ArticlePDF Available

Identification of Mutant K-Ras-dependent Phenotypes Using a Panel of Isogenic Cell Lines

Authors:

Abstract

To assess the consequences of endogenous mutant K-Ras, we analyzed the signaling and biological properties of a small panel of isogenic cell lines. These include the cancer cell lines DLD1, HCT116, and Hec1A, in which either the WT or mutant K-ras allele has been disrupted, and SW48 colorectal cancer cells and human mammary epithelial cells in which a single copy of mutant K-ras was introduced at its endogenous genomic locus. We find that single copy mutant K-Ras causes surprisingly modest activation of downstream signaling to ERK and Akt. In contrast, a negative feedback signaling loop to EGFR and N-Ras occurs in some, but not all, of these cell lines. Mutant K-Ras also had relatively minor effects on cell proliferation and cell migration but more dramatic effects on cell transformation as assessed by growth in soft agar. Surprisingly, knock-out of the wild type K-ras allele consistently increased growth in soft agar, suggesting tumor-suppressive properties of this gene under these conditions. Finally, we examined the effects of single copy mutant K-Ras on global gene expression. Although transcriptional programs triggered by mutant K-Ras were generally quite distinct in the different cell lines, there was a small number of genes that were consistently overexpressed, and these could be used to monitor K-Ras inhibition in a panel of human tumor cell lines. We conclude that there are conserved components of mutant K-Ras signaling and phenotypes but that many depend on cell context and environmental cues. Background: Many studies analyzing K-Ras function rely on overexpression. Results: Knock-out and knock-in of endogenous mutant K-ras have modest effects on downstream signaling but strong effects on gene expression and transformation. Conclusion: Genomic manipulation allows physiological determination of WT and mutant K-Ras consequences. Significance: Gene expression patterns can be used to monitor inhibition of mutant K-Ras.
Identification of Mutant K-Ras-dependent Phenotypes Using
a Panel of Isogenic Cell Lines
*
S
Received for publication, June 20, 2012, and in revised form, November 23, 2012 Published, JBC Papers in Press, November 27, 2012, DOI 10.1074/jbc.M112.394130
Steffan Vartanian
, Carolyn Bentley
, Matthew J. Brauer
§
,LiLi
§
, Senji Shirasawa
, Takehiko Sasazuki
,
Jung-Sik Kim**, Pete Haverty
§
, Eric Stawiski
‡‡
, Zora Modrusan
‡‡
, Todd Waldman**, and David Stokoe
‡1
From the Departments of
Discovery Oncology,
§
Bioinformatics, and
‡‡
Molecular Biology, Genentech Inc., South San Francisco,
California 94080, the
Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan, the
Institute for Advanced Study, Kyushu University, Fukuoka 812-8581, Japan, and the **Department of Oncology and Tumor
Biology Training Program, Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine,
Washington, D. C. 20037
Background: Many studies analyzing K-Ras function rely on overexpression.
Results: Knock-out and knock-in of endogenous mutant K-ras have modest effects on downstream signaling but strong effects
on gene expression and transformation.
Conclusion: Genomic manipulation allows physiological determination of WT and mutant K-Ras consequences.
Significance: Gene expression patterns can be used to monitor inhibition of mutant K-Ras.
To assess the consequences of endogenous mutant K-Ras, we
analyzed the signaling and biological properties of a small panel
of isogenic cell lines. These include the cancer cell lines DLD1,
HCT116, and Hec1A, in which either the WT or mutant K-ras
allele has been disrupted, and SW48 colorectal cancer cells and
human mammary epithelial cells in which a single copy of
mutant K-ras was introduced at its endogenous genomic locus.
We find that single copy mutant K-Ras causes surprisingly mod-
est activation of downstream signaling to ERK and Akt. In con-
trast, a negative feedback signaling loop to EGFR and N-Ras
occurs in some, but not all, of these cell lines. Mutant K-Ras also
had relatively minor effects on cell proliferation and cell migra-
tion but more dramatic effects on cell transformation as
assessed by growth in soft agar. Surprisingly, knock-out of the
wild type K-ras allele consistently increased growth in soft agar,
suggesting tumor-suppressive properties of this gene under
these conditions. Finally, we examined the effects of single copy
mutant K-Ras on global gene expression. Although transcrip-
tional programs triggered by mutant K-Ras were generally quite
distinct in the different cell lines, there was a small number of
genes that were consistently overexpressed, and these could be
used to monitor K-Ras inhibition in a panel of human tumor cell
lines. We conclude that there are conserved components of
mutant K-Ras signaling and phenotypes but that many depend
on cell context and environmental cues.
Ras was one of the first human oncogenes to be discovered
and remains one of the most frequently mutated genes across
multiple human tumors. Ras is a small membrane-bound GTP-
binding protein that recruits effector proteins such as Raf,
phosphoinositide 3-kinase, and RalGDS when bound to GTP,
resulting in activation of these proteins and their downstream
signaling events. Activation of Ras causes a wide variety of bio-
logical consequences depending on the cell type and stimuli,
including increased cell growth, proliferation, survival, differ-
entiation, and morphogenesis. Ras is able to hydrolyze GTP to
GDP, resulting in its self-inactivation, although this process is
slow and is accelerated by a family of proteins termed GTPase-
activating proteins (GAPs). Intrinsic exchange of GDP to GTP
is also quite slow, and this process is accelerated by a family of
proteins termed guanine nucleotide exchange factors (GEFs).
Through the regulation of GAP and GEF activities, the GTP
levels of Ras proteins are carefully adjusted depending on envi-
ronmental conditions, resulting in the maintenance of appro-
priate cellular homeostasis. Mutations of Ras that occur in
human tumors disrupt this process by preventing both intrinsic
and GAP-mediated hydrolysis, causing constitutive GTP asso-
ciation and uncontrolled signaling and proliferation (for
review, see Ref. 1).
Three ras genes are frequently mutated in human tumors,
with characteristic frequencies in different tissues. H-ras is
mutated in bladder cancer (10%) and salivary gland tumors
(15%), N-ras is mutated in melanoma (20%) and hemato-
poietic tumors (15%), and K-ras is predominantly mutated in
pancreatic ductal adenocarcinoma (60%), colorectal cancer
(30%), non-small cell lung adenocarcinoma (20%), and
endometrial cancer (15%) (frequencies as documented on the
Sanger Center COSMIC website). ras mutations can be both
prognostic for overall poor survival (2), as well as predictive for
lack of response to chemotherapy and targeted therapies (3),
showing the clinical importance of this gene. Genetically engi-
neered mouse models have also confirmed that a single point
mutation in K-ras or N-ras expressed in the relevant tissue is
sufficient to develop disease that strongly resembles human
tumors (4 6).
In addition to the well described pro-tumorigenic effects of
mutant Ras alleles, there is some suggestive evidence that the
* S. Vartanian, C. Bentley,L. Li, M. J. Brauer, P. Haverty, E. Stawiski, Z. Modrusan,
and D. Stokoe are paid employees of Genentech.
S
This article contains supplemental Table 1, Figs. 1– 8, and Materials and
Methods.
1
To whom correspondence should be addressed: Dept. of Discovery Oncol-
ogy, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080. Tel.: 650-
225-6031; Fax: 650-225-6412; E-mail: stokoe.david@gene.com.
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 288, NO. 4, pp. 2403–2413, January 25, 2013
© 2013 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2403
by guest on October 27, 2015http://www.jbc.org/Downloaded from
wild type (WT) copy of Ras may possess tumor-suppressive
effects. This was first noticed when mice expressing one copy of
K-Ras generated more and larger lung tumors following chemical
carcinogen treatment than mice expressing both alleles (7). A
tumor-suppressive effect for N-Ras has also been noted in some
mouse tumor models although this seems to be context-depen-
dent (for review, see Ref. 8). Such a tumor-suppressive effect of
WT Ras alleles is not as well characterized in human cancers.
The ability of mutant ras genes to activate downstream sig-
naling pathways that contribute to cell transformation is mostly
undisputed, although almost all of the studies drawing this con-
clusion have been performed using model systems that vastly
overexpress the mutant ras gene, usually involving H-Ras.
However, it has more recently become clear that overexpres-
sion of mutant ras has qualitatively different consequences
compared with single copy gene mutations that presumably
arise during the early stages of human tumorigenesis and that
cell type also plays an important role in determining cellular
responses. For example, overexpression of HRasV12 in immor-
talized mouse NIH3T3 cells causes transformation associated
with activation of Raf and PI3K pathways (9), whereas overex-
pression of HRas in normal fibroblasts causes a senescent-like
cell cycle arrest (10). In contrast, knock-in of a single copy of
mutant K-ras into nontransformed mouse or human cells
causes only very modest consequences on downstream signal-
ing (11–14). The consequences on cell transformation (15) or
tumor formation (11, 12) can also be surprisingly mild in the
absence of additional genetic alterations. Moreover, the assump-
tion of the strong transforming potential of mutant Ras needs to be
reassessed in light of recent discoveries that certain developmental
disorders such as Costello syndrome, cardiofacial cutaneous syn-
drome, and Noonans syndrome are caused by germ line-activating
mutations in ras (for review, see Ref. 16).
In this study, we examined the consequences of single copy
K-ras mutations in the context of human cancer cell lines, as
well as in immortalized but nontransformed human mammary
epithelial cells (HMECs).
2
We utilized cell line derivatives in
which the mutant or WT K-ras alleles had been deleted using
targeted homologous recombination, or in which a single copy
of mutant K-ras had been introduced using the same technol-
ogy. We found that although mutant K-Ras has strong effects
on cellular RasGTP levels, it has surprisingly mild conse-
quences on downstream signaling through Raf and PI3K path-
ways. Mutant K-Ras is also able to initiate negative feedback
signaling to the EGF receptor, which may have relevance in the
response of K-Ras mutant tumors to EGFR inhibitors. Despite
mild consequences on cellular signaling, flux through these
pathways is likely altered as demonstrated by robust regulation
of a small number of genes that seem to be consistently up-reg-
ulated by mutant K-Ras. We also demonstrate that the WT
K-Ras allele may behave in a tumor-suppressive manner in the
cancer cell lines analyzed.
EXPERIMENTAL PROCEDURES
Cell Culture and Lysate Preparation—Hec1A, HCT116, and
SW48 cells were cultured in McCoy’s 5A (Invitrogen) with 10%
FBS and 2 m
ML-glutamine. DLD1 cells were grown in RPMI
1640 medium with 10% FBS and 2 m
ML-glutamine, and
HMECs were cultured in 50:50 DMEM/F12 with 10% FBS, 20
ng/ml EGF, 0.01 mg/ml insulin, 500 ng/ml hydrocortisone, and
2m
ML-glutamine. Cells were cultured in a 37C/5% CO
2
incu
-
bator. For standard Western blotting and immunoprecipita-
tions, cells were harvested in radioimmuneprecipitation assay
buffer (50 m
M Tris, pH 7.4, 150 mM NaCl, 2 mM EDTA, 1%
Nonidet P-40, 0.1% SDS) with Complete Protease Inhibitor
(Roche Applied Science) and Phosphatase Inhibitor Mixture II
(Sigma). For Ras binding domain (RBD) pulldown experiments,
cells were harvested in 1 MgCl buffer (Millipore) with Com-
plete Protease Inhibitor.
siRNA Transfections—Cells were grown to 80% confluence in
10-cm dishes and transfected with nontargeting control or
K-Ras OTP siRNAs (Dharmacon). Briefly, 900 pmol of siRNA
and 20
l of RNAiMax (Invitrogen) were diluted separately in
1.5 ml of OptiMEM each. The siRNA and lipid were then mixed
together and incubated for 20 min at room temperature. The
complexes (3 ml) were added to the cells and incubated for 72 h.
