Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

JBC Papers in Press. Published on April 30, 2020 as Manuscript RA119.

012365
The latest version is at https://www.jbc.org/cgi/doi/10.1074/jbc.RA119.012365

The protein kinase MAP3K19 phosphorylates MAP2Ks and thereby activates ERK and JNK kinases and
increases viability of KRAS-mutant lung cancer cells

Van T. Hoang1, Katherine Nyswaner1, Pedro Torres-Ayuso1, John Brognard1*

From the 1Laboratory of Cell and Developmental Signaling, National Cancer Institute, Frederick, MD
21702

Running Title: MAP3K19 is a novel therapeutic target in RAS-mutant cancers

*To whom correspondence should be addressed: John Brognard, Laboratory of Cell and Developmental
Signaling, National Cancer Institute, Frederick, MD 21702; john.brognard@nih.gov; Tel. 301-846-1163;
Fax. 301-228-4863.

Keywords: kinase signaling, oncogenes, extracellular signal-regulated kinase (ERK), JUN N-terminal
kinase (JNK), KRAS, lung cancer, mitogen-activated protein kinase kinase kinase 19 (MAP3K19), YSK4,
kinome, cell proliferation
_____________________________________________________________________________________

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


Abstract JNK pathway activation. In summary, our results
Identifying additional mitogen-activated reveal that MAP3K19 directly activates the ERK
protein kinase (MAPK) pathway regulators is and JNK cascades and highlight a role for this
invaluable in aiding our understanding of the kinase in maintaining survival of KRAS-mutant
complex signaling networks that regulate cellular lung cancer cells.
processes, including cell proliferation and _______________________________________
survival. Here, using in vitro kinase assays and by
expressing wild-type (WT) or kinase-dead (KD) A large portion of the kinome remains
MAPK kinase kinase 19 (MAP3K19) in the poorly characterized, with the functions of
HEK293T cell line and assessing activation of the approximately 25% of kinases yet to be
extracellular signal-regulated kinase (ERK) and discovered (1). This includes several MAPKs.
JUN N-terminal kinase (JNK) signaling Elucidating the roles of understudied kinases in
pathways, we defined MAP3K19 as a novel cellular processes vital to oncogenesis will
regulator of MAPK signaling. We also observed provide potential candidates for therapeutic
that overexpression of WT MAP3K19 activates intervention (2–4). It has been well described that
both the ERK and JNK pathways in a panel of aberrant activation of kinase signaling
cancer cell lines. Furthermore, MAP3K19 contributes to cancer initiation and progression.
sustained ERK pathway activation in the The MAPK signaling pathways represent
presence of inhibitors targeting the RAF proto- essential signaling nodes that govern cell cycle
oncogene Ser/Thr protein kinase (RAF) and progression and cell survival, which underscores
MAPK/ERK kinase (MEK), indicating that the importance of elucidating the function of
MAP3K19 activates ERK via a RAF- novel MAPKs (5,6). A published study using an
independent mechanism. Findings from in vitro shRNA screen identified Mitogen-Activated
and in cell kinase assays demonstrate that Protein Kinase Kinase Kinase 19 (MAP3K19 or
MAP3K19 is a kinase that directly YSK4) as a genetic dependency in a panel of
phosphorylates both MEK and MAPK kinase 7 KRAS-mutant cancers (7). Consistent with a pro-
(MKK7). Results from an shRNA screen survival function for MAP3K19, shRNA-
indicated that MAP3K19 is essential for mediated knockdown of MAP3K19 suppressed
maintaining survival in KRAS-mutant cancers; HeLa cell proliferation and survival, further
therefore, we depleted or inhibited MAP3K19 in indicating that MAP3K19 plays a role in cancer
KRAS-mutant cancer cell lines and observed that cell survival (8). In addition, MAP3K19
this reduces viability and decreases ERK and overexpression was reported in patients with
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

chronic obstructive pulmonary disease (COPD), lines, band migration of WT MAP3K19 notably
a known risk factor for lung cancer (9,10). shifted following λ-PP treatment, minimizing the
MAP3K19 has been reported to regulate TGF-b- mobility gap between WT and KD MAP3K19
induced SMAD signaling and gene expression as (Figure 1E–F). Together, these results indicate
well as regulate NF-kB to promote the release of that MAP3K19 can activate both the JNK and
various cytokines, although the mechanisms ERK pathways upon overexpression.
underpinning these activities are unknown (9,10). We previously characterized the mixed-
Given that these large screening studies have lineage kinases (MLK1–4) as MEK kinases that
implicated MAP3K19 in promoting cancer cell can activate the ERK pathway in the presence of
growth, we aimed to determine the mechanism by RAF inhibitors. To investigate the mechanism by
which MAP3K19 promotes cancer cell viability. which MAP3K19 stimulates ERK pathway
Our data define MAP3K19 as a novel modulator activation, we overexpressed WT MAP3K19 in
of ERK and JNK signaling cascades and indicate HEK 293T cells and treated these cells with a
that this kinase may be an essential for cell pan-RAF inhibitor (L779450), a MEK inhibitor
survival in KRAS-mutant cancers. (AZD6244), or both the RAF and MEK
inhibitors. Overexpressed MLK1 was used as a
Results control. Expression of MAP3K19 resulted in an
MAP3K19 promotes ERK pathway activation increase in MEK phosphorylation that was

