SlideShare a Scribd company logo
1 of 11
Download to read offline
1 3
DOI 10.1007/s00018-014-1616-1 Cellular and Molecular Life Sciences
Cell. Mol. Life Sci. (2014) 71:3269–3279
Rev iew
Discoidin domain receptors: a proteomic portrait
Leo K. Iwai · Maciej T. Luczynski · Paul H. Huang 
Received: 29 January 2014 / Revised: 18 March 2014 / Accepted: 20 March 2014 / Published online: 5 April 2014
© Springer Basel 2014
activated by collagen [ 1]. These receptors regulate a num -
ber of fundamental biological processes including cell
adhesion, migration, proliferation, and extracellular remod-
eling [ 1–3]. Dysre gulation of DDR function has been
implicated in se veral diseases such as ἀbrosis, atheroscle -
rosis, arthritis, and cancer [1, 4]. There are two members in
this RTK class with DDR1 primarily expressed in epithelial
cells and DDR2 in mesench ymal cells. Correspondingly,
induction of epithelial–mesenchymal transition results in a
decrease in DDR1 e xpression and a reciprocal increase in
DDR2 levels [5, 6]. Their domain organization is composed
of an e xtracellular re gion containing tw o N-terminal dis -
coidin domains, an e xtracellular juxtamembrane domain,
a transmembrane domain, an intracellular juxtamembrane
domain, a kinase domain follo wed by a short C-terminal
tail. In addition, while DDR1 binds to both ἀbrillar and
basement membrane collagen, DDR2 preferentially binds
to ἀbrillar collagens and collagen type X [7–9].
Unlike classical growth factor RTKs like the epider-
mal growth f actor receptor (E GFR) and platelet-deri ved
growth f actor receptor (PDGFR), DDR eng agement by
collagen leads to a slo w and sustained acti vation proἀle in
which tyrosine phosphorylation levels start to increase only
hours after lig and stimulation and remains persistent o ver
several days [ 10, 11]. DDR1 and DDR2 contain 15 and
14 c ytoplasmic tyrosine residues, respecti vely, which are
potential sites for protein phosphorylation and binding to
downstream signaling adaptors and effectors. W hile a small
number of targeted studies have attempted to identify these
effectors, data from these studies have failed to comprehen-
sively deἀne the components of DDR signaling networks.
Advances have largely been hampered by challenges asso-
ciated with DDR biology including the unique receptor
activation kinetics and the inherent multi-lig and nature
of collagen. In addition, while there are an ab undance of
Abstract  The discoidin domain receptors (DDRs) are
collagen-binding receptor tyrosine kinases that ha ve been
implicated in a number of fundamental biological processes
ranging from growth and development to immunore gula-
tion. In this review, we examine how recent proteomic tech-
nologies ha ve enriched our understanding of DDR sign -
aling mechanisms. We provide an o verview on the use of
large-scale proteomic proἀling and chemical proteomics to
reveal novel insights into DDR therapeutics, signaling net -
works, and receptor crosstalk. A perspective of ho w prot-
eomics may be harnessed to answer outstanding fundamen-
tal questions including the dynamic re gulation of receptor
activation kinetics is presented. Collecti vely, these studies
present an emer ging molecular portrait of these unique
receptors and their functional role in health and disease.
Keywords  Discoidin domain receptors · Collagen ·
Cell signaling · Proteomics · Mass spectrometry
Introduction
The discoidin domain receptors (DDRs) are a class of
receptor tyrosine kinases (R TKs) that bind to and are
L. K. Iwai and M.T. Luczynski contributed equally to this work.
L. K. Iwai · M. T. Luczynski · P. H. Huang () 
Protein Networks Team, Division of Cancer Biology, Institute
of Cancer Research, 237 Fulham Road, London SW 3 6JB, UK
e-mail: paul.huang@icr.ac.uk
Present Address:
L. K. Iwai 
Laboratório E special de Toxinologia Aplicada/CeTICS, Instituto
Butantan, Av V ital Brasil 1500, São Paulo 05503‑000, Brazil
3270 L. K. Iwai et al.
1 3
studies that pro vide in vitro e vidence of the signaling and
functional properties of the DDRs in cell culture, there is
less data supporting the role of the DDRs as acti ve signal-
ing molecules in tissues and organs.
In the past 5 years, proteomics has undoubtedly emerged
as a po werful tool to further our understanding of DDR
biology. In this perspective, we will provide an overview
of how this technology has produced a molecular portrait
of DDR signaling mechanisms in human health and dis -
ease (outlined in Fig. 1). We further elaborate on the use
of proteomics to enrich our understanding of DDR struc -
ture–activity relationships as well as the reprogramming of
signaling in response to therapeutic intervention. Finally,
we offer a prospective view of the potential of proteomics
to pro vide future insights into une xplored areas of DDR
biology.
Proteomic proἀling of DDR activity in cancer
Phosphorylation is a re versible and highly dynamic signal
transduction mechanism that plays an important role in the
regulation of a myriad of cellular processes. The impor -
tance of this post-translational modiἀcation is e xempliἀed
by aberrant global phosphorylation e vents that ha ve been
reported in multiple pathologies such as cancer , and meta-
bolic and autoimmune diseases. In particular, dysregulation
of protein phosphorylation has been particularly well stud -
ied in cancer where multiple oncogenes and tumor suppres-
sors lie in protein phosphorylation pathw ays that directly
inḀuence cancer progression [12]. It is thus not surprising
that drugs tar geting protein kinases represent the lar gest
class of compounds currently a vailable to treat this disease
[13]. Le veraging on the dependenc y of cancer cells for
oncogenic signaling pathw ays, these kinase-speciἀc tar -
geted therapies ha ve led to dramatic clinical responses in
subsets of cancer patients [ 14–16]. Furthermore, monitor -
ing components in kinase signaling pathw ays are increas -
ingly being used as biomark ers for stratiἀcation of patient
response to therapies as well as recruitment criteria for clin-
ical trials. These developments in clinical practice to wards
personalized medicine ha ve made it necessary not only to
better understand the dynamic nature of kinase signaling
networks but also to de velop improved methods for moni -
toring phosphorylation levels in clinical specimens.
Ampliἀcation and mutations in the DDRs ha ve been
reported in multiple cancer types, including breast, o var-
ian, brain, and lung tumors [17–21] (a complete review of
the role of DDRs in cancer can be found in [3]). Previous
studies in E GFR-positive cancer patients suggest that total
Fig. 1  A timeline of proteomic studies that have informed our current understanding of discoidin domain receptor biology
3271Proteomics of discoidin domain receptors
1 3
E GFR protein levels is a poor predictor of patient response
to E GFR kinase inhibitors. In contrast, a high phospho-
E GFR level, the active form of the receptor, is a good prog-
nostic indicator of sensiti vity to E GFR-targeted therap y
[22, 23]. Along these lines, it w ould be vital to determine
if DDRs are in f act active in tumors where protein o verex-
pression/mutation is observ ed and if so, do the y represent
good biomarkers for patient response to kinase inhibitors.
Despite the functional importance of the DDR acti vity in
vitro, for about a decade follo wing the disco very of the
DDRs, there were no studies that conclusi vely demon -
strated that these receptors were indeed phosphorylated or
active in vivo. This paucity of e vidence was largely due to
the lack of phospho-speciἀc reagents (e.g., antibodies) tar -
geting this class of receptors.
The introduction of unbiased phosphoproteomic proἀl -
ing by mass spectrometry (MS) rapidly o vercame these
technical challenges. Rik ova et   al. [ 24] pro vided the ἀrst
study to demonstrate the scale of DDR phosphorylation in
vivo. The authors performed a lar ge-scale screen of tyros -
ine phosphorylation levels in 150 non-small cell lung carci -
noma (NSCLC) tumors and 41 cell lines. They were able to
identify more than 2,700 tyrosine-phosphorylated proteins,
which included over 50 different tyrosine kinases. This study
established that signiἀcant le vels of phosphorylated DDR1
and 2 were present in lung cancer . A comparison of phos -
phorylated tyrosine kinase proἀles in tumors versus cell lines
raised an interesting observ ation. W hile the analysis of cell
lines revealed the activation of common NSCLC targets such
as E GFR, c-Met, and E phA2 [ 25–27], the patient-deri ved
tumor proἀles sho wed that the DDRs w as by f ar the most
commonly activated tyrosine kinase class in vivo, accounting
for more than a third of the tumors analyzed. Importantly ,
very few DDR1 and no DDR2 phosphorylation e vents were
found in the cell lines. This data highlights the limited utility
of NSCLC cell lines in recapitulating in vivo tyrosine kinase
signaling events observed in patients. Furthermore, the pres -
ence of DDR acti vation only in clinical specimens suggests
that the lung tumor microen vironment, which contains high
levels of collagen [28], is critical for modulating signaling
networks in vivo. The same group performed a subsequent
screen of cholangiocarcinoma tumors from 23 patients [ 29].
The unique aspect of this study was the comparison of tumor
specimens with para-tumor (normal) tissues. Consistent with
the NSCLC study, high levels of activated DDRs was found
in more than 70  % of the patients studied, conἀrming that
these receptors are prevalent in multiple tumor types.
More recently, a phosphoproteomic analysis of ten cell
lines and three human tumors representing different forms
of the sarcoma identiἀed tyrosine phosphorylation of
DDR2 in 9/10 cell lines and 1/3 tumors [ 30]. These ἀnd-
ings are congruent with the restricted e xpression of DDR2
in cells of mesench ymal origin. Furthermore, sarcoma
cell lines are kno wn to secrete a lar ge amount of collagen,
which represents a source of autocrine lig and for the acti -
vation of DDR2 [ 10, 31, 32]. It should be noted that all of
the described studies above also identiἀed a large network
of other RTKs that are simultaneously co-acti vated in can-
cer cell lines and tumors that poses several important ques-
tions [33, 34]. Firstly, why do cells simultaneously activate
multiple tyrosine kinases in the context of cancer but not in
normal tissues? The second question is ho w does one de-
convolute these complex RTK networks and resolve which
downstream signals are speciἀc to particular R TKs such as
the DDRs? Finally, is there bi-directional crosstalk between
the DDRs and other tyrosine kinases embedded within
these RTK co-activation networks?
A recent proteomic proἀling in glioblastoma sheds some
light on the latter question. In this study , eight patient-
derived glioblastoma tumor x enografts e xpressing wild-
type or mutant forms of E GFR were subjected to both
quantitative proteomic and phosphoproteomic analysis
[35]. One of the intriguing ἀndings of this study w as that
well-established interactors of E GFR such as SHC, STAT3,
Cbl, Gab1, and PLC γ1 did not cluster with E GFR phos -
phorylation. In contrast, phosphorylation sites on the acti -
vation loop of DDR1 strongly correlated with the majority
of E GFR tyrosine phosphosites across all the tumors stud -
ied. This clustering analysis suggests that in GBM tumors,
crosstalk between E GFR and DDR1 is lik ely to be a sig -
niἀcant event, and that the resultant do wnstream pathways
from this interaction may be distinct from E GFR speciἀc
signaling networks. Taking an in vitro approach, our group
explored the potential for crosstalk between the insulin
receptor (IR) and DDR2. H E K293 cells e xpressing both
DDR2 and IR w as subjected to stimulation with insulin
and/or collagen [36]. Using iTRAQ labeling methodol-
ogy, we sho wed that insulin enhanced collagen-mediated
DDR2 activation. We also identiἀed SHIP-2 and SGK269
as potential do wnstream effectors that arise from the inte -
grated activation of DDR2 and IR. The exact mechanism
by which IR and E GFR interact with the DDRs is still
uncertain. Ho wever, the observ ed crosstalk between the
DDRs and other R TKs may represent a means for signal
diversiἀcation of collagen receptor signaling to achie ve
robustness despite e xposure to e xogenous insults, includ -
ing tar geted therap y [ 37]. Further in vestigation into the
extent and biological consequence of DDR-R TK crosstalk
is needed to resolve these questions.
The identiἀcation of acti ve DDRs in multiple cancers
conἀrms that DDR acti vation occurs in vi vo and may play
an important role in the tumor initiation and/or progres -
sion. These developments have led the w ay for the search
of small molecule inhibitors that would target the DDRs
and disrupt its acti vity and resultant signaling pathw ays in
cancer.
3272 L. K. Iwai et al.
1 3
Chemical proteomics and the DDRs
In order to better characterize the mode of action of kinase
inhibitors, MS-based chemical proteomics has successfully
been used to identify the tar get(s) of these small molecules
and their effects on tumor signaling netw orks. In a typical
chemical proteomics w orkḀow, an immobilized chemi -
cal probe or drug of interest is e xposed to a protein e xtract
and target proteins that bind to these probes are then iden -
tiἀed and quantiἀed by disco very-based MS proteomics
(Fig. 2a). This approach has been ef fective in identifying
the target proἀle of the multi-kinase inhibitors de veloped
for treatment of chronic myelogenous leuk emia (CML).
Kinase inhibitors such as dasatinib, nilotinib, and imatinib,
which tar get the constituti vely acti ve BCR-ABL fusion
oncoprotein, have been approved for the treatment of CML
since 2003. Imatinib is the ἀrst-line treatment for CML
positive for the Philadelphia chromosome, while dasatinib
and nilotinib are currently being used for imatinib-intol-
erant and -resistant CML patients, although the y still f ail
in patients harboring the g atekeeper mutation at T315I in
BCR-ABL [38].
Despite being rationally designed to target BCR-ABL,
several chemical proteomic studies ha ve since sho wn that
these drugs also inhibit other kinases including c-Src,
c-KIT, PDGFR, and the DDRs [ 39–41]. In f act, Imatinib
is no w appro ved for adv anced g astrointestinal stromal
tumor (GIST) patients with c-KIT e xpression as well as
other malignancies with PDGFR translocations [42–44].
Through an afἀnity-based chemical proteomic approach
using kinobeads (an immobilized non-selecti ve kinase
inhibitor matrix) in K562 CML cells, Bantscheff et al. [39]
identiἀed o ver 150 protein kinases that were capable of
binding to imatinib and dasatinib with varying afἀnities. In
addition to conἀrming the mode of action of these drugs
on the ABL and SRC f amily kinases, the study also deter -
mined that both DDR1 and 2 bound to these compounds
with nanomolar afἀnity . The authors v alidated these ἀnd -
ing using in vitro kinase assays, which demonstrated that
DDR1 is a bona ἀde tar get of imatinib . A further study
by Rix et  al. [41] on all three compounds immobilized to
a Sepharose matrix and incubated with K562 and CML
primary cell lysates sho wed that nilotinib and dasatinib
also bound strongly to DDR1. These studies corroborate
sequence alignment and molecular modeling experiments
which show striking similarities in the ATP-binding pocket
of the DDRs and other identiἀed tar get kinases such as
ABL and c-KIT [ 45]. Taken together, these chemical pro -
teomic studies demonstrate that these three compounds
are candidate drugs for malignancies dri ven by oncogenic
DDRs. Consistent with this idea, lung cancer cell lines with
oncogenic DDR2 mutations are susceptible to dasatinib
treatment [19].
Chemical proteomic analysis of cancer therapeutics
is not limited to kinase inhibitors. HSP90 is a chaper -
one protein that assists in the folding and stabilization of
multiple client proteins, including well-established onco -
proteins such as B-RAF , E RBB2, CDK4, and BCR-ABL
[46]. The essential function of HSP90 in maintaining
oncoprotein expression provides a therapeutic rationale
for the use of HSP90 inhibitors in cancer therap y. Using
stable isotope labeling with amino acids in cell culture
(SILAC) methodology , Sharma et   al. [ 47] treated HeLa
cervical cancer cells with the HSP90 inhibitor , 17-DMAG