Immunoprecipitations/Immunoblots—To check for siRNA
silencing of Ras, 2.5 mg of lysate was incubated with 10
lofPan
Ras antibody EP1125Y (Millipore 04-1039) at 4 ºC overnight. 50
l of protein A bead slurry was then added and incubated for 2 h
at 4 ºC with rotation. Beads were washed three times with lysis
buffer and resuspended in sample buffer. RBD pulldowns were
performed using a Ras Activation Assay kit (Millipore 17-218)
according to the manufacturer’s protocol. Western blot sam-
ples were run on 4 –20% Tris-glycine gels and transferred to
PVDF membranes using the iBlot system (Invitrogen). Pan Ras
immunoprecipitations were blotted for K-Ras (Santa Cruz
sc-30). RBD immunoprecipitations were blotted for Pan Ras
(Millipore 05-516), H-Ras, N-Ras, and K-Ras (Santa Cruz sc-29,
sc-31, and sc-30, respectively). Cell lysates were also blotted for
pERK (Cell Signaling 9101s), pAkt (Cell Signaling 9271s), pCbl
(Cell Signaling 3555s), pEGFR-Y1125 (Abgent AP3376a),
pEGFR-Y1069 (Upstate 07-715), total EGFR (Cell Signaling
4405s), total Cbl (Cell Signaling 2179s),
-actin (Sigma A2228),
total Ras (Millipore 05-516), total ERK (Cell Signaling 9102s),
and total Akt (Cell Signaling 4685s). In specified experiments,
cells were treated with GDC0941 (Fisher RG007-25MG) or
PD0325901 (Fisher RP02-10MG) diluted in dimethyl sulfoxide.
Soft Agar Assays—Base agar layer was prepared by mixing
equal parts 2 medium 20% FBS with 2.4% agarose. 1 ml of
this base agar was then added to each well of a 12-well tissue
culture plate (Grenier) and placed in a 4 ºC refrigerator to cool.
Cells were trypsinized and counted, then diluted to 5000 cells/
333
lof1 medium 10% FBS. A 1:1 mixture of 2 medium
20% FBS and 2.4% agarose was then prepared, and 667
l was
added to the cells, for a final agarose concentration of 0.8% in
the cell layer. 1 ml of cell mixture was then pipetted on top of
the base agar layer and placed at 4 ºC for 15 min to cool. The
plates were placed in a 37 ºC/5% CO
2
incubator overnight, and
2
The abbreviations used are: HMEC, human mammary epithelial cell; EGFR,
EGF receptor; RBD, Ras binding domain.
Mutant Ras-dependent Signaling and Gene Expression
2404 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 NUMBER 4 JANUARY 25, 2013
by guest on October 27, 2015http://www.jbc.org/Downloaded from
1 ml of medium was added on top of the cell layer the next day.
These assays were set up in triplicate.
Exome Alignment and Variant CallingIllumina fastq reads
were mapped to the UCSC human reference genome (GRCh37/
hg19) using BWA (17) set to default parameters. Duplicate mark-
ing, local realignment, and variant calling was done as described
previously (18). Variants included in dbSNP Build 131 (19) that
were also absent in COSMIC v56 (20) were excluded.
Proliferation/Migration Assays—For proliferation assays,
cells were trypsinized, counted, and plated at 2500 or 5000 cells
in 150
l/well, in a 96-well clear-bottomed black tissue culture
plate (BD Falcon). Cells were left to proliferate over 7 days in an
Incucyte instrument, which calculates cell density over speci-
fied intervals.
For migration assays, cells were plated the night before at
50,000 cells/well in a fibronectin- or collagen-coated Image-
Lock 96-well plate (Essen Bioscience). Cells were grown to con-
fluence, then a wound was created using the WoundMaker
apparatus (Essen Bioscience) according to the manufacturer’s
recommended protocol. The cells were then washed twice with
PBS and medium replaced to 100
l. Plates were imaged over
24 h at 2-h intervals.
Taqman Assays—RNA was isolated from cells using the
RNeasy kit (Qiagen 74106), and cDNA was synthesized using
the High Capacity cDNA Reverse Transcription Kit (Applied
Biosystems 4368814). Taqman assays were set up in multiplex,
using GAPDH as the endogenous control. The target probes
(DUSP5, DUSP6, ETV1, ETV5, NT5E, UPP1,K-ras, and IER3)
were tagged with FAM dye. Assays were run in 384-well format,
using 20 ng of input cDNA/reaction.
Additional procedures are described in the supplemental
Materials and Methods.
RESULTS
To assess the contribution of single copy K-Ras mutations
across multiple cell lines, we analyzed three previously pub-
lished tumor cell lines in which the wild type or mutant K-ras
allele was disrupted by homologous recombination. These cell
lines are the colorectal cancer lines DLD1 and HCT116 (21, 22),
which express heterozygous G13D mutations, and the endome-
trial cancer line Hec1A (23), which expresses a heterozygous
G12D mutation. In addition, we also analyzed two cell lines in
which a single copy of G13D mutant K-ras was introduced at its
endogenous locus, a colorectal cancer cell line SW48 which
evolved in the absence of mutant Ras, and nontransformed but
immortalized HMECs (15). Whole genome copy number anal-
ysis shows that for the lines analyzed (DLD1 and Hec1A),
although not truly “isogenic,” there are only very few and rela-
tively minor differences at the chromosomal level between the
parental lines and their isogenic derivatives (supplemental Fig.
1).
First we assessed the levels of RasGTP in these cell lines
growing in 10% FBS, under starved conditions, and following
short term stimulation with EGF. The RBD of cRaf was used as
an affinity probe for RasGTP, as described previously (24). In all
cases, knock-out and knock-in of mutant K-Ras decreased and
increased KRasGTP levels, respectively (Fig. 1A). Interestingly,
the presence or absence of mutant K-Ras affected NRasGTP
levels; in HCT116 and SW48 cells, the presence of mutant
K-Ras was associated with increased NRasGTP levels. This
could be due to the previously described positive feedback loop
caused by allosteric activation of the Ras exchange factor sos
(25). In contrast, in Hec1A, HMECs, and to a lesser extent
DLD1 cells, the presence of mutant K-Ras was associated with
decreased NRasGTP levels, suggesting the presence of a nega-
tive feedback loop. We also analyzed the levels of HRasGTP
using the same approach, but we did not detect any H-Ras pres-
ent in the RBD pulldowns under the conditions tested (data not
shown).
The effects of mutant K-Ras on downstream signaling to ERK
and Akt were examined next. In contrast to the dramatic effects
FIGURE 1. Consequences of mutant K-ras knock-out and knock-in on RasGTP levels and downstream signaling. A, cell lines of the indicated genotypes
were grown in 10% FBS, serum-starved for 18 h, or stimulated for 10 min with 100 ng/ml EGF. RasGTP was affinity-purified from 2.5 mg of cell lysate using
GST-Raf-RBD and analyzed by Western blotting with Ras isoform-specific antibodies. In addition, 40
g of lysate was used for Western blotting for total Ras
protein. B, the indicated cell lines were treated as above, and 40
g of protein lysates separated by SDS-PAGE and analyzed by Western blotting with
anti-phospho-ERK, phospho-Akt, total ERK, total Akt, and
-actin antibodies.
Mutant Ras-dependent Signaling and Gene Expression
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2405
by guest on October 27, 2015http://www.jbc.org/Downloaded from
on KRasGTP levels, the presence or absence of mutant K-Ras
had very modest effects on ERK and Akt phosphorylation,
under conditions of either basal proliferation in 10% FBS,
serum starvation, or acute stimulation with EGF (Fig. 1B).
Hec1A cells lacking mutant K-Ras actually showed slightly
increased phosphorylation of these proteins following EGF
stimulation, a paradoxical observation previously noted by
Waldman and colleagues (23).
To better understand the impact of single copy mutant K-Ras
on cell signaling, we performed a global phosphotyrosine anal-
ysis in the Hec1A
G12D/
and Hec1A
/WT
derivative cell lines
using Epitome phosphotyrosine arrays. Similar to the effects
seen by Western blot analysis, EGF-stimulated ERK1 and ERK2
phosphorylation was higher in the KRasG12D-deleted cells
(supplemental Fig. 2 A). The most dramatically altered proteins
between these cell lines were EGFR and its downstream sub-
strate Cbl (Fig. 2A). Both of these proteins showed increased
phosphorylation in response to EGF in the mutant K-Ras
knock-out clones compared with the WT K-Ras knock-out
clone. We confirmed these results by Western blot analysis
(Fig. 2B), which also analyzed the individual phosphorylation
sites, Tyr-1125 (a Grb2 binding site) and Tyr-1069 (a Cbl bind-
ing site). To determine whether the inhibitory effect of mutant
K-Ras on EGFR phosphorylation was a general phenomenon,
we examined the additional cell lines for basal and stimulated
EGFR and Cbl phosphorylation. DLD1 cells lacking mutant
K-Ras also showed increased EGFR phosphorylation at Tyr-
1125, even as total EGFR levels were decreased in this cell line
derivative (Fig. 2B). In addition, Cbl also showed increased
phosphorylation in the K-Ras knock-out DLD1 lines, although
this was more apparent under basal or starved conditions (Fig.
2B). HMECs expressing mutant K-Ras also showed decreased
EGFR phosphorylation (Fig. 2C). Increased EGFR phosphory-
lation was also seen in independently derived Hec1A WT and
mutant K-Ras knock-out clones (supplemental Fig. 2B), show-
ing that these effects are not due to clonal artifacts. However,
neither HCT116 nor SW48 cell isogenic lines showed any obvi-
ous differences in EGFR or Cbl phosphorylation in the K-Ras
knock-out or knock-in derivative cell lines (supplemental Fig. 2,
C and D). Therefore, mutant K-Ras negatively regulates EGFR
phosphorylation, as well as NRasGTP levels, in some cell lines.