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


To determine the role of MAP3K19 in maintained even in the presence of the RAF
regulating downstream MAP2Ks, we expressed inhibitor, indicating that MAP3K19-mediated
wild-type (WT) or kinase-dead (KD) MAP3K19 activation of MEK is independent of RAF,
in a HEK 293T cell line and assessed activation similar to MLK1 (Figure 2A). Interestingly,
of the ERK and JNK signaling pathways. MAP3K19 could sustain a lower level of ERK
Compared to protein levels in empty vector phosphorylation, even in the presence of the
control cells and KD-transfected cells, expression MEK inhibitor or the combination of RAF and
of WT MAP3K19 markedly enhanced MEK inhibitors, which raises the possibility that
phosphorylation of MEK, ERK, and JNK MAP3K19 may be able to activate ERK in a
proteins (Figure 1A). We also observed an MEK-independent manner (Figure 2A). MLK1
upward mobility shift in comparing WT to KD was not able to maintain ERK phosphorylation in
MAP3K19, consistent with WT MAP3K19 being the presence of a MEK inhibitor, concordant with
posttranslationally modified to an active our previous findings (11). To confirm these
conformation in HEK 293T cells. To verify that results and verify that kinase activity is essential
MAP3K19 is an ERK pathway activator, we for ERK pathway activation, we overexpressed
overexpressed MAP3K19 in additional cancer WT or KD MAP3K19 in HEK 293T cells treated
cell lines (LK2, MCF-7, and HCT-116) and with a combination of RAF and MEK inhibitors
monitored ERK pathway activation (Figure 1B– and monitored ERK phosphorylation.
D). In all three cell lines, overexpression of WT Overexpression of MAP3K19 WT, but not KD,
MAP3K19 led to an increase in both MEK and led to an increase in MEK and ERK
ERK activation, as assessed by increases in phosphorylation that was retained even in the
phosphorylation of both MEK and ERK (Figure presence of the RAF and MEK inhibitor
1B–D, increases in phosphorylation are treatment. These data indicate that MAP3K19
quantified in bar graphs below representative catalytic activity is critical for ERK pathway
immunoblots). MAP3K19 WT protein was activation and suggest that MAP3K19 is a MEK
detected at a higher molecular weight than KD in kinase (Figure 2B).
these cell lines, providing further support that
MAP3K19 is posttranslationally modified to MAP3K19 is a direct MAP2K kinase
assume an active conformation. To determine To determine whether MAP3K19 is a
whether the mobility shift between WT and KD direct MEK kinase, we performed in vitro kinase
MAP3K19 is phosphorylation dependent, we assays using purified KD MEK1 as a substrate.
pretreated HEK 293T and LK2 cell lysates with Full-length MAP3K19 that was
lambda protein phosphatase (λ-PP). In both cell immunoprecipitated from cells phosphorylated

2
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

MEK1 in a kinase-dependent manner (Figure MAP3K19 migrated at the same mobility as KD


3A). To confirm these results, we used purified MAP3K19 in A375 cells, suggesting that
MAP3K19 kinase domain in an in vitro kinase overexpressed MAP3K19 is likely to be
assay with KD MEK1. Purified MLK1 kinase maintained in an inactive conformation in BRAF-
domain was used as a control. MAP3K19 directly V600E-positive melanoma cells (Figure 4B).
phosphorylated MEK, indicating that MAP3K19 Similar results were observed in the Sk-Mel-28
is a direct MEK kinase, similar to MLK1 (Figure cell line (Figure 4C). Lastly, expression of
3B). To determine whether MAP3K19 can MAP3K19 did not promote ERK pathway
phosphorylate ERK, we performed an in vitro activation in the presence of RAF or MEK
kinase assay using KD ERK2 as a substrate. inhibitors in the A375 cells, indicating that
Purified MEK1, used as a positive control, increased expression of MAP3K19 is unlikely to
catalyzed phosphorylation of ERK, but neither be an acquired mechanism of resistance to ERK
MLK1 nor MAP3K19 are ERK kinases (Figure pathway inhibitors in melanoma cells (Figure
3C). To verify that MAP3K19 was not inhibited 4D).
by RAF or MEK inhibitors, we conducted an in
vitro kinase assay using KD MEK1 as a substrate MAP3K19 enhances KRAS-mediated ERK
with purified MAP3K19 kinase domain or MLK1 activation and is required to maintain viability
in the presence of MEK and/or RAF inhibitors. in KRAS-mutant lung cancer cells

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


MAP3K19-dependent MEK phosphorylation MAP3K19 was identified as a genetic
was preserved in the presence of all drugs, dependency in KRAS-mutant cancers. Therefore,
confirming that RAF and MEK inhibitors do not we investigated whether MAP3K19 would
inhibit MAP3K19 (Figure 3D). To assess enhance KRAS-mediated activation of the ERK
whether MAP3K19 can phosphorylate MKK7 pathway. Expression of KRAS G12C mutant
(MAP2K7), an upstream JNK kinase, we increased both ERK and MEK activation as
performed an in vitro kinase assay using KD expected. Co-expression of MAP3K19 and
MKK7 as a substrate. MAP3K19 directly KRAS G12C led to a marked increase in the
phosphorylates MKK7, which leads to activation expression levels of MAP3K19 and a correlated
of JNK (Figure 3E). To rule out the possibility increase in ERK pathway activation compared to
that MAP3K19 may directly phosphorylate JNK, either KRAS or MAP3K19 alone (Figure 5A). To
we used KD JNK1 or JNK2 as substrates in an in determine whether MAP3K19 is required to
vitro kinase assay and purified MKK7 as a maintain KRAS-mutant lung cancer cell viability,
control. MAP3K19 did not phosphorylate JNK we first assessed a panel of lung cancer cell lines
(Figure 3F). for expression of endogenous MAP3K19 and
observed that MAP3K19 is expressed in most
MAP3K19 does not promote resistance to ERK lung cancer cells at varying levels. We then
pathway inhibitors in melanoma depleted MAP3K19 in lung cancer cells with WT
Based on our data showing that KRAS (H3122) or mutant KRAS (H2030 and
MAP3K19 sustains MEK pathway activation in H2122) by using two unique siRNAs (Figure 5C).
the presence of RAF and MEK inhibitors, we Loss of MAP3K19 resulted in reduced viability
explored the possibility that MAP3K19 may play in KRAS-mutant lung cancer cells, but not KRAS
a role in promoting resistance to ERK pathway WT lung cancer cells (Figure 5C). To determine
inhibitors, similar to the MLKs. We assessed whether MAP3K19 depletion would alter MAPK
expression of MAP3K19 and observed an pathway activation, we assessed MEK, ERK, and
increase in MAP3K19 mRNA levels in melanoma JNK phosphorylation in H2122 cells and
cell lines resistant to RAF inhibitors (Figure 4A). observed a decrease in ERK and JNK pathway
To determine whether MAP3K19 overexpression activation (Figure 5D). These results indicate that
can promote increased MEK and ERK activation MAP3K19 plays a role in maintaining KRAS-
in BRAF-V600E-mutant melanoma cell lines, we mutant lung cancer cell viability by regulating the
expressed doxycycline (dox)-inducible ERK and JNK pathways.
MAP3K19 in A375 cells. There was no increase
in MEK or ERK phosphorylation, and WT

