You are on page 1of 9

Cutting Edge: Inhibiting TBK1 by

Compound II Ameliorates Autoimmune


Disease in Mice
This information is current as Maroof Hasan, Nicole Dobbs, Shaheen Khan, Michael A.
of January 5, 2022. White, Edward K. Wakeland, Quan-Zhen Li and Nan Yan
J Immunol 2015; 195:4573-4577; Prepublished online 2
October 2015;
doi: 10.4049/jimmunol.1500162
http://www.jimmunol.org/content/195/10/4573

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


Supplementary http://www.jimmunol.org/content/suppl/2015/10/02/jimmunol.150016
Material 2.DCSupplemental
References This article cites 18 articles, 7 of which you can access for free at:
http://www.jimmunol.org/content/195/10/4573.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Author Choice Freely available online through The Journal of Immunology
Author Choice option
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2015 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of
Cutting Edge Immunology
Cutting Edge: Inhibiting TBK1 by Compound II
Ameliorates Autoimmune Disease in Mice
Maroof Hasan,*,† Nicole Dobbs,*,† Shaheen Khan,‡ Michael A. White,x
Edward K. Wakeland,‡ Quan-Zhen Li,‡ and Nan Yan*,†
TANK-binding kinase 1 (TBK1) is a serine/threonine mulate inappropriately, leading to autoimmune and inflam-
protein kinase that plays a crucial role in innate immu- matory diseases through immune pathways that often depend
nity. Enhanced TBK1 function is associated with auto- on TBK1 (5, 6). TBK1 is a member of the IkB kinase (IKK)
immune diseases and cancer, implicating the potential family and is ubiquitously expressed. In addition to its role in
benefit of therapeutically targeting TBK1. In this article, innate immunity, TBK1 also plays an important role in onco-
we examined a recently identified TBK1 inhibitor Com- genic transformation through direct regulation of Akt survival
pound II on treating autoimmune diseases. We found signaling (7). Therefore, TBK1 represents an attractive thera-

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


that Compound II is a potent and specific inhibitor of peutic target for autoimmune diseases and cancer with under-
TBK1-mediated IFN response. Compound II inhibited lying hyperactive TBK1 signaling. The in vivo importance of
polyinosinic-polycytidylic acid–induced immune activa- TBK1 in autoimmune disease models has been hampered by
tion in vitro and in vivo. Compound II treatment also the apparent lethality of Tbk12/2 mouse (8, 9). Small molec-
ameliorated autoimmune disease phenotypes of Trex12/2 ular inhibitors of TBK1 (e.g., BX795) often lack specificity,
mice, increased mouse survival, and dampened the IFN thereby limiting their application (5). We have recently iden-
gene signature in TREX1 mutant patient lymphoblasts. In tified a 6-aminopyrazolopyrimidine derivative (Compound II)
addition, we found that TBK1 gene expression is elevated through a biochemical screen of small-molecule inhibitors of
in systemic lupus erythematosus patient cells, and sys- TBK1 and demonstrated its effectiveness on limiting cancer cell
temic lupus erythematosus cells with high IFN signature proliferation (7). In this study, we examined Compound II on
responded well to Compound II treatment. Together, treating autoimmune diseases.
our findings provided critical experimental evidence Trex1 deficiency causes autoimmune and inflammatory
for inhibiting TBK1 with Compound II as an effec- disease phenotypes in mice (10, 11), and we recently deter-
tive treatment for TREX1-associated autoimmune dis- mined that the underlying immune activation depends on
eases and potentially other interferonopathies. The TBK1 (12). TREX1 mutations in humans are also associated
Journal of Immunology, 2015, 195: 4573–4577. with a spectrum of autoimmune and inflammatory pheno-
types, including Aicardi–Goutières syndrome (AGS), familial
chilblain lupus, systemic lupus erythematosus (SLE), and ret-