To test the hypothesis that signaling downstream of mutant
K-Ras might be responsible for suppressing EGF-induced EGFR
phosphorylation, we utilized small molecule inhibitors of MEK
and PI3K in the mutant K-Ras-expressing cells. Fig. 3A shows that
PD0325901 was able to dramatically increase the response to EGF
in the mut/WT and mut/ Hec1A and DLD1 cells. A smaller
increase was also seen with GDC-0941. PD0325901 also increased
EGFR phosphorylation in the Hec1A
/WT
cell line, although this
was not seen with GDC-0941 or by either compound in the
DLD
/WT
cells. Interestingly, NRasGTP levels were also increased
with these inhibitors, suggesting a mechanistic link between EGFR
phosphorylation and NRasGTP levels. To determine the nature of
mutant K-Ras-induced EGFR inhibition, we asked whether condi-
tioned media from mut/ cells could inhibit EGFR phosphoryla-
tion in /WT cells. Supplemental Fig. 3 shows that there is a slight
inhibition of EGFR phosphorylation in cells in Hec1A
/WT
cells
using conditioned media from Hec1A
mut/
cells, but not in the
FIGURE 2. Mutant KRas negatively regulates EGFR signaling. A, Hec1A cells deleted for either WT or mutant KRas were serum-starved or starved and
stimulated with 100 ng/ml EGF for 10 min. 1 mg of lysate was reduced, alkylated, digested with Lys-C, and used to probe antibody microarrays. Signal for EGFR
and Cbl phosphorylation (mean S.D. (error bars, n 3)) is shown. B, the indicated cell lines were cultured in 10% FBS, 0% FBS, or stimulated with 100 ng/ml
EGF, and 40
g of protein lysate was separated by SDS-PAGE and probed by Western blotting using the antibodies shown. C, results are the same as for B but
using HMECs of the indicated genotypes.
Mutant Ras-dependent Signaling and Gene Expression
2406 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 NUMBER 4 JANUARY 25, 2013
by guest on October 27, 2015http://www.jbc.org/Downloaded from
similar DLD1 cell conditioned media swap. These results suggest
that activation of ERK downstream of mutant K-Ras results in a
signal that antagonizes response to EGF, which is partially (for
Hec1A cells) as a result of a secreted factor. The use of a cytokine
array did not help to clarify which secreted factor might be respon-
sible for this (data not shown).
We next examined the effects of endogenous mutant K-Ras
on cellular phenotypes. Cell proliferation was not dramatically
affected in any of the cell lines examined, although DLD1
/WT
cells showed slightly decreased proliferation relative to parental
and DLD1
G12D/
clones, and SW48 cells showed slightly
increased proliferation when mutant K-Ras was introduced
(Fig. 4A). We then analyzed cell migration using a classic
scratch wound-healing assay. DLD1 isogenic cell lines cells
showed little consistent differences in cell migration, whereas
Hec1A cells showed a slight decrease in migration upon loss of
the mutant K-ras allele, but increased migration upon loss of
the WT K-ras allele. Knock-in of mutant K-ras strongly
decreased the migration of HMECs, which showed the fastest
migration capacity of the cell types examined, with the wound
being completely closed in 20 h (Fig. 4B). The migration of
SW48 and HCT116 cells could not be assessed using this assay,
as HCT116 cells did not remain attached to the plate during the
wound-making process, and SW48 cells did not form a suffi-
ciently confluent monolayer.
We analyzed the transforming capacity of these cell lines
using anchorage-independent growth in soft agar. As expected,
and as previously reported, deletion of the mutant K-ras allele
strongly decreased the number of colonies in soft agar in DLD1,
HCT116, and Hec1A cells (Fig. 4C). There was also a modest
increase in the number of colonies in the HMECs expressing
mutant K-Ras, although the number of colonies in this line was
lower than seen in the cancer cell lines. Interestingly, the num-
ber of colonies was significantly increased in all three cell lines
lacking the wild type K-ras allele. SW48 cells did not form col-
onies under these conditions in our hands.
To attempt to understand why deletion of the WT K-ras
allele could enhance soft agar growth, we compared the signal-
ing pathways affected by this in Hec1A and HCT116 cells, the
cells that were most dramatically affected in this assay. Supple-
mental Fig. 4 shows that of 45 phosphorylation sites on signal-
ing proteins, only P-ERK (in Hec1A cells) and P-Akt (in
HCT116 cells) showed any differences. To determine the fre-
quency of WT K-ras allele loss in human tumor cell lines, we
assessed the K-ras mutant allele frequency in a panel of 624 cell
lines which had been subjected to exome-capture and deep
sequencing.
3
Of 130 cell lines with KRas variants, 35 showed
KRas variant DNA read frequencies consistent with homozy-
gous mutant K-ras status (Table 1). Moreover, even human
tumor cell lines classified as heterozygous for mutant K-ras
DNA frequently showed preferential expression of the mutant
K-ras allele, as assessed by combined exome-seq and RNAseq
analysis (Fig. 5). Combined, these data suggest that there is
selective pressure to lose or reduce expression of the WT K- ras
allele, at least in human cancer cell lines.
To address whether the signaling and phenotypic conse-
quences of chronic loss of mutant K-ras are contributed by
either clonal anomalies or long term adaptation, we also ana-
lyzed the effects of short term K-Ras knockdown using siRNA.
Knockdown of K-Ras (WT and mutant) in these cell lines
showed similar modest effects on ERK and/or Akt phosphory-
lation in the basal and starved settings (Fig. 6, A–C). Interest-
ingly, the increased EGFR phosphorylation seen in the K-Ras
knock-out DLD1 and Hec1A cells was also seen following
K-Ras knockdown, although the magnitude of this effect was
diminished (Fig. 6, A and C). Transient knockdown of K-Ras in
HCT116 cells also increased EGFR phosphorylation, despite
incomplete knockdown efficiency in this cell line (Fig. 6B). The
effects of K-Ras knockdown on proliferation and migration
were also assessed. K-Ras knockdown had little effect on pro-
liferation in any cell line (supplemental Fig. 5A), similar to the
results with the mutant K-Ras knock-out derivatives. Knock-
down of K-Ras in DLD1 cells had little effect on migration,
whereas K-Ras knockdown decreased migration of Hec1A cells
(supplemental Fig. 5B).
We then asked whether a combined analysis of K-Ras
isogenic cell lines could be used to identify genes that are
commonly regulated by mutant K-Ras expression. We pro-
filed the Hec1A, DLD1, and SW48 isogenic cell lines on
Affymetrix human genome U133 Plus microarrays. Knock-
out or knock-in of K-ras did not have strong effects on global
gene expression, cell lines clustered by parental cell line and
not K-Ras genotype (Fig. 7A). Analysis of multiple individual
clones of the same genotype (only performed for Hec1A
cells) showed that there were only small differences that
could be attributed to clone-specific alterations and that
independent genotypically matched lines clustered together.
3
C. Klijn, E. Stawiski, Z. Zhang, S. Seshagiri, and members of the Genentech
Cell Line Genomics Group, unpublished findings.
FIGURE 3. Signaling through the MEK and PI3K pathways negatively regulates EGFR and NRas activity. Hec1A and DLD1 cells of the indicated genotypes
were plated in 6-well dishes and at 70% confluence treated with the indicated concentration of the MEK inhibitor PD0325901 and the PI3K inhibitor GDC-0941.
18 h later, the cells were lysed, and 40
g was subjected to SDS-PAGE and Western blotting using the antibodies shown.
Mutant Ras-dependent Signaling and Gene Expression
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2407
by guest on October 27, 2015http://www.jbc.org/Downloaded from
Interestingly, WT K-ras-deleted lines clustered more closely
to parental cell lines than mutant Kras-deleted lines, sug-
gesting that in each cell line the mutant KRas allele has a
stronger effect on global gene expression than the WT allele.
The lists of genes affected by mutant K-Ras expression in
these three cell lines are shown in supplemental Table 1.In
general, the spectrum of genes regulated by the presence or
absence of mutant K-Ras in the three different cell lines was
quite distinct, as seen by overlaying the most significant up-
and down-regulated genes from DLD1 cells onto Volcano
FIGURE 4. K-Ras status has modest effects on cell proliferation and migration but more dramatic effects on colony formation. A, the indicated isogenic
cell lines were plated in 96-well plates at 5000 cells/well in 10% FBS, and confluence was tracked over 7 days using an Incucyte instrument. The means S.D.
(error bars, n 3) at 2-h intervals are shown. B, cell migration was analyzed by creating a scratch in the confluent cell monolayer and following wound closure
over 24 h using an Incucyte instrument. The left panel shows the percentage wound closure measured at 2-h intervals (mean S.D., n 3), and the right panels
show images of the wound at 0 and 24 h. C, the isogenic cell lines were seeded in a 0.8% agar/growth medium layer, and colony formation was analyzed after
30 days. The graphs show the average S.D. of triplicate wells counted using a GelCount instrument, and the images show representative sections of one well.
TABLE 1
Frequency of heterozygous (Het) and homozygous (Hom) K-ras mutations in a panel of cell lines
Gene Mutation Count Het/Hom (count) Tissue (counts)
Average variant
allele frequency
K-ras G12D 40 Het (37), Hom (3) Stomach (1), lung (2), pancreas (19),
uterus (10), ovary (2), colon (5),
breast (1)
0.52
K-ras G12V 22 Hom (9), Het (13) Lung (4), colon (4), pancreas (13),
breast (1)
0.75
K-ras G12C 16 Hom (9), Het (7) Lung (11), rectum (1), colon (2),
ovary (1), urinary bladder (1)
0.80
K-ras G13D 15 Het (14), Hom (1) Lung (4), breast (1), colon (6),
blood (1), ileum (1), lymph node (2)
0.43
K-ras G12R 7 Het (6), Hom (1) Pancreas (5), skin (1), breast (1) 0.65
K-ras G12A 6 Hom (1), Het (5) Blood (4), lung (1), colon (1) 0.54
K-ras Q61H 5 Hom (5) Lung (3), kidney (1), pancreas (1) 1
K-ras G13C 4 Hom (1), Het (3) lung (3), ovary (1) 0.61
K-ras A146T 3 Het (2), Hom (1) Blood (1), bone marrow (1), cecum (1) 0.67
K-ras G12S 2 Hom (1), Het (1) Lung (1), bone (1) 0.69
K-ras Q61K 2 Hom (1), Het (1) Stomach (1), lung (1) 0.83
K-ras Q61L 2 Hom (1), Het (1) Lung (1), colon (1) 0.79
K-ras A146V 1 Het (1) Blood (1) 0.75
K-ras A59T 1 Het (1) Bone (1) 0.45
K-ras K117N 1 Hom (1) Blood (1) 1
K-ras L19F 1 Het (1) Lung (1) 0.51
K-ras P121H 1 Het (1) Ovary (1) 0.52
K-ras Y64C 1 Het (1) Colon (1) 0.48
Mutant Ras-dependent Signaling and Gene Expression
2408 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 NUMBER 4 JANUARY 25, 2013
by guest on October 27, 2015http://www.jbc.org/Downloaded from
plots of gene expression in the three cell lines (Fig. 7B). Sim-
ilar disparate observations were also seen with the up- and
down-regulated genes from Hec1A and SW48 cells (supple-
mental Fig. 6). Nevertheless, we searched for genes that dis-
played common regulation in response to mutant K-Ras and
found two genes (NT5E and ETV1) that were commonly up-
regulated by mutant K-Ras in all three cell lines (Fig. 7C). We
also found several genes that were commonly up-regulated
by mutant K-Ras in Hec1A and DLD1 cells, including ETV5,
UPP1, IER3, DUSP5, and DUSP6.
