You are on page 1of 12

ARTHRITIS & RHEUMATOLOGY

Vol. 68, No. 2, February 2016, pp 347–358


DOI 10.1002/art.39447
C 2016, American College of Rheumatology
V

Regulation of Transforming Growth Factor b–Activated


Kinase 1 Activation by Epigallocatechin-3-Gallate
in Rheumatoid Arthritis Synovial Fibroblasts

Suppression of K63-Linked Autoubiquitination of


Tumor Necrosis Factor Receptor–Associated Factor 6

Anil K. Singh,1 Sadiq Umar,1 Sharayah Riegsecker,2


Mukesh Chourasia,3 and Salahuddin Ahmed1

Objective. Transforming growth factor b–activated Results. Inhibition of TAK1, but not IRAK-1 or
kinase 1 (TAK1) is a key MAPKKK family protein in TRAF6, completely abrogated IL-1b–induced IL-6 and
interleukin-1b (IL-1b), tumor necrosis factor (TNF), and IL-8 synthesis in RASFs. EGCG inhibited TAK1 phos-
Toll-like receptor signaling. This study was undertaken to phorylation at Thr184/187 and occupied the C174 position,
examine the posttranslational modification of TAK1 and an ATP-binding site, to inhibit its kinase activity. EGCG
its therapeutic regulation in rheumatoid arthritis (RA). pretreatment also inhibited K63-linked autoubiquitina-
Methods. The effect of TAK1, IL-1 receptor–asso- tion of TRAF6, a posttranslational modification essential
ciated kinase 1 (IRAK-1), and TNF receptor–associated for TAK1 autophosphorylation, by forming a stable H
factor 6 (TRAF6) inhibition was evaluated in IL-1b– bond at the K124 position on TRAF6. Furthermore,
stimulated human RA synovial fibroblasts (RASFs). EGCG enhanced proteasome-associated deubiquitinase
Western blotting, immunoprecipitation, and 20S protea- expression to rescue proteins from proteasomal degrada-
some assay were used to study the ubiquitination process tion. Western blot analyses of joint homogenates from
in RASFs. The efficacy of epigallocatechin-3-gallate rats with AIA showed a significant increase in K48-linked
(EGCG), a potent antiinflammatory molecule, in regu- polyubiquitination, TAK1 phosphorylation, and TRAF6
lating these processes in RASFs was evaluated. Molecu- expression when compared to naive rats. Administration
lar docking was performed to examine the interaction of of EGCG (50 mg/kg/day) for 10 days ameliorated AIA in
EGCG with human TAK1, IRAK-1, and TRAF6. These rats by reducing TAK1 phosphorylation and K48-linked
findings were confirmed using a rat model of adjuvant- polyubiquitination.
induced arthritis (AIA). Conclusion. Our findings provide a rationale for
targeting TAK1 for the treatment of RA with EGCG.
Supported by the NIH (grant AR-063104 to Dr. Ahmed), the
Arthritis Foundation (Innovative Research Grant to Dr. Ahmed), and
In rheumatoid arthritis (RA), the increased
Washington State University startup funding. expression of interleukin-1b (IL-1b), tumor necrosis
1
Anil K. Singh, PhD, Sadiq Umar, PhD, Salahuddin Ahmed, factor (TNF), and IL-6 in the synovial microenviron-
PhD: Washington State University College of Pharmacy, Spokane;
2
Sharayah Riegsecker, MS: University of Toledo College of Pharmacy
ment contributes to joint pain, inflammation, and tissue
and Pharmaceutical Sciences, Toledo, Ohio; 3Mukesh Chourasia, PhD: destruction (1). While the discovery of highly effective
National Institute of Pharmaceutical Education and Research, Hajipur, biologic therapies in the last decade has transformed
India.
Address correspondence to Salahuddin Ahmed, PhD, Depart- the therapeutic landscape for RA treatment, the ever-
ment of Pharmaceutical Sciences, Washington State University College increasing numbers of non- or partial responders, con-
of Pharmacy, SPBS Room 411, 412 East Spokane Falls Boulevard, comitant with the associated morbidity and mortality,
Spokane, WA 99210. E-mail: salah.ahmed@wsu.edu.
Submitted for publication March 6, 2015; accepted in revised pose a significant socioeconomic and clinical challenge.
form September 17, 2015. The success of pharmacologic approaches to clinically
347
348 SINGH ET AL

develop tofacitinib, a pan-JAK inhibitor, has opened a matory molecule, in regulating IL-1b–induced IL-6 and
new chapter of small-molecule therapeutics in the treat- chemokine production and matrix metalloproteinase 2
ment of RA (2). While the success of this inhibitor with (MMP-2) activation in human RA synovial fibroblasts
regard to clinical efficacy and safety remains untested, (RASFs) by inhibiting the activation of JNK, p38, and
research on novel small-molecule kinase-directed thera- NF-kB (17,18). These findings suggest that EGCG may
peutics in RA has accelerated (3,4). A target protein interfere with the proteins proximal to IL-1R to suppress
that is critical in cytokine signaling networks may yield multiple inflammatory signaling pathways. Thus, the pre-
higher efficacy and a better clinical response (4). sent study was carried out with a two-pronged approach,
Recent studies of intracellular signaling pathways aimed at defining the role and underlying mechanism of
have identified transforming growth factor b–activated TAK1 activation in RA and testing the selectivity of
kinase 1 (TAK1) as an interesting therapeutic target EGCG in regulating these events in RASFs and in a rat
for inflammatory diseases and cancer (5). TAK1 is an adjuvant-induced arthritis (AIA) model of RA. Our
MAPKKK that mediates activation of downstream results showed that EGCG selectively inhibits TAK1 acti-
MAPKs (JNK and p38) and NF-kB pathways in response vation by blocking its phosphorylation at the Thr184/187
to IL-1b, TNF, or Toll-like receptor (TLR) stimulation ATP-binding site and hinders the association of TRAF6
(6,7). Among these cytokines, IL-1b plays an important and TAK1 through down-regulation of TRAF6-associated
pathologic role in RA (8,9). Surprisingly, a recombinant K63-linked autoubiquitination. These findings suggest that
form of human IL-1 receptor (IL-1R) antagonist, ana- TAK1 is a potential therapeutic target in RA and that
kinra, lacked clinical efficacy in the treatment of RA and EGCG may be developed as a TAK1 inhibitor for the
elicited serious infections such as pneumonia and celluli- treatment of RA and other inflammatory diseases.
tis (10,11). While the biologic agents that target TNF
(etanercept) and IL-6 (tocilizumab) are successful in the MATERIALS AND METHODS
clinical management of RA, therapeutic approaches
Antibodies and reagents. Recombinant human IL-1b
aimed at developing small-molecule inhibitors with more and TNF were purchased from R&D Systems. EGCG (.95%
efficient blocking of IL-1b signaling pathways are desired pure) was purchased from Sigma (catalog no. 4143). A 20S protea-
for RA and other IL-1–driven diseases (12). some activity assay was purchased from Cayman Chemical. Goat
IL-1b binding to IL-1RI initiates autophosphor- anti-rabbit and goat anti-mouse horseradish peroxidase–linked
lyation of IL-1R–associated kinase 4 (IRAK-4), which secondary antibodies were purchased from Cell Signaling Technol-
ogy. Inhibitors of the signaling protein IRAK-1 (N-[2-morpholiny-
further recruits IRAK-1 to myeloid differentiation fac- lethyl]-2-[3-nitrobenzoylamido]-benzimidazole) and TAK1 ([5Z]-
tor 88 (MyD88) and subsequently autophosphorylates 7-oxozeaenol) were from EMD Millipore, and TRAF6 control
to trigger its activation and consequent degradation (6). and inhibitor peptides were from Imgenex/Novus. Human IL-6
TNF receptor–associated factor 6 (TRAF6), an E3 ubiq- and IL-8 enzyme-linked immunosorbent assay duo kits were pur-
uitin ligase, forms a complex with IRAK-1 that dissoci- chased from R&D Systems. Rabbit anti–IRAK-1 (sc-7883),
mouse monoclonal TRAF6 (sc-8409), and mouse monoclonal b-
ates from the receptor and translocates to the cytoplasm
actin (sc-47778) were purchased from Santa Cruz Biotechnology.
to recruit and activate TAK1 by K63-linked monoubiqui- Anti–FK-2 (catalog no. BML-PW8810) was purchased from Enzo
tination (Lys34) and polyubiquitination (13,14). TAK1 Life Sciences. Anti-pTAK1 Ser184/187 (catalog no. 4508), anti-
activation at its kinase domain threonine (Thr) 184/187 pIRAK-1 Thr345/Ser346 (catalog no. 11927), anti–IRAK-4 (cat-
results in phosphorylation of MAPKs and activation of alog no. 4363), anti-MyD88 (catalog no. 4238), anti-K63 polyu-
biquitin (catalog no. 5621), and anti-K48 polyubiquitin (catalog
IKK, leading to IkBa degradation and NF-kB activation
no. 8081) antibody were purchased from Cell Signaling Tech-
(15). nology. Anti-pTAK1 Ser439 (ab109404), anti-TAK1 (ab109526),
While TAK1 is a key signaling protein in IL-1b– and anti-TRAF6 (ab33915) were from Abcam.
induced gene expression, its highly regulated posttransla- Culture of human RASFs. SFs were isolated from
tional modifications, such as autophosphorylation, ubiqui- the synovium of patients with RA who had undergone total
tination, and deubiquitination by IRAK-1 or TRAF6, joint replacement surgery or synovectomy and were obtained
according to institutional review board (IRB)–approved proto-
define its role in disease pathogenesis (16). Despite estab- col and in compliance with the Declaration of Helsinki.
lished roles of MAPKs and NF-kB pathways in IL-1b RASFs were processed as described previously (17).
signaling in RA, significant gaps remain in our under- Treatment of RASFs. To evaluate the time-dependent
standing of the role of adaptor proteins proximal to IL- activation of IL-1b–induced signaling pathways and the pro-
1RI (IRAK-1, TRAF6, and TAK1) to create therapeutic tective effect of EGCG treatment, RASFs (2 3 105/well) were
plated in 6-well plates with or without EGCG (2.5–20 mM)
advantages for effectively inhibiting IL-1b signaling in RA. overnight (for 12–14 hours) and then stimulated with IL-1b
Studies from our laboratory have shown the efficacy of (10 ng/ml) for 30 minutes (for signaling studies) or 24 hours to
epigallocatechin-3-gallate (EGCG), a potent antiinflam- evaluate the production of IL-6 and IL-8 in the conditioned
TAK1 INHIBITION BY EGCG IN RA 349