T inal vasculopathy with cerebral leukodystrophy (13). Trex12/2


he innate immunity is the first line of defense against
invading pathogens. Our cell encodes several pattern or TREX1 mutant patient cells induce cell-intrinsic activation
recognition receptors that recognize specific compo- of IFN-stimulated genes (ISGs) that are critically dependent
nents of pathogens and activate appropriate immune responses on TBK1 and a downstream transcriptional factor IRF3 (12).
(1). Many innate immune signaling pathways converge to Besides the TREX1 disease model, TBK1 and its associated
a key protein TANK-binding kinase 1 (TBK1), which orches- IFN signaling pathway have also been implicated in the
trates the induction of IFN and inflammatory genes that are pathogenesis of complex SLE and other autoimmune diseases
critical mediators of immune defense (2, 3). Human TBK1 collectively called interferonopathies (14, 15). No effective
haploinsufficiency is associated with herpes simplex encephalitis treatment is available for these diseases. Thus, there is an ur-
(4), demonstrating its importance in antiviral response. Pattern gent need for developing novel therapeutics based on molec-
recognition receptors can also recognize self-ligands that accu- ular understandings of these diseases.

*Department of Internal Medicine, University of Texas Southwestern Medical Address correspondence and reprint requests to Dr. Nan Yan, University of Texas
Center, Dallas, TX 75390; †Department of Microbiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390.
Southwestern Medical Center, Dallas, TX 75390; ‡Department of Immunology, E-mail address: nan.yan@utsouthwestern.edu
University of Texas Southwestern Medical Center, Dallas, TX 75390; xDepartment
The online version of this article contains supplemental material.
of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
75390 Abbreviations used in this article: AGS, Aicardi–Goutières syndrome; AST, aspartate
aminotransferase; BMDM, bone marrow–derived macrophage; IKK, IkB kinase; ISG,
ORCIDs: 0000-0003-4640-6949 (M.H.); 0000-0002-1472-8129 (S.K.); 0000-0002-
IFN-stimulated gene; poly(I:C), polyinosinic-polycytidylic acid; qRT-PCR, quantita-
7107-0992 (E.K.W.).
tive RT-PCR; SLE, systemic lupus erythematosus; TBK1, TANK-binding kinase 1.
Received for publication February 2, 2015. Accepted for publication September 8, 2015.
This article is distributed under The American Association of Immunologists, Inc.,
This work was supported by the National Institutes of Health (Grants AI098569 and Reuse Terms and Conditions for Author Choice articles.
AR067135 to N.Y.) and the Alliance for Lupus Foundation (to N.Y.).
Copyright Ó 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1500162
4574 CUTTING EDGE: INHIBITING TBK1 AMELIORATES AUTOIMMUNE DISEASE

Materials and Methods Poly(I:C) transfection activates both IRF3 and NF-kB path-
Cells and mice ways in RAW264.7 cells as indicated by robust phosphory-
Trex1+/2 mice and TREX1-R114H patient lymphoblasts were described
lation of TBK1, IRF3, and NF-kB. Compound II completely
previously (12). SLE cell lines were obtained from Oklahoma Medical Re- inhibited TBK1 autophosphorylation (17) and significantly
search Foundation. Leukocyte subsets microarray analysis of healthy control delayed IRF3 phosphorylation without affecting either total
and SLEs were described by Becker et al. (14). Cells were maintained in protein level (Fig. 1B). In contrast, BX795 (at the same dose
DMEM or RPMI 1640 with 10% (v/v) heat-inactivated FCS, 2 mM
L-glutamine, 10 mM HEPES, and 1 mM sodium pyruvate (complete
as Compound II, 1 mM) only partially inhibited IRF3 phos-
DMEM) with the addition of 100 U/ml penicillin, 100 mg/ml streptomycin phorylation and had no effect on TBK1 autophosphorylation.
and cultured at 37˚C with 5% CO2. Bone marrow–derived macrophages Neither drug inhibited NF-kB phosphorylation. These data
(BMDMs) were generated as described previously (12). Experiments in WT suggest that Compound II is a potent and specific inhibitor of
mice with Compound II and polyinosinic-polycytidylic acid [poly(I:C)] were
done by i.p. injections of DMSO or Compound II (10 mg/kg) for 3 d fol- TBK1-mediated IFN response.
lowed by one i.p. injection of poly(I:C) (300 mg in 100 ml vol of PBS) and
isolated peritoneal cells after 2 h. Experiments with Trex12/2 mice were Compound II inhibits poly(I:C)-induced immune activation in vitro
started when the mice were 4 wk old. Trex12/2 mice were treated with and in vivo
DMSO or Compound II (i.p. injections 3 d/wk) for 7 wk. Stocks of DMSO
and Compound II were further diluted in PBS to obtain 100 ml total volume We next examined whether Compound II also inhibits im-
per injection for each mouse. Mouse serum and tissues were harvested after mune activation of TBK1 in BMDMs or mice stimulated
indicated weeks of treatment. Mouse tissues were fixed in 4% PFA followed
by standard H&E staining for histology analysis. Experiments involving
with poly(I:C) through the TLR pathway. Poly(I:C) activates
human and mouse materials were approved by the Institutional Animal Care TLR3, which signals through TBK1 and IRF3 to activate IFN
and Use Committee and Institutional Review Board of University of Texas and inflammatory cytokines. We pretreated BMDMs with

