants enhanced the ubiquitination of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged ubiquitin and empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S). They have proceeded to IP and immunoblot analysis with anti-Flag beads and were analyzed with anti-HA Ab by immunoblot. (C) Knockdown of TRIM25 reduced the ubiquitination of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged ubiquitin and TRIM25-specific siRNA, control siRNA, or Myc-TRIM25 (WT). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. (D) TRIM25 enhanced the K63-linked polyubiquitin on TRAF2. HEK293T cells were transfected with plasmids of Flag-TRAF2 and empty vector or Myc-TRIM25, together with HA-tagged ubiquitin, HA-tagged K6, K11, K27, K29, K33, K48, or K63 ubiquitin mutants. Whole cell extracts were immunoprecipitated with anti-Flag beads and blotted with anti-HA Ab. (E) Knockdown of TRIM25 reduced the K63-linked ubiquitin chains of TRAF2. HEK293T cells were transfected with Flag-TRAF2, HA-tagged K63 ubiquitin, along with TRIM25-specific siRNA or control siRNA (40 nM). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. The empty vector (pcDNA3.1) used to subclone was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. ***p < 0.001, Student t test.

Close modal

After TNF-α treatment, TRAF2 could recruit TAK1 and IKK complex to influence NF-κB signaling, and TRIM25 targeted TRAF2 through enhancement of its ubiquitination. Therefore, we reasoned that TRIM25 may bridge the TRAF2 and TAK1 or the IKK complex. As expected, we found that TRIM25 markedly enhanced the interaction between TRAF2 and TAK1 or IKKβ but did not affect the IKK complex (Fig. 6A–C). TRIM25 deficiency blocked the interaction between TRAF2 and TAK1 (Fig. 6D). Next, we found that knockdown of TRIM25 reduced the endogenous interaction between TRAF2 and TAK1 after TNF-α stimulation (Fig. 6E). We further found that TRIM25-∆RING or -CS mutant failed to bridge TRAF2 to TAK1 (Fig. 6F) or to facilitate TRAF2-mediated TAK1 activation (Fig. 6G). Collectively, these results indicated that the K63-linked ubiquitination of TRAF2 mediated by TRIM25 provided the platform to bridge TRAF2 to TAK1 or IKK complex.

FIGURE 6.

TRIM25 enhances the interaction between TRAF2 and TAK1 or IKKβ. (AC) HEK293T cells are transfected with different combinations of plasmids for HA-TAK1, HA-IKKβ, Flag-TRAF2, Flag-NEMO and Myc-TRIM25 or empty vector. The lysates have proceeded to IP and immunoblot analysis. (D) TRIM25 deficiency blocked the interaction between TRAF2 and TAK1. HEK293T cells were transfected with HA-TAK1 and Flag-TRAF2, along with TRIM25-specific siRNA or control siRNA (40 nM). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. (E) THP-1 cells were transfected with control siRNA or TRIM25-specific siRNAs (40 nM), then mock stimulated or stimulated the cells with TNF-α (5 ng/ml) for 2 h. IP of endogenous TRAF2 detected the association with TAK1 in THP-1 cell lysates. (F) TRIM25 inactive mutants reduced the interaction between TRAF2 and TAK1. HEK293T cells were transfected with HA-TAK1 and Flag-TRAF2, along with empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S) and have proceeded to IP and immunoblot analysis. (G) TRIM25 facilitated TRAF2-TAK1–mediated NF-κB activation. HEK293T cells were transfected with an NF-κB luciferase reporter, together with the vector encoding Flag-TRAF2 and HA-TAK1 plus HA-TAB1, along with empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S). The empty vector (pcDNA3.1) used to subclone was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, ***p < 0.001, Student t test.

FIGURE 6.