media. To study the effect of IL-1b stimulation on IRAK-1 Human phosphokinase antibody array. To evaluate
degradation, RASFs were stimulated with IL-1b (10 ng/ml) the effect of EGCG on other RASF kinases involved in impor-
for 0–120 minutes. To further study the effect of methotrexate tant pathophysiologic processes, we used a human antibody
(MTX) or dexamethasone (DEX) on IL-1b–induced IRAK-1 array kit (catalog no. ARY003B; R&D Systems). Further experi-
degradation, RASFs were pretreated with MTX (10 mM) or mental details of the assay are provided in the Supplementary
DEX (20 mM) for 1 hour and then stimulated with IL-1b for Methods, available on the Arthritis & Rheumatology web site at
30 minutes. http://onlinelibrary.wiley.com/doi/10.1002/art.39447/abstract.
IRAK-1 in vitro kinase activity. IRAK-1 kinase activ- Statistical analysis. A Kruskal-Wallis nonparametric
ity was determined using a fluorescence-based in vitro kinase test followed by a Mann-Whitney U test was used to evaluate
assay (ADAPTA kinase assay; Life Technologies). Briefly, the the statistical significance of group differences in measured
23 IRAK-1/histone H3 (1–20) peptide mixture was prepared parameters from IL-6 and IL-8 protein expression experi-
in 50 mM HEPES, pH 7.5, 0.01% Brij-35, 10 mM MgCl2, and ments or Western blotting studies of RASFs. Student’s t-test
1 mM EGTA. The final 10-ml kinase reaction was carried out was performed to calculate statistical differences between the
for 60 minutes and consisted of 100 mM ATP, EGCG (10 means of the different protein variables obtained from in vivo
nM220 mM), 3.17–30.5 ng IRAK-1, and 100 mM histone H3 findings. P values less than 0.05 (2-tailed) were considered
(1–20) peptide in 32.5 mM HEPES, pH 7.5, 0.005% Brij-35, significant.
5 mM MgCl2, and 0.5 mM EGTA. After 1 hour of incubation,
5 ml of detection mixture was added, and then emission was
read at 665 nM.
Assay of 20S proteasome activity. The 20S protea-
RESULTS
some activity in RASFs was determined using an assay kit as TAK1 regulation of IL-1b–induced IL-6 and
previously described (19).
IL-8 production. IL-1b is a master cytokine for local and
Western blot analysis. Western blot analysis was per-
formed as previously described (17,18). Specific details are systemic inflammation. Stimulation of RASFs with IL-1b
provided in the Supplementary Methods, available on the (10 ng/ml) resulted in a .180-fold and a .250-fold
Arthritis & Rheumatology web site at http://onlinelibrary.wiley. increase in IL-6 and IL-8 production, respectively (P ,
com/doi/10.1002/art.39447/abstract. 0.001). Pretreatment with 10 mM or 20 mM EGCG
Immunoprecipitation assay. RASFs were grown in
150-mm dishes up to 80% confluence, starved overnight with
resulted in inhibition of IL-6 and IL-8 (P , 0.01 versus
or without 20 mM EGCG, and then stimulated with IL-1b for stimulation with IL-1b alone, for all comparisons) (Figures
30 minutes. Cells were washed 2 times in ice-cold 13 phos- 1A and B). To test whether chronic exposure to EGCG at
phate buffered saline, lysed in 500 ml of radioimmunoprecipi- nanomolar concentrations produces similar inhibitory
tation assay buffer as previously described, and then used for effects on IL-1b–induced IL-6 and IL-8 production,
immunoprecipitation assays as described in the Supplementary
Methods, available on the Arthritis & Rheumatology web site at
RASFs were treated with EGCG at a daily dose of 1–
http://onlinelibrary.wiley.com/doi/10.1002/art.39447/abstract. 1,000 nM in 5% fetal bovine serum–RPMI 1640 for 7
AIA in rats. Female Lewis rats weighing ;100 gm days, followed by serum starvation, and stimulation with
(Harlan) were injected subcutaneously at the base of the tail IL-1b (10 ng/ml) for 8 or 24 hours (see Supplementary
with 300 ml (5 mg/ml) of lyophilized Mycobacterium butyricum Figure 1, available on the Arthritis & Rheumatology web
(Difco) in sterile mineral oil. The day of adjuvant injection was
considered day 0. Ankle circumferences were measured on site at http://onlinelibrary.wiley.com/doi/10.1002/art.39447/
day 17 by an investigator who was blinded with regard to treat- abstract). Even at nanomolar concentrations, EGCG was
ment group, as previously described (17). Healthy (adjuvant- effective in inhibiting IL-1b–induced IL-6 (20–35%) and
naive) rats were used as a control. In the treatment group, IL-8 (15–20%) in a dose-dependent manner (P , 0.01 for
EGCG (50 mg/kg) was administered intraperitoneally as previ- IL-6) (see Supplementary Figure 1, available on the Arth-
ously described (17). An EGCG dose of 50 mg/kg in rats cor-
responds to a human equivalent dose of 480 mg based on body ritis & Rheumatology web site at http://onlinelibrary.wiley.
surface area ratio (20). The ankle circumferences of both hind com/doi/10.1002/art.39447/abstract).
ankles from each animal were averaged. All animal studies To test the roles of IRAK-1, TAK1, and TRAF6
were approved by the University of Toledo Institutional Ani- in IL-1b–induced IL-6 and IL-8 production, RASFs
mal Care and Use Committee. were pretreated with inhibitors of IRAK-1 (N-[2-
Molecular modeling studies and ligand preparation.
EGCG ([2R,3R]-5,7-dihydroxy-2-[3,4,5-trihydroxyphenyl]-3,4- morpholinylethyl]-2-[3-nitrobenzoylamido]-benzimidazole;
dihydro-2H-1-benzopyran-3-yl 3,4,5-trihydroxybenzoate) was 50 mM), TAK1 ([5Z]-7-oxozeaenol; 5 mM), or TRAF6
first optimized as a ligand at a B3LYP/6-31111G** level of inhibitor peptide or control peptide (50 mM) and then stim-
calculation using jaguar2014.3 and then prepared using the Lig- ulated with IL-1b. Analysis of the conditioned media
Prep module for docking in the Glide module of Schrodinger showed that only TAK1 inhibition completely abrogated
suite 2014.3. Further details regarding the in silico modeling
and docking studies are provided in the Supplementary Meth- IL-1b–induced IL-6 and IL-8 production (P , 0.01 for
ods, available on the Arthritis & Rheumatology web site at http:// both) (Figures 1C and D). These results also underscore
onlinelibrary.wiley.com/doi/10.1002/art.39447/abstract. the therapeutic value of TAK1 regulation in RA and the
350 SINGH ET AL