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


Southwestern Medical Center. DMSO or Compound II overnight, then stimulated with
Reagents and Abs poly(I:C) for 2 h and measured IRF3 phosphorylation as a
marker for TBK1 activation. Compound II dramatically re-
TRI Reagent (Invitrogen) was used for RNA isolation. Compound II was
chemically synthesized (7). Abs used in this study include anti–phospho– duced poly(I:C)-induced IRF3 phosphorylation (Fig. 2A). We
IRF-3 (Ser396; Cell Signaling), anti-HMGB1 (Abcam), anti-Tubulin (Sigma), also treated wild type C57BL/6 mice with DMSO or Com-
anti–phospho-TBK1 (Cell Signaling), and anti–Phospho–NF-kB (Cell Sig- pound II by i.p. injections for 3 d, followed by poly(I:C)
naling); secondary Abs (GE Healthcare) were used for immunoblot analysis stimulation (i.p) for 2 h, and measured immune activation in
according to standard protocols.
peritoneal macrophages. Compound II significantly reduced
Quantitative PCR array, microarray, and serum analysis poly(I:C)-induced immune gene activation and IRF3 phos-
Quantitative PCR array analysis of immune gene profiles was performed as in phorylation compared with DMSO control (Fig. 2B, 2C).
Hasan et al. (12) using custom-ordered PCR array plates containing primer These data demonstrate that Compound II is a potent in-
sets prealiquoted (Bio-Rad). Each primer set was validated by Bio-Rad and in- hibitor of immune activation of TBK1 in vitro and in vivo.
house. Leukocyte subsets from healthy control and SLE subjects were ana-
lyzed by microarray (14). Mouse serum aspartate aminotransferase (AST)
level was measured by Vitros 250 at Mouse Metabolic and Phenotyping Core Compound II ameliorate autoimmune disease phenotypes of Trex12/2
(University of Texas Southwestern Medical Center). Autoantibody arrays mice and TREX1-mutant patient cells
were performed as described previously (16).
We previously showed that Trex1 deficiency induces cell-
Statistical methods intrinsic immune activation that depends on TBK1 and
Data are presented as the mean 6 SEM. GraphPad Prism 6 was used for
IRF3 (12). We then asked whether Compound II could
statistical analysis. Statistical tests performed are indicated in the figure leg- suppress the immune gene signature in Trex12/2 cells. We
ends: *p , 0.05, **p , 0.01, ***p , 0.001, ****p , 0.0001. treated wild type and Trex12/2 cells with DMSO or Com-
pound II and measured immune gene activation by quantita-
Results and Discussion tive RT-PCR (qRT-PCR). Ifit1 mRNA expression is elevated
Compound II is a potent and specific inhibitor of TBK1-mediated IFN 90-fold in Trex12/2 cells compared with wild type cells, and
response Compound II reduced Ifit1 mRNA level in Trex12/2 cells in
To evaluate the effect of Compound II in TBK1-mediated IFN a dose-dependent manner (Fig. 2D). Many other ISGs were
response, we first compared Compound II with another com- also elevated in Trex12/2 cells compared with wild type cells
monly used TBK1 inhibitor, BX795. We pretreated RAW264.7 (but not IFN or inflammatory genes), and the entire ISG sig-
(a mouse macrophage cell line) with either drug at a range of nature was completely suppressed by Compound II (Fig. 2E).
concentration (1 nM to 10 mM) followed by poly(I:C) trans- TREX1 patient lymphoblasts carrying R114H mutation (asso-
fection to induce IFN response. Both drugs inhibited poly(I:C)- ciated with both AGS and SLE) display a strong ISG signature
stimulated IRF3 activation (measured by Ifnb and Ifit1 mRNA similar to that of Trex12/2 mouse cells (12). We next treated
levels), but not NF-kB activation (measured by Tnfa mRNA TREX1-R114H patient lymphoblasts and a healthy control with
level), suggesting that Compound II specifically targets the DMSO or Compound II and measured expression of two ISGs,
TBK1/IRF3 pathway (Fig. 1A, Supplemental Fig. 1A). Com- CXCL10 and RSAD2 (Supplemental Fig. 1C). Both ISGs were
pound II also appeared to be significantly more potent, with significantly reduced after Compound II treatment, suggesting
a more gradual dose response and lower IC50 (20 nM) com- that inhibiting TBK1 by Compound II could be therapeutically
pared with BX795 (IC50 of 1 mM). We also found that BX795 useful for treating TREX1 patients with elevated ISG signature.
at high doses was toxic to RAW264.7 cells, whereas Compound We next treated Trex12/2 mice with Compound II and
II did not appear to be toxic across the entire the dose range examined its effect on autoimmune disease phenotypes asso-
(Supplemental Fig. 1B). ciated with Trex1 deficiency. Trex12/2 mice develop auto-
We also analyzed phosphorylation of TBK1 substrates to antibodies, splenomegaly, and liver dysfunction, and the
gain more molecular insights on the mechanism of inhibition. average survival of these mice is 8–10 wk (10, 11). We first
The Journal of Immunology 4575

