ArticlePDF Available

Ubiquitin-mediated degradation of active Src tyrosine kinase

Authors:

Abstract and Figures

Src family tyrosine kinases are involved in modulating various signal transduction pathways leading to the induction of DNA synthesis and cytoskeletal reorganization in response to cell-cell or cell-matrix adhesion. The critical role of these kinases in regulating cellular signaling pathways requires that their activity be tightly controlled. Src family proteins are regulated through reversible phosphorylation and dephosphorylation events that alter the conformation of the kinase. We have found evidence that Src also is regulated by ubiquitination. Activated forms of Src are less stable than either wild-type or kinase-inactive Src mutants and can be stabilized by proteasome inhibitors. In addition, poly-ubiquitinated forms of active Src have been detected in vivo. Taken together, our results establish ubiquitin-mediated proteolysis as a previously unidentified mechanism for irreversibly attenuating the effects of active Src kinase.
Content may be subject to copyright.
Ubiquitin-mediated degradation of active Src
tyrosine kinase
Kimya F. Harris, Ikuo Shoji, Eric M. Cooper, Sushant Kumar, Hideaki Oda, and Peter M. Howley*
Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115
Contributed by Peter M. Howley, August 5, 1999
Src family tyrosine kinases are involved in modulating various
signal transduction pathways leading to the induction of DNA
synthesis and cytoskeletal reorganization in response to cell-cell or
cell-matrix adhesion. The critical role of these kinases in regulating
cellular signaling pathways requires that their activity be tightly
controlled. Src family proteins are regulated through reversible
phosphorylation and dephosphorylation events that alter the
conformation of the kinase. We have found evidence that Src also
is regulated by ubiquitination. Activated forms of Src are less stable
than either wild-type or kinase-inactive Src mutants and can be
stabilized by proteasome inhibitors. In addition, poly-ubiquiti-
nated forms of active Src have been detected in vivo. Taken
together, our results establish ubiquitin-mediated proteolysis as a
previously unidentified mechanism for irreversibly attenuating the
effects of active Src kinase.
T
he Src family kinases are nonreceptor tyrosine kinases that
mediate a variety of cellular signaling pathways. Nine ver-
tebrate members of the Src kinase family are known: some are
expressed in a cell-type specific manner and others are expressed
more broadly. For example, Blk and Lck expression is limited to
lymphoid lineages. In contrast, Src, Fyn, and Yes are more
broadly expressed although their levels of expression do vary
among cell types. For instance, Src is abundant in brain and
platelets, whereas Fyn is abundant in brain, platelets, and T
lymphocytes (for review see ref. 1).
All Src family members share the same basic structural
features. Each possesses an amino-terminal Src homology (SH)
4 domain that contains a myristylation signal sequence required
for membrane localization. Adjacent to the SH4 domain is a
unique region followed by conserved SH3 and SH2 domains,
which mediate protein–protein interactions. The catalytic kinase
domain resides in the carboxyl-terminal half of the protein.
Finally, there is a short carboxyl-terminal tail domain that
functions to negatively regulate the kinase activity (for reviews
see refs. 1 and 2). An intramolecular interaction between the
SH2 domain and a phosphorylated tyrosine residue (amino acid
527 in chicken c-Src) within this tail domain alters the confor-
mation of Src such that the catalytic domain is confined to an
inactive state (closed conformation; ref. 3). The dephosphory-
lation of tyrosine 527 results in a release of the intramolecular
interaction with the SH2 domain relaxing conformational con-
straints. The molecule takes on an ‘‘open conformation’’ that
facilitates catalytic activation (3–5).
Src family kinases function in numerous signaling pathways
including those mediating DNA synthesis and proliferation.
Activated cell surface receptors interact with and signal through
the Src family kinases. For example, binding of ligand to the
platelet-derived growth factor receptor results in its association
with and activation of the Src family kinases Src, Fyn, and Yes
(6). In turn, the activated Src kinases trigger a cascade of events,
ultimately leading to entry into S phase and subsequent DNA
replication (7–9). Src kinases also have a role in progression
through the G
2
M transition of the cell cycle, suggesting that
they function in both the G
1
and mitotic checkpoints (10–12).
Several Src family members (Src, Fyn, and Yes) are also
important for transducing signals in response to cell-cell or
cell-matrix adhesion (13). Adherence of cells to the extracellular
matrix via integrin receptors results in the assembly of protein
complexes required to modify the cellular cytoskeleton (for
reviews see refs. 1, 14, and 15). One of the early responses to
integrin receptor engagement is the activation of Src family
kinases that phosphorylate substrates such as focal adhesion
kinase, paxillin, and p130
cas.