[17-(dimethylaminoethylamino)-17-demethoxygeldan-
amycin] and monitored changes in 6,000 proteins and
4,000 phosphopeptides. This study sho wed that inhibition
of HSP90 results in pleiotropic ef fects on multiple cellu -
lar processes including DNA metabolism, protein synthesis
and degradation, cell cycle, and apoptosis.
In another study performed by W u et al. [48], an inte -
grated proteomics strate gy comprising multiple dif fer-
ent techniques was used to study the ef fects of the HSP90
inhibitor geldanamycin on four dif ferent cancer cell lines.
These techniques comprise SILAC quantiἀcation and
chemical precipitation of kinases using kinobeads, HSP90
immunoprecipitation, and immobilized geldanamycin on
Sepharose beads. They identiἀed o ver 1,600 proteins that
were signiἀcantly altered upon drug treatment. Of these,
98 were protein kinases that were do wn-regulated and
classiἀed as HSP90 clients. DDR1 was among this list of
kinases and a protein turno ver-rate analysis using pulsed
SILAC e xperiments at 6, 12, and 24   h combined with
kinobeads enrichment sho wed that, upon drug treatment,
DDR1 underwent a more rapid de gradation compared
to E GFR, E RK1, or E PHA2. This observ ation pro vides
important pharmacokinetic information for drug response
where targeting HSP90 w ould lead to a more rapid ef fect
on DDR1 compared to other kinases and client proteins.
Fig. 2  Proteomic strate gies used to elucidate discoidin domain
receptor signaling netw orks and chemical interactions. a Schematic
of general chemical proteomic approach for identiἀcation of drug/
probe-binding target proteins. Cell or tissue lysate is incubated with
immobilized probes (e.g., kinobeads) or drug to isolate interacting
proteins. Subsequent elution and mass spectrometry analysis result
in the identiἀcation of probe/drug tar gets. b Different methodologies
used for the identiἀcation of DDR signaling. (i) Proἀling of activated/
phosphorylated kinases in cancer cell lines and tumors using global
phosphoproteomic approaches. ( ii) Catalog ef fects of drug treatment
(kinase or HSP90 inhibitors) in cancer cell lines on cellular pro -
teome and phosphoproteome can inform potential no vel combination
regimens for disease. ( iii) Treatment of DDR-e xpressing cells with
phosphatase inhibitor results in global upre gulation of cellular phos -
phorylation. Subsequent DDR immunoprecipitation and mass spec-
trometry analysis identiἀes phosphorylation-mediated protein inter -
actions. (iv) Stimulation of DDRs with exogenous collagen leads to
the activation of receptor -speciἀc signaling netw orks, which can be
proἀled with global phosphoproteomics
▸
3273Proteomics of discoidin domain receptors
1 3
A
B
3274 L. K. Iwai et al.
1 3
Another study employing a similar approach also identiἀed
DDR2 as client protein of HSP90 [ 49]. Collectively, these
data demonstrate that the DDRs are major client proteins
of HSP90, and HSP90 inhibitors may ha ve utility ag ainst
tumors that are DDR positive.
These studies also raise an interesting question as to
whether a combination approach of using both kinase and
HSP90 inhibitors would be more effective in eliminating
DDR oncogenic signaling compared to either drug alone
(Fig. 2b). This combination would be particularly important
since gatekeeper mutants of DDRs are resistant to dasat -
inib inhibition [ 19, 30, 45, 50]. W hile such DDR muta -
tions have yet to be reported in patients, based on reports
in other oncogenic kinases [ 51, 52], it is lik ely that g ate-
keeper mutations will arise with prolonged use of imatinib
or dasatinib. Indeed, in vitro modeling of dasatinib resist -
ance in lung cancer cell lines results in the de velopment of
the T654I gatekeeper mutation in DDR2 as a mechanism
of acquired resistance [53]. In a recent study of HSP90 and
its recruitment to protein kinases via CDC37, Polier et  al.
[54] analyzed the mode of action of dif ferent ATP-compet-
itive kinase inhibitors on HSP90-CDC37-kinase chaper -
one complex formation. Their study revealed a dual mode
of action for this class of inhibitors. These drugs not only
acted as con ventional ATP-competitive kinase inhibitors
by competing for ATP binding b ut also serv ed as antago -
nists depriving client kinase recruitment to the HSP90 and
CDC37 chaperone system f acilitating kinase de gradation
by proteasomes. These data suggest that a combination
with HSP90 inhibition may boost the client protein degra-
dation capability of kinase inhibitors, which will enhance
therapeutic efἀcac y and reduce the lik elihood of de vel-
oping drug-resistant gatekeeper mutations. Furthermore,
multi-target kinase inhibitors, such as dasatinib described
above, inhibit the DDRs as an unanticipated “off-target”
effect. More recently, DDR selective inhibitors, which are
capable of inhibiting cancer cell proliferation and migra -
tion, have been identiἀed by several groups [55–59]. These
compounds have the potential to progress to become potent
anti-cancer agents in DDR-driven cancers.
Cellular signaling is a dynamic process that constantly
adapts to both endogenous and e xogenous stimuli. Another
interesting facet of DDR biology is the ability of tumor cells
to modulate receptor expression levels in response to thera-
peutic challenge. This “kinase reprogramming” phenom -
enon was studied in depth by Duncan et  al. using a chemi -
cal proteomics approach. E mploying multiple xed kinase
inhibitor beads and mass spectrometry (MIB/MS) to enrich
for kinases in triple-ne gative breast cancer cells, treatment
with ME K inhibitors AZD6244 and U0126 resulted in sig -
niἀcant reprogramming of the kinome induced by the pro -
teasomal c-Myc de gradation [ 60]. Initial treatment with
the drugs inhibited cell gro wth and elevated RNA levels of
RTKs including DDR1 and DDR2. Continuous drug e xpo-
sure o ver 30   days generated therap y-resistant cells with
increased protein levels of ME K2, VE GFR2, and PDGFRβ
and elevated tyrosine phosphorylation of ME K, AXL, RAF,
and AKT. The individual depletion of some of the repro -
grammed R TKs such as DDR1, DDR2, AXL, VE GFR2,
and PDGFR β using siRN A in combination with U0126
were able restore gro wth arrest. This data indicates that a
single knockdown of DDR1 or DDR2 in the presence of a
ME K inhibitor is able to kill triple-negative breast cancer
cells. Interestingly , combination therap y using AZD6244
with a low dose of RTK inhibitors (sorafenib, a PDGFRα/β,
VE GFR, DDR1/2, and RAF inhibitor; and foretinib, a cMet
and VE GFR inhibitor), which were inef fective as single
agents, inhibited phosphorylation of multiple R TKs and
efἀciently arrested growth of SUM159 triple-negative breast
cancer cells. Importantly , in vi vo experiments using breast
cancer C3Tag mouse model treated orally with AZD6244
also showed kinome reprogramming with increased expres-
sion of M E K2, E RK1, PDGFRβ, and DDR1 recapitulating
the reprogramming response observ ed in vitro. Treatment
of C3Tag mice with AZD6244 and Sorafenib led to tumor
growth arrest, increased apoptosis, and tumor regression
in the majority of mice. These ἀndings show that a deἀned
combination therapy based on the a priori knowledge of the
kinome pattern of resistance is ef fective in producing sig -
niἀcant therapeutic beneἀt.
Although the role of DDR in cancer is still poorly
characterized, MS-based chemical proteomics has been
exploited to monitor changes in the composition of the
cancer kinome, including DDR1 and DDR2, upon M E K
inhibitor treatment. Cancer cells circumv ent the inhibition
of oncogenic signaling pathways by upregulating the DDRs
and additional survival routes, highlighting the importance
of this class of receptors in conferring resistance to kinase
inhibitor therapy. On a more global scale, multiple cancer
types are driven by Myc activation [61] and this study sug-
gests that upre gulation of DDRs in the conte xt of a R TK
co-activation netw ork may be a general mechanism by
which cancer cells evade elimination of the Myc pathway.
DDR signaling networks
Perhaps the most substantial contrib ution of proteomics to
DDR biology is the lar ge-scale elucidation of its signaling
Fig. 3  Reported intracellular protein–protein interactions and do wn-
stream signaling components of a DDR1 and b DDR2. The ἀgure
depicts known tyrosine phosphorylation sites ( black squares) in the
cytoplasmic domains of both receptors and the do wnstream effector
proteins (white squares) that have been shown to either interact with
or be phosphorylated by the DDRs. Associated references for the
reported interactions are indicated in parentheses
▸
3275Proteomics of discoidin domain receptors
1 3
A
B
3276 L. K. Iwai et al.
1 3
networks. There is a wealth of kno wledge of the collagen-
binding properties of the DDRs [ 2]; in contrast, the sign -
aling pathw ays acti vated by these receptors are lar gely
unknown. A decade of biochemical studies on the DDRs
has identiἀed a handful of signaling ef fectors including
SHC, CSK, PI3K, SHP2, R UNX2, and NCK1/2 [ 11, 62–
67]. E merging data from proteomics studies ha ve revealed
complex interactions in DDR do wnstream signaling net -
works. F or instance, in order to characterize the global
landscape of possible DDR1 interacting proteins, Lemeer
et  al. emplo yed a strate gy where DDR1-o verexpressing
glioblastoma cells were treated with a tyrosine phosphatase
inhibitor pervanadate, and subjected to co-immunoprecip -
itation with anti-DDR1 antibodies (Fig.   2b) [ 68]. Utiliz -
ing quantitative MS based on SILA C methodology, more
than 30 proximal signaling proteins interacting with DDR1
were identiἀed. These proteins include some pre viously
described interactors b ut also highlighted ne w interactions
such as GRB2, E PHA2, RASGAP, SHIP1, SHIP2, and
STATs. Furthermore, in order to identify the speciἀc tyros -
ine phosphorylation residues in DDR1 that were responsi -
ble for mediating these interactions, the authors performed
phosphopeptide pull-down experiments incubating placenta
lysates with all 15 DDR1 tyrosine phosphorylated synthetic
peptides immobilized on Sepharose beads. They addition-
ally validated the identiἀed interactors by performing quan-
titative competition-binding experiments with phosphoryl-
ated peptides and non-phosphorylated controls using TMT
6-plex chemical labeling follo wed by MS analysis. RAS -
GAP was shown to interact with Y484, Y543, and Y586
while STAT1a/b, STAT3, and ST AT5b with the juxtam -
embrane Y703 and the acti vation loop Y796 sites. In addi -
tion, they found that both activators (PI3K was shown to
bind to pY881) and ne gative regulators (SHIP1 and SHIP2
were shown to bind to pY740) of the PI3K pathw ay were
arranged in close proximity on DDR1, indicating a poten -
tial mechanism for compartmentalized pathw ay regulation.
Taken together, this work characterizes the cellular interac-
tome of DDR1 and pro vides a roadmap for future mecha -
nistic studies on the roles of these effectors on DDR1 sign-
aling and biology . The binding partners to the DDR1 and
its speciἀc phosphorylation sites are summarized in Fig. 3a
[11, 48, 62–65, 68–77].
W hile the interactome study pro vides a catalogue of all
possible DDR1 interactions, it may not accurately repre -
sent the situation in cells since both temporal and spatial
effects are o verlooked in these e xperiments. Furthermore,
it is unclear as to which of the tyrosine residues in DDR1
are actually phosphorylated in cells in response to collagen
activation since the global tyrosine phosphatase inhibitor
pervanadate was used in this study. Using a complementary
strategy to identify DDR signaling effectors, our group has
performed an iTRAQ-based quantitative phosphoproteomic
analyses in DDR2-overexpressing cells to map the tempo -
ral activation of signaling networks in response to collagen
stimulation over the course of 24  h (Fig. 2b) [78]. We ἀnd
that speciἀc phosphorylation sites on the DDR2 recep -
tor were dif ferentially re gulated upon collagen eng age-
ment in cells. For instance, the activation loop sites Y736
and Y740 as well as the no vel Y684 and Y813 sites pre -
sented a delayed acti vation with maximal phosphorylation
at 24   h, while Y481 in the juxtamembrane re gion of the
receptor displayed a distinct constitutively phosphorylated
proἀle. These ἀndings support the idea that the re gulation
of RTK signaling is achie ved through site-speciἀc dif fer-
ences in both magnitude and kinetics of receptor phospho -
rylation [79]. Bioinformatic analysis identiἀed 45 proteins
including SHP-2, SHIP-2, PIK3C2A, E RK1, and PLCL2
that clustered strongly with DDR2, implicating these pro -
teins as downstream effectors of DDR2 signaling netw ork.
Importantly, these phosphorylation e vents were sho wn to
be speciἀc to DDR2 activation and independent of the col-
lagen-binding inte grins. These DDR2 signaling ef fectors
are summarized in Fig. 3b [36, 66, 67, 80–82].
Perspectives
An outstanding fundamental question in DDR biology is
how collagen binding promotes the delayed and sustained
kinetics of DDR acti vation. DDRs e xist as preformed
dimers [83–85] that under go a series of autophosphoryla -
tion e vents upon lig and eng agement. Understanding the
sequential order of receptor phosphorylation may shed
light on the molecular basis of the unique kinetic proἀle of
the DDRs. W hile proteomic studies have shown that differ-
ent sites on the DDRs have distinct activation kinetics [78],
the initial phosphorylation and subsequent dephosphoryla -
tion dynamics of these receptors are unknown. MS in com-
bination with quenching is a useful tool for elucidating the
site-speciἀc order of such R TK phosphorylation e vents.
E mploying this approach, Furdui et  al. ele gantly sho wed
that ἀbroblast growth factor receptor 1 (FGFR1) autophos -
phorylation is dictated by a precisely ordered sequence of
events [79]. In this method, FGFR1 and ATP are mixed and
quenched prior to sequencing by MS, which allo wed the
authors to monitor each distinct tyrosine phosphorylation
site on FGFR1 re vealing a unique receptor acti vation pro-
ἀle. They showed that initially , asymmetric dimer forma -
tion of the receptor f acilitates the transphosphorylation of
an activation loop tyrosine residue, followed by subsequent
phosphorylation of other docking site tyrosines. In the last
step, a dif ferent tyrosine residue in the acti vation loop is
phosphorylated, increasing the overall kinase activity of the
receptor, f acilitating do wnstream signal transduction [ 79,
86]. A similar characterization of temporal DDR receptor
3277Proteomics of discoidin domain receptors
1 3
phosphorylation will identify the key tyrosine residues that
are critical for kinase activity and the mechanisms that gov-
ern the observed delayed and sustained activation kinetics.
The DDRs and their lig ands are subjected to dynamic
regulation in vivo [87]. For instance, receptor acti vation is
modulated by matrix metalloproteinases either directly by
cleavage of its e xtracellular domain or indirectly through
degradation of the collagen matrix [ 88]. In addition, ligand
competition by matrisomal proteins such as SPARC and
crosstalk with other transmembrane receptors including
the integrins may also inḀuence DDR acti vation and con -
sequent downstream signaling [7, 89, 90]. E lucidating this
complex biology will require the acquisition of dynamic
signaling maps that detail netw ork changes in response to
cellular perturbation. Building on previous DDR interac-
tome studies, tar geted proteomic approaches such as mul -
tiple reaction monitoring (MRM) offers the powerful capa-
bility of reproducible acquisition of dynamic data across
multiple conditions, replicates and protein species. A recent
example of this technology by Zheng et   al. [ 91] showed
that the SHC1 adaptor protein is a highly dynamic regula-
tor of E GFR signaling netw orks. In this study , ἀbroblasts
were stimulated with E GF and dif ferent phosphorylation
sites within SHC1 as well as 41 dif ferent SHC1-protein
binding partners were monitored o ver 16 time points by
MRM. This high-resolution analysis of the assembly/dis -
assembly dynamics of protein-binding partners to SHC1
revealed a stage-speciἀc function of this scaf fold protein.
Upon initial activation by E GF, proteins directing cell divi-
sion and survi val were enriched, switching to an interme -