TBK1-mediated IFN signaling, or it may require an earlier and


higher dose treatment. Collectively, our data suggest that tar-
geting TBK1 by Compound II is an effective treatment at
ameliorating autoimmune disease phenotypes associated with
Trex1 deficiency.
Compound II reduces immune gene signature in SLE patient cells
To further explore the potential application of Compound II
in complex autoimmune diseases such as SLE, we first ana-
lyzed the expression of TBK1 and other kinases in the IkB
family (i.e., IKKa, IKKb, and IKKe) in SLE and healthy
control primary leukocyte subsets. Three leukocyte pop-
ulations, CD4+CD3+ (T cells), CD19+CD32 (B cells), and
CD33+CD32 (myeloid cells), were sorted from PBMCs and
analyzed by microarray (14). We found that TBK1 expression

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


FIGURE 1. Compound II is a potent and specific inhibitor of TBK1-
mediated IFN response. (A) qRT-PCR analysis of Ifnb and Tnfa mRNA
expression. RAW264.7 cells were pretreated with Compound II (CmpII,
same throughout) or BX795 at indicated doses (nM) overnight and trans-
fected with 0.5 mg poly(I:C) for 2 h. (B) Immunoblot analysis of TBK1,
IRF3, and NF-kB phosphorylation. RAW267.4 cells were pretreated with
Compound II or BX795 overnight at 1 mM followed by poly(I:C) trans-
fection as in (A). Protein lysates were collected at indicated time post-
transfection (top) and analyzed by immunoblot with indicated Ab (right).
Data are representative of at least two independent experiments. Error bars
represent SEM. *p , 0.05.