3
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

A MAP3K19 inhibitor suppresses ERK current study describes MAP3K19 as a novel


activation and inhibits viability in RAS-mutant activator of the ERK and JNK pathways (Figure
lung cancer 7).
To identify possible pharmacological Mutations that activate the
inhibitors, we evaluated three kinase inhibitors RAF/MEK/ERK pathway occur in RAF proteins,
that were previously identified to inhibit such as the BRAF V600E mutation, and in the
MAP3K19 (AT-9283, NVP-TAE226, and GSK- RAS family of proteins, which are upstream
269962A) in an internal pan-kinase inhibitor activators of RAF. Although KRAS gain-of-
screen (12,13). In HEK 293T cells function mutations are detected in approximately
overexpressing MAP3K19, AT-9283 was the 25% of lung adenocarcinoma (LUAD) patients,
only compound that suppressed activation of the direct inhibition of all KRAS mutants, aside from
ERK pathway and promoted a downward shift in G12/13C, has not been therapeutically possible,
MAP3K19 migration, comparable to KD and resistance often develops to inhibitors of the
MAP3K19, indicating that this compound RAF/MEK/ERK pathway. An alternative
inhibits MAP3K19 (Figure 6A). To validate that approach for targeting KRAS-mutant tumors is
AT-9283 targets MAP3K19, we performed an in identification of targetable vulnerabilities
vitro kinase assay where MAP3K19 was through synthetic lethal screens (7). Our interest
pretreated with AT-9283. We observed a in MAP3K19 arises from two studies indicating

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


reduction in phosphorylation of inactive MEK at that this MAP3K may be involved in lung cancer.
both 1 µM and 5 µM of drug treatment, which One study identified MAP3K19 as a genetic
verifies that this compound is a MAP3K19 dependency in a panel of KRAS-mutant cancers,
inhibitor. Treatment of the KRAS-mutant lung including LUAD samples (7). The authors of the
cancer cell lines with AT-9283 suppressed second study observed that MAP3K19 is more
viability and promoted a decrease in ERK abundant in patients with COPD, a known risk
pathway activation (Figure 6C–D). However, factor for lung cancer (9,10). Consistent with
there was also a decrease in cell viability in the these observations, our data demonstrate that
H3122 cell line (KRAS WT) treated with AT- siRNA-mediated depletion or pharmacological
9283, likely due to off-target effects. Overall, inhibition of MAP3K19 significantly reduces cell
these results suggest that MAP3K19 could serve viability and decreases ERK and JNK pathway
as a novel therapeutic target in KRAS-mutant activation in KRAS-mutant lung cancer cells.
lung cancers. However, the AT-9283 inhibitor also decreased
the viability of KRAS WT LUAD cells,
Discussion indicating the need for additional studies using
MAP3Ks are Ser/Thr kinases that, upon more specific MAP3K19 inhibitors to further
activation, phosphorylate and activate MAP2Ks, validate MAP3K19 as a genetic vulnerability and
ultimately leading to activation of MAPKs (14). potential therapeutic target in KRAS-mutant lung
The RAFs (MAP3Ks)àMEK1/2 cancer cells.
(MAP2Ks)àERK1/2 (MAPKs) cascade is a Reactivation of MAPK signaling is a
classic and cancer-relevant example (15). In prominent mechanism of resistance to BRAF
addition to the RAF/MEK/ERK cascade, there inhibitors (17–23). From our observation that
are others, including those that activate JNKs, MAP3K19 maintained ERK activation in the
p38s, and ERK5 (16). Identifying novel presence of RAF and MEK inhibitors, we
regulators of MAPK signaling cascades is critical explored the role of MAP3K19 in regulating
to fully understand how dysregulation of these resistance to MAPK-targeted therapy. Although
pathways occurs and contributes to cancer and we observed that expression of MAP3K19 was
resistance to targeted therapies. Many MAP3Ks elevated in vemurafenib-resistant BRAF-mutant
are unexplored. Our identification of the melanoma cell lines, induced expression of
MAP3Ks MLK1–4 as kinases that confer MAP3K19 could not sustain ERK pathway
resistance to RAF inhibitors in melanoma activation in the presence of RAF or MEK
provides proof that analysis of these understudied inhibitors in melanoma cells. Thus, MAP3K19
kinases has therapeutic implications (11). Our likely does not play a role in promoting resistance

4
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

to ERK pathway inhibitors in these cells. Our (I/II) (Invitrogen). The following primary
findings establishing MAP3K19 as a novel direct antibodies were used: MAP3K19 (antibody was
kinase of MEK may be relevant in other cancer generated at the Dundee antibody production
models. Response to BRAF inhibitor therapy has facility in collaboration with Dr. James Hastie);
been limited in patients with BRAF-V600E- MLK1, MEK1/2, p-MEK1/2 (S217/221),
mutant metastatic colorectal cancer, and it is ERK1/2, p-ERK1/2 (T202/Y204), p-MKK7
possible that MAP3K19 plays a role in resistance (S271/T275), JNK, p-JNK (T183/Y185), GST,
in tumors of different origins (24). In addition, Tubulin (Cell Signaling Technology); and V5
MEK inhibitor therapy has limited efficacy (Bio-Rad). Tubulin antibody was used at 1:5,000
against NRAS-mutant melanoma in the clinic dilution. All other antibodies were used at
(25). Future studies examining MAP3K19 1:1,000 dilution. Sheep HRP-conjugated
activity in the context of BRAF-mutant colorectal antibody (Cell Signaling Technology) was used
cancer and NRAS-mutant melanoma may shed as a secondary antibody at 1:5,000 dilution in 5%
light on MAP3K19’s potential role in promoting bovine serum albumin for MAP3K19 detection.
resistance to ERK pathway inhibitors in these After incubation with anti-rabbit DyLight™ 680
cancers. Conjugate or anti-mouse DyLight™ 800
Overall, our findings indicate that Conjugate (Cell Signaling Technology) at
MAP3K19 is an upstream regulator of the ERK 1:10,000 dilution in Odyssey Blocking Buffer