Figure 1. Regulation of interleukin-1b (IL-1b)–induced IL-6 and IL-8 production in rheumatoid arthritis synovial fibroblasts (RASFs) by trans-
forming growth factor b–activated kinase 1 (TAK1). A and B, IL-6 (A) and IL-8 (B) production in RASFs pretreated with epigallocatechin-3-
gallate (EGCG; 2.5–20 mM) overnight, and then stimulated with IL-1b (10 ng/ml) for 24 hours. Production of IL-6 and IL-8 in the conditioned
media was determined using commercially available enzyme-linked immunosorbent assay kits. C and D, IL-6 (C) and IL-8 (D) production in
RASFs preincubated with signaling inhibitors (i) for IL-1 receptor–associated kinase 1 (IRAK; N-[2-morpholinylethyl]-2-[3-nitrobenzoylamido]-
benzimidazole; 50 mM), TAK1 ([5Z]-7-oxozeaenol; 5 mM), control peptide for tumor necrosis factor receptor–associated factor 6 (TRAF6;
50 mM), or TRAF6 inhibitor peptide (50 mM) for 2 hours, and then stimulated with IL-1b (10 ng/ml) for 24 hours. Bars show the mean 6 SEM
(n 5 4 experiments using different donors). ** 5 P , 0.01 versus IL-1b alone.

effectiveness of EGCG in suppressing IL-1b–induced IL-6 in IL-1b–induced signaling mechanisms. Further evalu-
and IL-8 production. ation of the effect of EGCG pretreatment (10–20 mM)
EGCG inhibition of IRAK-1 kinase activity but on IRAK-1 protein expression showed that EGCG was
not its proteasomal degradation. IL-1b–induced acti- unable to rescue IL-1b–induced spontaneous IRAK-1
vation of NF-kB and MAPK pathways is tightly regulat- degradation in RASFs (Figure 2B). Interestingly, pre-
ed by molecules such as IRAK-1, TRAF6, and TAK1 treatment with MTX or DEX also had no effect on IL-
(6,21). Since not much is known in terms of the thera- 1b–induced degradation of IRAK-1 at 30 minutes.
peutic value of these signal transducers in RA, we per- To further investigate the mechanism of IRAK-1
formed a time-dependent study with IL-1b stimulation proteasomal degradation in RASFs, we immunoprecipi-
in human RASFs. IL-1b induced a rapid degradation of tated global ubiquitinated proteins from cell lysates
IRAK-1, followed by the phosphorylation of TAK1 at using an FK-2 monoclonal antibody and probed for
the active site (Thr184/187), which reached a peak after IRAK-1 expression. IRAK-1 was heavily ubiquitinated
30 minutes of stimulation with IL-1b (Figure 2A). How- in response to IL-1b stimulation, and EGCG had no
ever, TRAF6 expression remained unchanged, suggest- influence on this process (Figure 2C). Studies suggest
ing that its autoubiquitination, not expression, is critical that IRAK-1 phosphorylation and subsequent degrada-
TAK1 INHIBITION BY EGCG IN RA 351

Figure 2. Inhibition of IRAK-1 kinase activity, but not its proteasomal degradation, by EGCG. A, Western immunoblot (WB) showing time-
dependent activation of the proximal signaling proteins (IRAK-1, pTAK1 Thr184/187, and TRAF6) in RASFs upon stimulation with IL-1b for 5–
120 minutes. B, Western immunoblot of IRAK-1 protein degradation in RASFs pretreated with EGCG (10–20 mM overnight), methotrexate
(MTX; 10 mM for 2 hours), or dexamethasone (Dexa; 20 mM for 2 hours) and then stimulated with IL-1b for 30 minutes. C, Ubiquitinated
IRAK-1 (Ub-IRAK-1) in RASFs treated with IL-1b and/or EGCG (20 mM) and immunoprecipitated (IP) with global ubiquitin (FK-2) or IgG
(M2 Flag). D, Inhibition of IRAK-1 kinase activity in vitro by EGCG, tested using an ADAPTA kinase assay according to the manufacturer’s
instructions. Values are the mean 6 SEM. Western immunoblots are representative of 3–4 experiments repeated with RASFs from different
donors. NS 5 no stimulation (see Figure 1 for other definitions).

tion, but not IRAK-1 kinase activity, is an important a dose-dependent manner (P , 0.01 versus IL-1b–treated
event in the activation of downstream signaling cascades RASFs, for 20 mM) (Figure 3A). To obtain further insights
(22–24). This prompted us to confirm the effect of into molecular mechanisms, we studied the effect of EGCG
EGCG on IRAK-1 activity using an ADAPTA kinase on other IL-1b signaling proteins proximal to IL-1, such as
assay in vitro, which showed an ;66% inhibition of MyD88, IRAK-4, and pIRAK-4. MyD88 and IRAK-4
IRAK-1 activity by EGCG at a 1 mM concentration expression remained unchanged with different treatments,
(Figure 2D). while IL-1b–induced phosphorylation of IRAK-4 at Thr345/
Selective inhibition of TAK1 phosphorylation at Ser346 was modestly inhibited by EGCG in RASFs (Figure
the Thr184/187 ATP-binding site by EGCG. TAK1 is a 3B). However, this partial reduction in the phosphorylation
central mediator of TNF, IL-1b, and TLR signal trans- of IRAK-4 by EGCG was unable to rescue IRAK-1 degra-
duction pathways (25). Studies also suggest that TAK1 dation in these samples (Figure 3B).
phosphorylation at Thr184/187 is a critical determinant To confirm the selectivity of EGCG for the
of its kinase activity (26,27). We evaluated the effect Thr184/187 active site of TAK1, we determined the effect
of EGCG in regulating IL-1b–induced pTAK1 (Thr184/187) of EGCG on the serine 439 phosphorylation site of the
in RASFs (Figure 3A). Pretreatment with EGCG (2.5–20 TAK1 protein (Figure 3B). Interestingly, EGCG had no
mM) inhibited pTAK1 (Thr184/187) expression in RASFs in effect on IL-1b–induced activation of pTAK1 (Ser439)
352 SINGH ET AL