treated Trex12/2 mice i.p. with DMSO or Compound II


(three injections per week, 10 mg/kg per injection) and found
that overall survival of Trex12/2 mice was significantly im-
proved after 7 wk of Compound II treatment compared with
DMSO controls (Fig. 3A). Compound II–treated Trex12/2
mice also have reduced AST level in the serum (indicating
improved liver function) and reduced number of splenocytes
and spleen size compared with DMSO-treated Trex12/2 mice
(Fig. 3B, 3C). We also analyzed autoantibodies in the serum
using an autoantigen microarray that contains 95 autoanti-
gens that are commonly found in a variety of autoimmune
diseases (16). We found that Trex12/2 mice produce a wide
array of autoantibodies compared with wild type littermates
and Compound II treatment significantly reduced most of the
autoantibodies in Trex12/2 mice (Fig. 3D, 3E). Other phe-
notypes of Trex12/2 mice, such as diminished hair and reduced
mobility, were also dramatically improved after Compound II FIGURE 2. Compound II inhibits TBK1-mediated IFN signaling in vitro
treatment (data not shown). Trex12/2 mice develop severe
and in vivo. (A) Immunoblot analysis of phospho-IRF3 (pIRF3) in wild type
BMDMs stimulated with poly(I:C) after pretreatment with DMSO or
systemic inflammation in multiple organs and succumb to Compound II (1 mM). HMGB1 is the loading control. (B) Immunoblot
disease mostly caused by inflammatory myocarditis (10, 11). analysis of phospho-IRF3 (pIRF3) in peritoneal cells. C57BL/6 mice were
We found that Compound II–treated Trex12/2 mouse heart pretreated with DMSO or Compound II (10 mg/kg per injection per day) for
showed reduced inflammation and pathology compared with 3 d followed by one i.p. injection of poly(I:C). Peritoneal cells were isolated
DMSO-treated or untreated Trex12/2 heart, although inflam- 2 h after poly(I:C) stimulation. (C) qRT-PCR analysis of indicated mRNAs
mation in the liver did not appear to improve at the point of in peritoneal cells from (B). Expression value of each gene was normalized
to housekeeping gene Gapdh (same throughout). Untreated value for each
our analysis (Supplemental Fig. 2). These histological findings gene was set to 1. (D and E) qRT-PCR analysis of Ifit1 (D) and other im-
are consistent with the partial rescue we have observed, and mune genes (E) in WT and Trex12/2 MEFs treated with DMSO or Com-
suggest that some of the tissue damage caused by the inborn pound II (CmpII). Data are representative of at least three independent
genetic defect may not be completely reversible by blocking experiments. Error bars represent SEM. ****p , 0.001.
4576 CUTTING EDGE: INHIBITING TBK1 AMELIORATES AUTOIMMUNE DISEASE

inhibiting TBK1 by Compound II could be potentially useful


for treating SLEs with high IFN signature.
In conclusion, our data demonstrated that inhibiting
TBK1 by Compound II is an effective treatment option for
a variety of autoimmune diseases with elevated IFN sig-
nature. Elevated TBK1 expression or function has been
associated with many important human immune disorders
and cancer (5–7, 14, 18). Thus, TBK1 represents an at-
tractive therapeutic target, and a potent and specific in-
hibitor of TBK1 could have broad applications. Several
small-molecule inhibitors of TBK1 including Compound
II are in various stages of development and clinical trials
for treating cancer. These drugs are potentially useful for
repurposing to treat autoimmune diseases. Prolonged in-
hibition of TBK1 may increase the possibility of viral
infections; thus, such treatments should proceed with cau-
tion or be used in combination with other antiviral thera-
pies. Our findings provided important experimental
evidence and a rational for using TBK1 inhibitors such as

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


Compound II for treating autoimmune diseases such as
AGS, SLE, and potentially other interferonopathies, many
of which are life threatening and currently lack effective
treatment.

FIGURE 3. Compound II ameliorates autoimmune diseases of Trex12/2


mice. (A) Survival study of Trex12/2 mice treated with DMSO or CmpII
(n = 13, mice were treated with DMSO or CmpII, 10 mg/kg per injection,
three injections per week for 7 wk). (B and C) Serum AST levels (B) and
splenocyte counts (C) from WT or Trex12/2 mice treated with DMSO or
CmpII (n= 4). (D) Serum autoantibody array analysis of WT or Trex12/2
mice treated with DMSO or CmpII (n = 4). (E) Representative autoantibody
titer data from (D). Mantel–Cox log-rank test (A). Unpaired t test (B, C, and
E). Error bars represent SEM. *p , 0.05, **p , 0.01.

was consistently elevated in SLE patient cells compared with


healthy controls (Fig. 4A), and the elevation is significant in
CD4+ and CD33+ populations. Despite that IKK family
kinases often act in similar signaling pathways, we did not
observe elevated expression of other IKK genes (IKKA, IKKB,
and IKKE) in SLEs compared with healthy controls, sug-
gesting that TBK1 or the underlying signaling pathway(s) FIGURE 4. TBK1 expression is elevated in SLE leukocytes, and Compound
may be uniquely associated with SLE (data not shown). II reduces IFN signature in SLEs. (A) TBK1 expression in healthy control and
To examine whether Compound II is effective at reducing SLE leukocyte subsets (CD4+, CD19+, or CD33+, as indicated on the y-axis).
IFN signature in SLE patient cells, we selected several SLE Individual population was isolated from whole PBMCs. Gene expression was
lymphoblast cell lines with low, intermediate, and high ISG measured by microarray. The p values are shown on the top of each panel. (B)
signature. We did not observe elevated expression of IFN qRT-PCR analysis of CXCL10 in SLE lymphoblasts treated with DMSO or
Compound II (CmpII). Five SLE lymphoblast cell lines were selected based
genes likely due to immortalization of these cell lines. SLE
on their low (SLE1, SLE2), intermediate (SLE3, SLE4), or high (SLE5) IFN
cells with intermediate and high ISG signature responded well gene signature. Each cell line was treated with DMSO or CmpII for 24 h
to Compound II treatment as indicated by reduced CXCL10 followed by qRT-PCR analysis of CXCL10. Data are representative of at least
mRNA expression (Fig. 4B). Together, our results identify three independent experiments. Error bars represent SEM. ns, not significant.
TBK1 as a potential target in SLE and demonstrate that *p , 0.05, **p , 0.01, unpaired t test.
The Journal of Immunology 4577