TRIM25 enhances the interaction between TRAF2 and TAK1 or IKKβ. (AC) HEK293T cells are transfected with different combinations of plasmids for HA-TAK1, HA-IKKβ, Flag-TRAF2, Flag-NEMO and Myc-TRIM25 or empty vector. The lysates have proceeded to IP and immunoblot analysis. (D) TRIM25 deficiency blocked the interaction between TRAF2 and TAK1. HEK293T cells were transfected with HA-TAK1 and Flag-TRAF2, along with TRIM25-specific siRNA or control siRNA (40 nM). Flag-tagged proteins were immunoprecipitated with anti-Flag beads and analyzed with anti-HA Ab by immunoblot. (E) THP-1 cells were transfected with control siRNA or TRIM25-specific siRNAs (40 nM), then mock stimulated or stimulated the cells with TNF-α (5 ng/ml) for 2 h. IP of endogenous TRAF2 detected the association with TAK1 in THP-1 cell lysates. (F) TRIM25 inactive mutants reduced the interaction between TRAF2 and TAK1. HEK293T cells were transfected with HA-TAK1 and Flag-TRAF2, along with empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S) and have proceeded to IP and immunoblot analysis. (G) TRIM25 facilitated TRAF2-TAK1–mediated NF-κB activation. HEK293T cells were transfected with an NF-κB luciferase reporter, together with the vector encoding Flag-TRAF2 and HA-TAK1 plus HA-TAB1, along with empty vector, Myc-TRIM25 (WT), Myc-TRIM25 (ΔRING), or Myc-TRIM25 (C53S). The empty vector (pcDNA3.1) used to subclone was transfected as the “–” for all dates. All results were representative of three independent experiments. The graphs showed the mean ± SD of three independent experiments. **p < 0.01, ***p < 0.001, Student t test.

Close modal

TNF-α, as one of the pleiotropic cytokines that activate NF-κB signaling, leads to the production of several proinflammatory cytokines, such as IL-1β, IL-6, and TNF-α, and further promotes inflammation and immune responses. Increasing evidence indicates that dysregulation of NF-κB leads to several inflammation-associated diseases. Further, proinflammatory cytokines secreted by innate immune cells have been shown to play a pivotal role in tumor development and progression in chronic inflammation conditions, so it is critical for the healthy host to tightly regulate TNF-α–induced NF-κB pathway (1, 3). Our study identified TRIM25 as a positive regulator in TNF-α–induced NF-κB signaling pathway, which might shed light on identifying a potential therapeutic target for inflammation-associated diseases and tumors in the future.

The posttranslation modification of target proteins is important in the regulation of the NF-κB pathway. Increasing numbers of proteins that contain many members of TRIM family were reported to participate in the regulation of the TNF-α–induced NF-κB pathway. Just as reported, TRIM8 associates with TAK1, enhances the binding of K63-linked polyubiquitination to TAK1, and positively regulates TNF-α– and IL-1β–triggered NF-κB activation (11). TRIM13 regulates ubiquitination and turnover of NEMO at the level of IKK complex to suppress the TNF-α–induced NF-κB pathway and also potentiates TLR2-mediated NF-κB activation via K29-linked polyubiquitination of TRAF2 (20, 22). TRIM12c, as a mouse homologue of TRIM5, forms a complex with TRAF6 to enhance the activation of the IFN and NF-κB pathways (23). After stimulation by TNF-α, TRIM22, TRIM38, and TRIM39 inhibit NF-κB pathways by targeting different proteins through different manners of ubiquitination (21, 24, 25). There are still many proteins, such as TRIM45, TRIM52, RACK1, and so on, that have regulated functions in TNF-α–induced NF-κB pathway (2628). The involvement of so many molecules in the regulation of NF-κB signaling suggests the importance of maintaining its fine regulation. That is to say, the complicated regulatory network of NF-κB signaling is required to ensure an appropriate immune response.