These Src-mediated phosphoryla-
tion events have been associated with changes in cell adhesion,
motility, and shape (13, 16).
Because the Src family kinases affect both cell cycle progres-
sion and cytoskeletal organization, dysregulation may lead to
constitutive activation and cellular transformation (2). For ex-
ample, the viral derivative of Src, v-Src, and the point mutant
SrcY527F both lack the negative regulatory Tyr-527, leaving
them in the ‘‘open,’’ active conformation (17–21). Additional Src
variants bearing mutations in the SH2, SH3, and kinase domains
are also constitutively active presumably because of disruption of
the ‘‘closed’’ conformation of the protein (22, 23).
Although the reversible phosphorylation of the regulatory
tyrosine within the carboxyl terminus plays an important regu-
latory function, additional mechanisms may exist for controlling
the activity of Src family kinases. Indeed, our laboratory recently
has demonstrated that the Src family member Blk is regulated by
ubiquitin-mediated degradation (24). Specifically, the active
form of Blk is recognized by E6AP, an E3 ubiquitin-protein
ligase that promotes its ubiquitination and subsequent degrada-
tion (24–26). In this study, we show that Src itself is degraded in
a ubiquitin-dependent manner and that the active form is
specifically targeted for degradation. Taken together, these
results suggest that targeted degradation of active forms of the
Src family of tyrosine kinases may constitute an additional
mechanism for restricting the activity of these important signal-
ing proteins.
Materials and Methods
Plasmid Constructs. The pLNCX vectors encoding the chicken
c-Src, v-Src, c-Src(Y416F), and c-Src(K295R) were kind gifts of
Joan Brugge, Harvard Medical School (18, 20, 21, 27). pLNCX
vectors encoding c-Src(Y527F) and c-Src(E378G) were gener-
ated by subcloning ClaI fragments into pLNCX from plasmids
also given by Dr. Brugge (19, 21–23). pCMV4c-Src and pCMV4-
c-Src(E378G) were generated by subcloning KpnI–HindIII frag-
ments into pCMV4.
Cell Culture and Transfection. All cell lines were maintained in
DMEM (GIBCOBRL), supplemented with 100 unitsml of
penicillin, 100
gml of streptomycin, and 10% FBS, at 37° in a
5% CO
2
incubator. csk, csk, src, and src
mouse embryo fibroblasts (MEFs; refs. 28 and 31) were kind
gifts of Sheila Thomas, Harvard Medical School. Transfections
Abbreviations: SH, Src homology; MEF, mouse embryo fibroblast; PVDF, poly(vinylidene
difluoride).
*To whom reprint requests should be addressed. E-mail: peterhowley@hms.harvard.edu.
The publication costs of this article were defrayed in part by page charge payment. This
article must therefore be hereby marked advertisement in accordance with 18 U.S.C.
§1734 solely to indicate this fact.
13738–13743
PNAS
November 23, 1999
vol. 96
no. 24
were performed by using standard electroporation procedures at
0.25 V for Cos-7 cells and 0.30 V for src MEFs.
Antibodies. mAb 327 (anti-Src antibody; Oncogene Science and
generous gift of Dr. Thomas), polyclonal antibody PAb N16
(anti-Src antibody; Santa Cruz Biotechnology), polyclonal anti-
body PAb C11 (anti-actin antibody; Santa Cruz Biotechnology),
mAb 9E10 (anti-Myc antibody; Oncogene Science), and mAb
Ubi-1 (anti-ubiquitin antibody; Zymed) were used.
Cellular Lysate Preparation. Forty eight hours posttransfection,
cells were washed twice with PBS and incubated in 1 ml of
ice-cold RIPA buffer (150 mM NaCl1% NP400.5% deoxy-
cholate0.1% SDS50 mM TrisHCl, pH 7.55
g/ml PMSF5
g/ml aprotinin5
g/ml leupeptin) or NP-40 lysis buffer (100
mM TrisHCl, pH 7.4120 mM NaCl1% NP-405
g/ml
PMSF5
g/ml aprotinin5
g/ml leupeptin) as indicated. Ly-
sates were sonicated and cleared of debris by centrifuging for 5
min at 4°C. For experiments requiring proteasome inhibitor
treatment, 48 hr posttransfection, cells were treated with 50
M
Z-L
3
VS. (generous gift of Hidde Ploegh, Harvard Medical
School; ref. 30) for 8–14 hr. Control cells were treated with an
equal amount of DMSO and harvested as described above.