Acknowledgments 9. Perry, A. K., E. K. Chow, J. B. Goodnough, W.-C. Yeh, and G. Cheng. 2004. Dif-
ferential requirement for TANK-binding kinase-1 in type I interferon responses to toll-
We thank Rolf Brekken and members of the Yan Laboratory for helpful like receptor activation and viral infection. J. Exp. Med. 199: 1651–1658.
discussions. 10. Gall, A., P. Treuting, K. B. Elkon, Y.-M. Loo, M. Gale, Jr., G. N. Barber, and
D. B. Stetson. 2012. Autoimmunity initiates in nonhematopoietic cells and pro-
gresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity
36: 120–131.
Disclosures 11. Morita, M., G. Stamp, P. Robins, A. Dulic, I. Rosewell, G. Hrivnak, G. Daly,
The authors have no financial conflicts of interest. T. Lindahl, and D. E. Barnes. 2004. Gene-targeted mice lacking the Trex1 (DNase
III) 39–.59 DNA exonuclease develop inflammatory myocarditis. Mol. Cell. Biol.
24: 6719–6727.
12. Hasan, M., J. Koch, D. Rakheja, A. K. Pattnaik, J. Brugarolas, I. Dozmorov,
References B. Levine, E. K. Wakeland, M. A. Lee-Kirsch, and N. Yan. 2013. Trex1 regulates
1. Bowie, A. G., and L. Unterholzner. 2008. Viral evasion and subversion of pattern- lysosomal biogenesis and interferon-independent activation of antiviral genes. Nat.
recognition receptor signalling. Nat. Rev. Immunol. 8: 911–922. Immunol. 14: 61–71.
2. Häcker, H., and M. Karin. 2006. Regulation and function of IKK and IKK-related 13. Crow, Y. J., and J. Rehwinkel. 2009. Aicardi-Goutieres syndrome and related
kinases. Sci. STKE 2006: re13. phenotypes: linking nucleic acid metabolism with autoimmunity. Hum. Mol. Genet.
3. Fitzgerald, K. A., S. M. McWhirter, K. L. Faia, D. C. Rowe, E. Latz, D. T. Golenbock, 18(R2): R130–R136.
A. J. Coyle, S. M. Liao, and T. Maniatis. 2003. IKKepsilon and TBK1 are essential 14. Becker, A. M., K. H. Dao, B. K. Han, R. Kornu, S. Lakhanpal, A. B. Mobley,
components of the IRF3 signaling pathway. Nat. Immunol. 4: 491–496. Q.-Z. Li, Y. Lian, T. Wu, A. M. Reimold, et al. 2013. SLE peripheral blood
4. Herman, M., M. Ciancanelli, Y. H. Ou, L. Lorenzo, M. Klaudel-Dreszler, B cell, T cell and myeloid cell transcriptomes display unique profiles and each
E. Pauwels, V. Sancho-Shimizu, R. Pérez de Diego, A. Abhyankar, E. Israelsson, subset contributes to the interferon signature. PLoS One 8: e67003.
et al. 2012. Heterozygous TBK1 mutations impair TLR3 immunity and underlie 15. Crow, Y. J. 2011. Type I interferonopathies: a novel set of inborn errors of im-
herpes simplex encephalitis of childhood. J. Exp. Med. 209: 1567–1582. munity. Ann. N. Y. Acad. Sci. 1238: 91–98.
5. Yu, T., Y.-S. Yi, Y. Yang, J. Oh, D. Jeong, and J. Y. Cho. 2012. The pivotal role of TBK1 16. Subramanian, S., K. Tus, Q.-Z. Li, A. Wang, X.-H. Tian, J. Zhou, C. Liang,
in inflammatory responses mediated by macrophages. Mediators Inflamm. 2012: 979105. G. Bartov, L. D. McDaniel, X. J. Zhou, et al. 2006. A Tlr7 translocation accelerates