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


and JNK cascades and is required to maintain (LI-COR Biosciences), bands were detected
lung cancer cell survival in KRAS-mutant using the ChemiDoc MP Imaging System (Bio-
cancers. Rad). All western blots are representative of at
least three independent experiments.
Experimental procedures
Cell lines and reagents Lambda protein phosphatase treatment
Lung cancer cell lines NCI-H2122 and NCI- HEK 293T or LK2 cells were lysed in RIPA
H2030 were acquired from American Type buffer and centrifuged at 4 °C. After MnCl2 (2
Culture Collection (ATCC). NCI-H3122 cells mM) was added to the lysates, samples were
were obtained from the National Cancer Institute treated with 0 or 1 µL of λ phosphatase (New
(NCI) Repository. All LUAD cell lines were England Biolabs) for 1.5 hours at 30 °C. Laemmli
maintained in RPMI-1640 medium (Quality buffer was added to terminate the reaction, and
Biological) supplemented with 10% fetal bovine samples were boiled for five minutes.
serum (FBS) (Atlanta Biologicals), 1%
penicillin-streptomycin (Invitrogen), and 1% qRT-PCR
GlutaMAX (Invitrogen). HEK 293T cells were Cells were collected in cold phosphate-bufferd
cultured in DMEM supplemented with 10% FBS, saline and pelleted by centrifugation at 4 °C.
1% penicillin-streptomycin, and 1% GlutaMAX. Total RNA was extracted using the RNeasy Mini
Inhibitors were purchased as follows: PLX4032 Kit in accordance with the manufacturer’s
(vemurafenib), U0126, AZD6244 (selumetinib), protocol (Qiagen). The quality and concentration
and GDC-0973 (cobimetinib) from Selleck of RNA were determined using the NanoDrop
Chemicals; L779450 from Abcam; and AT-9283 One spectrophotometer (Thermo Scientific).
from Cayman Chemical. All inhibitors were Total RNA (1 μg) was reverse-transcribed using
dissolved in DMSO, and aliquots were stored at the iScript kit (Bio-Rad), and qPCR was
−20 °C. performed using SYBR Green (Bio-Rad). The
following primer sequences were used:
Western blot MAP3K19 F:
Forty-eight hours after transient transfection, GCATCAGCAGAAGTGAGGAG,
cells were treated with inhibitors or DMSO MAP3K19 R: TCAACACCTTCTGTCCTGGG,
vehicle control for one hour before lysis with M- Actin F: GGCACCCAGCACAATGAAGA,
PER mammalian protein extraction reagent Actin R: ACTCCTGCTTGCTGATCCAC.
(Thermo Scientific) supplemented with 1% Cycle numbers were normalized to β-actin, and
protease inhibitor and 1% phosphatase inhibitors control conditions were scaled to one.

5
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

Experiments were conducted in triplicate with 10-cm-plate format. Knockdown of mRNA was
internal duplicates. assessed by qRT-PCR 48 hours after transfection.

Cell viability assays Immunoprecipitation and kinase assays


Cells were treated with inhibitor at the indicated Forty-eight hours after transfection, cells were
concentrations and time points or were reverse- treated with inhibitors or DMSO and lysed with
transfected with siRNA for 72 hours, in a 96- Triton X-100 lysis buffer (Cell Signaling
well-plate format. Cells were fixed with Technology) supplemented with protease
glutaraldehyde for 20 minutes, then stained with inhibitors (Roche) dissolved in sterile water. Cell
1% crystal violet in 10% methanol solution for 15 lysates were incubated overnight (4 °C) with anti-
minutes. Wells were washed thoroughly and V5 antibody (1:100), then incubated with 50 μL
allowed to air dry. For quantification, 100 μL of of a 50% slurry of Protein G Dynabeads (Thermo
33% acetic acid was added to each well and Scientific) for one hour. Beads were washed three
incubated for at least one hour with shaking. times, first with lysis buffer, then kinase buffer
Absorbance values were read at 595 nm. Data are (Cell Signaling Technology). The kinase assay
represented as mean cell viability normalized to was performed in the presence of ATP (50 µM)
control ± SEM of triplicate experiments, each and 100 ng of kinase-inactive MEK1 as substrate
consisting of three technical replicates. (Carna Biosciences) at 30 °C for 30 minutes. The

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


kinase assay was terminated after adding SDS
Transient transfections sample buffer and boiling for 15 minutes.
For ectopic expression of MAP3K19, HEK 293T Proteins were resolved by SDS–PAGE and
cells were seeded at 50% confluency and allowed analyzed by Western blotting.
to adhere overnight. The following day, cells
were transfected with the designated amount of In vitro kinase assay
plasmid DNA using jetPRIME transfection Recombinant human (GST)-tagged MAP3K19
reagent (Polyplus Transfection) for the indicated (YSK4) kinase domain (Thermo Fisher
time points. MAP3K19 and MLK1 wild-type Scientific) or human MLK1 kinase domain
constructs were purchased as entry vector (Carna Biosciences) was incubated at room
(OriGene). The KD mutation was introduced by temperature with kinase-inactive MEK1, ERK2,
site-directed mutagenesis (QuikChange II kit, MKK7, JNK1, or JNK2 (Carna Biosciences) in
Stratagene) in the critical lysine required to bind the absence or presence of MAPK pathway
ATP (within the VAIK motif) (K1089M- inhibitors for five minutes. The kinase reaction
MAP3K19). The following primer sequences was initiated with the addition of ATP (50 µM) at
were used: 30 °C for 30 minutes, and the reaction was
K1089M F: GGTATCCAAAGCCACCTGCAT stopped by adding 4× SDS sample buffer. After
TACAGCTATTAGCTGTCCT, the samples were boiled for 15 minutes, the
K1089M R: AGGACAGCTAATAGCTGTAAT proteins were resolved by SDS–PAGE and
GCAGGTGGCTTTGGATACC. analyzed by Western blotting.
MAP3K19 constructs were cloned into
pLenti6.3/TO/V5-DEST vector (Thermo Generation of tetracycline-inducible cell lines
Scientific) with the Gateway system according to WT or KD MAP3K19 plasmids cloned into
the manufacturer’s instructions. KRAS G12C pLenti6.3/TO/V5-DEST and pLenti3.3/TR
plasmid (pCDH-CMV-HA backbone) was kindly vector (for tetracycline repressor expression)
provided by Dr. Deborah Morrison (National were transfected into HEK 293FT cells using
Cancer Institute, USA). Lipofectamine 2000, and lentiviral supernatant
For RNAi experiments, cells were was collected. Parental A375 melanoma cells
reverse-transfected using Lipofectamine were transduced with lentiviral stocks and treated
RNAiMAX (Thermo Scientific) and 40 nM of with antibiotics for selection (blasticidin
siRNA oligos (OriGene) diluted with Opti-MEM [Invitrogen] and Geneticin [Gibco]).
I Reduced Serum Medium (Thermo Scientific) in Doxycycline (Sigma) was used to induce
expression of MAP3K19.