diate stage involving functions like vesicle trafἀcking, and
ἀnally progressing to a ἀnal stage characterized by proteins
involved on cytoskeletal reorganization and downregulation
of the initial cell di vision signals. Applying this strate gy
to the dynamic interactome and signaling netw orks of the
DDRs may provide a molecular explanation of the intrigu -
ing activation kinetics of the DDRs and the functional rel -
evance of its slow and prolonged signaling network proἀle.
Conclusions
Proteomics has contributed to the unraveling of DDR biol-
ogy and their functional impact in health and disease. W hile
biochemical, structural, genetic, and physiological studies
performed by multiple groups in last decade have undoubt-
edly led to a better understanding of their biological prop -
erties, there is a still much to learn about these atypical
RTKs. In an era of collaborati ve science and inte grative
biology, we anticipate that “Omic” technologies (including
proteomics) in combination with classical approaches hold
the exciting potential to pro vide a comprehensive molecu-
lar portrait of these receptors.
Acknowledgments  The work in the authors’ laboratory is funded
by the Wellcome Trust (W T089028) and the Biotechnology and Bio -
logical Sciences Research Council (BB/I014276/1).
References
	 1.	 Fu HL et   al (2013) Discoidin domain receptors: unique recep -
tor tyrosine kinases in collagen-mediated signaling. J Biol Chem
288(11):7430–7437
	 2.	 Leitinger B (2011) Transmembrane collagen receptors. Annu Rev
Cell Dev Biol 27:265–290
	 3.	Valiathan RR et   al (2012) Discoidin domain receptor tyrosine
kinases: ne w players in cancer progression. Cancer Metastasis
Rev 31(1–2):295–321
	 4.	Vogel W F, Abdulhussein R, F ord CE (2006) Sensing e xtracellu-
lar matrix: an update on discoidin domain receptor function. Cell
Signal 18(8):1108–1116
	 5.	 Maeyama M et  al (2008) Switching in discoid domain receptor
expressions in SLUG-induced epithelial–mesenchymal transition.
Cancer 113(10):2823–2831
	 6.	Walsh LA, Na wshad A, Medici D (2011) Discoidin domain
receptor 2 is a critical re gulator of epithelial–mesenchymal tran-
sition. Matrix Biol 30(4):243–247
	 7.	 Carafoli F, Hohenester E (2013) Collagen recognition and trans -
membrane signalling by discoidin domain receptors. Biochim
Biophys Acta 1834(10):2187–2194
	 8.	 Leitinger B, Hohenester E (2007) Mammalian collagen receptors.
Matrix Biol 26(3):146–155
	 9.	 Leitinger B, Kw an AP (2006) The discoidin domain recep -
tor DDR2 is a receptor for type X collagen. Matrix Biol
25(6):355–364
	10.	 Shrivastava A et   al (1997) An orphan receptor tyrosine kinase
family whose members serv e as noninte grin collagen receptors.
Mol Cell 1(1):25–34
	11.	Vogel W et  al (1997) The discoidin domain receptor tyrosine
kinases are activated by collagen. Mol Cell 1(1):13–23
	12.	 Blume-Jensen P, Hunter T (2001) Oncogenic kinase signalling.
Nature 411(6835):355–365
	13.	 Cohen P, Alessi DR (2013) Kinase drug disco very—what’s next
in the ἀeld? ACS Chem Biol 8(1):96–104
	14.	 Lynch TJ et   al (2004) Activating mutations in the epidermal
growth factor receptor underlying responsi veness of non-small-
cell lung cancer to geἀtinib. N E ngl J Med 350(21):2129–2139
	15.	 Gerber DE , Minna JD (2010) ALK inhibition for non-small cell
lung cancer: from disco very to therap y in record time. Cancer
Cell 18(6):548–551
	16.	 Druker BJ et al (2001) E fἀcacy and safety of a speciἀc inhibitor
of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N
E ngl J Med 344(14):1031–1037
	17.	Weiner HL et  al (2000) Consistent and selective expression of
the discoidin domain receptor -1 tyrosine kinase in human brain
tumors. Neurosurgery 47(6):1400–1409
	18.	 Yamanaka R et al (2006) Identiἀcation of e xpressed genes char-
acterizing long-term survival in malignant glioma patients. Onco-
gene 25(44):5994–6002
	19.	 Hammerman PS et al (2011) Mutations in the DDR2 kinase gene
identify a no vel therapeutic target in squamous cell lung cancer .
Cancer Discov 1(1):78–89
	20.	 Ren T et   al (2013) Increased e xpression of discoidin domain
receptor 2 (DDR2): a novel independent prognostic marker of
worse outcome in breast cancer patients. Med Oncol 30(1):397
	21.	 Ford CE et   al (2007) E xpression and mutation analysis of the
discoidin domain receptors 1 and 2 in non-small cell lung carci -
noma. Br J Cancer 96(5):808–814
3278 L. K. Iwai et al.
1 3
	22.	Van Schaeybroeck S et al (2005) E pidermal growth factor recep-
tor activity determines response of colorectal cancer cells to geἀ -
tinib alone and in combination with chemotherap y. Clin Cancer
Res 11(20):7480–7489
	23.	 Thomson S et al (2005) E pithelial to mesenchymal transition is a
determinant of sensiti vity of non-small-cell lung carcinoma cell
lines and xenografts to epidermal growth factor receptor inhibi-
tion. Cancer Res 65(20):9455–9462
	24.	 Rikova K et   al (2007) Global surv ey of phosphotyrosine
signaling identiἀes oncogenic kinases in lung cancer. Cell
131(6):1190–1203
	25.	 Amann J et al (2005) Aberrant epidermal growth factor receptor
signaling and enhanced sensiti vity to E GFR inhibitors in lung
cancer. Cancer Res 65(1):226–235
	26.	 Garofalo M et   al (2012) E GFR and M E T receptor tyrosine
kinase-altered microRNA expression induces tumorigenesis and
geἀtinib resistance in lung cancers. Nat Med 18(1):74–82
	27.	 Ishikawa M et al (2012) Higher expression of E phA2 and ephrin-
A1 is related to f avorable clinicopathological features in patho -
logical stage I non-small cell lung carcinoma. Lung Cancer
76(3):431–438
	28.	 van K empen LC et   al (2003) The tumor microen vironment:
a critical determinant of neoplastic e volution. E ur J Cell Biol
82(11):539–548
	29.	 Gu TL et al (2011) Surv ey of tyrosine kinase signaling re veals
ROS kinase fusions in human cholangiocarcinoma. PLoS One
6(1):e15640
	30.	 Bai Y et   al (2012) Phosphoproteomics identiἀes dri ver tyros -
ine kinases in sarcoma cell lines and tumors. Cancer Res
72(10):2501–2511
	31.	 Alitalo K et   al (1982) Biosynthesis of type V procollagen by
A204, a human rhabdomyosarcoma cell line. J Biol Chem
257(15):9016–9024
	32.	 Kleman JP et  al (1992) The human rhabdomyosarcoma cell line
A204 lays do wn a highly insoluble matrix composed mainly of
alpha 1 type-XI and alpha 2 type- V collagen chains. E ur J Bio -
chem 210(1):329–335
	33.	 Xu AM, Huang PH (2010) Receptor tyrosine kinase coacti vation
networks in cancer. Cancer Res 70(10):3857–3860
	34.	 Huang PH (2012) Phosphoproteomic studies of receptor tyrosine
kinases: future perspectives. Mol BioSyst 8(4):1100–1107
	35.	 Johnson H et al (2012) Molecular characterization of E GFR and
E GFRvIII signaling networks in human glioblastoma tumor x en-
ografts. Mol Cell Proteomics 11(12):1724–1740
	36.	 Iwai LK, Chang F , Huang PH (2013) Phosphoproteomic analy -
sis identiἀes insulin enhancement of discoidin domain receptor 2
phosphorylation. Cell Adh Migr 7(2):161–164
	37.	 Meyer AS et al (2013) The Receptor AXL diversiἀes E GFR sign-
aling and limits the response to E GFR-targeted inhibitors in tri-
ple-negative breast cancer cells. Sci Signal 6(287):ra66
	38.	Vajpai N et  al (2008) Solution conformations and dynamics of
ABL kinase-inhibitor comple xes determined by NMR substanti -
ate the different binding modes of imatinib/nilotinib and dasat-
inib. J Biol Chem 283(26):18292–18302
	39.	 Bantscheff M et   al (2007) Quantitati ve chemical proteomics
reveals mechanisms of action of clinical ABL kinase inhibitors.
Nat Biotech 25(9):1035–1044
	40.	Weisberg E et al (2005) Characterization of AMN107, a selective
inhibitor of native and mutant Bcr-Abl. Cancer Cell 7(2):129–141
	41.	 Rix U et al (2007) Chemical proteomic proἀles of the BCR-ABL
inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase
and nonkinase targets. Blood 110(12):4055–4063
	42.	 Joensuu H et   al (2001) E ffect of the tyrosine kinase inhibitor
STI571 in a patient with a metastatic g astrointestinal stromal
tumor. N E ngl J Med 344(14):1052–1056
	43.	 Demetri GD et al (2002) E fἀcacy and safety of imatinib mesylate
in adv anced g astrointestinal stromal tumors. N E ngl J Med
347(7):472–480
	44.	 Heinrich MC et al (2003) Kinase mutations and imatinib response
in patients with metastatic gastrointestinal stromal tumor. J Clin
Oncol 21(23):4342–4349
	45.	 Day E et   al (2008) Inhibition of collagen-induced discoidin
domain receptor 1 and 2 activation by imatinib, nilotinib and
dasatinib. E ur J Pharmacol 599(1–3):44–53
	46.	 Taipale M, Jarosz DF, Lindquist S (2010) HSP90 at the hub of
protein homeostasis: emer ging mechanistic insights. Nat Re v
Mol Cell Biol 11(7):515–528
	47.	 Sharma K et   al (2012) Quantitati ve proteomics re veals that
Hsp90 inhibition preferentially tar gets kinases and the DN A
damage response. Mol Cell Proteomics 11(3):M111.014654.
doi:10.1074/mcp.M111.014654
	48.	W u Z, Moghaddas Gholami A, Kuster B (2012) Systematic iden-
tiἀcation of the HSP90 candidate re gulated proteome. Mol Cell
Proteomics 11(6):M111.016675. doi:10.1074/mcp.M111.016675
	49.	 Haupt A et al (2012) Hsp90 inhibition dif ferentially destabilises
MAP kinase and TGF-beta signalling components in cancer cells
revealed by kinase-tar geted chemoproteomics. BMC Cancer
12:38
	50.	 Li J et   al (2010) A chemical and phosphoproteomic charac -
terization of dasatinib action in lung cancer . Nat Chem Biol
6(4):291–299
	51.	 Gorre ME et al (2001) Clinical resistance to STI-571 cancer ther-
apy caused by BCR-ABL gene mutation or ampliἀcation. Sci -
ence 293(5531):876–880
	52.	 Pao W et al (2005) Acquired resistance of lung adenocarcinomas
to geἀtinib or erlotinib is associated with a second mutation in
the E GFR kinase domain. PLoS Med 2(3):e73
	53.	 Beauchamp E M et al (2013) Acquired resistance to dasatinib in
lung cancer cell lines conferred by DDR2 g atekeeper mutation
and NF1 loss. Mol Cancer Ther 13(2):475–482
	54.	 Polier S et   al (2013) ATP-competitive inhibitors block protein
kinase recruitment to the Hsp90-Cdc37 system. Nat Chem Biol
9(5):307–312
	55.	 Siddiqui K et al (2009) Actinomycin D identiἀed as an inhibitor
of discoidin domain receptor 2 interaction with collagen through
an insect cell-based screening of a drug compound library . Biol
Pharm Bull 32(1):136–141
	56.	 Hu Y et al (2013) Discoipyrroles A-D: isolation, structure deter -
mination, and synthesis of potent migration inhibitors from
Bacillus hunanensis. J Am Chem Soc 135(36):13387–13392
	57.	 Potts MB et   al (2013) Using functional signature ontology
(FUSION) to identify mechanisms of action for natural products.
Sci Signal 6(297):ra90
	58.	 Gao M et   al (2013) Disco very and optimization of
3-(2-(Pyrazolo[1,5-a]pyrimidin-6-yl)ethynyl)benzamides as
novel selective and orally bioavailable discoidin domain receptor
1 (DDR1) inhibitors. J Med Chem 56(8):3281–3295
	59.	 Kim HG et   al (2013) Disco very of a potent and selecti ve
DDR1 receptor tyrosine kinase inhibitor . ACS Chem Biol
8(10):2145–2150
	60.	 Duncan JS et  al (2012) Dynamic reprogramming of the kinome
in response to tar geted ME K inhibition in triple-negative breast
cancer. Cell 149(2):307–321
	61.	 Dang CV (2012) MYC on the path to cancer. Cell 149(1):22–35
	62.	 L’Hote CG, Thomas PH, Ganesan TS (2002) Functional analy -
sis of discoidin domain receptor 1: ef fect of adhesion on DDR1
phosphorylation. FASE B J 16(2):234–236
	63.	 Koo DH et  al (2006) Pinpointing phosphotyrosine-dependent
interactions downstream of the collagen receptor DDR1. F E BS
Lett 580(1):15–22
3279Proteomics of discoidin domain receptors
1 3
	64.	Wang CZ et   al (2006) A discoidin domain receptor 1/SHP-2
signaling complex inhibits alpha2beta1-inte grin-mediated signal
transducers and activators of transcription 1/3 acti vation and cell
migration. Mol Biol Cell 17(6):2839–2852
	65.	 Yang G et al (2009) Proteomic, functional and motif-based analy-
sis of C-terminal Src kinase-interacting proteins. Proteomics
9(21):4944–4961
	66.	 Lin KL et   al (2010) Transcriptional upre gulation of DDR2 by
ATF4 facilitates osteoblastic differentiation through p38 MAPK-
mediated Runx2 activation. J Bone Miner Res 25(11):2489–2503
	67.	 Zhang Y et al (2011) An essential role of discoidin domain recep-
tor 2 (DDR2) in osteoblast dif ferentiation and chondroc yte mat-
uration via modulation of Runx2 acti vation. J Bone Miner Res
26(3):604–617
	68.	 Lemeer S et   al (2012) Phosphotyrosine-mediated protein
interactions of the discoidin domain receptor 1. J Proteomics
75(12):3465–3477
	69.	 Faraci-Orf E , McFadden C, Vogel W F (2006) DDR1 signaling is
essential to sustain Stat5 function during lactogenesis. J Cell Bio-
chem 97(1):109–121
	70.	 Kim HG et  al (2011) DDR1 receptor tyrosine kinase promotes
prosurvival pathw ay through Notch1 acti vation. J Biol Chem
286(20):17672–17681
	71.	 Dejmek J et al (2005) E xpression and signaling activity of W nt-
5a/discoidin domain receptor -1 and Syk plays distinct b ut deci-
sive roles in breast cancer patient survi val. Clin Cancer Res 11(2
Pt 1):520–528
	72.	 Huang Y et al (2009) The collagen receptor DDR1 re gulates cell
spreading and motility by associating with myosin IIA. J Cell Sci
122(Pt 10):1637–1646
	73.	 Shintani Y et   al (2008) Collagen I-mediated up-re gulation of
N-cadherin requires cooperati ve signals from inte grins and dis -
coidin domain receptor 1. J Cell Biol 180(6):1277–1289
	74.	 Hidalgo-Carcedo C et al (2011) Collective cell migration requires
suppression of actomyosin at cell–cell contacts mediated by
DDR1 and the cell polarity re gulators Par3 and P ar6. Nat Cell
Biol 13(1):49–58
	75.	 Hansen C et  al (2006) Phosphorylation of D ARPP-32 regulates
breast cancer cell migration do wnstream of the receptor tyrosine
kinase DDR1. E xp Cell Res 312(20):4011–4018
	76.	 Hilton HN et  al (2008) KIBRA interacts with discoidin domain
receptor 1 to modulate collagen-induced signalling. Biochim Bio-
phys Acta 1783(3):383–393
	77.	 Das S et al (2006) Discoidin domain receptor 1 receptor tyrosine
kinase induces cyclooxygenase-2 and promotes chemoresistance
through nuclear f actor-kappaB pathw ay acti vation. Cancer Res
66(16):8123–8130
	78.	 Iwai LK et al (2013) Phosphoproteomics of collagen receptor net-
works reveals SHP-2 phosphorylation do wnstream of wild-type
DDR2 and its lung cancer mutants. Biochem J 454(3):501–513
	79.	 Furdui CM et al (2006) Autophosphorylation of FGFR1 kinase is
mediated by a sequential and precisely ordered reaction. Mol Cell
21(5):711–717
	80.	 Yang K et  al (2005) Tyrosine 740 phosphorylation of discoidin
domain receptor 2 by Src stimulates intramolecular autophos-
phorylation and Shc signaling comple x formation. J Biol Chem
280(47):39058–39066
	81.	 Ikeda K et al (2002) Discoidin domain receptor 2 interacts with
Src and Shc follo wing its acti vation by type I collagen. J Biol
Chem 277(21):19206–19212
	82.	 Su J et   al (2009) Discoidin domain receptor 2 is associated
with the increased e xpression of matrix metalloproteinase-13 in
synovial ἀbroblasts of rheumatoid arthritis. Mol Cell Biochem
330(1–2):141–152
	83.	 Noordeen NA et   al (2006) A transmembrane leucine zipper is
required for acti vation of the dimeric receptor tyrosine kinase
DDR1. J Biol Chem 281(32):22744–22751
	84.	 Mihai C et   al (2009) Mapping of DDR1 distrib ution and oli -
gomerization on the cell surf ace by FR E T microscop y. J Mol
Biol 385(2):432–445
	85.	 Fu, HL et al (2014) Glycosylation at ASN211 regulates the acti-
vation state of the discoidin domain receptor 1 (DDR1). J Biol
Chem
	86.	 Lew E D et   al (2009) The precise sequence of FGF receptor
autophosphorylation is kinetically dri ven and is disrupted by
oncogenic mutations. Sci Signal 2(58):ra6
	87.	 Payne LS, Huang PH (2013) The pathobiology of collagens in
glioma. Mol Cancer Res 11(10):1129–1140
	88.	 Fu HL et   al (2013) Shedding of discoidin domain receptor 1
by membrane-type matrix metalloproteinases. J Biol Chem
288(17):12114–12129
	89.	 Carafoli F et   al (2009) Crystallographic insight into col -
lagen recognition by discoidin domain receptor 2.
Structure 17(12):1573–1581
	90.	 Xu H et   al (2012) Discoidin domain receptors promote alpha -
1beta1- and alpha2beta1-inte grin-mediated cell adhesion to col -
lagen by enhancing integrin activation. PLoS One 7(12):e52209
	91.	 Zheng Y et al (2013) Temporal regulation of E GF signalling net-
works by the scaffold protein Shc1. Nature 499(7457):166–171