Downloaded from http://www.jimmunol.org/ by guest on January 5, 2022


6. Hammaker, D., D. L. Boyle, and G. S. Firestein. 2012. Synoviocyte innate immune systemic autoimmunity in murine lupus. Proc. Natl. Acad. Sci. USA 103: 9970–
responses: TANK-binding kinase-1 as a potential therapeutic target in rheumatoid 9975.
arthritis. Rheumatology (Oxford) 51: 610–618. 17. Ma, X., E. Helgason, Q. T. Phung, C. L. Quan, R. S. Iyer, M. W. Lee,
7. Ou, Y.-H., M. Torres, R. Ram, E. Formstecher, C. Roland, T. Cheng, R. Brekken, K. K. Bowman, M. A. Starovasnik, and E. C. Dueber. 2012. Molecular basis of
R. Wurz, A. Tasker, T. Polverino, et al. 2011. TBK1 directly engages Akt/PKB Tank-binding kinase 1 activation by transautophosphorylation. Proc. Natl. Acad.
survival signaling to support oncogenic transformation. Mol. Cell 41: 458–470. Sci. USA 109: 9378–9383.
8. Hemmi, H., O. Takeuchi, S. Sato, M. Yamamoto, T. Kaisho, H. Sanjo, T. Kawai, 18. Reilly, S. M., S.-H. Chiang, S. J. Decker, L. Chang, M. Uhm, M. J. Larsen,
K. Hoshino, K. Takeda, and S. Akira. 2004. The roles of two IkappaB kinase- J. R. Rubin, J. Mowers, N. M. White, I. Hochberg, et al. 2013. An inhibitor of the
related kinases in lipopolysaccharide and double stranded RNA signaling and viral protein kinases TBK1 and IKK- improves obesity-related metabolic dysfunctions in
infection. J. Exp. Med. 199: 1641–1650. mice. Nat. Med. 19: 313–321.
Supplemental Figures

Inhibiting TBK1 by Compound II ameliorates autoimmune disease in


mice

Maroof Hasan1,2, Nicole Dobbs1,2, Shaheen Khan3, Michael A. White4, Edward


Wakeland3, Quan-Zhen Li3, & Nan Yan1,2,*
1
Departments of Internal Medicine, 2Departments of Microbiology, 3Departments of
Immunology, 4Departments of Cell Biology, University of Texas Southwestern Medical
Center, Dallas, Texas, USA.

Correspondence to: N.Y. (nan.yan@utsouthwestern.edu)

  1  
Supplemental Figure 1. Compound II is more potent and less toxic in RAW264.7 cells
compared to BX795, and reduces IFN gene signature in TREX1R114H/R114H patient
lymphoblasts. (A) Quantitative RT-PCR analysis of Ifit1 mRNA expression (as in Figure
1). RAW264.7 cells were pre-treated with Compound II or BX795 at indicated doses
(bottom) overnight, and transfected with 0.5 μg poly(I:C) for 2 hours. (B) Microscopic
images of RAW264.7 cells after overnight treatment with Compound II or BX795 at dose
indicated on the top. Arrows indicate dying cells. (C) Quantitative RT-PCR analysis of
indicated genes in healthy control (HC) and TREX1R114H/R114H lymphoblasts treated with
DMSO or Cmp II. Healthy control treated with DMSO value was set to 1. ***P < 0.005.
**** P < 0.001. Data are representative of at least three independent experiments. Error
bars, SEM.

  2  
 

Supplemental Figure 2. Histology analysis of WT and Trex1-/- mice with or without


treatment. Two representative H & E staining images of heart and liver from untreated
WT or Trex1-/- mice (8-week old) or Trex1-/- mice treated with DMSO or Compound II
for 4 weeks (8-week old, as in Figure 3). Reduced inflammation and tissue pathology was
observed in Trex1-/- mice heart after Compound II treatment, but not in the liver.

  3  

You might also like