6
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

Generation of vemurafenib-resistant cell lines


Melanoma cell lines A375 and MDA-MB-435
were generated by chronic treatment with 1 μM
vemurafenib, as described previously (11). Cells
were cultured in media with fresh vemurafenib
added 1.

Statistical analysis
Statistical analyses were performed using
unpaired Student’s t-test or one-way ANOVA
(Dunnett’s multiple comparisons test) on
GraphPad Prism software (GraphPad Software
Inc.). *, p < 0.05; **, p < 0.01; ***, p < 0.001.

Downloaded from http://www.jbc.org/ by guest on June 2, 2020

7
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

All data reported are contained within the manuscript.

Acknowledgments: We thank Dr. Deborah Morrison and her lab (NCI) for providing reagents as well as
advice for our studies and Roger Liang for his analysis of MAP3K19 constructs. We are also grateful to
Dr. Craig Thomas (NCI) for his helpful suggestions regarding MAP3K19 inhibitors.

Conflict of interest: The authors declare that they have no conflicts of interest with the contents of this
article.

References

1. Fedorov, O., Muller, S., and Knapp, S. (2010) The (un)targeted cancer kinome. Nat Chem Biol 6,
166-169
2. Knapp, S. (2018) New opportunities for kinase drug repurposing and target discovery. Br J Cancer
118, 936-937
3. Bhullar, K. S., Lagaron, N. O., McGowan, E. M., Parmar, I., Jha, A., Hubbard, B. P., and
Rupasinghe, H. P. V. (2018) Kinase-targeted cancer therapies: progress, challenges and future
directions. Mol Cancer 17, 48

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


4. Cohen, P., and Alessi, D. R. (2013) Kinase drug discovery--what's next in the field? ACS Chem
Biol 8, 96-104
5. Atay, O., and Skotheim, J. M. (2017) Spatial and temporal signal processing and decision making
by MAPK pathways. J Cell Biol 216, 317-330
6. Chang, L., and Karin, M. (2001) Mammalian MAP kinase signalling cascades. Nature 410, 37-40
7. Barbie, D. A., Tamayo, P., Boehm, J. S., Kim, S. Y., Moody, S. E., Dunn, I. F., Schinzel, A. C.,
Sandy, P., Meylan, E., Scholl, C., Frohling, S., Chan, E. M., Sos, M. L., Michel, K., Mermel, C.,
Silver, S. J., Weir, B. A., Reiling, J. H., Sheng, Q., Gupta, P. B., Wadlow, R. C., Le, H., Hoersch,
S., Wittner, B. S., Ramaswamy, S., Livingston, D. M., Sabatini, D. M., Meyerson, M., Thomas, R.
K., Lander, E. S., Mesirov, J. P., Root, D. E., Gilliland, D. G., Jacks, T., and Hahn, W. C. (2009)
Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature
462, 108-112
8. Grueneberg, D. A., Degott, S., Pearlberg, J., Li, W., Davies, J. E., Baldwin, A., Endege, W.,
Doench, J., Sawyer, J., Hu, Y., Boyce, F., Xian, J., Munger, K., and Harlow, E. (2008) Kinase
requirements in human cells: I. Comparing kinase requirements across various cell types.
Proceedings of the National Academy of Sciences of the United States of America 105, 16472-
16477
9. Boehme, S. A., Franz-Bacon, K., Ludka, J., DiTirro, D. N., Ly, T. W., and Bacon, K. B. (2016)
MAP3K19 Is Overexpressed in COPD and Is a Central Mediator of Cigarette Smoke-Induced
Pulmonary Inflammation and Lower Airway Destruction. PLoS One 11, e0167169
10. Durham, A. L., and Adcock, I. M. (2015) The relationship between COPD and lung cancer. Lung
Cancer 90, 121-127
11. Marusiak, A. A., Edwards, Z. C., Hugo, W., Trotter, E. W., Girotti, M. R., Stephenson, N. L., Kong,
X., Gartside, M. G., Fawdar, S., Hudson, A., Breitwieser, W., Hayward, N. K., Marais, R., Lo, R.
S., and Brognard, J. (2014) Mixed lineage kinases activate MEK independently of RAF to mediate
resistance to RAF inhibitors. Nat Commun 5, 3901
12. Anastassiadis, T., Deacon, S. W., Devarajan, K., Ma, H., and Peterson, J. R. (2011) Comprehensive
assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotechnol 29,
1039-1045
13. Davis, M. I., Hunt, J. P., Herrgard, S., Ciceri, P., Wodicka, L. M., Pallares, G., Hocker, M., Treiber,
D. K., and Zarrinkar, P. P. (2011) Comprehensive analysis of kinase inhibitor selectivity. Nat
Biotechnol 29, 1046-1051

8
MAP3K19 is a novel therapeutic target in RAS-mutant cancers

14. Gallo, K. A., and Johnson, G. L. (2002) Mixed-lineage kinase control of JNK and p38 MAPK
pathways. Nature reviews. Molecular cell biology 3, 663-672
15. Morrison, D. K. (2012) MAP kinase pathways. Cold Spring Harb Perspect Biol 4
16. Hoang, V. T., Yan, T. J., Cavanaugh, J. E., Flaherty, P. T., Beckman, B. S., and Burow, M. E.
(2017) Oncogenic signaling of MEK5-ERK5. Cancer Lett 392, 51-59
17. Arozarena, I., and Wellbrock, C. (2017) Overcoming resistance to BRAF inhibitors. Ann Transl
Med 5, 387
18. Nazarian, R., Shi, H., Wang, Q., Kong, X., Koya, R. C., Lee, H., Chen, Z., Lee, M. K., Attar, N.,
Sazegar, H., Chodon, T., Nelson, S. F., McArthur, G., Sosman, J. A., Ribas, A., and Lo, R. S.
(2010) Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS
upregulation. Nature 468, 973-977
19. Shi, H., Moriceau, G., Kong, X., Lee, M. K., Lee, H., Koya, R. C., Ng, C., Chodon, T., Scolyer, R.
A., Dahlman, K. B., Sosman, J. A., Kefford, R. F., Long, G. V., Nelson, S. F., Ribas, A., and Lo,
R. S. (2012) Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-
mediated acquired B-RAF inhibitor resistance. Nat Commun 3, 724
20. Su, F., Viros, A., Milagre, C., Trunzer, K., Bollag, G., Spleiss, O., Reis-Filho, J. S., Kong, X.,
Koya, R. C., Flaherty, K. T., Chapman, P. B., Kim, M. J., Hayward, R., Martin, M., Yang, H.,
Wang, Q., Hilton, H., Hang, J. S., Noe, J., Lambros, M., Geyer, F., Dhomen, N., Niculescu-Duvaz,