More Related Content

What's hot

Personalizing Oncology with Genomics
Personalizing Oncology with GenomicsPersonalizing Oncology with Genomics
Personalizing Oncology with GenomicsJeff Fitzgerald
 
Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Gul Muneer
 
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...asclepiuspdfs
 
Next-Generation Sequencing Clinical Research Milestones Infographic
Next-Generation Sequencing Clinical Research Milestones InfographicNext-Generation Sequencing Clinical Research Milestones Infographic
Next-Generation Sequencing Clinical Research Milestones InfographicQIAGEN
 
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...Gul Muneer
 
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapHadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapLoubna Abdel Hadi
 
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...Ken Rogan
 
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...CrimsonpublishersCancer
 
Heinrich_et_al-2003-International_Journal_of_Cancer
Heinrich_et_al-2003-International_Journal_of_CancerHeinrich_et_al-2003-International_Journal_of_Cancer
Heinrich_et_al-2003-International_Journal_of_CancerBianca Heinrich
 
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...Nicholas Vaughn
 
Next Generation Sequencing
Next Generation SequencingNext Generation Sequencing
Next Generation SequencingJoshuaLee309
 
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2Victor Lobanenkov
 
Nanodroplet processing platform for deep and quantitative proteome profiling ...
Nanodroplet processing platform for deep and quantitative proteome profiling ...Nanodroplet processing platform for deep and quantitative proteome profiling ...
Nanodroplet processing platform for deep and quantitative proteome profiling ...Gul Muneer
 
Lecaut et al 2012
Lecaut et al 2012Lecaut et al 2012
Lecaut et al 2012Fran Flores
 
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...Gul Muneer
 

What's hot (20)

Personalizing Oncology with Genomics
Personalizing Oncology with GenomicsPersonalizing Oncology with Genomics
Personalizing Oncology with Genomics
 
Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...Assessing the clinical utility of cancer genomic and proteomic data across tu...
Assessing the clinical utility of cancer genomic and proteomic data across tu...
 
sequencing-methods-review
sequencing-methods-reviewsequencing-methods-review
sequencing-methods-review
 
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...
Pluripotent Stem Cell Markers and microRNA Expression May Correlate with Dent...
 
Next-Generation Sequencing Clinical Research Milestones Infographic
Next-Generation Sequencing Clinical Research Milestones InfographicNext-Generation Sequencing Clinical Research Milestones Infographic
Next-Generation Sequencing Clinical Research Milestones Infographic
 
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...
Proteogenomic analysis of human colon cancer reveals new therapeutic opportun...
 
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChapHadiDiVitoMarfiaNavoneCampanellaRiboniChap
HadiDiVitoMarfiaNavoneCampanellaRiboniChap
 
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...
Bioengineered 3D Co culture Lung In Vitro Models: Platforms to Integrate Cell...
 
paper
paperpaper
paper
 
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...
Hitting the Bullseye: Are Cell Penetrating Peptides (CPP) the Future of Targe...
 