We confirmed the mutant K-Ras-dependent regulation of
these genes by Taqman analysis in these cell lines (Fig. 8A). All
genes showed similar regulation by Taqman compared with the
array; in addition although IER3, ETV5, and UPP1 were not
identified as up-regulated by mutant K-Ras in SW48 cells by
microarray, they did show up-regulation by Taqman. We next
asked whether these genes could more generally be used as
markers for K-Ras inhibition in a panel of K-Ras mutant cell
lines and additionally asked whether they were also affected by
K-Ras knockdown in WT K-Ras cell lines. K-Ras siRNA effec-
tively decreased K-Ras expression in all of the lines examined
(although knockdown was modest in HupT3 and HT55 cells)
and decreased P-ERK in the mutant K-Ras-expressing cell lines
(Fig. 8B). P-ERK was not affected in the K-Ras WT cell lines, nor
was P-Akt decreased in any cell line. The basal expression of
these genes in this panel of cell lines did not correlate with
mutant K-Ras status (data not shown). However, all of the
mutant K-Ras-specific genes identified from the isogenic cell
lines were generally decreased following K-Ras knockdown in
the mutant K-Ras-expressing cell lines. In contrast, their
expression in the WT K-Ras cells was much less affected, con-
firming their usefulness as general markers for inhibition of
mutant K-Ras signaling (supplemental Fig. 7 and summarized
in Fig. 8C). This effect was statistically significant for all genes
apart from ETV1, which had low/undetectable expression in
this experiment in two cell lines.
To determine whether any of the genes induced by mutant
K-Ras contributes to its transformation properties, we knocked
down ETV1 and NT5E using siRNA pools in the same panel of
11 cell lines, and monitored proliferation in the second dimen-
sion on plastic and in the third dimension in hanging drop cul-
tures (26). Knockdown relative to a nontargeting control is
shown in supplemental Fig. 8, A–C. Whereas K-Ras knock-
down inhibited proliferation in both the second and third
dimensions selectively in the mutant K-Ras-expressing cell
lines, there was little selective inhibition of proliferation follow-
ing knockdown of either NT5E or ETV1 (supplemental Fig. 8, D
and E).
DISCUSSION
K-Ras is widely recognized as an important human oncogene
that is mutated in a large percentage of human epithelial
tumors. However, most of the analyses of the cellular conse-
quences of Ras expression have been performed using overex-
pression studies, often using H-Ras, and frequently using cell
types that are either mesenchymal or murine. In this study, we
have documented the effects of the endogenous mutated K-ras
allele, using cell line derivatives generated by homologous
recombination targeting. These studies have demonstrated
that there are both common phenotypes to mutant K-Ras
expression across multiple cell lines, as well as consequences
that are unique to the cell line, and therefore likely also unique
to the tumor tissue from which these were originally derived.
The use of isogenic cell lines has several advantages over com-
monly utilized overexpression and knockdown approaches.
Overexpression of mutant ras alleles results in a senescent-like
phenotype that has been attributed to increased production of
reactive oxygen species and associated stresses (27, 28) and is
likely unrelated to the normal functions of single copy mutant
ras, which results in tumor initiation without senescence
(11). The use of RNAi against ras (similar to any gene knock-
down) is problematic due to incomplete knockdown and the
possibility of off-target effects associated with siRNA or shRNA
sequences. Moreover, subtleties associated with activities of the
remaining WT allele would also not be revealed using a conven-
tional knockdown approach. The concerns of artifactual results
due to cell line selection and clonal anomalies were partially
alleviated in this study by the use of multiple clones (where
available) and the use of transient knockdown to show similar
effects on signaling and biological phenotypes.
One common consequence of mutant K-ras deletion was the
detrimental effects on soft agar colony growth, an often used
surrogate assay for tumorigenesis. This shows that, even with
the multitude of genetic alterations present in these cell lines,
deletion of a single allele has substantial effects on transforma-
tion. In contrast to the strong effect in transformation assays,
deletion of mutant K-ras has mild or nonexistent effects on cell
proliferation under the conditions assessed. This is unlikely to
be a trait selected for during the generation of these lines, as
knockdown of K-Ras in the parental lines also had only modest
effects on cell proliferation. Recently, there has been an appre-
ciation that K-Ras mutant cell lines vary in their response to
K-Ras knockdown, with some cell lines showing a strong
decrease in cell number due to enhanced apoptosis and/or
FIGURE 5. WT K-ras is either deleted or underexpressed in the majority of
mutant KRas-expressing cell lines. The read counts of WT (y axis) and
mutant (x axis) K-ras using RNA-seq data from 88 mutant K-Ras cell lines are
plotted. Lines designated as homozygous mutant K-Ras are shown in blue;
lines designated as heterozygous mutant K-Ras are shown in red.
Mutant Ras-dependent Signaling and Gene Expression
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2409
by guest on October 27, 2015http://www.jbc.org/Downloaded from
growth arrest, and other cell lines showing little effect (29, 30).
In these published studies, Hec1 (of which Hec1A is a deriva-
tive), HCT116, and DLD1 cells were all predicted and empiri-
cally shown to be insensitive to K-Ras knockdown in prolifera-
tion assays, consistent with the data shown here. It perhaps is
not surprising that the isogenic K-Ras lines that exist are “insen-
sitive,” as K-Ras-“sensitive” lines would likely be unable to
expand following deletion of the mutant allele. Nevertheless,
we would suggest that even though a cell line is classified as
Ras-independent by proliferation criteria, it remains Ras-de-
pendent by more stringent growth conditions that may more
closely reflect the tumor environment.
FIGURE 6. K-Ras siRNA knockdown in parental cell lines shows similar effects as chronic mutant K-Ras deletion. A, 100 nM K-Ras siRNA pools were
transfected into DLD1 cells and treated as shown. 72 h following transfection, 2.5 mg of protein lysates was subjected to pulldown with GST-RBD glutathione
bead slurry, and total RasGTP and NRasGTP were analyzed by Western blotting with the indicated antibodies. Additionally, 40
g of protein lysate was
separated by SDS-PAGE and analyzed by Western blotting using the indicated antibodies. B and C, HCT116 and Hec1A cells were treated as described in A.
FIGURE 7. Mutant K-Ras initiates distinct gene transcription programs that are generally cell line-dependent. A, RNA from triplicate cultures of the
indicated cell lines was hybridized onto Affymetrix U133 microarrays. The 403 genes that were differentially expressed in at least two cell lines were clustered
using the complete linkage method as implemented in the function hclust. B, genes that were regulated by mutant K-Ras in DLD1 cells were overlaid onto all
genes differentially regulated by mutant K-Ras in DLD1 cells (left plot), Hec1A cells (middle plot), and SW48 cells (right plot). C, genes up-regulated or down-
regulated by mutant K-Ras expression 2-fold in different cell lines are shown.
Mutant Ras-dependent Signaling and Gene Expression
2410 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 NUMBER 4 JANUARY 25, 2013
by guest on October 27, 2015http://www.jbc.org/Downloaded from
One aspect of mutant K-Ras signaling that was common to
some, but not all, of the cell lines studied here, was the cross-
talk between K-Ras and N-Ras. This was most dramatically seen
in the Hec1A cells and HMECs, but was also seen in DLD1 cells
to a lesser extent. This also correlated with the ability of mutant
K-Ras to down-regulate EGFR phosphorylation, an observation
also recently made by in lung cancer cell lines (31) and in colo-
rectal cancer cell lines (32). This could explain the relatively
mild effects on downstream signaling in the different cell lines
upon mutant K-ras deletion or expression, i.e. increased NRas-
GTP might compensate for the loss of KRasGTP and in the case
of Hec1A cells, could explain the paradoxical increase in EGF-
stimulated ERK phosphorylation upon mutant K-ras deletion.
This observation may also contribute to the lack of effect of
EGFR inhibitors in mutant K-Ras lung and colorectal tumor
patients (33, 34). Although activated Ras could maintain down-
stream signaling in the presence of EGFR inhibition, it could
also shut off EGFR signaling in these tumors, rendering them
independent of EGF and therefore refractory to EGFR inhibi-
tion. Here we show that the decreased EGFR phosphorylation
and NRasGTP levels in mutant K-Ras-expressing cell lines are
largely due to negative feedback initiated by MEK signaling.
This could be mediated, at least in part, by production of
secreted factors that antagonize EGF-dependent signaling,
although we were unable to define the exact factor that causes
this.
Deletion or introduction of mutant K-ras caused surprisingly
diverse effects on gene expression, showing that mutant K-Ras
has specific functions depending on the tissue in which it is
expressed. There have previously been several publications
using “Ras gene signatures” to classify human tumors. Sweet-
Cordero et al. (35) analyzed mutant K-Ras-driven lung tumors
and compared them with normal mouse lung tissue and human
K-Ras mutant lung cancers to derive 89 genes up-regulated in
mutant K-Ras lung cancer. Bild et al. (36) overexpressed H-Ras
in HMECs to derive 177 genes up-regulated by H-Ras. Arena et
al. (37) used single copy K-Ras knock-in in mouse liver progen-
itor cells to derive 149 genes that were up-regulated upon this
condition. Mutant K-Ras results in highly divergent conse-
quences in different tissues and environments, as exemplified
by the fact that only a single gene (DUSP6) is in common among
these three sets of signatures. Although the approach used here
to identify mutant K-Ras-regulated genes is likely no better or
no worse, it confirms that only a small number of genes are
consistently altered by mutant K-Ras. Notably, the seven genes
identified and validated in this study have all been found asso-
ciated with Ras expression or signaling in previous studies;
DUSP6, DUSP5 (36, 38), NT5E, ETV1 (39), IER3 (36, 39), UPP1
(36, 40), and ETV5 (39, 41). The functions of these genes are
quite diverse and represent mechanisms involved in negative
feedback signaling (DUSP5 and 6), nucleoside metabolism
(NT5E and UPP), transcriptional responses activated down-
stream of ERK signaling (ETV1 and ETV5), and protection from
apoptosis (IER3). Knockdown of NT5E has previously been
shown to decrease transformation properties of K-Ras mutant
MDA-MB-231 cells (42). Although knockdown of NT5E did
decrease proliferation in the second and third dimensions in
K-Ras mutant A427 cells, there was no statistically significant
correlation between the effects in mutant versus WT K-Ras
cells (supplemental Fig. 6). It is likely that K-Ras uses multiple
target genes to fully elicit transformation properties.