TRIM25, a member of the TRIM proteins family, is involved in cell proliferation, innate immunity, and cancer. It was reported that TRIM25 affected RIG-I–mediated antiviral activity by delivering the K63-linked ubiquitin moiety to the N-terminal CARDs of RIG-I or releasing NEMO and TBK1 complex into the cytosol through the proteasome degradation of scaffold protein MAVS (17, 18). However, there was no report about TRIM25 directly regulating TNF-α–induced NF-κB signaling. Unlike TRIM25 regulates NF-κB signaling by TRAF6 through MDA5-MAVS antiviral signaling axis (18), we found TRIM25 facilitated NF-κB activity induced by TNF-α, TRAF2, and TRAF5 but not TAK1-TAB1, IKKβ, or p65. Also, it enhanced the degradation of IκBα and the transcription of proinflammatory cytokines, such as IL-1β, IL-6, and TNF-α, in response to TNF-α treatment. Immunoprecipitation analyses showed that TRIM25 directly interacted with TRAF2 but not with TRAF5 or TAK1 complex and increased the K63-linked ubiquitination of TRAF2. Conversely, knockdown of TRIM25 reduced the ubiquitination reaction to TRAF2. We also confirmed the endogenous interaction of TRAF2 and TRIM25 by IP analyses in THP-1 cells and PBMCs, and the interaction was further enhanced with the treatment of TNF-α. To identify the detailed mechanism that TRIM25 potentiates TNF-α–induced NF-κB signaling by targeting TRAF2, we confirmed TRIM25 reinforced the interaction between TRAF2 and TAK1 or IKKβ; it merely had no function to the interaction between IKKβ and NEMO. The enhanced interaction potentiated the activation of TAK1 and IKKβ to heighten TNF-α–induced NF-κB signaling, whereas TRIM25-∆RING or TRIM25-CS mutant could affect the interaction of TRAF2 and TAK1, and knockdown of TRIM25 reduced the endogenous interaction between TRAF2 and TAK1 after TNF-α stimulation. In this context, it has been reported that TRIM8 upregulates TNF-α– and IL-1β–triggered NF-κB signaling by targeting TAK1. USP18 inhibits NF-κB signaling by targeting TAK1 and NEMO (11, 29). From this study, we also found that TRIM25 had the interaction with itself by IP, which might be helpful to its stabilization, and then strengthened immune reaction. Previous studies showed that TRAF2 participated in the apoptosis pathway, just as TRAF2 is a negative regulator of TNF-α–induced apoptosis (30), RIPK1 inhibits LPS/TNF–mediated apoptosis through a TRAF2- and caspase-8–dependent pathway (31). Hence TRIM25 may influence the apoptosis pathway by targeting TRAF2, which may link the cross-talk between the NF-κB and apoptosis signaling pathway for a further study.

In conclusion, to our knowledge, the evidence in this study identified an undiscovered role for TRIM25 in the regulating of TNF-α–induced NF-κB signaling. In the current study, TRIM25 mainly acted at the level of TRAF2 by K63-linked ubiquitination, then strengthened the interaction between TRAF2 and TAK1 or IKKβ. Thus, our findings gain an insight into the molecular mechanisms by which TRIM25 upregulates NF-κB signaling and provide a potential therapeutic target for inflammatory diseases and cancers in the future.

We thank Dr. Xiao-Feng Qin and Dr. Jun Cui for providing plasmids and reagents as a gift. We also thank Dr. Weicheng Liang for providing language support.

This work was supported by the National Natural Science Foundation of China (81970509, 31601135, and 31970419), and Scientific and Technological Innovation Leaders in Central Plains (194200510002). Y.Q. is partially supported by the Startup Research Fund of Sun-Yet Sen University.

Abbreviations used in this article:

AntiSeq

antisense sequencing

HEK293T

human embryonic kidney 293T

IP

immunoprecipitation

KO

knockout

RT-PCR

real-time PCR

SenseSeq

sense sequencing

siRNA

small interfering RNA

TRIM

tripartite motif

WT

wild type.