Figure 3. EGCG selectively inhibits phosphorylation of TAK1 at the Thr184/187 site to inhibit its kinase activity. A, Expression of pTAK1
(Thr184/187), TAK1, TRAF6, and b-actin in the cell lysates of RASFs pretreated with EGCG (2.5–20 mM) and then stimulated with IL-1b for 30
minutes. Bars show the mean 6 SEM. ## 5 P , 0.01 versus no stimulation; ** 5 P , 0.01 versus stimulation with IL-1b alone. B, Expression of
myeloid differentiation factor 88 (MyD88), pIRAK-4 (Thr345/Ser346), IRAK-4, IRAK-1, pTAK1 (Ser439), total TAK1, and b-actin in RASFs
treated with IL-1b and/or EGCG (20 mM), analyzed to confirm the selectivity of EGCG for pTAK1 (Thr184/187). C, Expression of IRAK-1,
pTAK1 (Thr184/187), pTAK1 (Ser439), total TAK1, TRAF6, and b-actin in cell lysates of RASFs pretreated with EGCG (20 mM), Toll-like recep-
tor 4 (TLR-4) agonist (lipopolysaccharide [LPS]; 1 mg/ml), or TLR-2 agonist (Pam3Cys; 1 mg/ml) followed by stimulation with tumor necrosis
factor (TNF; 20 ng/ml) for 30 minutes. Results are representative of 4 experiments with RASFs from different donors. See Figure 1 for other
definitions.

in RASFs, suggesting its selectivity in regulating TAK1 ings from the antibody array are qualitative and open to
activation by inhibiting phosphorylation at the Thr184/187 interpretation, further quantitative analysis on the kin-
site within the kinase activation loop of TAK1 (28). To ases of interest may provide clearer and specific insights.
further confirm whether this TAK1 inhibitory activity of The results of the present study suggest that EGCG
EGCG was selective for IL-1b, we performed similar preferentially inhibits IL-1b–induced phosphorylation
experiments with TNF, TLR-2 agonist (Pam3Cys), and and activation of TAK1 (Thr184/187) to suppress down-
TLR-4 agonist (lipopolysaccharide) alone or in combi- stream signaling pathways such as p38, JNK, and NF-kB
nation with EGCG (Figure 3C). Only TNF was a potent in RASFs, as observed previously (17,18).
inducer of pTAK1 (Thr184/187), and EGCG was effective Inhibition of 20S proteasome activity and enhance-
in blocking TNF-induced pTAK1 (Thr184/187) in RASFs ment of deubiquitinase expression in RASFs treated with
(Figure 3C). Overall, the findings suggest a novel mech- EGCG. The ubiquitin–proteasome system plays an
anism of cytokine-activated TAK1 kinase regulation by important role in the posttranslational modification of
EGCG in RASFs. proteins that are either destined to be degraded or sta-
To test the effect of EGCG on other known kin- bilized for further signaling (16). K48-linked Lys chains
ases involved in several pathophysiologic processes, we “prime” protein to be recognized by 26S proteasome for
performed a kinome assay on RASF lysates treated with degradation, whereas K63-linked Lys chains are tagged
EGCG and/or IL-1b using a human phosphokinase to the proteins that are required for stability and sus-
antibody array (R&D Systems). This array simulta- tained signaling (29). However, the ubiquitination pro-
neously detects the relative site phosphorylation of 43 cess has never been studied in depth with regard to RA
kinases and 2 related proteins (see Supplementary Fig- pathogenesis. To explore this, we immunoprecipitated
ures 2A and B, available on the Arthritis & Rheumatolo- total polyubiquitinated proteins using an FK-2 antibody
gy web site at http://onlinelibrary.wiley.com/doi/10.1002/ in RASFs treated with IL-1b and/or EGCG and ana-
art.39447/abstract). The kinase array showed that IL-1b lyzed them for K63-linked and K48-linked ubiquitination
induced the majority of kinases in RASFs within 30 (Figure 4). EGCG had no effect on K63-linked or K48-
minutes of stimulation. However, EGCG pretreatment linked ubiquitination by itself, but markedly enhanced
had modest to no effect on most of the kinases (see Sup- K63-linked, but not K48-linked, ubiquitination in the
plementary Figures 2A and B, available on the Arthritis presence of IL-1b (Figure 4A). A similar trend in the
& Rheumatology web site at http://onlinelibrary.wiley. cell lysates from the remaining input suggests that
com/doi/10.1002/art.39447/abstract). Since these find- EGCG may enhance the global K63-linked ubiquitina-
TAK1 INHIBITION BY EGCG IN RA 353

Figure 4. Inhibition of K63 autoubiquitination of TRAF6 and up-regulation of the expression of deubiquitinases in RASFs by EGCG. A, K63-
linked and K48-linked ubiquitination patterns in RASFs treated with IL-1b and/or EGCG (20 mM) and immunoprecipitated (IP) with global
ubiquitin (FK-2) or IgG. B, Global K63-linked and K48-linked polyubiquitination in RASFs treated as described in A. C, Levels of 20S proteasome
activity in RASFs (0.5 3 104/well in a 96-well plate) treated with IL-1b and/or EGCG (20 mM) for 30 minutes. Proteasome activity was analyzed using
a commercially available kit. Bars show the mean 6 SEM from 3 experiments performed using RASFs from different donors. D, Expression of
proteasome-associated and unassociated deubiquitinases in RASFs treated with IL-1b and/or EGCG (20 mM) for 30 minutes. E, TRAF6 and TAK1
ubiquitination in RASFs (5 3 106/150-mm dish) treated with IL-1b and/or EGCG (20 mM). RASFs were immunoprecipitated with a K63 antibody
and probed. Bars show the mean 6 SEM from 4 experiments performed using RASFs from different donors. F, Effect of EGCG in interfering with
the association of TRAF6 and TAK1 in RASFs. Cell lysates from RASFs treated as described in E were immunoprecipitated with a TRAF6 antibody
and probed with TAK1. Bars show the mean 6 SEM from 4 experiments performed using RASFs from different donors. # 5 P , 0.05; ## 5 P ,
0.01, versus no stimulation; * 5 P , 0.05; ** 5 P , 0.01, versus stimulation with IL-1b alone. WB 5 Western blotting (see Figure 1 for other
definitions).