FIGURE 8. Expression of 7 genes are commonly regulated by mutant K-Ras. A, RNA was purified from the indicated cell lines and subjected to Taqman
quantitative PCR analysis against the genes shown in each graph. Expression was normalized to the control gene GAPDH and expressed (mean S.D. (error
bars), n 3) relative to the levels seen in the parental cell line. B, a panel of 11 cell lines expressing WT or mutant K-Ras was treated with 100 n
M NTC or K-Ras
siRNA pools, and protein and RNA were harvested 72 h later from identically treated plates. K-Ras knockdown was assessed by immunoprecipitation-Western
blotting using total and K-Ras-specific antibodies, and expression of P-ERK and P-Akt was assessed by Western blotting. C, RNA isolated from NTC or K-Ras
siRNA-treated cells was subjected to Taqman quantitative PCR using the indicated probes, and the effects on gene expression in each cell line are indicated as
a ratio of K-Ras:NTC siRNA treatment. The horizontal bar indicates the average expression of the gene across all cell lines. The open symbols represent WT
K-Ras-expressing cells, and the filled symbols represent mutant K-Ras-expressing cells. p values are the results of an unpaired t test comparing the effects on
mutant versus WT K-Ras-expressing cells.
Mutant Ras-dependent Signaling and Gene Expression
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2411
by guest on October 27, 2015http://www.jbc.org/Downloaded from
The increased transformation properties of Hec1A, HCT116, and
DLD1 cells lacking the WT K-ras allele was unexpected but is
reminiscent of studies in mice suggesting that the WT K-ras
allele can display tumor-suppressive effects. This was first
demonstrated in chemical carcinogen-induced tumors that
result in K-ras mutation, that were more aggressive when one
copy of K-ras was deleted in the germ line (7). Loss of the wild
type K-ras allele was also recently observed in high grade, but
not low grade lung tumors driven by conditional mutant K-Ras
(43). A tumor-suppressive function for WT N-Ras has also been
observed in mouse tumors driven by both chemical carcino-
gens, as well as by transgenic expression of mutant N-Ras (44).
The potential for WT K-Ras to act as a tumor suppressor in
human tumors has not been as well characterized, although our
analysis of a large panel of human cancer cell lines shows that
one-quarter of mutant K-Ras-expressing cells have allelic
imbalance/homozygous expression of the mutant allele. Even
in cells that have heterozygous expression, a majority show
enhanced expression of the mutant allele. These results clearly
suggest a selective advantage for the tumor cells to lose the WT
K-ras allele or to decrease its expression. The model systems
used in this study, or other similar ones, should allow further
dissection of this unexpected phenomenon.
Acknowledgments—We thank the members of the sequencing, bioin-
formatics, cell culture, and microarray core facilities at Genentech for
expertise and timely analysis; Chris Klijn and Zemin Zhang for K-Ras
RNA-seq information; Fred de Sauvage and Jeff Settleman for critical
input during this project; and David Davis for many useful and
thoughtful discussions.
REFERENCES
1. Karnoub, A. E., and Weinberg, R. A. (2008) Ras oncogenes: split person-
alities. Nat. Rev. Mol. Cell Biol. 9, 517–531
2. Mascaux, C., Iannino, N., Martin, B., Paesmans, M., Berghmans, T.,
Dusart, M., Haller, A., Lothaire, P., Meert, A. P., Noel, S., Lafitte, J. J., and
Sculier, J. P. (2005) The role of RAS oncogene in survival of patients with
lung cancer: a systematic review of the literature with meta-analysis. Br. J.
Cancer 92, 131–139
3. Linardou, H., Dahabreh, I. J., Kanaloupiti, D., Siannis, F., Bafaloukos, D.,
Kosmidis, P., Papadimitriou, C. A., and Murray, S. (2008) Assessment of
somatic K-RAS mutations as a mechanism associated with resistance to
EGFR-targeted agents: a systematic review and meta-analysis of studies in
advanced non-small-cell lung cancer and metastatic colorectal cancer.
Lancet Oncol. 9, 962–972
4. Jackson, E. L., Willis, N., Mercer, K., Bronson, R. T., Crowley, D., Montoya,
R., Jacks, T., and Tuveson, D. A. (2001) Analysis of lung tumor initiation
and progression using conditional expression of oncogenic K-ras. Genes
Dev. 15, 3243–3248
5. Hingorani, S. R., Petricoin, E. F., Maitra, A., Rajapakse, V., King, C., Jaco-
betz, M. A., Ross, S., Conrads, T. P., Veenstra, T. D., Hitt, B. A., Kawaguchi,
Y., Johann, D., Liotta, L. A., Crawford, H. C., Putt, M. E., Jacks, T., Wright,
C. V., Hruban, R. H., Lowy, A. M., and Tuveson, D. A. (2003) Preinvasive
and invasive ductal pancreatic cancer and its early detection in the mouse.
Cancer Cell 4, 437– 450
6. Li, Q., Haigis, K. M., McDaniel, A., Harding-Theobald, E., Kogan, S. C.,
Akagi, K., Wong, J. C., Braun, B. S., Wolff, L., Jacks, T., and Shannon, K.
(2011) Hematopoiesis and leukemogenesis in mice expressing oncogenic
NRasG12D from the endogenous locus. Blood 117, 2022–2032
7. Zhang, Z., Wang, Y., Vikis, H. G., Johnson, L., Liu, G., Li, J., Anderson,
M. W., Sills, R. C., Hong, H. L., Devereux, T. R., Jacks, T., Guan, K. L., and
You, M. (2001) Wildtype K-ras2 can inhibit lung carcinogenesis in mice.
Nat. Genet. 29, 25–33
8. Diaz, R., Lopez-Barcons, L., Ahn, D., Garcia-Espana, A., Yoon, A., Mat-
thews, J., Mangues, R., Perez-Soler, R., and Pellicer, A. (2004) Complex
effects of ras proto-oncogenes in tumorigenesis. Carcinogenesis 25,
535–539
9. White, M. A., Nicolette, C., Minden, A., Polverino, A., Van Aelst, L., Karin,
M., and Wigler, M. H. (1995) Multiple Ras functions can contribute to
mammalian cell transformation. Cell 80, 533–541
10. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D., and Lowe, S. W.
(1997) Oncogenic ras provokes premature cell senescence associated with
accumulation of p53 and p16INK4a. Cell 88, 593–602
11. Tuveson, D. A., Shaw, A. T., Willis, N. A., Silver, D. P., Jackson, E. L.,
Chang, S., Mercer, K. L., Grochow, R., Hock, H., Crowley, D., Hingorani,
S. R., Zaks, T., King, C., Jacobetz, M. A., Wang, L., Bronson, R. T., Orkin,
S. H., DePinho, R. A., and Jacks, T. (2004) Endogenous oncogenic
K-ras(G12D) stimulates proliferation and widespread neoplastic and de-
velopmental defects. Cancer Cell 5, 375–387
12. Guerra, C., Mijimolle, N., Dhawahir, A., Dubus, P., Barradas, M., Serrano,
M., Campuzano, V., and Barbacid, M. (2003) Tumor induction by an
endogenous K-ras oncogene is highly dependent on cellular context. Can-
cer Cell 4, 111–120
13. Konishi, H., Karakas, B., Abukhdeir, A. M., Lauring, J., Gustin, J. P., Garay,
J. P., Konishi, Y., Gallmeier, E., Bachman, K. E., and Park, B. H. (2007)
Knock-in of mutant K-ras in nontumorigenic human epithelial cells as a
new model for studying K-ras-mediated transformation. Cancer Res. 67,
8460 8467
14. Braun, B. S., Tuveson, D. A., Kong, N., Le, D. T., Kogan, S. C., Rozmus, J.,
Le Beau, M. M., Jacks, T. E., and Shannon, K. M. (2004) Somatic activation
of oncogenic K-ras in hematopoietic cells initiates a rapidly fatal my-
eloproliferative disorder. Proc. Natl. Acad. Sci. U.S.A. 101, 597–602
15. Di Nicolantonio, F., Arena, S., Gallicchio, M., Zecchin, D., Martini, M.,
Flonta, S. E., Stella, G. M., Lamba, S., Cancelliere, C., Russo, M., Geuna, M.,
Appendino, G., Fantozzi, R., Medico, E., and Bardelli, A. (2008) Replace-
ment of normal with mutant alleles in the genome of normal human cells
unveils mutation-specific drug responses. Proc. Natl. Acad. Sci. U.S.A.
105, 20864 –20869
16. Schubbert, S., Shannon, K., and Bollag, G. (2007) Hyperactive Ras in de-
velopmental disorders and cancer. Nat. Rev. Cancer 7, 295–308
17. Li, H., and Durbin, R. (2009) Fast and accurate short read alignment with
Burrows-Wheeler transform. Bioinformatics 25, 1754 –1760
18. DePristo, M. A., Banks, E., Poplin, R., Garimella, K. V., Maguire, J. R., Hartl,
C., Philippakis, A. A., del Angel, G., Rivas, M. A., Hanna, M., McKenna, A.,
Fennell, T. J., Kernytsky, A. M., Sivachenko, A. Y., Cibulskis, K., Gabriel,
S. B., Altshuler, D., and Daly, M. J. (2011) A framework for variation dis-
covery and genotyping using next-generation DNA sequencing data. Nat.
Genet. 43, 491– 498
19. Sherry, S. T., Ward, M. H., Kholodov, M., Baker, J., Phan, L., Smigielski,
E. M., and Sirotkin, K. (2001) dbSNP: the NCBI database of genetic varia-
tion. Nucleic Acids Res. 29, 308 –311
20. Forbes, S. A., Bindal, N., Bamford, S., Cole, C., Kok, C. Y., Beare, D., Jia, M.,
Shepherd, R., Leung, K., Menzies, A., Teague, J. W., Campbell, P. J., Strat-
ton, M. R., and Futreal, P. A. (2011) COSMIC: mining complete cancer
genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids
Res. 39, D945–50
21. Shirasawa, S., Furuse, M., Yokoyama, N., and Sasazuki, T. (1993) Altered
growth of human colon cancer cell lines disrupted at activated Ki-ras.
Science 260, 85– 88
22. Yun, J., Rago, C., Cheong, I., Pagliarini, R., Angenendt, P., Rajagopalan, H.,
Schmidt, K., Willson, J. K., Markowitz, S., Zhou, S., Diaz, L. A., Jr., Vel-
culescu, V. E., Lengauer, C., Kinzler, K. W., Vogelstein, B., and Papado-
poulos, N. (2009) Glucose deprivation contributes to the development of
K-ras pathway mutations in tumor cells. Science 325, 1555–1559
23. Kim, J. S., Lee, C., Foxworth, A., and Waldman, T. (2004) B-Raf is dispen-
sable for K-ras-mediated oncogenesis in human cancer cells. Cancer Res.
64, 1932–1937
24. Taylor, S. J., and Shalloway, D. (1996) Cell cycle-dependent activation of
Ras. Curr. Biol. 6, 1621–1627
25. Boykevisch, S., Zhao, C., Sondermann, H., Philippidou, P., Halegoua, S.,
Mutant Ras-dependent Signaling and Gene Expression
2412 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 NUMBER 4 JANUARY 25, 2013
by guest on October 27, 2015http://www.jbc.org/Downloaded from
Kuriyan, J., and Bar-Sagi, D. (2006) Regulation of ras signaling dynamics by
Sos-mediated positive feedback. Curr. Biol. 16, 2173–2179
26. Del Duca, D., Werbowetski, T., and Del Maestro, R. F. (2004) Spheroid
preparation from hanging drops: characterization of a model of brain
tumor invasion. J. Neurooncol. 67, 295–303
27. Lee, A. C., Fenster, B. E., Ito, H., Takeda, K., Bae, N. S., Hirai, T., Yu, Z. X.,
Ferrans, V. J., Howard, B. H., and Finkel, T. (1999) Ras proteins induce
senescence by altering the intracellular levels of reactive oxygen species.