Immunoblotting. For each experiment, equal amounts of total
protein were resolved by 8% SDSPAGE. After SDSPAGE,
proteins were transferred to poly(vinylidene difluoride) (PVDF)
membrane in 12.5 mM TrisHCl-100 mM glycine for 2 hr at 60
V. The membrane then was incubated in a blocking solution of
5% nonfat dried milk in TNET (10 mM TrisHCl2.5 mM
EDTA50 mM NaCl0.1% Tween20) with rocking. After wash-
ing three times in TNET, the membrane was incubated in
primary antibody for at least 2 hr, washed three times, and
incubated in secondary antibody (horseradish peroxidase linked;
Amersham Pharmacia) for 1 hr. Membranes then were washed
three times in TNET and developed with enhanced chemilumi-
nescence reagents (NEN).
Pulse–Chase Experiments. src cells were transiently trans-
fected with the pLNCX-Src constructs described above. After
plasmid transfection, the cells were pooled and divided equally
among five 6-cm
2
dishes. Forty eight hours posttransfection, cells
were washed twice in PBS and then incubated in DMEM lacking
methionine and cysteine for 1 hr at 37°C. Each 6-cm
2
dish was
labeled with 100
Ci of
35
S methioninecysteine (Express Pro-
tein Labeling Mix; NEN) for a 45-min pulse, washed three times
with PBS, and chased with DMEM containing 10% FCS and
100-fold excess
L-methionine. At each time point, cells were
harvested in 0.5 ml of NP-40 lysis buffer as described above, and
equivalent amounts of cell lysate were immunoprecipitated by
using mAb 327. Immune complexes bound to the Sepharose
beads were washed three times in NP-40 lysis buffer and released
into gel loading buffer (2% SDS60 mM TrisHCl, pH 6.810%
glycerol0.1% bromophenol blue292 mM
-mercaptoethanol)
by boiling for 3 min. Samples were resolved by 8% SDSPAGE,
dried, and exposed to film. Results were quantitated by using a
Molecular Dynamics PhosphorImager (Storm860).
In Vivo
Ubiquitination Assay. Cos-7 cells were cotransfected with
pCMV4 expressing either c-Src or c-SrcE378G and pCMV4Myc-
Ub. Where indicated, cells were treated with proteasome inhib-
itors as described above. Lysates were harvested in 1 ml of RIPA
buffer and immunoprecipitated with N16 anti-Src antibodies.
Immunoprecipitates were separated by SDSPAGE, transferred
to PVDF membrane, and immunoblotted as described above by
using mAb 9E10. For csk cells, lysates were harvested and
immunoprecipitated as described above. Immunoprecipitates
were separated and transferred as above and immunoblotted
with mAb Ubi-1.
Results
Previous work from our laboratory has demonstrated that the
Src family tyrosine kinase Blk is degraded via the ubiquitin
pathway (24). Specifically, we found that activated Blk is ubi-
quitinated and is a substrate for E6AP, an E3 ubiquitin-protein
ligase. Our studies with E6AP and Blk suggested to us a model
in which the ubiquitin-mediated degradation of activated forms
of the Src family kinase might be a more general mechanism by
which this family of signaling proteins is regulated. To test this
model, we chose to investigate whether Src itself was regulated
by ubiquitination and proteolysis.
Activated Forms of Src Have Reduced Steady-State Protein Levels. To
test whether activated Src is less stable, steady-state Src levels
were analyzed in csk and csk MEF cells (29, 31). Csk
(C-terminal Src kinase) is responsible for the negative regulatory
tyrosine phosphorylation of Src at Tyr-527 (32). In cells from
Csk-deficient mice, Src activity is increased 11-fold as compared
with cells from a wild-type littermate (31). However, the steady-
state levels of Src were reduced 5.4-fold from the levels observed
in wild-type cells (Fig. 1), suggesting that steady-state levels of
Src are inversely proportional to their activity (29, 31). Half-life
experiments using cyclohexamide treatment to block protein
synthesis demonstrated that whereas Src protein from csk
cells remained stable for the duration of the experiment (2 hr),
Src protein levels from csk cells were undetectable after 90
min (data not shown). These results demonstrated that the
difference in steady-state levels was caused by a decrease in the
protein stability of endogenous Src in cskcells, presumably
because it is activated.