Heinrich_et_al-2003-International_Journal_of_Cancer
Heinrich_et_al-2003-International_Journal_of_CancerHeinrich_et_al-2003-International_Journal_of_Cancer
Heinrich_et_al-2003-International_Journal_of_Cancer
 
Vu-2015_tissue-plasticity-PNET
Vu-2015_tissue-plasticity-PNETVu-2015_tissue-plasticity-PNET
Vu-2015_tissue-plasticity-PNET
 
Metanalisys
MetanalisysMetanalisys
Metanalisys
 
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...
Tryptophan Scanning Reveals Dense Packing of Connexin Transmembrane Domains i...
 
Next Generation Sequencing
Next Generation SequencingNext Generation Sequencing
Next Generation Sequencing
 
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2
Pugacheva et al. COMPLETE GB_16.1_p.161_publ.online_08_14_2015 2
 
Nanodroplet processing platform for deep and quantitative proteome profiling ...
Nanodroplet processing platform for deep and quantitative proteome profiling ...Nanodroplet processing platform for deep and quantitative proteome profiling ...
Nanodroplet processing platform for deep and quantitative proteome profiling ...
 
Lecaut et al 2012
Lecaut et al 2012Lecaut et al 2012
Lecaut et al 2012
 
nanomedicine and nanotechnology
nanomedicine and nanotechnologynanomedicine and nanotechnology
nanomedicine and nanotechnology
 
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...
Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neu...
 

Similar to Discoidin receptors a proteomic portret

Biomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyBiomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyTrustlife
 
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...semualkaira
 
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...semualkaira
 
Methods, Challenges and Future Directions of Radiogenomics-Crimson Publishers
Methods, Challenges and Future Directions of Radiogenomics-Crimson PublishersMethods, Challenges and Future Directions of Radiogenomics-Crimson Publishers
Methods, Challenges and Future Directions of Radiogenomics-Crimson PublishersCrimsonpublishersCancer
 
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...Lichchavi Dhananjaya Rajasinghe
 
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...CrimsonpublishersCancer
 
Identification of the Enrichment Pathways of Catalase in Multiple Primary Tumors
Identification of the Enrichment Pathways of Catalase in Multiple Primary TumorsIdentification of the Enrichment Pathways of Catalase in Multiple Primary Tumors
Identification of the Enrichment Pathways of Catalase in Multiple Primary Tumorsdaranisaha
 
全人關懷獎的簡報
全人關懷獎的簡報全人關懷獎的簡報
全人關懷獎的簡報bgbgbg
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...semualkaira
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...semualkaira
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...semualkaira
 
Seminario biologia molecular- Maria Upegui
Seminario biologia molecular- Maria UpeguiSeminario biologia molecular- Maria Upegui
Seminario biologia molecular- Maria Upeguiisabellabuitrago4
 
Protocol nlr and plr version 1.0
Protocol nlr and plr version 1.0Protocol nlr and plr version 1.0
Protocol nlr and plr version 1.0gwendolynliow
 
Seminario biologia molecular
Seminario biologia molecular Seminario biologia molecular
Seminario biologia molecular elianatorres34
 
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...dr.Ihsan alsaimary
 
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...JohnJulie1
 
Recent developments in p53 and nano oncology
Recent developments in p53 and nano oncologyRecent developments in p53 and nano oncology
Recent developments in p53 and nano oncologytazib rahaman
 
Notch signalling
Notch signallingNotch signalling
Notch signallingSAIMA BARKI
 

Similar to Discoidin receptors a proteomic portret (20)

Biomedicine & Pharmacotherapy
Biomedicine & PharmacotherapyBiomedicine & Pharmacotherapy
Biomedicine & Pharmacotherapy
 
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
 
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
PTPRC as a Predictive Marker Related to PD-L1 for Prognosis and Immunotherapy...
 
Methods, Challenges and Future Directions of Radiogenomics-Crimson Publishers
Methods, Challenges and Future Directions of Radiogenomics-Crimson PublishersMethods, Challenges and Future Directions of Radiogenomics-Crimson Publishers
Methods, Challenges and Future Directions of Radiogenomics-Crimson Publishers
 
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...
(VITAMIN e)Tocotrienol-rich mixture inhibits cell proliferation and induces a...
 
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...
The Immunosuppressive Significance of Lactate Dehydrogenase (LDH) Blood Level...
 
Identification of the Enrichment Pathways of Catalase in Multiple Primary Tumors
Identification of the Enrichment Pathways of Catalase in Multiple Primary TumorsIdentification of the Enrichment Pathways of Catalase in Multiple Primary Tumors
Identification of the Enrichment Pathways of Catalase in Multiple Primary Tumors
 
全人關懷獎的簡報
全人關懷獎的簡報全人關懷獎的簡報
全人關懷獎的簡報
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
 
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
IGF2BP3 Is Associated with HPV Status and Tight Junction in HPV-Related Cervi...
 
Seminario biologia molecular- Maria Upegui
Seminario biologia molecular- Maria UpeguiSeminario biologia molecular- Maria Upegui
Seminario biologia molecular- Maria Upegui
 
Nrgastro.2012.208
Nrgastro.2012.208Nrgastro.2012.208
Nrgastro.2012.208
 
Protocol nlr and plr version 1.0
Protocol nlr and plr version 1.0Protocol nlr and plr version 1.0
Protocol nlr and plr version 1.0
 
Seminario biologia molecular
Seminario biologia molecular Seminario biologia molecular
Seminario biologia molecular
 
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...
Assessment of immunomolecular_expression_and_prognostic_role_of_tlr7_among_pa...
 
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
Increased Expression of GNL3L is Associated with Aggressive Phenotypes in Col...
 
Recent developments in p53 and nano oncology
Recent developments in p53 and nano oncologyRecent developments in p53 and nano oncology
Recent developments in p53 and nano oncology
 