J. Biol. Chem. 274, 7936 –7940
28. DeNicola, G. M., Karreth, F. A., Humpton, T. J., Gopinathan, A., Wei, C.,
Frese, K., Mangal, D., Yu, K. H., Yeo, C. J., Calhoun, E. S., Scrimieri, F.,
Winter, J. M., Hruban, R. H., Iacobuzio-Donahue, C., Kern, S. E., Blair,
I. A., and Tuveson, D. A. (2011) Oncogene-induced Nrf2 transcription
promotes ROS detoxification and tumorigenesis. Nature 475, 106–109
29. Singh, A., Greninger, P., Rhodes, D., Koopman, L., Violette, S., Bardeesy,
N., and Settleman, J. (2009) A gene expression signature associated with
“K-Ras addiction” reveals regulators of EMT and tumor cell survival. Can-
cer Cell 15, 489 –500
30. Singh, A., Sweeney, M. F., Yu, M., Burger, A., Greninger, P., Benes, C.,
Haber, D. A., and Settleman, J. (2012) TAK1 inhibition promotes apopto-
sis in K-Ras-dependent colon cancers. Cell 148, 639650
31. Sunaga, N., Shames, D. S., Girard, L., Peyton, M., Larsen, J. E., Imai, H.,
Soh, J., Sato, M., Yanagitani, N., Kaira, K., Xie, Y., Gazdar, A. F., Mori, M.,
and Minna, J. D. (2011) Knockdown of oncogenic K-Ras in non-small cell
lung cancers suppresses tumor growth and sensitizes tumor cells to tar-
geted therapy. Mol. Cancer Ther. 10, 336 –346
32. van Houdt, W. J., Hoogwater, F. J., de Bruijn, M. T., Emmink, B. L., Ni-
jkamp, M. W., Raats, D. A., van der Groep, P., van Diest, P., Borel Rinkes,
I. H., and Kranenburg, O. (2010) Oncogenic K-ras desensitizes colorectal
tumor cells to epidermal growth factor receptor inhibition and activation.
Neoplasia 12, 443– 452
33. De Roock, W., Jonker, D. J., Di Nicolantonio, F., Sartore-Bianchi, A., Tu,
D., Siena, S., Lamba, S., Arena, S., Frattini, M., Piessevaux, H., Van Cutsem,
E., O’Callaghan, C. J., Khambata-Ford, S., Zalcberg, J. R., Simes, J., Kara-
petis, C. S., Bardelli, A., and Tejpar, S. (2010) Association of K-Ras p.G13D
mutation with outcome in patients with chemotherapy-refractory meta-
static colorectal cancer treated with cetuximab. JAMA 304, 1812–1820
34. Mao, C., Qiu, L. X., Liao, R. Y., Du, F. B., Ding, H., Yang, W. C., Li, J., and
Chen, Q. (2010) K-Ras mutations and resistance to EGFR-TKIs treatment
in patients with non-small cell lung cancer: a meta-analysis of 22 studies.
Lung Cancer 69, 272–278
35. Sweet-Cordero, A., Mukherjee, S., Subramanian, A., You, H., Roix, J. J.,
Ladd-Acosta, C., Mesirov, J., Golub, T. R., and Jacks, T. (2005) An onco-
genic K-ras2 expression signature identified by cross-species gene-ex-
pression analysis. Nat. Genet. 37, 48 –55
36. Bild, A. H., Yao, G., Chang, J. T., Wang, Q., Potti, A., Chasse, D., Joshi,
M. B., Harpole, D., Lancaster, J. M., Berchuck, A., Olson, J. A., Jr., Marks,
J. R., Dressman, H. K., West, M., and Nevins, J. R. (2006) Oncogenic path-
way signatures in human cancers as a guide to targeted therapies. Nature
439, 353–357
37. Arena, S., Isella, C., Martini, M., de Marco, A., Medico, E., and Bardelli, A.
(2007) Knock-in of oncogenic K-ras does not transform mouse somatic
cells but triggers a transcriptional response that classifies human cancers.
Cancer Res. 67, 8468 8476
38. Tchernitsa, O. I., Sers, C., Zuber, J., Hinzmann, B., Grips, M., Schramme,
A., Lund, P., Schwendel, A., Rosenthal, A., and Schäfer, R. (2004) Tran-
scriptional basis of K-ras oncogene-mediated cellular transformation in
ovarian epithelial cells. Oncogene 23, 4536 4555
39. Packer, L. M., East, P., Reis-Filho, J. S., and Marais, R. (2009) Identification
of direct transcriptional targets of (V600E)BRAF/MEK signalling in
melanoma. Pigment Cell Melanoma Res. 22, 785–798
40. Guerrero, I., Villasante, A., Corces, V., and Pellicer, A. (1985) Loss of the
normal N-ras allele in a mouse thymic lymphoma induced by a chemical
carcinogen. Proc. Natl. Acad. Sci. U.S.A. 82, 7810–7814
41. Pratilas, C. A., Taylor, B. S., Ye, Q., Viale, A., Sander, C., Solit, D. B., and
Rosen, N. (2009) (V600E)BRAF is associated with disabled feedback inhi-
bition of RAF-MEK signaling and elevated transcriptional output of the
pathway. Proc. Natl. Acad. Sci. U.S.A. 106, 45194524
42. Zhi, X., Wang, Y., Zhou, X., Yu, J., Jian, R., Tang, S., Yin, L., and Zhou, P.
(2010) RNAi-mediated CD73 suppression induces apoptosis and cell-cy-
cle arrest in human breast cancer cells. Cancer Sci. 101, 2561–2569
43. Junttila, M. R., Karnezis, A. N., Garcia, D., Madriles, F., Kortlever, R. M.,
Rostker, F., Brown Swigart, L., Pham, D. M., Seo, Y., Evan, G. I., and
Martins, C. P. (2010) Selective activation of p53-mediated tumour sup-
pression in high-grade tumours. Nature 468, 567–571
44. Diaz, R., Ahn, D., Lopez-Barcons, L., Malumbres, M., Perez de Castro, I.,
Lue, J., Ferrer-Miralles, N., Mangues, R., Tsong, J., Garcia, R., Perez-Soler,
R., and Pellicer, A. (2002) The N-ras proto-oncogene can suppress the
malignant phenotype in the presence or absence of its oncogene. Cancer
Res. 62, 4514 4518
Mutant Ras-dependent Signaling and Gene Expression
JANUARY 25, 2013 VOLUME 288 NUMBER 4 JOURNAL OF BIOLOGICAL CHEMISTRY 2413
by guest on October 27, 2015http://www.jbc.org/Downloaded from
David Stokoe
andStawiski, Zora Modrusan, Todd Waldman
Sasazuki, Jung-Sik Kim, Pete Haverty, Eric
J. Brauer, Li Li, Senji Shirasawa, Takehiko
Steffan Vartanian, Carolyn Bentley, Matthew
Lines
Phenotypes Using a Panel of Isogenic Cell
Identification of Mutant K-Ras-dependent
Signal Transduction:
doi: 10.1074/jbc.M112.394130 originally published online November 27, 2012
2013, 288:2403-2413.J. Biol. Chem.
10.1074/jbc.M112.394130Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted
When this article is cited
to choose from all of JBC's e-mail alertsClick here
Supplemental material:
http://www.jbc.org/content/suppl/2012/11/27/M112.394130.DC1.html
http://www.jbc.org/content/288/4/2403.full.html#ref-list-1
This article cites 44 references, 18 of which can be accessed free at
by guest on October 27, 2015http://www.jbc.org/Downloaded from
... Kirsten Rat Sarcoma Viral Oncogene Homolog (Kras or kras2) was one of the earliest human oncogenes to be described and is one of the most commonly mutated genes in different human cancers, including colorectal cancer (Dinu et al., 2014;Tsuchida et al., 2016). Activation of Ras is associated with a wide range of biological responses, such as cell growth, proliferation, survival, differentiation and tissue morphogenesis, depending on the cell type and stimuli (Vartanian et al., 2013). KRAS mutations are associated with poor prognosis and resistance to targeted therapies, demonstrating the clinical importance of this gene (Arrington et al., 2012). ...
... Zhang et al. showed that following exposure to carcinogenic chemicals, mice harbouring only one copy of wild type Kras2 produced larger lung tumors than mice harbouring both wild type alleles (Zhongqiu Zhang et al., 2001). To investigate the effect of a single copy mutant KRAS, Vartanian et al. analysed the signalling and biological properties of a panel of isogenic cancer cell lines (DLD1, HCT116, and Hec1A) (Vartanian et al., 2013). They demonstrated that a single copy of mutant KRAS results in only modest activation of downstream signalling pathways involving extracellular signal-regulated kinase (ERK) and AKT. ...
... One copy of mutant KRAS had a small effect on cell proliferation and cell migration, but a greater effect on cell transformation, as evaluated by growth in soft agar. Interestingly, elimination of the wild type KRAS allele led to increased growth in soft agar, indicating tumor-suppressive characteristics of the wild type KRAS allele under these conditions (Vartanian et al., 2013). ...
Article
Full-text available
Introduction KRAS was one of the earliest human oncogenes to be described and is one of the most commonly mutated genes in different human cancers, including colorectal cancer. Despite KRAS mutants being known driver mutations, KRAS has proved difficult to target therapeutically, necessitating a comprehensive understanding of the molecular mechanisms underlying KRAS -driven cellular transformation. Objectives To investigate the metabolic signatures associated with single copy mutant KRAS in isogenic human colorectal cancer cells and to determine what metabolic pathways are affected. Methods Using NMR-based metabonomics, we compared wildtype (WT)- KRAS and mutant KRAS effects on cancer cell metabolism using metabolic profiling of the parental KRAS G13D/+ HCT116 cell line and its isogenic, derivative cell lines KRAS +/– and KRAS G13D/– . Results Mutation in the KRAS oncogene leads to a general metabolic remodelling to sustain growth and counter stress, including alterations in the metabolism of amino acids and enhanced glutathione biosynthesis. Additionally, we show that KRAS G13D/ + and KRAS G13D/− cells have a distinct metabolic profile characterized by dysregulation of TCA cycle, up-regulation of glycolysis and glutathione metabolism pathway as well as increased glutamine uptake and acetate utilization. Conclusions Our study showed the effect of a single point mutation in one KRAS allele and KRAS allele loss in an isogenic genetic background, hence avoiding confounding genetic factors. Metabolic differences among different KRAS mutations might play a role in their different responses to anticancer treatments and hence could be exploited as novel metabolic vulnerabilities to develop more effective therapies against oncogenic KRAS. Graphical abstract
... In contrast, R15 did not inhibit ERK-MAPK activation ( Figures 6E and S4E), proliferation, or anchorageindependent growth of G12V or G12C mutant lines ( Figures 6J, 6O, and S4J), consistent with the inability of R15 to appreciably bind or inhibit these RAS mutants in cells ( Figure 2). Although R15 did not reduce pERK-MAPK levels in Hec1A R15 and HCT-116 R15 lines, there was a significant reduction in anchorage-independent growth of these lines, consistent with an apparent difference in signaling in 2D versus 3D growth conditions reported by others (Janes et al., 2018;Khan et al., 2019;Vartanian et al., 2013;Figures S4B, S4C, S4G, and S4H). Similar results were observed for H1299 R15 ( Figures S4D and S4I). ...