1
Hayden
,
M. S.
,
S.
Ghosh
.
2011
.
NF-κB in immunobiology.
Cell Res.
21
:
223
244
.
2
Oeckinghaus
,
A.
,
M. S.
Hayden
,
S.
Ghosh
.
2011
.
Crosstalk in NF-κB signaling pathways.
Nat. Immunol.
12
:
695
708
.
3
Liang
,
Y.
,
Y.
Zhou
,
P.
Shen
.
2004
.
NF-kappaB and its regulation on the immune system.
Cell. Mol. Immunol.
1
:
343
350
.
4
Ghosh
,
S.
,
M. S.
Hayden
.
2008
.
New regulators of NF-kappaB in inflammation.
Nat. Rev. Immunol.
8
:
837
848
.
5
Acconcia
,
F.
,
S.
Sigismund
,
S.
Polo
.
2009
.
Ubiquitin in trafficking: the network at work.
Exp. Cell Res.
315
:
1610
1618
.
6
Ebner
,
P.
,
G. A.
Versteeg
,
F.
Ikeda
.
2017
.
Ubiquitin enzymes in the regulation of immune responses.
Crit. Rev. Biochem. Mol. Biol.
52
:
425
460
.
7
Gallo
,
L. H.
,
J.
Ko
,
D. J.
Donoghue
.
2017
.
The importance of regulatory ubiquitination in cancer and metastasis.
Cell Cycle
16
:
634
648
.
8
Baldwin
,
A. S.
 Jr
.
1996
.
The NF-kappa B and I kappa B proteins: new discoveries and insights.
Annu. Rev. Immunol.
14
:
649
683
.
9
Vallabhapurapu
,
S.
,
M.
Karin
.
2009
.
Regulation and function of NF-kappaB transcription factors in the immune system.
Annu. Rev. Immunol.
27
:
693
733
.
10
Chen
,
Z. J.
2005
.
Ubiquitin signalling in the NF-kappaB pathway.
Nat. Cell Biol.
7
:
758
765
.
11
Li
,
Q.
,
J.
Yan
,
A. P.
Mao
,
C.
Li
,
Y.
Ran
,
H. B.
Shu
,
Y. Y.
Wang
.
2011
.
Tripartite motif 8 (TRIM8) modulates TNFα- and IL-1β-triggered NF-κB activation by targeting TAK1 for K63-linked polyubiquitination.
Proc. Natl. Acad. Sci. USA
108
:
19341
19346
.
12
Chen
,
Z. J.
2012
.
Ubiquitination in signaling to and activation of IKK.
Immunol. Rev.
246
:
95
106
.
13
Wertz
,
I. E.
,
V. M.
Dixit
.
2010
.
Signaling to NF-kappaB: regulation by ubiquitination.
Cold Spring Harb. Perspect. Biol.
2
: a003350.
14
Nisole
,
S.
,
J. P.
Stoye
,
A.
Saïb
.
2005
.
TRIM family proteins: retroviral restriction and antiviral defence.
Nat. Rev. Microbiol.
3
:
799
808
.
15
Gack
,
M. U.
,
Y. C.
Shin
,
C. H.
Joo
,
T.
Urano
,
C.
Liang
,
L.
Sun
,
O.
Takeuchi
,
S.
Akira
,
Z.
Chen
,
S.
Inoue
,
J. U.
Jung
.
2007
.
TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity.
Nature
446
:
916
920
.
16
Cadena
,
C.
,
S.
Ahmad
,
A.
Xavier
,
J.
Willemsen
,
S.
Park
,
J. W.
Park
,
S. W.
Oh
,
T.
Fujita
,
F.
Hou
,
M.
Binder
,
S.
Hur
.
2019
.
Ubiquitin-dependent and -independent roles of E3 ligase RIPLET in innate immunity.
Cell
177
:
1187
1200.e16
.
17
Castanier
,
C.
,
N.
Zemirli
,
A.
Portier
,
D.
Garcin
,
N.
Bidère
,
A.
Vazquez
,
D.
Arnoult
.
2012
.
MAVS ubiquitination by the E3 ligase TRIM25 and degradation by the proteasome is involved in type I interferon production after activation of the antiviral RIG-I-like receptors.
BMC Biol.
10
:
44
.
18
Lee
,
N. R.
,
H. I.
Kim
,
M. S.
Choi
,
C. M.
Yi
,
K. S.
Inn
.
2015
.
Regulation of MDA5-MAVS antiviral signaling axis by TRIM25 through TRAF6-mediated NF-κB activation.
Mol. Cells
38
:
759
764
.
19
Zhu
,
Z.
,
Y.
Wang
,
C.
Zhang
,
S.
Yu
,
Q.
Zhu
,
K.
Hou
,
B.
Yan
.
2016
.