Notch signalling
Notch signallingNotch signalling
Notch signalling
 
Genomics
GenomicsGenomics
Genomics
 

Discoidin receptors a proteomic portret

  • 1. 1 3 DOI 10.1007/s00018-014-1616-1 Cellular and Molecular Life Sciences Cell. Mol. Life Sci. (2014) 71:3269–3279 Rev iew Discoidin domain receptors: a proteomic portrait Leo K. Iwai · Maciej T. Luczynski · Paul H. Huang  Received: 29 January 2014 / Revised: 18 March 2014 / Accepted: 20 March 2014 / Published online: 5 April 2014 © Springer Basel 2014 activated by collagen [ 1]. These receptors regulate a num - ber of fundamental biological processes including cell adhesion, migration, proliferation, and extracellular remod- eling [ 1–3]. Dysre gulation of DDR function has been implicated in se veral diseases such as ἀbrosis, atheroscle - rosis, arthritis, and cancer [1, 4]. There are two members in this RTK class with DDR1 primarily expressed in epithelial cells and DDR2 in mesench ymal cells. Correspondingly, induction of epithelial–mesenchymal transition results in a decrease in DDR1 e xpression and a reciprocal increase in DDR2 levels [5, 6]. Their domain organization is composed of an e xtracellular re gion containing tw o N-terminal dis - coidin domains, an e xtracellular juxtamembrane domain, a transmembrane domain, an intracellular juxtamembrane domain, a kinase domain follo wed by a short C-terminal tail. In addition, while DDR1 binds to both ἀbrillar and basement membrane collagen, DDR2 preferentially binds to ἀbrillar collagens and collagen type X [7–9]. Unlike classical growth factor RTKs like the epider- mal growth f actor receptor (E GFR) and platelet-deri ved growth f actor receptor (PDGFR), DDR eng agement by collagen leads to a slo w and sustained acti vation proἀle in which tyrosine phosphorylation levels start to increase only hours after lig and stimulation and remains persistent o ver several days [ 10, 11]. DDR1 and DDR2 contain 15 and 14 c ytoplasmic tyrosine residues, respecti vely, which are potential sites for protein phosphorylation and binding to downstream signaling adaptors and effectors. W hile a small number of targeted studies have attempted to identify these effectors, data from these studies have failed to comprehen- sively deἀne the components of DDR signaling networks. Advances have largely been hampered by challenges asso- ciated with DDR biology including the unique receptor activation kinetics and the inherent multi-lig and nature of collagen. In addition, while there are an ab undance of Abstract  The discoidin domain receptors (DDRs) are collagen-binding receptor tyrosine kinases that ha ve been implicated in a number of fundamental biological processes ranging from growth and development to immunore gula- tion. In this review, we examine how recent proteomic tech- nologies ha ve enriched our understanding of DDR sign - aling mechanisms. We provide an o verview on the use of large-scale proteomic proἀling and chemical proteomics to reveal novel insights into DDR therapeutics, signaling net - works, and receptor crosstalk. A perspective of ho w prot- eomics may be harnessed to answer outstanding fundamen- tal questions including the dynamic re gulation of receptor activation kinetics is presented. Collecti vely, these studies present an emer ging molecular portrait of these unique receptors and their functional role in health and disease. Keywords  Discoidin domain receptors · Collagen · Cell signaling · Proteomics · Mass spectrometry Introduction The discoidin domain receptors (DDRs) are a class of receptor tyrosine kinases (R TKs) that bind to and are L. K. Iwai and M.T. Luczynski contributed equally to this work. L. K. Iwai · M. T. Luczynski · P. H. Huang ()  Protein Networks Team, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW 3 6JB, UK e-mail: paul.huang@icr.ac.uk Present Address: L. K. Iwai  Laboratório E special de Toxinologia Aplicada/CeTICS, Instituto Butantan, Av V ital Brasil 1500, São Paulo 05503‑000, Brazil
  • 2. 3270 L. K. Iwai et al. 1 3 studies that pro vide in vitro e vidence of the signaling and functional properties of the DDRs in cell culture, there is less data supporting the role of the DDRs as acti ve signal- ing molecules in tissues and organs. In the past 5 years, proteomics has undoubtedly emerged as a po werful tool to further our understanding of DDR biology. In this perspective, we will provide an overview of how this technology has produced a molecular portrait of DDR signaling mechanisms in human health and dis - ease (outlined in Fig. 1). We further elaborate on the use of proteomics to enrich our understanding of DDR struc - ture–activity relationships as well as the reprogramming of signaling in response to therapeutic intervention. Finally, we offer a prospective view of the potential of proteomics to pro vide future insights into une xplored areas of DDR biology. Proteomic proἀling of DDR activity in cancer Phosphorylation is a re versible and highly dynamic signal transduction mechanism that plays an important role in the regulation of a myriad of cellular processes. The impor - tance of this post-translational modiἀcation is e xempliἀed by aberrant global phosphorylation e vents that ha ve been reported in multiple pathologies such as cancer , and meta- bolic and autoimmune diseases. In particular, dysregulation of protein phosphorylation has been particularly well stud - ied in cancer where multiple oncogenes and tumor suppres- sors lie in protein phosphorylation pathw ays that directly inḀuence cancer progression [12]. It is thus not surprising that drugs tar geting protein kinases represent the lar gest class of compounds currently a vailable to treat this disease [13]. Le veraging on the dependenc y of cancer cells for oncogenic signaling pathw ays, these kinase-speciἀc tar - geted therapies ha ve led to dramatic clinical responses in subsets of cancer patients [ 14–16]. Furthermore, monitor - ing components in kinase signaling pathw ays are increas - ingly being used as biomark ers for stratiἀcation of patient response to therapies as well as recruitment criteria for clin- ical trials. These developments in clinical practice to wards personalized medicine ha ve made it necessary not only to better understand the dynamic nature of kinase signaling networks but also to de velop improved methods for moni - toring phosphorylation levels in clinical specimens. Ampliἀcation and mutations in the DDRs ha ve been reported in multiple cancer types, including breast, o var- ian, brain, and lung tumors [17–21] (a complete review of the role of DDRs in cancer can be found in [3]). Previous studies in E GFR-positive cancer patients suggest that total Fig. 1  A timeline of proteomic studies that have informed our current understanding of discoidin domain receptor biology
  • 3. 3271Proteomics of discoidin domain receptors 1 3 E GFR protein levels is a poor predictor of patient response to E GFR kinase inhibitors. In contrast, a high phospho- E GFR level, the active form of the receptor, is a good prog- nostic indicator of sensiti vity to E GFR-targeted therap y [22, 23]. Along these lines, it w ould be vital to determine if DDRs are in f act active in tumors where protein o verex- pression/mutation is observ ed and if so, do the y represent good biomarkers for patient response to kinase inhibitors. Despite the functional importance of the DDR acti vity in vitro, for about a decade follo wing the disco very of the DDRs, there were no studies that conclusi vely demon - strated that these receptors were indeed phosphorylated or active in vivo. This paucity of e vidence was largely due to the lack of phospho-speciἀc reagents (e.g., antibodies) tar - geting this class of receptors. The introduction of unbiased phosphoproteomic proἀl - ing by mass spectrometry (MS) rapidly o vercame these technical challenges. Rik ova et   al. [ 24] pro vided the ἀrst study to demonstrate the scale of DDR phosphorylation in vivo. The authors performed a lar ge-scale screen of tyros - ine phosphorylation levels in 150 non-small cell lung carci - noma (NSCLC) tumors and 41 cell lines. They were able to identify more than 2,700 tyrosine-phosphorylated proteins, which included over 50 different tyrosine kinases. This study established that signiἀcant le vels of phosphorylated DDR1 and 2 were present in lung cancer . A comparison of phos - phorylated tyrosine kinase proἀles in tumors versus cell lines raised an interesting observ ation. W hile the analysis of cell lines revealed the activation of common NSCLC targets such as E GFR, c-Met, and E phA2 [ 25–27], the patient-deri ved tumor proἀles sho wed that the DDRs w as by f ar the most commonly activated tyrosine kinase class in vivo, accounting for more than a third of the tumors analyzed. Importantly , very few DDR1 and no DDR2 phosphorylation e vents were found in the cell lines. This data highlights the limited utility of NSCLC cell lines in recapitulating in vivo tyrosine kinase signaling events observed in patients. Furthermore, the pres - ence of DDR acti vation only in clinical specimens suggests that the lung tumor microen vironment, which contains high levels of collagen [28], is critical for modulating signaling networks in vivo. The same group performed a subsequent screen of cholangiocarcinoma tumors from 23 patients [ 29]. The unique aspect of this study was the comparison of tumor specimens with para-tumor (normal) tissues. Consistent with the NSCLC study, high levels of activated DDRs was found in more than 70  % of the patients studied, conἀrming that these receptors are prevalent in multiple tumor types. More recently, a phosphoproteomic analysis of ten cell lines and three human tumors representing different forms of the sarcoma identiἀed tyrosine phosphorylation of DDR2 in 9/10 cell lines and 1/3 tumors [ 30]. These ἀnd- ings are congruent with the restricted e xpression of DDR2 in cells of mesench ymal origin. Furthermore, sarcoma cell lines are kno wn to secrete a lar ge amount of collagen, which represents a source of autocrine lig and for the acti - vation of DDR2 [ 10, 31, 32]. It should be noted that all of the described studies above also identiἀed a large network of other RTKs that are simultaneously co-acti vated in can- cer cell lines and tumors that poses several important ques- tions [33, 34]. Firstly, why do cells simultaneously activate multiple tyrosine kinases in the context of cancer but not in normal tissues? The second question is ho w does one de- convolute these complex RTK networks and resolve which downstream signals are speciἀc to particular R TKs such as the DDRs? Finally, is there bi-directional crosstalk between the DDRs and other tyrosine kinases embedded within these RTK co-activation networks? A recent proteomic proἀling in glioblastoma sheds some light on the latter question. In this study , eight patient- derived glioblastoma tumor x enografts e xpressing wild- type or mutant forms of E GFR were subjected to both quantitative proteomic and phosphoproteomic analysis [35]. One of the intriguing ἀndings of this study w as that well-established interactors of E GFR such as SHC, STAT3, Cbl, Gab1, and PLC γ1 did not cluster with E GFR phos - phorylation. In contrast, phosphorylation sites on the acti - vation loop of DDR1 strongly correlated with the majority of E GFR tyrosine phosphosites across all the tumors stud - ied. This clustering analysis suggests that in GBM tumors, crosstalk between E GFR and DDR1 is lik ely to be a sig - niἀcant event, and that the resultant do wnstream pathways from this interaction may be distinct from E GFR speciἀc signaling networks. Taking an in vitro approach, our group explored the potential for crosstalk between the insulin receptor (IR) and DDR2. H E K293 cells e xpressing both DDR2 and IR w as subjected to stimulation with insulin and/or collagen [36]. Using iTRAQ labeling methodol- ogy, we sho wed that insulin enhanced collagen-mediated DDR2 activation. We also identiἀed SHIP-2 and SGK269 as potential do wnstream effectors that arise from the inte - grated activation of DDR2 and IR. The exact mechanism by which IR and E GFR interact with the DDRs is still uncertain. Ho wever, the observ ed crosstalk between the DDRs and other R TKs may represent a means for signal diversiἀcation of collagen receptor signaling to achie ve robustness despite e xposure to e xogenous insults, includ - ing tar geted therap y [ 37]. Further in vestigation into the extent and biological consequence of DDR-R TK crosstalk is needed to resolve these questions. The identiἀcation of acti ve DDRs in multiple cancers conἀrms that DDR acti vation occurs in vi vo and may play an important role in the tumor initiation and/or progres - sion. These developments have led the w ay for the search of small molecule inhibitors that would target the DDRs and disrupt its acti vity and resultant signaling pathw ays in cancer.
  • 4. 3272 L. K. Iwai et al. 1 3 Chemical proteomics and the DDRs In order to better characterize the mode of action of kinase inhibitors, MS-based chemical proteomics has successfully been used to identify the tar get(s) of these small molecules and their effects on tumor signaling netw orks. In a typical chemical proteomics w orkḀow, an immobilized chemi - cal probe or drug of interest is e xposed to a protein e xtract and target proteins that bind to these probes are then iden - tiἀed and quantiἀed by disco very-based MS proteomics (Fig. 2a). This approach has been ef fective in identifying the target proἀle of the multi-kinase inhibitors de veloped for treatment of chronic myelogenous leuk emia (CML). Kinase inhibitors such as dasatinib, nilotinib, and imatinib, which tar get the constituti vely acti ve BCR-ABL fusion oncoprotein, have been approved for the treatment of CML since 2003. Imatinib is the ἀrst-line treatment for CML positive for the Philadelphia chromosome, while dasatinib and nilotinib are currently being used for imatinib-intol- erant and -resistant CML patients, although the y still f ail in patients harboring the g atekeeper mutation at T315I in BCR-ABL [38]. Despite being rationally designed to target BCR-ABL, several chemical proteomic studies ha ve since sho wn that these drugs also inhibit other kinases including c-Src, c-KIT, PDGFR, and the DDRs [ 39–41]. In f act, Imatinib is no w appro ved for adv anced g astrointestinal stromal tumor (GIST) patients with c-KIT e xpression as well as other malignancies with PDGFR translocations [42–44]. Through an afἀnity-based chemical proteomic approach using kinobeads (an immobilized non-selecti ve kinase inhibitor matrix) in K562 CML cells, Bantscheff et al. [39] identiἀed o ver 150 protein kinases that were capable of binding to imatinib and dasatinib with varying afἀnities. In addition to conἀrming the mode of action of these drugs on the ABL and SRC f amily kinases, the study also deter - mined that both DDR1 and 2 bound to these compounds with nanomolar afἀnity . The authors v alidated these ἀnd - ing using in vitro kinase assays, which demonstrated that DDR1 is a bona ἀde tar get of imatinib . A further study by Rix et  al. [41] on all three compounds immobilized to a Sepharose matrix and incubated with K562 and CML primary cell lysates sho wed that nilotinib and dasatinib also bound strongly to DDR1. These studies corroborate sequence alignment and molecular modeling experiments which show striking similarities in the ATP-binding pocket of the DDRs and other identiἀed tar get kinases such as ABL and c-KIT [ 45]. Taken together, these chemical pro - teomic studies demonstrate that these three compounds are candidate drugs for malignancies dri ven by oncogenic DDRs. Consistent with this idea, lung cancer cell lines with oncogenic DDR2 mutations are susceptible to dasatinib treatment [19]. Chemical proteomic analysis of cancer therapeutics is not limited to kinase inhibitors. HSP90 is a chaper - one protein that assists in the folding and stabilization of multiple client proteins, including well-established onco - proteins such as B-RAF , E RBB2, CDK4, and BCR-ABL [46]. The essential function of HSP90 in maintaining oncoprotein expression provides a therapeutic rationale for the use of HSP90 