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


I., Zambon, A., Niculescu-Duvaz, D., Preece, N., Robert, L., Otte, N. J., Mok, S., Kee, D., Ma, Y.,
Zhang, C., Habets, G., Burton, E. A., Wong, B., Nguyen, H., Kockx, M., Andries, L., Lestini, B.,
Nolop, K. B., Lee, R. J., Joe, A. K., Troy, J. L., Gonzalez, R., Hutson, T. E., Puzanov, I.,
Chmielowski, B., Springer, C. J., McArthur, G. A., Sosman, J. A., Lo, R. S., Ribas, A., and Marais,
R. (2012) RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF
inhibitors. N Engl J Med 366, 207-215
21. Villanueva, J., Vultur, A., Lee, J. T., Somasundaram, R., Fukunaga-Kalabis, M., Cipolla, A. K.,
Wubbenhorst, B., Xu, X., Gimotty, P. A., Kee, D., Santiago-Walker, A. E., Letrero, R., D'Andrea,
K., Pushparajan, A., Hayden, J. E., Brown, K. D., Laquerre, S., McArthur, G. A., Sosman, J. A.,
Nathanson, K. L., and Herlyn, M. (2010) Acquired resistance to BRAF inhibitors mediated by a
RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer
Cell 18, 683-695
22. Wilson, T. R., Fridlyand, J., Yan, Y., Penuel, E., Burton, L., Chan, E., Peng, J., Lin, E., Wang, Y.,
Sosman, J., Ribas, A., Li, J., Moffat, J., Sutherlin, D. P., Koeppen, H., Merchant, M., Neve, R., and
Settleman, J. (2012) Widespread potential for growth-factor-driven resistance to anticancer kinase
inhibitors. Nature 487, 505-509
23. Sun, C., Wang, L., Huang, S., Heynen, G. J., Prahallad, A., Robert, C., Haanen, J., Blank, C.,
Wesseling, J., Willems, S. M., Zecchin, D., Hobor, S., Bajpe, P. K., Lieftink, C., Mateus, C.,
Vagner, S., Grernrum, W., Hofland, I., Schlicker, A., Wessels, L. F., Beijersbergen, R. L., Bardelli,
A., Di Nicolantonio, F., Eggermont, A. M., and Bernards, R. (2014) Reversible and adaptive
resistance to BRAF(V600E) inhibition in melanoma. Nature 508, 118-122
24. Prahallad, A., Sun, C., Huang, S., Di Nicolantonio, F., Salazar, R., Zecchin, D., Beijersbergen, R.
L., Bardelli, A., and Bernards, R. (2012) Unresponsiveness of colon cancer to BRAF(V600E)
inhibition through feedback activation of EGFR. Nature 483, 100-103
25. Thumar, J., Shahbazian, D., Aziz, S. A., Jilaveanu, L. B., and Kluger, H. M. (2014) MEK targeting
in N-RAS mutated metastatic melanoma. Mol Cancer 13, 45

9
Figure 1
A B C D

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


HEK293T
8 2.5 * EV
EV *
Relative protein density
Relative protein density

* WT
* WT 2.0
6 KD
KD 1.5
4
* 1.0
* *
2 ** **
0.5

0 0.0
p-MEK:MEK p-ERK:ERK p-JNK:JNK p-MEK:MEK p-ERK:ERK p-MEK:MEK p-ERK:ERK p-MEK:MEK p-ERK:ERK

HEK293T LK2 MCF7 HCT-116

Figure 1. MAP3K19 activates MAPK family members. A,


HEK293T; B, LK2; C, MCF7; or D, HCT-116 cells were
E F transiently transfected with empty vector (EV), wild-type (WT),
or kinase dead (KD-K1089M) MAP3K19. Phosphorylation
status of MAPK family members was assessed by Western
blot. Band density was quantified by ImageJ software. Data are
shown as mean phospho:total protein density normalized to EV
control ± SD. * p < 0.05; ** p < 0.01. E, HEK293T, or F, LK2
cells were transfected with EV, WT, or KD MAP3K19, and
lysates were treated with 0 or 1 μL of lambda protein
phosphatase (λ-PP). Western blot was performed.
Figure 2 Fig2B

A HEK293T B HEK293T
vehicle
1.6 ***

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


*** ***

Relative protein density,


BRAFi + S
1.2 BRAFi + C

p-MEK:MEK
0.8

0.4

0.0 Fig2B
50 kDa
EV MAP3K19-WT MAP3K19-KD

1.6 vehicle

Relative protein density,


BRAFi +
***
1.2 BRAFi +

p-ERK:ERK
37 kDa
37 kDa

Fig2A 0.8

1.5
vehicle
Relative protein density,

*** *** 0.4


37 kDa
** ** L779450 *** ***
*** ***
p-MEK:MEK

1.0 * AZD6244 0.0


combination EV MAP3K19-WT MAP3K19-KD

0.5

Figure 2. MAP3K19 maintains MEK phosphorylation in the presence of RAF inhibitors. A, 48


0.0
EV Fig2A
MAP3K19 MLK1
h after transfection with empty vector (EV), MAP3K19-WT, or MLK1-WT, HEK293T cells were
2.0 treated with DMSO vehicle control, 1 μM L779450, 1 μM AZD6244, or a combination of both RAF
vehicle
Relative protein density,

and MEK inhibitors for 1 h, and Western blot was performed on cell lysates. B, 48 h after
*** *** L779450
1.5 transfection with EV, wild-type (WT), or kinase dead (KD) MAP3K19, HEK293T cells were treated
p-ERK:ERK