tion process to stabilize certain proteins required to as UCHL2 (Figure 4D). However, the expression of
block IL-1b signaling in RASFs (Figure 4B). However, PSMD13, a 26S proteasome non-ATP regulatory core
further analysis of 20S proteasome activity in similarly subunit, remained unchanged after EGCG treatment,
treated lysates showed that IL-1b–induced proteasome suggesting that it selectively inhibits 20S proteasome
activity in RASFs was increased by ;40%, an effect that activity without influencing the degradation process.
was significantly reduced in the presence of EGCG, by EGCG interferes with TRAF6–TAK1 associa-
;50% (P , 0.05) (Figure 4C). tion by inhibiting K63-linked autoubiquitination of
These findings suggest that despite being ineffec- TRAF6. Autoubiquitination of TRAF6 at K124 is directly
tive in down-regulating K48-linked ubiquitination, EGCG linked to NF-kB activation and its nuclear translocation
may block further processing of these proteins in the (31). To examine the effect of EGCG on K63-linked
26S proteasome tunnel. One possible mechanism could autoubiquitination of TAK1 and TRAF6 in response to
be deubiquitinase enzymes that hydrolyze polyubiquitin IL-1b, we immunoprecipitated K63-linked proteins from
chains on the tagged proteins, thus rescuing them from cell lysates treated with IL-1b and/or EGCG and probed
degradation (30). Analysis of the associated and unasso- for TAK1 and TRAF6 proteins (Figure 4E). Although
ciated deubiquitinases, such as UCHL2, USP14, UCH37, we did not observe a marked change in K63-mediated
UBC9, and deubiquitinase associated with core 20S pro- ubiquitination of TAK1, there was an ;30% increase in
teasome (PSMD13), in RASFs showed that in the pres- K63-mediated ubiquitination of TRAF6 with IL-1b treat-
ence of IL-1b, EGCG increased the expression of ment, which was inhibited by ;30% after treatment with
UCH37 and USP14, deubiquitinases known to preferen- EGCG (Figure 4E). To further understand the impact of
tially hydrolyze K48-linked polyubiquitin chains, and had the inhibition of TRAF6 autoubiquitination, we immuno-
a marginal effect on unassociated deubiquitinase such precipitated cell lysates with TRAF6 and probed for its
354 SINGH ET AL

association with TAK1. Interestingly, we found that IL-


1b–treated samples exhibited ;2.5-fold higher TRAF6–
TAK1 association compared to the untreated group (P ,
0.01) (Figure 4F). However, treatment with EGCG alone
or in the presence of IL-1b inhibited TRAF6 association
with TAK1 in RASFs, in part by preventing its K63-medi-
ated autoubiquitination (Figure 4F).
EGCG occupies ATP-binding sites of IRAK-1
and TAK1 to inhibit kinase activity. Molecular model-
ing of the IRAK-1 protein based on the known 3-
dimensional structure of IRAK-4 was used for conduct-
ing in silico molecular docking studies (Figure 5A). We
found that the hydroxyl group of a trihydroxyphenyl ring
of EGCG is perfectly positioned (;1.6Å) to make an H
bond with D358 and N345, and form a p–p interaction at
F223 to stabilize EGCG in the binding pocket, resulting in
its inhibitory activity. D358 is a conserved aspartate resi-
due and an ATP-binding site for IRAK-1, as obtained
from PROSITE ProRule: PRU00159 (http://www.uni
prot.org). The hydroxyl group of trihydroxybenzoate ring
shows a strong H bond with E214 and a weak H bond
with K239. The benzoate nucleus fits well in a pocket
formed by various lipophilic or aromatic residues. It is in
interaction with hydrophobic residues L347 and V226 and
perfectly positioned to form a p–p interaction with Y288.
Thus, binding of EGCG at these sites results in a revers-
ible inhibition of IRAK-1 activity, as further confirmed
earlier with an ADAPTA in vitro kinase assay.
Molecular docking studies of EGCG with a
TAK1–transforming growth factor b–activated kinase 1
and MAP3K-7 binding protein 1 (TAB-1) complex
revealed an interesting finding. The hydroxyl group of a
benzopyran ring in EGCG completely occupied the posi-
tion in the cavity that facilitates strong H bond formation
with the C174 residue (Figure 5B). Importantly, (5Z)27- Figure 5. Unique insights from in silico molecular docking studies
oxozeaenol, a known TAK1 inhibitor, acts on C174 to of EGCG binding on IRAK-1, TAK1–transforming growth factor b–
epitomize irreversible covalent binding that results in a activated kinase 1 and MAP3K-7 binding protein 1 (TAB-1) com-
plex, and TRAF6. A, Molecular docking of EGCG on a constructed
permanent abrogation of TAK1 kinase activity (32). In IRAK-1 structure shows that the hydroxyl group of the trihydroxy-
addition to binding with C174, EGCG stabilizes its posi- phenyl ring of EGCG is perfectly positioned (1.6Å) to make an H
tion in the binding pockets by forming hydrophobic links bond with D358, a conserved aspartate residue and ATP-binding
to distal V42, V50, and L163 residues that happen to be site, and N345, which inhibits IRAK-1 kinase activity. B, EGCG
TAK1 nucleotide-binding regions, and forming a p–p forms an H bond at C174, an ATP-binding site, on the TAK1–TAB-
1 complex and stabilizes its position in the binding pockets by form-
interaction at Y113 with the trihydroxybenzoate ring of ing hydrophobic links to distal V42, V50, and L163 residues that
EGCG. The trihydroxyphenyl ring fits well in the outer happen to be TAK1 nucleotide-binding regions and by forming a p–
pocket formed by various lipophilic residues and forms p interaction at Y113, thus serving as a potent and reversible TAK1
an H bond with the K158, N161, and N188 residues by its inhibitor. C, Side chains of E79 and K124 residues on TRAF6 play
hydroxyl groups and stabilizes its position by hydrophobic significant roles in fastening the position of the trihydroxyphenyl ring
of EGCG through the H bond network, thereby inhibiting its K63
interaction with the P160 site, thus acting as a potent autoubiquitination. See Figure 1 for other definitions.
reversible TAK1 inhibitor.
In terms of TRAF6, the almost linear structure of with EGCG (Figure 5C). The hydroxyl group of the tri-
this molecule provides a limited opportunity due to its hydroxybenzoate ring of ECGC forms the H bond net-
shallow binding pockets for hydrophobic interactions work with the side chain of R78 and E149, p–p interaction
TAK1 INHIBITION BY EGCG IN RA 355

Figure 6. EGCG modulates the in vivo ubiquitination process to inhibit adjuvant-induced arthritis (AIA) in rats. A, Amelioration of AIA in rats
treated with EGCG (50 mg/kg/day) administered intraperitoneally for 10 days (starting on day 7 after arthritis induction), as indicated by
reduced ankle circumferences. The ankle circumferences of both hind ankles from each animal were averaged. Bars show the mean 6 SEM.
## 5 P , 0.01 versus naive rats; * 5 P , 0.05 versus rats with AIA. B and C, Global K63-linked ubiquitination (K63-Ub) (B) and global K48-
linked ubiquitination (C) in joint homogenates (30 mg per sample) from naive rats, rats with AIA, and EGCG-treated rats with AIA. D, Expres-
sion of pTAK1 (Thr184/187), pTAK1 (Ser439), total TAK1, TRAF6, and b-actin in the same joint homogenates as described in C. E, Effect of
EGCG administration on the modulation of in vivo deubiquitinase enzymes associated with the removal of ubiquitin chains. See Figure 1 for
other definitions.