inhibitors in cancer therap y. Using stable isotope labeling with amino acids in cell culture (SILAC) methodology , Sharma et   al. [ 47] treated HeLa cervical cancer cells with the HSP90 inhibitor , 17-DMAG [17-(dimethylaminoethylamino)-17-demethoxygeldan- amycin] and monitored changes in 6,000 proteins and 4,000 phosphopeptides. This study sho wed that inhibition of HSP90 results in pleiotropic ef fects on multiple cellu - lar processes including DNA metabolism, protein synthesis and degradation, cell cycle, and apoptosis. In another study performed by W u et al. [48], an inte - grated proteomics strate gy comprising multiple dif fer- ent techniques was used to study the ef fects of the HSP90 inhibitor geldanamycin on four dif ferent cancer cell lines. These techniques comprise SILAC quantiἀcation and chemical precipitation of kinases using kinobeads, HSP90 immunoprecipitation, and immobilized geldanamycin on Sepharose beads. They identiἀed o ver 1,600 proteins that were signiἀcantly altered upon drug treatment. Of these, 98 were protein kinases that were do wn-regulated and classiἀed as HSP90 clients. DDR1 was among this list of kinases and a protein turno ver-rate analysis using pulsed SILAC e xperiments at 6, 12, and 24   h combined with kinobeads enrichment sho wed that, upon drug treatment, DDR1 underwent a more rapid de gradation compared to E GFR, E RK1, or E PHA2. This observ ation pro vides important pharmacokinetic information for drug response where targeting HSP90 w ould lead to a more rapid ef fect on DDR1 compared to other kinases and client proteins. Fig. 2  Proteomic strate gies used to elucidate discoidin domain receptor signaling netw orks and chemical interactions. a Schematic of general chemical proteomic approach for identiἀcation of drug/ probe-binding target proteins. Cell or tissue lysate is incubated with immobilized probes (e.g., kinobeads) or drug to isolate interacting proteins. Subsequent elution and mass spectrometry analysis result in the identiἀcation of probe/drug tar gets. b Different methodologies used for the identiἀcation of DDR signaling. (i) Proἀling of activated/ phosphorylated kinases in cancer cell lines and tumors using global phosphoproteomic approaches. ( ii) Catalog ef fects of drug treatment (kinase or HSP90 inhibitors) in cancer cell lines on cellular pro - teome and phosphoproteome can inform potential no vel combination regimens for disease. ( iii) Treatment of DDR-e xpressing cells with phosphatase inhibitor results in global upre gulation of cellular phos - phorylation. Subsequent DDR immunoprecipitation and mass spec- trometry analysis identiἀes phosphorylation-mediated protein inter - actions. (iv) Stimulation of DDRs with exogenous collagen leads to the activation of receptor -speciἀc signaling netw orks, which can be proἀled with global phosphoproteomics ▸
  • 5. 3273Proteomics of discoidin domain receptors 1 3 A B
  • 6. 3274 L. K. Iwai et al. 1 3 Another study employing a similar approach also identiἀed DDR2 as client protein of HSP90 [ 49]. Collectively, these data demonstrate that the DDRs are major client proteins of HSP90, and HSP90 inhibitors may ha ve utility ag ainst tumors that are DDR positive. These studies also raise an interesting question as to whether a combination approach of using both kinase and HSP90 inhibitors would be more effective in eliminating DDR oncogenic signaling compared to either drug alone (Fig. 2b). This combination would be particularly important since gatekeeper mutants of DDRs are resistant to dasat - inib inhibition [ 19, 30, 45, 50]. W hile such DDR muta - tions have yet to be reported in patients, based on reports in other oncogenic kinases [ 51, 52], it is lik ely that g ate- keeper mutations will arise with prolonged use of imatinib or dasatinib. Indeed, in vitro modeling of dasatinib resist - ance in lung cancer cell lines results in the de velopment of the T654I gatekeeper mutation in DDR2 as a mechanism of acquired resistance [53]. In a recent study of HSP90 and its recruitment to protein kinases via CDC37, Polier et  al. [54] analyzed the mode of action of dif ferent ATP-compet- itive kinase inhibitors on HSP90-CDC37-kinase chaper - one complex formation. Their study revealed a dual mode of action for this class of inhibitors. These drugs not only acted as con ventional ATP-competitive kinase inhibitors by competing for ATP binding b ut also serv ed as antago - nists depriving client kinase recruitment to the HSP90 and CDC37 chaperone system f acilitating kinase de gradation by proteasomes. These data suggest that a combination with HSP90 inhibition may boost the client protein degra- dation capability of kinase inhibitors, which will enhance therapeutic efἀcac y and reduce the lik elihood of de vel- oping drug-resistant gatekeeper mutations. Furthermore, multi-target kinase inhibitors, such as dasatinib described above, inhibit the DDRs as an unanticipated “off-target” effect. More recently, DDR selective inhibitors, which are capable of inhibiting cancer cell proliferation and migra - tion, have been identiἀed by several groups [55–59]. These compounds have the potential to progress to become potent anti-cancer agents in DDR-driven cancers. Cellular signaling is a dynamic process that constantly adapts to both endogenous and e xogenous stimuli. Another interesting facet of DDR biology is the ability of tumor cells to modulate receptor expression levels in response to thera- peutic challenge. This “kinase reprogramming” phenom - enon was studied in depth by Duncan et  al. using a chemi - cal proteomics approach. E mploying multiple xed kinase inhibitor beads and mass spectrometry (MIB/MS) to enrich for kinases in triple-ne gative breast cancer cells, treatment with ME K inhibitors AZD6244 and U0126 resulted in sig - niἀcant reprogramming of the kinome induced by the pro - teasomal c-Myc de gradation [ 60]. Initial treatment with the drugs inhibited cell gro wth and elevated RNA levels of RTKs including DDR1 and DDR2. Continuous drug e xpo- sure o ver 30   days generated therap y-resistant cells with increased protein levels of ME K2, VE GFR2, and PDGFRβ and elevated tyrosine phosphorylation of ME K, AXL, RAF, and AKT. The individual depletion of some of the repro - grammed R TKs such as DDR1, DDR2, AXL, VE GFR2, and PDGFR β using siRN A in combination with U0126 were able restore gro wth arrest. This data indicates that a single knockdown of DDR1 or DDR2 in the presence of a ME K inhibitor is able to kill triple-negative breast cancer cells. Interestingly , combination therap y using AZD6244 with a low dose of RTK inhibitors (sorafenib, a PDGFRα/β, VE GFR, DDR1/2, and RAF inhibitor; and foretinib, a cMet and VE GFR inhibitor), which were inef fective as single agents, inhibited phosphorylation of multiple R TKs and efἀciently arrested growth of SUM159 triple-negative breast cancer cells. Importantly , in vi vo experiments using breast cancer C3Tag mouse model treated orally with AZD6244 also showed kinome reprogramming with increased expres- sion of M E K2, E RK1, PDGFRβ, and DDR1 recapitulating the reprogramming response observ ed in vitro. Treatment of C3Tag mice with AZD6244 and Sorafenib led to tumor growth arrest, increased apoptosis, and tumor regression in the majority of mice. These ἀndings show that a deἀned combination therapy based on the a priori knowledge of the kinome pattern of resistance is ef fective in producing sig - niἀcant therapeutic beneἀt. Although the role of DDR in cancer is still poorly characterized, MS-based chemical proteomics has been exploited to monitor changes in the composition of the cancer kinome, including DDR1 and DDR2, upon M E K inhibitor treatment. Cancer cells circumv ent the inhibition of oncogenic signaling pathways by upregulating the DDRs and additional survival routes, highlighting the importance of this class of receptors in conferring resistance to kinase inhibitor therapy. On a more global scale, multiple cancer types are driven by Myc activation [61] and this study sug- gests that upre gulation of DDRs in the conte xt of a R TK co-activation netw ork may be a general mechanism by which cancer cells evade elimination of the Myc pathway. DDR signaling networks Perhaps the most substantial contrib ution of proteomics to DDR biology is the lar ge-scale elucidation of its signaling Fig. 3  Reported intracellular protein–protein interactions and do wn- stream signaling components of a DDR1 and b DDR2. The ἀgure depicts known tyrosine phosphorylation sites ( black squares) in the cytoplasmic domains of both receptors and the do wnstream effector proteins (white squares) that have been shown to either interact with or be phosphorylated by the DDRs. Associated references for the reported interactions are indicated in parentheses ▸
  • 7. 3275Proteomics of discoidin domain receptors 1 3 A B
  • 8. 3276 L. K. Iwai et al. 1 3 networks. There is a wealth of kno wledge of the collagen- binding properties of the DDRs [ 2]; in contrast, the sign - aling pathw ays acti vated by these receptors are lar gely unknown. A decade of biochemical studies on the DDRs has identiἀed a handful of signaling ef fectors including SHC, CSK, PI3K, SHP2, R UNX2, and NCK1/2 [ 11, 62– 67]. E merging data from proteomics studies ha ve revealed complex interactions in DDR do wnstream signaling net - works. F or instance, in order to characterize the global landscape of possible DDR1 interacting proteins, Lemeer et  al. emplo yed a strate gy where DDR1-o verexpressing glioblastoma cells were treated with a tyrosine phosphatase inhibitor pervanadate, and subjected to co-immunoprecip - itation with anti-DDR1 antibodies (Fig.   2b) [ 68]. Utiliz - ing quantitative MS based on SILA C methodology, more than 30 proximal signaling proteins interacting with DDR1 were identiἀed. These proteins include some pre viously described interactors b ut also highlighted ne w interactions such as GRB2, E PHA2, RASGAP, SHIP1, SHIP2, and STATs. Furthermore, in order to identify the speciἀc tyros - ine phosphorylation residues in DDR1 that were responsi - ble for mediating these interactions, the authors performed phosphopeptide pull-down experiments incubating placenta lysates with all 15 DDR1 tyrosine phosphorylated synthetic peptides immobilized on Sepharose beads. They addition- ally validated the identiἀed interactors by performing quan- titative competition-binding experiments with phosphoryl- ated peptides and non-phosphorylated controls using TMT 6-plex chemical labeling follo wed by MS analysis. RAS - GAP was shown to interact with Y484, Y543, and Y586 while STAT1a/b, STAT3, and ST AT5b with the juxtam - embrane Y703 and the acti vation loop Y796 sites. In addi - tion, they found that both activators (PI3K was shown to bind to pY881) and ne gative regulators (SHIP1 and SHIP2 were shown to bind to pY740) of the PI3K pathw ay were arranged in close proximity on DDR1, indicating a poten - tial mechanism for compartmentalized pathw ay regulation. Taken together, this work characterizes the cellular interac- tome of DDR1 and pro vides a roadmap for future mecha - nistic studies on the roles of these effectors on DDR1 sign- aling and biology . The binding partners to the DDR1 and its speciἀc phosphorylation sites are summarized in Fig. 3a [11, 48, 62–65, 68–77]. W hile the interactome study pro vides a catalogue of all possible DDR1 interactions, it may not accurately repre - sent the situation in cells since both temporal and spatial effects are o verlooked in these e xperiments. Furthermore, it is unclear as to which of the tyrosine residues in DDR1 are actually phosphorylated in cells in response to collagen activation since the global tyrosine phosphatase inhibitor pervanadate was used in this study. Using a complementary strategy to identify DDR signaling effectors, our group has performed an iTRAQ-based quantitative phosphoproteomic analyses in DDR2-overexpressing cells to map the tempo - ral activation of signaling networks in response to collagen stimulation over the course of 24  h (Fig. 2b) [78]. We ἀnd that speciἀc phosphorylation sites on the DDR2 recep - tor were dif ferentially re gulated upon collagen eng age- ment in cells. For instance, the activation loop sites Y736 and Y740 as well as the no vel Y684 and Y813 sites pre - sented a delayed acti vation with maximal phosphorylation at 24   h, while Y481 in the juxtamembrane re gion of the receptor displayed a distinct constitutively phosphorylated proἀle. These ἀndings support the idea that the re gulation of RTK signaling is achie ved through site-speciἀc dif fer- ences in both magnitude and kinetics of receptor phospho - rylation [79]. Bioinformatic analysis identiἀed 45 proteins including SHP-2, SHIP-2, PIK3C2A, E RK1, and PLCL2 that clustered strongly with DDR2, implicating these pro - teins as downstream effectors of DDR2 signaling netw ork. Importantly, these phosphorylation e vents were sho wn to be speciἀc to DDR2 activation and independent of the col- lagen-binding inte grins. These DDR2 signaling ef fectors are summarized in Fig. 3b [36, 66, 67, 80–82]. Perspectives An outstanding fundamental question in DDR biology is how collagen binding promotes the delayed and sustained kinetics of DDR acti vation. DDRs e xist as preformed dimers [83–85] that under go a series of autophosphoryla - tion e vents upon lig and eng agement. Understanding the sequential order of receptor phosphorylation may shed light on the molecular basis of the unique kinetic proἀle of the DDRs. W hile proteomic studies have shown that differ- ent sites on the DDRs have distinct activation kinetics [78], the initial phosphorylation and subsequent dephosphoryla - tion dynamics of these receptors are unknown. MS in com- bination with quenching is a useful tool for elucidating the site-speciἀc order of such R TK phosphorylation e vents. E mploying this approach, Furdui et  al. ele gantly sho wed that ἀbroblast growth factor receptor 1 (FGFR1) autophos - phorylation is dictated by a precisely ordered sequence of events [79]. In this method, FGFR1 and ATP are mixed and quenched prior to sequencing by MS, which allo wed the authors to monitor each distinct tyrosine phosphorylation site on FGFR1 re vealing a unique receptor acti vation pro- ἀle. They showed that initially , asymmetric dimer forma - tion of the receptor f acilitates the transphosphorylation of an activation loop tyrosine residue, followed by subsequent phosphorylation of other docking site tyrosines. In the last step, a dif ferent tyrosine residue in the acti vation loop is phosphorylated, increasing the overall kinase activity of the receptor, f acilitating do wnstream signal transduction [ 79, 86]. A similar characterization of temporal DDR receptor