TRIM25 blockade by RNA interference inhibited migration and invasion of gastric cancer cells through TGF-β signaling.
Sci. Rep.
6
:
19070
.
20
Huang
,
B.
,
S. H.
Baek
.
2017
.
Trim13 potentiates toll-like receptor 2-mediated nuclear factor κB activation via K29-linked polyubiquitination of tumor necrosis factor receptor-associated factor 6.
Mol. Pharmacol.
91
:
307
316
.
21
Hu
,
M. M.
,
Q.
Yang
,
J.
Zhang
,
S. M.
Liu
,
Y.
Zhang
,
H.
Lin
,
Z. F.
Huang
,
Y. Y.
Wang
,
X. D.
Zhang
,
B.
Zhong
,
H. B.
Shu
.
2014
.
TRIM38 inhibits TNFα- and IL-1β-triggered NF-κB activation by mediating lysosome-dependent degradation of TAB2/3.
Proc. Natl. Acad. Sci. USA
111
:
1509
1514
.
22
Tomar
,
D.
,
R.
Singh
.
2014
.
TRIM13 regulates ubiquitination and turnover of NEMO to suppress TNF induced NF-κB activation.
Cell. Signal.
26
:
2606
2613
.
23
Chang
,
T. H.
,
R.
Yoshimi
,
K.
Ozato
.
2015
.
Tripartite motif (TRIM) 12c, a mouse homolog of TRIM5, is a ubiquitin ligase that stimulates type I IFN and NF-κB pathways along with TNFR-associated factor 6.
J. Immunol.
195
:
5367
5379
.
24
Qiu
,
H.
,
F.
Huang
,
H.
Xiao
,
B.
Sun
,
R.
Yang
.
2013
.
TRIM22 inhibits the TRAF6-stimulated NF-κB pathway by targeting TAB2 for degradation.
Virol. Sin.
28
:
209
215
.
25
Suzuki
,
M.
,
M.
Watanabe
,
Y.
Nakamaru
,
D.
Takagi
,
H.
Takahashi
,
S.
Fukuda
,
S.
Hatakeyama
.
2016
.
TRIM39 negatively regulates the NFκB-mediated signaling pathway through stabilization of Cactin.
Cell. Mol. Life Sci.
73
:
1085
1101
.
26
Shibata
,
M.
,
T.
Sato
,
R.
Nukiwa
,
T.
Ariga
,
S.
Hatakeyama
.
2012
.
TRIM45 negatively regulates NF-κB-mediated transcription and suppresses cell proliferation.
Biochem. Biophys. Res. Commun.
423
:
104
109
.
27
Fan
,
W.
,
T.
Liu
,
X.
Li
,
Y.
Zhou
,
M.
Wu
,
X.
Cui
,
H.
Chen
,
P.
Qian
.
2017
.
TRIM52: a nuclear TRIM protein that positively regulates the nuclear factor-kappa B signaling pathway.
Mol. Immunol.
82
:
114
122
.
28
Yao
,
F.
,
L. Y.
Long
,
Y. Z.
Deng
,
Y. Y.
Feng
,
G. Y.
Ying
,
W. D.
Bao
,
G.
Li
,
D. X.
Guan
,
Y. Q.
Zhu
,
J. J.
Li
,
D.
Xie
.
2014
.
RACK1 modulates NF-κB activation by interfering with the interaction between TRAF2 and the IKK complex.
Cell Res.
24
:
359
371
.
29
Yang
,
Z.
,
H.
Xian
,
J.
Hu
,
S.
Tian
,
Y.
Qin
,
R. F.
Wang
,
J.
Cui
.
2015
.
USP18 negatively regulates NF-κB signaling by targeting TAK1 and NEMO for deubiquitination through distinct mechanisms.
Sci. Rep.
5
:
12738
.
30
Karl
,
I.
,
M.
Jossberger-Werner
,
N.
Schmidt
,
S.
Horn
,
M.
Goebeler
,
M.
Leverkus
,
H.
Wajant
,
T.
Giner
.
2014
.
TRAF2 inhibits TRAIL- and CD95L-induced apoptosis and necroptosis. [Published erratum appears in 2014 Cell Death Dis. 5: e1556.]
Cell Death Dis.
5
: e1444.
31
Schneider
,
A. T.
,
J.
Gautheron
,
M.
Feoktistova
,
C.
Roderburg
,
S. H.
Loosen
,
S.
Roy
,
F.
Benz
,
P.
Schemmer
,
M. W.
Büchler
,
U.
Nachbur
, et al
.
2017
.
RIPK1 suppresses a TRAF2-dependent pathway to liver cancer.
Cancer Cell
31
:
94
109
.

The authors have no financial conflicts of interest.