... This result is consistent with prior results in which the inhibition of RAS by NS1 resulted in increased pAKT levels in CFPAC-1 R15 and Hec1a R15 cells (Khan et al., 2019). These data indicate that certain cells are more RAS dependent under 2D conditions, consistent with previously published studies (Janes et al., 2018;Khan et al., 2019;Vartanian et al., 2013). R15 expression did not impair the active AKT levels in KRAS(G12V) mutant CFPAC-1 R15 cells ( Figure S5E), consistent with the lack of effect of R15 on KRAS(G12V) in prior assays. ...
Article
Full-text available
RAS guanosine triphosphatases (GTPases) are mutated in nearly 20% of human tumors, making them an attractive therapeutic target. Following our discovery that nucleotide-free RAS (apo RAS) regulates cell signaling, we selectively target this state as an approach to inhibit RAS function. Here, we describe the R15 monobody that exclusively binds the apo state of all three RAS isoforms in vitro, regardless of the mutation status, and captures RAS in the apo state in cells. R15 inhibits the signaling and transforming activity of a subset of RAS mutants with elevated intrinsic nucleotide exchange rates (i.e., fast exchange mutants). Intracellular expression of R15 reduces the tumor-forming capacity of cancer cell lines driven by select RAS mutants and KRAS(G12D)-mutant patient-derived xenografts (PDXs). Thus, our approach establishes an opportunity to selectively inhibit a subset of RAS mutants by targeting the apo state with drug-like molecules.
... G12D, G12V, and G13D) exhibit variations in their phenotypic manifestations, such as transforming potential and migration properties. [18][19][20][21][22][23][24][25][26] Furthermore, (K)RAS mutant-and copy numberspecific alterations in lipid, nucleotide, and glycolytic metabolism have been established in both in vitro and in vivo systems. [27][28][29][30] We predicted that all KRAS mutant MEF cell lines have increased capacity to produce ATP, but there are also modest shifts between energy-requiring processes, possibly arising because the total capacity is limited. ...
Article
Full-text available
Cellular utilization of available energy flows to drive a multitude of forms of cellular ‘work’ is a major biological constraint. Cells steer metabolism to address changing phenotypic states but little is known as to how bioenergetics couples to the richness of processes in a cell as a whole. Here, we outline a whole-cell energy framework that is informed by proteomic analysis and an energetics-based gene ontology. We separate analysis of metabolic supply and the capacity to generate high-energy phosphates from a representation of demand that is built on the relative abundance of ATPases and GTPases that deliver cellular work. We employed mouse embryonic fibroblast cell lines that express wildtype KRAS or oncogenic mutations and with distinct phenotypes. We observe shifts between energy-requiring processes. Calibrating against Seahorse analysis, we have created a whole-cell energy budget with apparent predictive power, for instance in relation to protein synthesis.
... S2 and S3). Although HEC-1A and H1944 cell lines did not show a reduction of pERK level upon JAM20 induction in two-dimensional (2D) culture, their colony-formation activity was inhibited by JAM20 expression (SI Appendix, Fig. S3), consistent with published studies (36)(37)(38). JAM20 did not impair the MAPK signaling in A375 melanoma cells harboring the BRAF(V600E) mutant, as expected (SI Appendix, Fig. S4). These results demonstrate JAM20's selectivity to RAS in cancer cell lines, consistent with its effects on model cellular systems, such as HEK293 and NIH/3T3 cells (Fig. 2). ...
Article
Full-text available
RAS mutants are major therapeutic targets in oncology with few efficacious direct inhibitors available. The identification of a shallow pocket near the Switch II region on RAS has led to the development of small-molecule drugs that target this site and inhibit KRAS(G12C) and KRAS(G12D). To discover other regions on RAS that may be targeted for inhibition, we have employed small synthetic binding proteins termed monobodies that have a strong propensity to bind to functional sites on a target protein. Here, we report a pan-RAS monobody, termed JAM20, that bound to all RAS isoforms with nanomolar affinity and demonstrated limited nucleotide-state specificity. Upon intracellular expression, JAM20 potently inhibited signaling mediated by all RAS isoforms and reduced oncogenic RAS-mediated tumorigenesis in vivo. NMR and mutation analysis determined that JAM20 bound to a pocket between Switch I and II, which is similarly targeted by low-affinity, small-molecule inhibitors, such as BI-2852, whose in vivo efficacy has not been demonstrated. Furthermore, JAM20 directly competed with both the RAF(RBD) and BI-2852. These results provide direct validation of targeting the Switch I/II pocket for inhibiting RAS-driven tumorigenesis. More generally, these results demonstrate the utility of tool biologics as probes for discovering and validating druggable sites on challenging targets.
... Activating mutations in KRAS support the decoupling of glycolysis and TCA metabolism, with glutamine supplying increased carbon to drive the TCA cycle [4]. Several studies have compared the effects of KRAS and BRAF mutations in genome edited cell systems [5,6]. An analysis of KRAS G12D, G12V and G13D in human SW48 CRC cells revealed differential reprogramming of proteomes between KRAS codon 12 and 13 mutant cell lines. ...
Preprint
Full-text available
Background Genes in the Ras pathway have somatic mutations in at least 60 % of colorectal cancers. Despite activating the same pathway, the BRAF V600E mutation and the prevalent mutations in codon 12 and 13 of KRAS have all been linked to different clinical outcomes, but the molecular mechanisms behind these differences largely remain to be clarified. Methods To characterize the similarities and differences between common activating KRAS mutations and between KRAS and BRAF mutations, we used genome editing to engineer KRAS G12C/D/V and G13D mutations in colorectal cancer cells that had their mutant BRAF V600E allele removed and subjected them to transcriptome sequencing, global proteomics and metabolomics analyses. Results By intersecting differentially expressed genes, proteins and metabolites, we uncovered (i) two-fold more regulated genes and proteins when comparing KRAS to BRAF mutant cells to those lacking Ras pathway mutation, (ii) five differentially expressed proteins in KRAS mutants compared to cells lacking Ras pathway mutation (IFI16, S100A10, CD44, GLRX and AHNAK2) and 6 (CRABP2, FLNA, NXN, LCP1, S100A10 and S100A2) compared to BRAF mutant cells, (iii) 19 proteins expressed differentially in a KRAS mutation specific manner versus BRAF V600E cells, (iv) regulation of the Integrin Linked Kinase pathway by KRAS but not BRAF mutation, (v) regulation of amino acid metabolism, particularly of the tyrosine, histidine, arginine and proline pathways, the urea cycle and purine metabolism by Ras pathway mutations, (vi) increased free carnitine in KRAS and BRAF mutant RKO cells. Conclusions This comprehensive integrative-omics analysis confirms known and adds novel genes, proteins and metabolic pathways regulated by mutant KRAS and BRAF signaling in colorectal cancer. The results from the new model systems presented here can inform future development of diagnostic and therapeutic approaches targeting tumors with KRAS and BRAF mutations.
... In addition to their catalytic activity towards ERK1/2, both phosphatases bind tightly to ERK and can retain the inactive kinase in either the nucleus (DUSP5) or the cytoplasm (DUSP6), indicating that they act to regulate the spatiotemporal activity of this key growth factor regulated signalling pathway [3,6,7]. Because abnormal activation of RAS/ERK signalling is frequently observed in human cancers [8] and both DUSP5 and DUSP6 are often up-regulated in tumours and cancer cell lines, which harbour activating mutations in the RAS/ERK pathway [9][10][11][12][13], these enzymes have been presumed to negatively regulate the oncogenic potential of signalling and to be potential tumour suppressors. ...
Article
Full-text available
The cytoplasmic phosphatase DUSP6 and its nuclear counterpart DUSP5 are negative regulators of RAS/ERK signalling. Here we use deletion of either Dusp5 or Dusp6 to explore the roles of these phosphatases in a murine model of KRASG12D-driven pancreatic cancer. By 56-days, loss of either DUSP5 or DUSP6 causes a significant increase in KRASG12D-driven pancreatic hyperplasia. This is accompanied by increased pancreatic acinar to ductal metaplasia (ADM) and the development of pre-neoplastic pancreatic intraepithelial neoplasia (PanINs). In contrast, by 100-days, pancreatic hyperplasia is reversed with significant atrophy of pancreatic tissue and weight loss observed in animals lacking either DUSP5 or DUSP6. On further ageing, Dusp6−/− mice display accelerated development of metastatic pancreatic ductal adenocarcinoma (PDAC), while in Dusp5−/− animals, although PDAC development is increased this process is attenuated by atrophy of pancreatic acinar tissue and severe weight loss in some animals before cancer could progress. Our data suggest that despite a common target in the ERK MAP kinase, DUSP5 and DUSP6 play partially non-redundant roles in suppressing oncogenic KRASG12D signalling, thus retarding both tumour initiation and progression. Our data suggest that loss of either DUSP5 or DUSP6, as observed in certain human tumours, including the pancreas, could promote carcinogenesis.
Article
Eph receptors, the largest known family of receptor tyrosine kinases, and ephrin ligands have been implicated in a variety of human cancers. The novel bidirectional signaling events initiated by binding of Eph receptors to their cognate ephrin ligands modulate many cellular processes such as proliferation, metastasis, angiogenesis, invasion, and apoptosis. The relationships between the abundance of a unique subset of Eph receptors and ephrin ligands with associated cellular processes indicate a key role of these molecules in tumorigenesis. The combinatorial expression of these molecules converges on MAP kinase and/or AKT/mTOR signaling pathways. The intracellular target proteins of the initial signal may, however, vary in some cancers. Furthermore, we have also described the commonality of up- and down-regulation of individual receptors and ligands in various cancers. The current state of research in Eph receptors illustrates MAP kinase and mTOR pathways as plausible targets for therapeutic interventions in various cancers.