AZD6244
***
*** combination
with DMSO vehicle control or indicated inhibitors: 1 μM vemurafenib (BRAFi), 1 μM selumetinib (S
1.0
** – MEKi), or 500 nM cobimetinib (C – MEKi) for 1 h, and Western blot was performed on cell
*
0.5 lysates. Band density was quantified by ImageJ software. Data are shown as mean phospho:total
protein density ± SD. Dunnett's multiple comparisons test was used for statistical analysis, with EV-
0.0
EV MAP3K19 MLK1 DMSO group as control. * p < 0.05; ** p < 0.01; *** p < 0.001.
Figure 3
A HEK293T B C D

ERK2 + MAP3K19
MEK1 + MAP3K19

ERK2 + MEK1
ERK2 + MLK1
MEK1 + MLK1
250 kDa

◇ ◇ ◇ ◇ ◇ ◇ ◇

ERK2
MEK1
150 kDa

75 k Da
75 k Da
p-MEK1/2 p-ERK1/2

GST GST
MAP3K19 75 k Da MAP3K19 75 k Da
MEK1 ERK2

75 k Da 75 k Da
MEK1/2 ERK1/2
KA pMEK ◇ ◇ ◇ ◇ ◇ ◇ ◇ ◇ ◇
IP pMEK KA pERK
1.0
** 1.5
Relative protein density,
0.4 ***

Relative protein density,


Relative protein density,

** 0.8
p-MEK:MEK

p-ERK:ERK
0.3 0.6 1.0
p-MEK:MEK

0.4
0.2 0.5
0.2

0.1 0.0 0.0

19

1
-

19

LK

EK
LK

K
K

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


P3

M
0.0
P3

A
A

M
EV WT KD
M

KA
E F
JNK1 + MAP3K19

JNK2 + MAP3K19
JNK1 + MKK7

JNK2 + MKK7
6

Relative protein density,


◇ ◇ ◇ ◇ ◇ ◇ ◇ ◇ ◇ ***
JNK1

JNK2
***

p-MEK:MEK
75 k Da 75 k Da 4 ***
p-JNK p-JNK
** ***
**
GST GST *
MAP3K19/MKK7 75 k Da MAP3K19/MKK7 *
J NK1
75 k Da
2
J NK2
KA pMKK7 KA pJNK1 KA pJNK2
1.5 1.2 0.8
*** *
Relative protein density,

Relative protein density,

Relative protein density,

0
*
0.6
p-MKK7:MKK7

79 e

U 2
45 ZD 6

45 ZD 4

44
ZD 4

ZD 4
PL 50
p-JNK:JNK

1.0 0.8
p-JNK:JNK

L7 cl

03

79 A 012

79 A 624
**

4
62
hi

0+ 62

0+ 62
4
X4
ve
0.4

A
0.5 0.4
0.2

L7

L7
0.0 0.0 0.0
-

19

19

7
-

19

K
LK

K
K

K
P3

P3
P3

M
A

A
A

M
M

Figure 3. MAP3K19 directly phosphorylates MAP2Ks. A, MAP3K19 was immunoprecipitated from HEK293T cells and subjected to a kinase assay
with kinase-inactive MEK1. B and C, In vitro kinase assay using recombinant MAP3K19 protein and kinase-inactive MEK1 or ERK2, respectively. D,
Kinase-inactive MEK1 and purified GST-MAP3K19 or GST-MLK1 kinase domain were subjected to in vitro kinase assay in the presence or absence
of inhibitors: 5 μM L779450, 1 μM PLX4032, 5 μM U0126, or 2 μM AZD6244. E and F, In vitro kinase assay using recombinant MAP3K19 protein and
kinase-inactive MKK7 or JNK1/2, respectively. Data are shown as mean phospho:total protein density ± SD. Dunnett's multiple comparisons test was
used for statistical analysis, with samples in first lane as control. * p < 0.05; ** p < 0.01; *** p < 0.001. ◇denotes kinase-inactive
Figure 4
A RT-qPCR:A375
vemurafenib-resistant (VR) melanoma cells
MDA-MB-435 A375 ESprimer MDA-MB-435
B A375 C

Relative Expression, MAP3K19


Relative Expression, MAP3K19

2.5 3
**
2.0 *

2
1.5

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


1.0 50 kDa
1
0.5

0.0 0
ctrl C2 C4 ctrl 1E 2D
VR clones VR clones

D A375 MAP3K19-WT MAP3K19-KD


A375
37 kDa

Sk-Mel-28
- - - - + + + + + + + + doxycycline 1.2
- dox
1.5 EV

Relative protein density


+ dox

Relative protein density


- + - + - + - + - + - + PLX4032 WT

- - + + - - + + - - + + AZD6244 KD
0.8 1.0

MAP3K19 150kDa
150 k Da
0.4 0.5
p-ERK1/2
37kDa
37 k Da

0.0 0.0
ERK1/2 WT KD WT KD p:t MEK p:t ERK
50 kDa
p:t MEK p:t ERK
50 kDa
tubulin
Figure 4. MAP3K19 expression does not promote resistance to ERK pathway inhibitors in
melanoma cell lines. A, RT-(q)PCR analysis of MAP3K19 expression in vemurafenib-resistant
0.04 WT KD
clones derived from A375 (C2, C4) or MDA-MB-435 (1E, 2D) melanoma cells. B, Parental A375
Relative protein density,

melanoma cell line was used to generate cells with doxycycline (dox)-inducible expression of
0.03 MAP3K19. Protein expression of MAP3K19 was confirmed, and phosphorylation of MEK1/2 and
ERK1/2 was assessed by Western blot 24 h after dox treatment. Data are shown as mean
p:t ERK

0.02 phospho:total (p:t) protein density ± SD. C, Sk-Mel-28 cells were transiently transfected with empty
*
* vector (EV), wild-type (WT), or kinase dead (KD) MAP3K19. Phosphorylation status of MAPK family
** *
0.01 ** ** ** ** ** members was assessed by Western blot. Data are shown as mean p:t protein density ± SD. D, Dox
was added to A375-TR cells for 24 h to induce expression of MAP3K19, then cells were treated with
0.00 DMSO vehicle control, 500 nM PLX4032 (BRAFi), 500 nM AZD6244 (MEKi), or a combination of both
- - - - + + + + + + + + dox
- + - + - + - + - + - + PLX4032 BRAF and MEK inhibitors for 1 h. Western blot was performed on cell lysates. Data are shown as
- - + + - - + + - - + + AZD6244 mean p:t protein density ± SD, with MAP3K19-WT (-dox) DMSO as control. * p < 0.05; ** p < 0.01.
Figure 5
B
300