with Y56, and hydrophobic interaction with L145 to pro- blot analysis of the joint homogenates showed a modest
vide stability. The backbones of R146 and H147 are also decrease in K63-linked ubiquitinated proteins in the
involved in the formation of an H bond with the hydroxyl AIA group compared to the naive group, which was
group of the benzopyran ring. However, of mechanistic reversed with EGCG pretreatment (Figure 6B). How-
importance, the side chains of E79 and K124 residues play ever, a marked increase in K48-linked ubiquitinated
a significant role in fastening the position of a trihydroxy- proteins was observed in joint homogenates from rats
phenyl ring through the H bond network. Since K124 is an with AIA, which was effectively inhibited to levels near
important site for K63 autoubiquitination of TRAF6, those in the control group after EGCG administration
these findings provide insights into the mechanisms of (P , 0.05) (Figure 6C). These findings clearly indicate
the observed inhibitory effect of EGCG on TRAF6 K63- that EGCG differentially modulates K63-linked and
linked autoubiquitination in RASFs. K48-linked ubiquitination, which results in the amelio-
Amelioration of AIA in rats by EGCG modula- ration of AIA in rats.
tion of the ubiquitination process in vivo. A rat model Further correlating these findings with the
of AIA was used to study in vivo ubiquitination associ- expression of signaling intermediates, we found that the
ated with RA and the modulatory effect of EGCG. It levels of pTAK1 (Thr184/187), pTAK1 (Ser439), total
has previously been shown that administration of TAK1, TRAF6, UCH37, and USP14 were markedly
100 mg/kg of EGCG daily for 10 days inhibits IL-6 pro- increased in joints from rats with AIA as compared to
duction in serum and joint homogenates from rats with joints from control rats (Figures 6D and E) (P , 0.05 or
AIA (17). In the present study, we observed that the P , 0.01 for all of the proteins studied except K63) (see
administration of EGCG (50 mg/kg/day intraperitone- Supplementary Figures 3A–G, available on the Arthritis &
ally) during disease onset was effective in ameliorating Rheumatology web site at http://onlinelibrary.wiley.
AIA, as evident from a significant reduction in ankle com/doi/10.1002/art.39447/abstract for statistical analysis).
circumference (P , 0.05; n 5 6) (Figure 6A). Western Interestingly, joint homogenates from EGCG-treated rats
356 SINGH ET AL

showed a marked decrease in the expression of these impaired innate immune responses with IRAK-1/-4
proteins, to levels comparable to those observed in inhibitors (36). In addition, transcriptome analyses using
the naive group (P , 0.05 for pTAK1 [Ser439], total IL-1R antagonist and 13 gene partners, including
TAK1, and TRAF6) (Figure 6 and Supplementary Fig- MyD88, IRAK-1/-4, and TRAF6, suggest that identifying
ures 3A–G, available on the Arthritis & Rheumatology a protein target proximal to the IL-1 receptor may elicit
web site at http://onlinelibrary.wiley.com/doi/10.1002/art. serious adverse effects, as observed with anakinra treat-
39447/abstract). These findings suggest that EGCG may ment (37). This opens therapeutic avenues for validating
blunt cytokine-signaling pathways in the arthritic rat other signaling proteins involved in IL-1b–mediated
ankles by regulating TAK1 and other associated proteins inflammation and tissue destruction.
via interference with posttranslational modifications to The ideal position of TAK1 in the inflammatory
inhibit RA pathogenesis. signaling cascades triggered by IL-1b and by TNF makes
it an attractive therapeutic target (28,38). Although TAK1
DISCUSSION inhibition using a small interfering RNA approach has
shown some promise for RA (39), there is a complete
In this study, we identified a mechanism of lack of understanding of its posttranslational regulation
TAK1 regulation in human RASFs and AIA in rats in in RA pathogenesis. Binding of IL-1b to its receptor IL-
which the ubiquitin–proteasome system is modulated to 1R leads to the recruitment of adaptor proteins and kin-
inhibit IL-1b–induced signaling pathways. Importantly, ases such as MyD88, IRAK-1, and IRAK-4. IRAK-4
our study provides insights into the mechanisms of the phosphorylates IRAK-1 and facilitates TRAF6 recruit-
posttranslational ubiquitination processes that govern ment to the complex and IRAK-1 degradation (40).
TAK1 activation in RASFs and the mechanism of the Being an E3 ubiquitin ligase, TRAF6 undergoes K63-
interaction of EGCG with IRAK-1, TRAF6, and TAK1 linked autoubiquitination that follows the recruitment of
that results in the inhibition of TAK1 activity. Given the TAK1–TAB-1 or the TAB-2/3 complex and the induction
safety profile of EGCG in humans (33), our results pro- of TAK1 kinase activity. TAK1 eventually phosphorylates
vide a platform to design TAK1 inhibitors that can effec- MKK4/7–JNK and MKK3/6–p38 MAPK and IKK,
tively disrupt the integrated cytokine signaling to limit resulting in nuclear translocation of the transcription fac-
their role in RA pathogenesis. tors activator protein 1 and NF-kB (5). Like MTX and
IL-1b is a master regulator of inflammation and DEX, EGCG pretreatment was unable to block IRAK-1
tissue destruction in several autoimmune diseases, such degradation; however, it markedly inhibited in vitro
as RA, gout, and type 2 diabetes mellitus (8). From a IRAK-1 kinase activity. Molecular docking studies using
therapeutic standpoint, only IL-1R antagonists have been IRAK-1 homology structural modeling provide evidence
pursued for clinical testing to block the effect of IL-1 in that EGCG can tightly occupy ATP-binding sites, result-
the treatment of RA. Despite the failure of anakinra and ing in complete blockade of its kinase activity. However,
given the pathologic significance of IL-1 in RA, further several studies provide evidence that IRAK-1 phosphory-
therapeutic approaches are warranted with the aim of lation and subsequent degradation, not its kinase activity,
testing agents that are more potent inhibitors of IL-1 sig- is an important event for the activation of downstream
naling pathways. In this regard, IRAK-4 kinase inhibitors signaling cascades (22–24).
have shown promise in preclinical testing (12). However, Ubiquitination is an important posttranslational
recent studies suggest the differential role of IRAK-4 in modification that regulates various cellular processes
regulating cytokine production in different cell types including cell survival, apoptosis, and signaling (16,41).
(34). Of particular interest, the inhibition of IRAK-4 acti- It is carried out by the ubiquitin–proteasome system,
vation in human dermal fibroblasts was not effective in which involves E1, E2, and E3 ubiquitin ligases that
regulating IL-1R– or TLR agonist–induced IL-6 produc- identify the proteins for ubiquitin ligation, proteases
tion, whereas its inhibition in human monocytes was that are involved in degrading proteins, and the protea-
effective in reducing IL-6 levels (34). These findings sug- some that safeguards the highly specific nature of the
gest that the kinase activities of IRAK-1/-4 are redundant degradation system and prevents nonspecific protein
in regulating inflammatory cytokine production in human degradation. The process is tightly regulated by 2 key
cells, including RASFs (35). chain-linking lysine residues, K48 and K63 (5,16). K48-
Given the role of IRAK-1/-4 in controlling linked polyubiquitination of the protein primes it for
immune alterations and innate responses to pathogens, proteasomal degradation, whereas K63-linked ubiquiti-
studies using several multiomics and computational nation of the protein stabilizes it for further cell signal-
intensive approaches provide evidence of possible ing and function. Another class of cysteine proteases,
TAK1 INHIBITION BY EGCG IN RA 357