Article
Full-text available
Introduction Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer, and photodynamic therapy (PDT) is a promising modality against cSCC. This study investigated the impact of PDT on the MAPK pathway and cell cycle alternation of cSCC as well as the related molecular mechanisms. Method Expressing mRNA profile data sets GSE98767, GSE45216, and GSE84758 were acquired from the GEO database. The functions of differently expressed genes (DEGs) were enriched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Least absolute shrinkage and selection operator (Lasso) analysis were used to establish a diagnosis model based on GSE98767. A correlation analysis and a protein–protein interaction (PPI) network were used to evaluate the relationship between cSCC-PDT-related genes and the MAPK pathway. Single-sample gene set enrichment analysis (ssGSEA) was performed on GSE98767 to estimate MAPK activation and cell cycle activity. Finally, the effect of MAPK activation on the cell cycle was explored in vitro. Result Four cSCC-PDT-related genes, DUSP6, EFNB2, DNAJB1, and CCNL1, were identified as diagnostic markers of cSCC, which were upregulated in cSCC or LC50 PDT-protocol treatment and negatively correlated with the MAPK promoter. Despite having a smaller MAPK activation score, cSCC showed higher cell cycle activity. The PDT treatment suppressed the G1 to G2/M phase in JNK overexpressed A431 cells. Conclusion CCNL1, DNAJB1, DUSP6, and EFNB2 were identified as potential PDT target genes in cSCC treatment, whose potential therapeutic mechanism was inhibiting the MAPK pathway and inducing cell cycle alternation.
Article
KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.
Article
Full-text available
Reactive oxygen species (ROS) are mutagenic and may thereby promote cancer. Normally, ROS levels are tightly controlled by an inducible antioxidant program that responds to cellular stressors and is predominantly regulated by the transcription factor Nrf2 (also known as Nfe2l2) and its repressor protein Keap1 (refs 2-5). In contrast to the acute physiological regulation of Nrf2, in neoplasia there is evidence for increased basal activation of Nrf2. Indeed, somatic mutations that disrupt the Nrf2-Keap1 interaction to stabilize Nrf2 and increase the constitutive transcription of Nrf2 target genes were recently identified, indicating that enhanced ROS detoxification and additional Nrf2 functions may in fact be pro-tumorigenic. Here, we investigated ROS metabolism in primary murine cells following the expression of endogenous oncogenic alleles of Kras, Braf and Myc, and found that ROS are actively suppressed by these oncogenes. K-Ras(G12D), B-Raf(V619E) and Myc(ERT2) each increased the transcription of Nrf2 to stably elevate the basal Nrf2 antioxidant program and thereby lower intracellular ROS and confer a more reduced intracellular environment. Oncogene-directed increased expression of Nrf2 is a new mechanism for the activation of the Nrf2 antioxidant program, and is evident in primary cells and tissues of mice expressing K-Ras(G12D) and B-Raf(V619E), and in human pancreatic cancer. Furthermore, genetic targeting of the Nrf2 pathway impairs K-Ras(G12D)-induced proliferation and tumorigenesis in vivo. Thus, the Nrf2 antioxidant and cellular detoxification program represents a previously unappreciated mediator of oncogenesis.
Article
Full-text available
Recent advances in sequencing technology make it possible to comprehensively catalog genetic variation in population samples, creating a foundation for understanding human disease, ancestry and evolution. The amounts of raw data produced are prodigious, and many computational steps are required to translate this output into high-quality variant calls. We present a unified analytic framework to discover and genotype variation among multiple samples simultaneously that achieves sensitive and specific results across five sequencing technologies and three distinct, canonical experimental designs. Our process includes (i) initial read mapping; (ii) local realignment around indels; (iii) base quality score recalibration; (iv) SNP discovery and genotyping to find all potential variants; and (v) machine learning to separate true segregating variation from machine artifacts common to next-generation sequencing technologies. We here discuss the application of these tools, instantiated in the Genome Analysis Toolkit, to deep whole-genome, whole-exome capture and multi-sample low-pass (∼4×) 1000 Genomes Project datasets.
Article
Full-text available
Oncogenic KRAS is found in more than 25% of lung adenocarcinomas, the major histologic subtype of non-small cell lung cancer (NSCLC), and is an important target for drug development. To this end, we generated four NSCLC lines with stable knockdown selective for oncogenic KRAS. As expected, stable knockdown of oncogenic KRAS led to inhibition of in vitro and in vivo tumor growth in the KRAS-mutant NSCLC cells, but not in NSCLC cells that have wild-type KRAS (but mutant NRAS). Surprisingly, we did not see large-scale induction of cell death and the growth inhibitory effect was not complete. To further understand the ability of NSCLCs to grow despite selective removal of mutant KRAS expression, we conducted microarray expression profiling of NSCLC cell lines with or without mutant KRAS knockdown and isogenic human bronchial epithelial cell lines with and without oncogenic KRAS. We found that although the mitogen-activated protein kinase pathway is significantly downregulated after mutant KRAS knockdown, these NSCLCs showed increased levels of phospho-STAT3 and phospho-epidermal growth factor receptor, and variable changes in phospho-Akt. In addition, mutant KRAS knockdown sensitized the NSCLCs to p38 and EGFR inhibitors. Our findings suggest that targeting oncogenic KRAS by itself will not be sufficient treatment, but may offer possibilities of combining anti-KRAS strategies with other targeted drugs.
Article
Full-text available
Non-small cell lung carcinoma (NSCLC) is the leading cause of cancer-related death worldwide, with an overall 5-year survival rate of only 10–15% 1. Deregulation of the Ras pathway is a frequent hallmark of NSCLC, often through mutations that directly activate Kras 2. p53 is also frequently inactivated in NSCLC and, since oncogenic Ras can be a potent trigger of p53 3, it seems likely that oncogenic Ras signalling plays a major and persistent part in driving the selection against p53. Hence, pharmacological restoration of p53 is an appealing therapeutic strategy for treating this disease 4. Here, we model the likely therapeutic impact of p53 restoration in a spontaneously evolving mouse model of NSCLC initiated by sporadic oncogenic activation of endogenous Kras 5. Surprisingly, p53 restoration failed to induce significant regression of established tumours although it did result in a significant decrease in the relative proportion of tumours classed as high grade. This is due to selective activation of p53 only in the more aggressive tumour cells within each tumour. Such selective activation of p53 correlates with marked up regulation in Ras signal intensity and induction of the oncogenic signalling sensor p19ARF 6. Our data indicate that p53-mediated tumour suppression is triggered only when oncogenic Ras signal flux exceeds a critical threshold. Importantly, the failure of low-level oncogenic Kras to engage p53 reveals inherent limits in the capacity of p53 to restrain early tumour evolution and to the efficacy of therapeutic p53 restoration to eradicate cancers.
Article
Ras genes are the most common targets for somatic gain-of-function mutations in human cancer. Recently, germline mutations that affect components of the Ras-Raf-mitogen-activated and extracellular-signal regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway were shown to cause several developmental disorders, including Noonan, Costello and cardio-facio-cutaneous syndromes. Many of these mutant alleles encode proteins with aberrant biochemical and functional properties. Here we will discuss the implications of germline mutations in the Ras-Raf-MEK-ERK pathway for understanding normal developmental processes and cancer pathogenesis.
Article
Oncogenic mutations in B-Raf and Kirsten-Ras (K-Ras) are mutually exclusive during human cancer pathogenesis. In an effort to study the biological basis of this epistasis, gene targeting was used to create isogenic sets of human cancer cells differing only in presence or absence of endogenous oncogenic K-Ras or wild-type B-Raf. Whereas cells lacking the K-Ras oncogene were unable to efficiently form xenograft tumors, isogenic cells retaining activated K-Ras but deleted for B-Raf remained highly tumorigenic. Deletion of oncogenic K-Ras failed to reduce the activation state of B-Raf or ERK1/2, despite the requirement of oncogenic K-Ras for tumorigenesis. Genechip analysis revealed numerous genes in which the regulation by oncogenic K-Ras did not require B-Raf. These studies suggest that despite the mutual exclusivity of K-Ras and B-Raf mutations in human cancer and the well-described role for Raf proteins as Ras effectors, B-Raf is dispensable for K-Ras-mediated oncogenesis in a human cancer cell line. Additional studies are required to demonstrate the generalizability of these unexpected findings.
Article
Oncogenic ras can transform most immortal rodent cells to a tumorigenic state. However, transformation of primary cells by ras requires either a cooperating oncogene or the inactivation of tumor suppressors such as p53 or p16. Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest. The arrest induced by ras is accompanied by accumulation of p53 and p16, and is phenotypically indistinguishable from cellular senescence. Inactivation of either p53 or p16 prevents ras-induced arrest in rodent cells, and E1A achieves a similar effect in human cells. These observations suggest that the onset of cellular senescence does not simply reflect the accumulation of cell divisions, but can be prematurely activated in response to an oncogenic stimulus. Negation of ras-induced senescence may be relevant during multistep tumorigenesis.
Article
Colon cancers frequently harbor KRAS mutations, yet only a subset of KRAS mutant colon cancer cell lines are dependent upon KRAS signaling for survival. In a screen for kinases that promote survival of KRAS-dependent colon cancer cells, we found that the TAK1 kinase (MAP3K7) is required for tumor cell viability. The induction of apoptosis by RNAi-mediated depletion or pharmacologic inhibition of TAK1 is linked to its suppression of hyperactivated Wnt signaling, evident in both endogenous and genetically reconstituted cells. In APC mutant/KRAS-dependent cells, KRAS stimulates BMP-7 secretion and BMP signaling, leading to TAK1 activation and enhancement of Wnt-dependent transcription. An in vitro-derived "TAK1 dependency signature" is enriched in primary human colon cancers with mutations in both APC and KRAS, suggesting potential clinical utility in stratifying patient populations. Together, these findings identify TAK1 inhibition as a potential therapeutic strategy for a treatment-refractory subset of colon cancers exhibiting aberrant KRAS and Wnt pathway activation.
Article
NRAS is frequently mutated in hematologic malignancies. We generated Mx1-Cre, Lox-STOP-Lox (LSL)-Nras(G12D) mice to comprehensively analyze the phenotypic, cellular, and biochemical consequences of endogenous oncogenic Nras expression in hematopoietic cells. Here we show that Mx1-Cre, LSL-Nras(G12D) mice develop an indolent myeloproliferative disorder but ultimately die of a diverse spectrum of hematologic cancers. Expressing mutant Nras in hematopoietic tissues alters the distribution of hematopoietic stem and progenitor cell populations, and Nras mutant progenitors show distinct responses to cytokine growth factors. Injecting Mx1-Cre, LSL-Nras(G12D) mice with the MOL4070LTR retrovirus causes acute myeloid leukemia that faithfully recapitulates many aspects of human NRAS-associated leukemias, including cooperation with deregulated Evi1 expression. The disease phenotype in Mx1-Cre, LSL-Nras(G12D) mice is attenuated compared with Mx1-Cre, LSL-Kras(G12D) mice, which die of aggressive myeloproliferative disorder by 4 months of age. We found that endogenous Kras(G12D) expression results in markedly elevated Ras protein expression and Ras-GTP levels in Mac1(+) cells, whereas Mx1-Cre, LSL-Nras(G12D) mice show much lower Ras protein and Ras-GTP levels. Together, these studies establish a robust and tractable system for interrogating the differential properties of oncogenic Ras proteins in primary cells, for identifying candidate cooperating genes, and for testing novel therapeutic strategies.