  • 9. 3277Proteomics of discoidin domain receptors 1 3 phosphorylation will identify the key tyrosine residues that are critical for kinase activity and the mechanisms that gov- ern the observed delayed and sustained activation kinetics. The DDRs and their lig ands are subjected to dynamic regulation in vivo [87]. For instance, receptor acti vation is modulated by matrix metalloproteinases either directly by cleavage of its e xtracellular domain or indirectly through degradation of the collagen matrix [ 88]. In addition, ligand competition by matrisomal proteins such as SPARC and crosstalk with other transmembrane receptors including the integrins may also inḀuence DDR acti vation and con - sequent downstream signaling [7, 89, 90]. E lucidating this complex biology will require the acquisition of dynamic signaling maps that detail netw ork changes in response to cellular perturbation. Building on previous DDR interac- tome studies, tar geted proteomic approaches such as mul - tiple reaction monitoring (MRM) offers the powerful capa- bility of reproducible acquisition of dynamic data across multiple conditions, replicates and protein species. A recent example of this technology by Zheng et   al. [ 91] showed that the SHC1 adaptor protein is a highly dynamic regula- tor of E GFR signaling netw orks. In this study , ἀbroblasts were stimulated with E GF and dif ferent phosphorylation sites within SHC1 as well as 41 dif ferent SHC1-protein binding partners were monitored o ver 16 time points by MRM. This high-resolution analysis of the assembly/dis - assembly dynamics of protein-binding partners to SHC1 revealed a stage-speciἀc function of this scaf fold protein. Upon initial activation by E GF, proteins directing cell divi- sion and survi val were enriched, switching to an interme - diate stage involving functions like vesicle trafἀcking, and ἀnally progressing to a ἀnal stage characterized by proteins involved on cytoskeletal reorganization and downregulation of the initial cell di vision signals. Applying this strate gy to the dynamic interactome and signaling netw orks of the DDRs may provide a molecular explanation of the intrigu - ing activation kinetics of the DDRs and the functional rel - evance of its slow and prolonged signaling network proἀle. Conclusions Proteomics has contributed to the unraveling of DDR biol- ogy and their functional impact in health and disease. W hile biochemical, structural, genetic, and physiological studies performed by multiple groups in last decade have undoubt- edly led to a better understanding of their biological prop - erties, there is a still much to learn about these atypical RTKs. In an era of collaborati ve science and inte grative biology, we anticipate that “Omic” technologies (including proteomics) in combination with classical approaches hold the exciting potential to pro vide a comprehensive molecu- lar portrait of these receptors. Acknowledgments  The work in the authors’ laboratory is funded by the Wellcome Trust (W T089028) and the Biotechnology and Bio - logical Sciences Research Council (BB/I014276/1). References 1. Fu HL et   al (2013) Discoidin domain receptors: unique recep - tor tyrosine kinases in collagen-mediated signaling. J Biol Chem 288(11):7430–7437 2. Leitinger B (2011) Transmembrane collagen receptors. Annu Rev Cell Dev Biol 27:265–290 3. Valiathan RR et   al (2012) Discoidin domain receptor tyrosine kinases: ne w players in cancer progression. Cancer Metastasis Rev 31(1–2):295–321 4. Vogel W F, Abdulhussein R, F ord CE (2006) Sensing e xtracellu- lar matrix: an update on discoidin domain receptor function. Cell Signal 18(8):1108–1116 5. Maeyama M et  al (2008) Switching in discoid domain receptor expressions in SLUG-induced epithelial–mesenchymal transition. Cancer 113(10):2823–2831 6. Walsh LA, Na wshad A, Medici D (2011) Discoidin domain receptor 2 is a critical re gulator of epithelial–mesenchymal tran- sition. Matrix Biol 30(4):243–247 7. Carafoli F, Hohenester E (2013) Collagen recognition and trans - membrane signalling by discoidin domain receptors. Biochim Biophys Acta 1834(10):2187–2194 8. Leitinger B, Hohenester E (2007) Mammalian collagen receptors. Matrix Biol 26(3):146–155 9. Leitinger B, Kw an AP (2006) The discoidin domain recep - tor DDR2 is a receptor for type X collagen. Matrix Biol 25(6):355–364 10. Shrivastava A et   al (1997) An orphan receptor tyrosine kinase family whose members serv e as noninte grin collagen receptors. Mol Cell 1(1):25–34 11. Vogel W et  al (1997) The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell 1(1):13–23 12. Blume-Jensen P, Hunter T (2001) Oncogenic kinase signalling. Nature 411(6835):355–365 13. Cohen P, Alessi DR (2013) Kinase drug disco very—what’s next in the ἀeld? ACS Chem Biol 8(1):96–104 14. Lynch TJ et   al (2004) Activating mutations in the epidermal growth factor receptor underlying responsi veness of non-small- cell lung cancer to geἀtinib. N E ngl J Med 350(21):2129–2139 15. Gerber DE , Minna JD (2010) ALK inhibition for non-small cell lung cancer: from disco very to therap y in record time. Cancer Cell 18(6):548–551 16. Druker BJ et al (2001) E fἀcacy and safety of a speciἀc inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N E ngl J Med 344(14):1031–1037 17. Weiner HL et  al (2000) Consistent and selective expression of the discoidin domain receptor -1 tyrosine kinase in human brain tumors. Neurosurgery 47(6):1400–1409 18. Yamanaka R et al (2006) Identiἀcation of e xpressed genes char- acterizing long-term survival in malignant glioma patients. Onco- gene 25(44):5994–6002 19. Hammerman PS et al (2011) Mutations in the DDR2 kinase gene identify a no vel therapeutic target in squamous cell lung cancer . Cancer Discov 1(1):78–89 20. Ren T et   al (2013) Increased e xpression of discoidin domain receptor 2 (DDR2): a novel independent prognostic marker of worse outcome in breast cancer patients. Med Oncol 30(1):397 21. Ford CE et   al (2007) E xpression and mutation analysis of the discoidin domain receptors 1 and 2 in non-small cell lung carci - noma. Br J Cancer 96(5):808–814
  • 10. 3278 L. K. Iwai et al. 1 3 22. Van Schaeybroeck S et al (2005) E pidermal growth factor recep- tor activity determines response of colorectal cancer cells to geἀ - tinib alone and in combination with chemotherap y. Clin Cancer Res 11(20):7480–7489 23. Thomson S et al (2005) E pithelial to mesenchymal transition is a determinant of sensiti vity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibi- tion. Cancer Res 65(20):9455–9462 24. Rikova K et   al (2007) Global surv ey of phosphotyrosine signaling identiἀes oncogenic kinases in lung cancer. Cell 131(6):1190–1203 25. Amann J et al (2005) Aberrant epidermal growth factor receptor signaling and enhanced sensiti vity to E GFR inhibitors in lung cancer. Cancer Res 65(1):226–235 26. Garofalo M et   al (2012) E GFR and M E T receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and geἀtinib resistance in lung cancers. Nat Med 18(1):74–82 27. Ishikawa M et al (2012) Higher expression of E phA2 and ephrin- A1 is related to f avorable clinicopathological features in patho - logical stage I non-small cell lung carcinoma. Lung Cancer 76(3):431–438 28. van K empen LC et   al (2003) The tumor microen vironment: a critical determinant of neoplastic e volution. E ur J Cell Biol 82(11):539–548 29. Gu TL et al (2011) Surv ey of tyrosine kinase signaling re veals ROS kinase fusions in human cholangiocarcinoma. PLoS One 6(1):e15640 30. Bai Y et   al (2012) Phosphoproteomics identiἀes dri ver tyros - ine kinases in sarcoma cell lines and tumors. Cancer Res 72(10):2501–2511 31. Alitalo K et   al (1982) Biosynthesis of type V procollagen by A204, a human rhabdomyosarcoma cell line. J Biol Chem 257(15):9016–9024 32. Kleman JP et  al (1992) The human rhabdomyosarcoma cell line A204 lays do wn a highly insoluble matrix composed mainly of alpha 1 type-XI and alpha 2 type- V collagen chains. E ur J Bio - chem 210(1):329–335 33. Xu AM, Huang PH (2010) Receptor tyrosine kinase coacti vation networks in cancer. Cancer Res 70(10):3857–3860 34. Huang PH (2012) Phosphoproteomic studies of receptor tyrosine kinases: future perspectives. Mol BioSyst 8(4):1100–1107 35. Johnson H et al (2012) Molecular characterization of E GFR and E GFRvIII signaling networks in human glioblastoma tumor x en- ografts. Mol Cell Proteomics 11(12):1724–1740 36. Iwai LK, Chang F , Huang PH (2013) Phosphoproteomic analy - sis identiἀes insulin enhancement of discoidin domain receptor 2 phosphorylation. Cell Adh Migr 7(2):161–164 37. Meyer AS et al (2013) The Receptor AXL diversiἀes E GFR sign- aling and limits the response to E GFR-targeted inhibitors in tri- ple-negative breast cancer cells. Sci Signal 6(287):ra66 38. Vajpai N et  al (2008) Solution conformations and dynamics of ABL kinase-inhibitor comple xes determined by NMR substanti - ate the different binding modes of imatinib/nilotinib and dasat- inib. J Biol Chem 283(26):18292–18302 39. Bantscheff M et   al (2007) Quantitati ve chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat Biotech 25(9):1035–1044 40. Weisberg E et al (2005) Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 7(2):129–141 41. Rix U et al (2007) Chemical proteomic proἀles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 110(12):4055–4063 42. Joensuu H et   al (2001) E ffect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic g astrointestinal stromal tumor. N E ngl J Med 344(14):1052–1056 43. Demetri GD et al (2002) E fἀcacy and safety of imatinib mesylate in adv anced g astrointestinal stromal tumors. N E ngl J Med 347(7):472–480 44. Heinrich MC et al (2003) Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 21(23):4342–4349 45. Day E et   al (2008) Inhibition of collagen-induced discoidin domain receptor 1 and 2 activation by imatinib, nilotinib and dasatinib. E ur J Pharmacol 599(1–3):44–53 46. Taipale M, Jarosz DF, Lindquist S (2010) HSP90 at the hub of protein homeostasis: emer ging mechanistic insights. Nat Re v Mol Cell Biol 11(7):515–528 47. Sharma K et   al (2012) Quantitati ve proteomics re veals that Hsp90 inhibition preferentially tar gets kinases and the DN A damage response. Mol Cell Proteomics 11(3):M111.014654. doi:10.1074/mcp.M111.014654 48. W u Z, Moghaddas Gholami A, Kuster B (2012) Systematic iden- tiἀcation of the HSP90 candidate re gulated proteome. Mol Cell Proteomics 11(6):M111.016675. doi:10.1074/mcp.M111.016675 49. Haupt A et al (2012) Hsp90 inhibition dif ferentially destabilises MAP kinase and TGF-beta signalling components in cancer cells revealed by kinase-tar geted chemoproteomics. BMC Cancer 12:38 50. Li J et   al (2010) A chemical and phosphoproteomic charac - terization of dasatinib action in lung cancer . Nat Chem Biol 6(4):291–299 51. Gorre ME et al (2001) Clinical resistance to STI-571 cancer ther- apy caused by BCR-ABL gene mutation or ampliἀcation. Sci - ence 293(5531):876–880 52. Pao W et al (2005) Acquired resistance of lung adenocarcinomas to geἀtinib or erlotinib is associated with a second mutation in the E GFR kinase domain. PLoS Med 2(3):e73 53. Beauchamp E M et al (2013) Acquired resistance to dasatinib in lung cancer cell lines conferred by DDR2 g atekeeper mutation and NF1 loss. Mol Cancer Ther 13(2):475–482 54. Polier S et   al (2013) ATP-competitive inhibitors block protein kinase recruitment to the Hsp90-Cdc37 system. Nat Chem Biol 9(5):307–312 55. Siddiqui K et al (2009) Actinomycin D identiἀed as an inhibitor of discoidin domain receptor 2 interaction with collagen through an insect cell-based screening of a drug compound library . Biol Pharm Bull 32(1):136–141 56. Hu Y et al (2013) Discoipyrroles A-D: isolation, structure deter - mination, and synthesis of potent migration inhibitors from Bacillus hunanensis. J Am Chem Soc 135(36):13387–13392 57. Potts MB et   al (2013) Using functional signature ontology (FUSION) to identify mechanisms of action for natural products. Sci Signal 6(297):ra90 58. Gao M et   al (2013) Disco very and optimization of 3-(2-(Pyrazolo[1,5-a]pyrimidin-6-yl)ethynyl)benzamides as novel selective and orally bioavailable discoidin domain receptor 1 (DDR1) inhibitors. J Med Chem 56(8):3281–3295 59. Kim HG et   al (2013) Disco very of a potent and selecti ve DDR1 receptor tyrosine kinase inhibitor . ACS Chem Biol 8(10):2145–2150 60. Duncan JS et  al (2012) Dynamic reprogramming of the kinome in response to tar geted ME K inhibition in triple-negative breast cancer. Cell 149(2):307–321 61. Dang CV (2012) MYC on the path to cancer. Cell 149(1):22–35 62. L’Hote CG, Thomas PH, Ganesan TS (2002) Functional analy - sis of discoidin domain receptor 1: ef fect of adhesion on DDR1 phosphorylation. FASE B J 16(2):234–236 63. Koo DH et  al (2006) Pinpointing phosphotyrosine-dependent interactions downstream of the collagen receptor DDR1. F E BS Lett 580(1):15–22
  • 11. 3279Proteomics of discoidin domain receptors 1 3 64. Wang CZ et   al (2006) A discoidin domain receptor 1/SHP-2 signaling complex inhibits alpha2beta1-inte grin-mediated signal transducers and activators of transcription 1/3 acti vation and cell migration. Mol Biol Cell 17(6):2839–2852 65. Yang G et al (2009) Proteomic, functional and motif-based analy- sis of C-terminal Src kinase-interacting proteins. Proteomics 9(21):4944–4961 66. Lin KL et   al (2010) Transcriptional upre gulation of DDR2 by ATF4 facilitates osteoblastic differentiation through p38 MAPK- mediated Runx2 activation. J Bone Miner Res 25(11):2489–2503 67. Zhang Y et al (2011) An essential role of discoidin domain recep- tor 2 (DDR2) in osteoblast dif ferentiation and chondroc yte mat- uration via modulation of Runx2 acti vation. J Bone Miner Res 26(3):604–617 68. Lemeer S et   al (2012) Phosphotyrosine-mediated protein interactions of the discoidin domain receptor 1. J Proteomics 75(12):3465–3477 69. Faraci-Orf E , McFadden C, Vogel W F (2006) DDR1 signaling is essential to sustain Stat5 function during lactogenesis. J Cell Bio- chem 97(1):109–121 70. Kim HG et  al (2011) DDR1 receptor tyrosine kinase promotes prosurvival pathw ay through Notch1 acti vation. J Biol Chem 286(20):17672–17681 71. Dejmek J et al (2005) E xpression and signaling activity of W nt- 5a/discoidin domain receptor -1 and Syk plays distinct b ut deci- sive roles in breast cancer patient survi val. Clin Cancer Res 11(2 Pt 1):520–528 72. Huang Y et al (2009) The collagen receptor DDR1 re gulates cell spreading and motility by associating with myosin IIA. J Cell Sci 122(Pt 10):1637–1646 73. Shintani Y et   al (2008) Collagen I-mediated up-re gulation of N-cadherin requires cooperati ve signals from inte grins and dis - coidin domain receptor 1. J Cell Biol 180(6):1277–1289 74. Hidalgo-Carcedo C et al (2011) Collective cell migration requires suppression of actomyosin at cell–cell contacts mediated by DDR1 and the cell polarity re gulators Par3 and P ar6. Nat Cell Biol 13(1):49–58 75. Hansen C et  al (2006) Phosphorylation of D ARPP-32 regulates breast cancer cell migration do wnstream of the receptor tyrosine kinase DDR1. E xp Cell Res 312(20):4011–4018 76. Hilton HN et  al (2008) KIBRA interacts with discoidin domain receptor 1 to modulate collagen-induced signalling. Biochim Bio- phys Acta 1783(3):383–393 77. Das S et al (2006) Discoidin domain receptor 1 receptor tyrosine kinase induces cyclooxygenase-2 and promotes chemoresistance through nuclear f actor-kappaB pathw ay acti vation. Cancer Res 66(16):8123–8130 78. Iwai LK et al (2013) Phosphoproteomics of collagen receptor net- works reveals SHP-2 phosphorylation do wnstream of wild-type DDR2 and its lung cancer mutants. Biochem J 454(3):501–513 79. Furdui CM et al (2006) Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol Cell 21(5):711–717 80. Yang K et  al (2005) Tyrosine 740 phosphorylation of discoidin domain receptor 2 by Src stimulates intramolecular autophos- phorylation and Shc signaling comple x formation. J Biol Chem 280(47):39058–39066 81. Ikeda K et al (2002) Discoidin domain receptor 2 interacts with Src and Shc follo wing its acti vation by type I collagen. J Biol Chem 277(21):19206–19212 82. Su J et   al (2009) Discoidin domain receptor 2 is associated with the increased e xpression of matrix metalloproteinase-13 in synovial ἀbroblasts of rheumatoid arthritis. Mol Cell Biochem 330(1–2):141–152 83. Noordeen NA et   al (2006) A transmembrane leucine zipper is required for acti vation of the dimeric receptor tyrosine kinase DDR1. J Biol Chem 281(32):22744–22751 84. Mihai C et   al (2009) Mapping of DDR1 distrib ution and oli - gomerization on the cell surf ace by FR E T microscop y. J Mol Biol 385(2):432–445 85. Fu, HL et al (2014) Glycosylation at ASN211 regulates the acti- vation state of the discoidin domain receptor 1 (DDR1). J Biol Chem 86. Lew E D et   al (2009) The precise sequence of FGF receptor autophosphorylation is kinetically dri ven and is disrupted by oncogenic mutations. Sci Signal 2(58):ra6 87. Payne LS, Huang PH (2013) The pathobiology of collagens in glioma. Mol Cancer Res 11(10):1129–1140 88. Fu HL et   al (2013) Shedding of discoidin domain receptor 1 by membrane-type matrix metalloproteinases. J Biol Chem 288(17):12114–12129 89. Carafoli F et   al (2009) Crystallographic insight into col - lagen recognition by discoidin domain receptor 2. Structure 17(12):1573–1581 90. Xu H et   al (2012) Discoidin domain receptors promote alpha - 1beta1- and alpha2beta1-inte grin-mediated cell adhesion to col - lagen by enhancing integrin activation. PLoS One 7(12):e52209 91. Zheng Y et al (2013) Temporal regulation of E GF signalling net- works by the scaffold protein Shc1. Nature 499(7457):166–171