deubiquitinases, can hydrolyze and remove these ubiq- ACKNOWLEDGMENTS


uitin chains to reverse the fate of the protein (30). In The authors thank the National Disease Research Inter-
this study, we found that IL-1b–induced K63 autoubiqui- change and the Cooperative Human Tissue Network for provid-
tination of TRAF6 was markedly inhibited by EGCG. ing RA synovial tissue, Dr. David A. Fox (University of Michigan
Further molecular insights obtained using molecular Medical School) for critical review of the manuscript and helpful
docking studies confirmed that EGCG forms H bond suggestions, and Ms Maria Beamer for technical support in ani-
mal studies.
networks with TRAF6 at the E79 and K124 positions,
and p–p interactions with Y56 that create an electron
AUTHOR CONTRIBUTIONS
cloud and exceptional stability of EGCG for holding on
to critical TRAF6 sites. Site-specific K63 autoubiquitina- All authors were involved in drafting the article or
tion of TRAF6 is a critical determinant of IKK activity revising it critically for important intellectual content, and all
authors approved the final version to be published. Dr. Ahmed
(31,42). Although it has previously been shown that had full access to all of the data in the study and takes respon-
EGCG inhibits NF-kB activation in a variety of cell sibility for the integrity of the data and the accuracy of the data
types (17,18,43–45), the present study provides evidence analysis.
of TAK1 as the actual molecular target for its antirheu- Study conception and design. Singh, Chourasia, Ahmed.
Acquisition of data. Singh, Umar, Riegsecker, Chourasia.
matic activity. Analysis and interpretation of data. Singh, Umar, Chourasia,
Only autoubiquitinated TRAF6 is capable of asso- Ahmed.
ciating with and autophosphorylating TAK1 (46,47).
Thus, blocking K63 autoubiquitination of TRAF6 showed REFERENCES
a profound effect on this process and a marked decrease
1. Bottini N, Firestein GS. Duality of fibroblast-like synoviocytes in
in the association of TRAF6 with TAK1 and the subse- RA: passive responders and imprinted aggressors. Nat Rev
quent inhibition of pTAK1 (Thr184/187) in RASFs. While Rheumatol 2013;9:24–33.
TRAF6 has been an attractive therapeutic target for 2. Clark JD, Flanagan ME, Telliez JB. Discovery and development
of Janus kinase (JAK) inhibitors for inflammatory diseases.
inflammatory diseases, it is required for optimal physio- J Med Chem 2014;57:5023–38.
logic signaling mechanisms, making it a riskier target. 3. Fox DA. Kinase inhibition: a new approach to the treatment of
This study provides a unique mechanism of therapeuti- rheumatoid arthritis. N Engl J Med 2012;367:565–7.
4. Meier FM, McInnes IB. Small-molecule therapeutics in rheuma-
cally regulating TRAF6 via reversible inhibition of its toid arthritis: scientific rationale, efficacy and safety. Best Pract
function through stable H bond formation by EGCG Clin Res Rheumatol 2014;28:605–24.
without jeopardizing its physiologic functions. Rationaliz- 5. Sakurai H. Targeting of TAK1 in inflammatory disorders and
cancer. Trends Pharmacol Sci 2012;33:522–30.
ing drug design approaches in light of these findings may 6. Wang C, Deng L, Hong M, Akkaraju GR, Inoue J, Chen ZJ.
yield more potent and safer drugs for the treatment of TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nature
2001;412:346–51.
RA and other inflammatory diseases. However, further 7. Yamaguchi K, Shirakabe K, Shibuya H, Irie K, Oishi I, Ueno N,
studies are warranted to understand the effect of EGCG et al. Identification of a member of the MAPKKK family as a
on E1 or E2 ubiquitin ligases, which associate with potential mediator of TGF-b signal transduction. Science 1995;
270:2008–11.
TRAF6 to activate MAPK and IKK through TAK1. 8. Dinarello CA, van der Meer JW. Treating inflammation by block-
The continued work to develop new RA thera- ing interleukin-1 in humans. Semin Immunol 2013;25:469–84.
pies suggests that the existing drugs for RA are expen- 9. Horai R, Saijo S, Tanioka H, Nakae S, Sudo K, Okahara A,
et al. Development of chronic inflammatory arthropathy resem-
sive, immunosuppressive, and sometimes unsuitable for bling rheumatoid arthritis in interleukin 1 receptor antagonist-
long-term use (48,49). Additionally, existing RA thera- deficient mice. J Exp Med 2000;191:313–20.
pies are B cell–, T cell–, and cytokine-directed. No exist- 10. Salliot C, Dougados M, Gossec L. Risk of serious infections dur-
ing rituximab, abatacept and anakinra treatments for rheumatoid
ing medication for RA targets SFs, despite an arthritis: meta-analyses of randomised placebo-controlled trials.
established body of molecular and clinical evidence sug- Ann Rheum Dis 2009;68:25–32.
gesting their role in RA pathogenesis (50–52). This may 11. Buch MH, Bingham SJ, Seto Y, McGonagle D, Bejarano V,
White J, et al. Lack of response to anakinra in rheumatoid arth-
be attributed to our limited understanding of the molec- ritis following failure of tumor necrosis factor a blockade. Arth-
ular mechanisms that govern their invasive and destruc- ritis Rheum 2004;50:725–8.
tive potential. In this study, we have provided a 12. Bahia MS, Kaur M, Silakari P, Silakari O. Interleukin-1 receptor
associated kinase inhibitors: potential therapeutic agents for
rationale for targeting RASF TAK1 in RA and identi- inflammatory- and immune-related disorders. Cell Signal 2015;27:
fied a unique mechanism through which EGCG inhibits 1039–55.
the interaction between signaling molecules important 13. Cui W, Xiao N, Xiao H, Zhou H, Yu M, Gu J, et al. b-TrCP-
mediated IRAK1 degradation releases TAK1-TRAF6 from the
in cytokine signaling, ultimately inhibiting inflammation membrane to the cytosol for TAK1-dependent NF-kB activation.
and tissue destruction in RA. Mol Cell Biol 2012;32:3990–4000.
358 SINGH ET AL