A HEK293T

Relative protein density,


200

MAP3K19

MW ladder
100

0
- - WT KD WT KD MAP3K19
- + - - + + KRAS-G12C
1.5

Relative protein density,


***

1.0 * C

p:t MEK
** 200 KRASWT KRASG12C G12C

Fold Change, MAP3K19 mRNA


1.5 KRASWT KRAS
0.5

Cell Viability (% ctrl)


150
ctrl
ctrl
si-A

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


0.0 1.0
si-A
- - WT KD WT KD MAP3K19
100 si-B
- + - - + + KRAS-G12C
** * * si-B
2.5 *
*** *** * *
Relative protein density, *** 50 0.5
2.0 ** *
*
p:t ERK
1.5
0
0.0
H3122 H2030 H2122
1.0 H3122 H2030 H2122

D H2122 0.5
200 KRASWT
LUAD Cell Lines
LUAD Cell Lines
KRASG12C
0.0
- - WT KD WT KD MAP3K19

Cell Viability (% ctrl)


- + - - + + KRAS-G12C
150 200 ctrl
ctrl si-A

Cell Viability (% ctrl)


1.2
Relative protein density

si-A 100 150 si-B


* *
* si-B ** * *
0.8
50 100
* * *
*** * ** *
0.4 50
0
H3122 H2030 H2122
0.0 0 LUAD Cell Lines
MAP3K19 p-MEK p-ERK p-JNK H3122 H2030 H2122
LUAD Cell Lines
Figure 5. MAP3K19 cooperates with KRAS G12C to enhance ERK activation and plays a role in maintaining cell viability in KRAS-mutant lung
cancer cells. A, Western blot analysis of MAPK phosphorylation changes due to transient expression of MAP3K19-WT, MAP3K19-KD, or KRAS G12C.
Data are shown as mean protein density (phospho:total for MEK and ERK) ± SD. B, Western blot of endogenous MAP3K19 in lung cancer cell lines. C,
(Top) RT-(q)PCR validation of siRNA-mediated MAP3K19 knockdown in LUAD cell lines 48 h after transfection. (Bottom) Cells were seeded in 96-well
plates 24 h after transfection, and cell viability was evaluated after 48 h by crystal violet assay. Data are represented as percent cell viability normalized
to control ± SD. D, Phosphorylation status of MAPK family members following depletion of MAP3K19 expression in H2122 cells. Data are shown as
mean phosphorylated protein density normalized to control (set to 1) ± SD. * p < 0.05; ** p < 0.01; *** p < 0.001.
Figure 6
A HEK293T B KA
1.5

Relative protein density,


1.0

p:t MEK
**
*
0.5

0.0
vehicle 1 µM 5 µM 1 µM
AT-9283 AZD6244

vehicle

C H2122
1.5
5 µM AT-9283
10 µM AT-9283

Relative protein density


1.0 *
* * *

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


6A pMEK **
0.5 ** **
5 KD EV MAP3K19-WT
Relative protein density,

4
*
* 0.0
MAP3K19 p:t MEK p:t ERK p:t JNK
p:t MEK

D
3 ***
***
150 H31
2 150 H3122

Cell Viability (% control)


Cell Viability (% control)
* H20
H2030
1 ***
100 H21
H2122
** ** 100
0
*
vehicle

vehicle

1 µM AT

2.5 µM AT

1 µM NVP

vehicle
1 µM GSK

100 nM AT

1 µM AT

2.5 µM AT

100 nM NVP

1 µM NVP

1 µM GSK

100 nM GSK

** ***
50 **
50 **
***
*** ******
Treatment ***
***
0
0
vehicle 1 µM 5 µM
KD EV MAP3K19-WT vehicle 1 µM 5 µM
3 AT-9283
AT-9283
***
Relative protein density,

Figure 6. Analysis of MAP3K19 inhibitor. A, HEK293T cells were transiently transfected with
**
empty vector (EV) or wild-type (WT) MAP3K19. After 48 h, cells were treated with DMSO
2 * vehicle control or indicated doses of inhibitors for 1 h. Western blot was then performed on cell
*
p:t ERK

** lysates. Data are shown as mean phospho:total (p:t) protein density ± SD with EV vehicle-
treated control set to 1. B, Kinase-inactive MEK1 and purified GST-MAP3K19 kinase domain
1 ** were subjected to an in vitro kinase assay in the presence or absence of inhibitors. Data are
** shown as mean protein density (p:t) ± SD with vehicle-treated control set to 1. C, Western blot
*** *** of MAPK pathway expression and activation in H2122 cells treated with AT-9283 for 24 h. Data
0
are shown as mean protein density (p:t for MEK, ERK, and JNK) ± SD with vehicle-treated
vehicle

vehicle

1 µM AT

2.5 µM AT

1 µM NVP

vehicle
1 µM GSK

100 nM AT

1 µM AT

2.5 µM AT

100 nM NVP

1 µM NVP

1 µM GSK

100 nM GSK

control set to 1. D, Viability of lung adenocarcinoma cells was evaluated by crystal violet assay
after 72-h treatment with AT-9283. Data are represented as percent cell viability normalized to
Treatment vehicle-treated control ± SD. * p < 0.05; ** p < 0.01; *** p < 0.001.
Figure 7

Downloaded from http://www.jbc.org/ by guest on June 2, 2020


Figure 7. Schematic representation of MAP3K19
regulating MAPK pathways. MAP3K19 can directly
phosphorylate MEK1/2 and MKK7, leading to ERK
and JNK activation.
The protein kinase MAP3K19 phosphorylates MAP2Ks and thereby activates ERK
and JNK kinases and increases viability of KRAS-mutant lung cancer cells
Van T Hoang, Katherine Nyswaner, Pedro Torres-Ayuso and John Brognard
J. Biol. Chem. published online April 30, 2020

Access the most updated version of this article at doi: 10.1074/jbc.RA119.012365

Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

Downloaded from http://www.jbc.org/ by guest on June 2, 2020

You might also like