14. Zhou H, Yu M, Fukuda K, Im J, Yao P, Cui W, et al. IRAK-M 33. Chow HH, Cai Y, Hakim IA, Crowell JA, Shahi F, Brooks CA,
mediates Toll-like receptor/IL-1R-induced NFkB activation and et al. Pharmacokinetics and safety of green tea polyphenols after
cytokine production. EMBO J 2013;32:583–96. multiple-dose administration of epigallocatechin gallate and poly-
15. Sorrentino A, Thakur N, Grimsby S, Marcusson A, von Bulow phenon E in healthy individuals. Clin Cancer Res 2003;9:3312–9.
V, Schuster N, et al. The type I TGF-b receptor engages 34. Cushing L, Stochaj W, Siegel M, Czerwinski R, Dower K, Wright
TRAF6 to activate TAK1 in a receptor kinase-independent man- Q, et al. Interleukin 1/Toll-like receptor-induced autophosphoryla-
ner. Nat Cell Biol 2008;10:1199–207. tion activates interleukin 1 receptor-associated kinase 4 and con-
16. Chen ZJ. Ubiquitination in signaling to and activation of IKK. trols cytokine induction in a cell type-specific manner. J Biol
Immunol Rev 2012;246:95–106. Chem 2014;289:10865–75.
17. Ahmed S, Marotte H, Kwan K, Ruth JH, Campbell PL, Rabquer 35. Song KW, Talamas FX, Suttmann RT, Olson PS, Barnett JW, Lee SW,
BJ, et al. Epigallocatechin-3-gallate inhibits IL-6 synthesis and et al. The kinase activities of interleukin-1 receptor associated kinase
suppresses transsignaling by enhancing soluble gp130 production. (IRAK)-1 and 4 are redundant in the control of inflammatory cytokine
Proc Natl Acad Sci U S A 2008;105:14692–7. expression in human cells. Mol Immunol 2009;46:1458–66.
18. Ahmed S, Pakozdi A, Koch AE. Regulation of interleukin-1b– 36. Tieri P, Zhou X, Zhu L, Nardini C. Multi-omic landscape of
induced chemokine production and matrix metalloproteinase 2 rheumatoid arthritis: re-evaluation of drug adverse effects. Front
activation by epigallocatechin-3-gallate in rheumatoid arthritis Cell Dev Biol 2014;2:59.
synovial fibroblasts. Arthritis Rheum 2006;54:2393–401. 37. Cao Y, Jiao Y, Wang L, Huang Y, Postlethwaite A, Stuart J,
19. Ahmed S, Silverman MD, Marotte H, Kwan K, Matuszczak N, et al. Anakinra as an interleukin 1 receptor antagonist, compli-
Koch AE. Down-regulation of myeloid cell leukemia 1 by cated genetics and molecular impacts: from the point of view of
epigallocatechin-3-gallate sensitizes rheumatoid arthritis synovial mouse genomics. Int Immunopharmacol 2012;13:28–36.
fibroblasts to tumor necrosis factor a–induced apoptosis. Arth- 38. Hammaker DR, Boyle DL, Inoue T, Firestein GS. Regulation of
ritis Rheum 2009;60:1282–93. the JNK pathway by TGF-b activated kinase 1 in rheumatoid
20. Freireich EJ, Gehan EA, Rall DP, Schmidt LH, Skipper HE. arthritis synoviocytes. Arthritis Res Ther 2007;9:R57.
Quantitative comparison of toxicity of anticancer agents in 39. Courties G, Seiffart V, Presumey J, Escriou V, Scherman D,
mouse, rat, hamster, dog, monkey, and man. Cancer Chemother Zwerina J, et al. In vivo RNAi-mediated silencing of TAK1
Rep 1966;50:219–44. decreases inflammatory Th1 and Th17 cells through targeting of
21. Ninomiya-Tsuji J, Kishimoto K, Hiyama A, Inoue J, Cao Z, myeloid cells. Blood 2010;116:3505–16.
Matsumoto K. The kinase TAK1 can activate the NIK-I kB as 40. Dai L, Aye Thu C, Liu XY, Xi J, Cheung PC. TAK1, more than
well as the MAP kinase cascade in the IL-1 signalling pathway. just innate immunity. IUBMB Life 2012;64:825–34.
41. Chen ZJ. Ubiquitin signalling in the NF-kB pathway. Nat Cell
Nature 1999;398:252–6.
Biol 2005;7:758–65.
22. Knop J, Martin MU. Effects of IL-1 receptor-associated kinase
42. Nathan JA, Kim HT, Ting L, Gygi SP, Goldberg AL. Why do
(IRAK) expression on IL-1 signaling are independent of its
cellular proteins linked to K63-polyubiquitin chains not associate
kinase activity. FEBS Lett 1999;448:81–5.
with proteasomes? EMBO J 2013;32:552–65.
23. Li X, Commane M, Burns C, Vithalani K, Cao Z, Stark GR.
43. Ahmed S, Wang N, Lalonde M, Goldberg VM, Haqqi TM. Green tea
Mutant cells that do not respond to interleukin-1 (IL-1) reveal a
polyphenol epigallocatechin-3-gallate (EGCG) differentially inhibits
novel role for IL-1 receptor-associated kinase. Mol Cell Biol
interleukin-1b-induced expression of matrix metalloproteinase-1 and
1999;19:4643–52. –13 in human chondrocytes. J Pharmacol Exp Ther 2004;308:767–73.
24. Maschera B, Ray K, Burns K, Volpe F. Overexpression of an 44. Hafeez BB, Ahmed S, Wang N, Gupta S, Zhang A, Haqqi TM.
enzymically inactive interleukin-1-receptor-associated kinase acti- Green tea polyphenols-induced apoptosis in human osteosarco-
vates nuclear factor-kB Biochem J 1999;339:227–31. ma SAOS-2 cells involves a caspase-dependent mechanism with
25. Ajibade AA, Wang HY, Wang RF. Cell type-specific function of downregulation of nuclear factor-kB. Toxicol Appl Pharmacol
TAK1 in innate immune signaling. Trends Immunol 2013;34:307–16. 2006;216:11–9.
26. Singhirunnusorn P, Suzuki S, Kawasaki N, Saiki I, Sakurai H. 45. Singh R, Ahmed S, Islam N, Goldberg VM, Haqqi TM.
Critical roles of threonine 187 phosphorylation in cellular stress- Epigallocatechin-3-gallate inhibits interleukin-1b–induced expres-
induced rapid and transient activation of transforming growth sion of nitric oxide synthase and production of nitric oxide in
factor-b-activated kinase 1 (TAK1) in a signaling complex con- human chondrocytes: suppression of nuclear factor kB activation
taining TAK1-binding protein TAB1 and TAB2. J Biol Chem by degradation of the inhibitor of nuclear factor kB. Arthritis
2005;280:7359–68. Rheum 2002;46:2079–86.
27. Yu Y, Ge N, Xie M, Sun W, Burlingame S, Pass AK, et al. 46. Deng L, Wang C, Spencer E, Yang L, Braun A, You J, et al.
Phosphorylation of Thr-178 and Thr-184 in the TAK1 T-loop is Activation of the IkB kinase complex by TRAF6 requires a
required for interleukin (IL)21-mediated optimal NFkB and dimeric ubiquitin-conjugating enzyme complex and a unique poly-
AP-1 activation as well as IL-6 gene expression. J Biol Chem ubiquitin chain. Cell 2000;103:351–61.
2008;283:24497–505. 47. Wu H, Arron JR. TRAF6, a molecular bridge spanning adaptive
28. Kilty I, Jones LH. TAK1 selective inhibition: state of the art and immunity, innate immunity and osteoimmunology. Bioessays
future opportunities. Future Med Chem 2015;7:23–33. 2003;25:1096–105.
29. Adhikari A, Xu M, Chen ZJ. Ubiquitin-mediated activation of 48. Furst DE. The risk of infections with biologic therapies for rheu-
TAK1 and IKK. Oncogene 2007;26:3214–26. matoid arthritis. Semin Arthritis Rheum 2010;39:327–46.
30. Komander D, Clague MJ, Urbe S. Breaking the chains: structure 49. Khanna D, McMahon M, Furst DE. Safety of tumour necrosis
and function of the deubiquitinases. Nat Rev Mol Cell Biol factor-a antagonists. Drug Saf 2004;27:307–24.
2009;10:550–63. 50. Chang SK, Noss EH, Chen M, Gu Z, Townsend K, Grenha R,
31. Lamothe B, Besse A, Campos AD, Webster WK, Wu H, Darnay et al. Cadherin-11 regulates fibroblast inflammation. Proc Natl
BG. Site-specific Lys-63-linked tumor necrosis factor receptor- Acad Sci U S A 2011;108:8402–7.
associated factor 6 auto-ubiquitination is a critical determinant 51. Lee DM, Kiener HP, Agarwal SK, Noss EH, Watts GF, Chisaka
of IkB kinase activation. J Biol Chem 2007;282:4102–12. O, et al. Cadherin-11 in synovial lining formation and pathology
32. Wu J, Powell F, Larsen NA, Lai Z, Byth KF, Read J, et al. in arthritis. Science 2007;315:1006–10.
Mechanism and in vitro pharmacology of TAK1 inhibition by 52. Bartok B, Firestein GS. Fibroblast-like synoviocytes: key effector
(5Z)-7-Oxozeaenol. ACS Chem Biol 2013;8:643–50. cells in rheumatoid arthritis. Immunol Rev 2010;233:233–55.

You might also like