To test this hypothesis further, we analyzed the steady-state
levels of a series of active and inactive mutants of Src kinase
transfected into src MEFs (28). The Rous sarcoma virus
derivative of Src, v-Src, encodes a protein with 10–20 times the
activity of c-Src (18, 20). c-SrcY527F lacks the negative regula-
tory Tyr-527 and exhibits 5–10 times the kinase activity of c-Src
(19, 21). c-SrcE378G contains a mutation in the kinase domain
and is 20 times more active than c-Src (22, 23). We also
characterized the steady-state levels of two inactive mutants of
c-Src: c-SrcY416F, which lacks a positive regulatory tyrosine
required for maximal kinase activity, and c-SrcK295R, which
lacks the critical lysine in the ATP-binding pocket, rendering it
completely inactive (20, 21, 27).
Western blot analysis performed on src cells transfected
with the above constructs demonstrated that the levels of the
activated forms of Src, namely v-Src, c-SrcY527F, and
Fig. 1. Steady-state levels of endogenous Src protein. Equivalent amounts of
whole-cell lysate from src and csk MEFs and matched wild-type MEFs
were immunoprecipitated with mAb 327, separated by SDSPAGE, transferred
to PVDF membrane, and probed with mAb 327.
*
indicates the antibody heavy
chain.
Harris et al. PNAS
November 23, 1999
vol. 96
no. 24
13739
BIOCHEMISTRY
c-SrcE378G, were much lower than the levels of wild-type c-Src
protein (Fig. 2A, compare lanes 1–4). Quantitation of the Src
levels indicated a reduction in the steady-state protein by 3-fold,
4-fold, and 10-fold for c-SrcY527F, c-SrcE378G, and v-Src,
respectively, compared with c-Src levels. This inverse correlation
of protein levels to kinase activity has been noted previously for
v-Src and c-SrcY527F (20). The less active c-SrcY416F and the
completely inactive c-SrcK295R both showed steady-state levels
equivalent to or greater than c-Src (Fig. 2A, lanes 5 and 6). There
was a 1.4-fold increase in the steady-state levels of c-SrcY416F
and a 1.3-fold increase in the levels of c-SrcK295R compared
with c-Src (Fig. 2B). These results were confirmed in multiple
separate experiments using either RIPA or NP-40 lysis buffers
(data not shown). Each blot was probed for actin to control for
protein loading. The comparable levels of c-Src, Y416F, and
K295R forms of Src are consistent with our model because c-Src
is found predominantly in the inactive form (20, 21).
Activated Forms of Src Kinase Are Rapidly Degraded. We next tested
whether the decreased steady-state levels of the activated forms
of Src were caused by degradation and a shortened protein
half-life. Pulsechase analyses therefore were performed on
individual Src proteins. The half-life of wild-type c-Src was
approximately 5–6 hr. Previous reports also have indicated that
c-Src is a relatively stable protein (33, 34). In contrast to the
stability of c-Src, we found that the half-lives of the active forms
of the kinase, c-SrcE378G and c-SrcY527F, were 1 hr and 3 hr,
respectively. The decreased stability observed for the active
forms of Src is consistent with the reduction in steady-state levels
being caused by increased protein turnover. Furthermore, the
less active and inactive forms of Src were more stable than the
wild-type c-Src. Both c-SrcY416F and c-SrcK295R proteins had
half-lives greater than 7.5 hr (Fig. 3).
Active Src Kinase Is a Target for the Ubiquitin Degradation Machinery.
We next examined whether the differential stabilities of the various
forms of the Src kinase were the result of the susceptibility of the
activated forms to ubiquitination. Our previous results with Blk
indicated that the preferential proteolysis of the activated forms of
the kinase involved ubiquitination and subsequent degradation by
the proteasome (24). To determine whether activated Src was being
degraded by the proteasome, we examined the effect of the
proteasome inhibitor, Z-L
3
VS, on the levels of Src protein (30). We
compared the effects of proteasome inhibitor on the steady-state
levels of c-Src (predominantly inactive) and the active form
c-SrcE378G. As shown in Fig. 4, there was a minimal 1.2-fold
increase in c-Src protein level in the presence of proteasome
inhibitor; however, there was a dramatic 3.3-fold increase in the
c-SrcE378G protein levels. This result, demonstrating the stabili-
zation of active Src by a proteasome inhibitor, confirmed the
involvement of the proteasome in the degradation of activated Src.
The slight stabilization of c-Src might be caused by a low level of
active wild-type c-Src or the normal turnover of inactive c-Src. The
proteasome inhibitor had no effect on actin levels.
To investigate further the role of the ubiquitin pathway in Src
proteolysis, we examined whether ubiquitinated forms of Src
Fig. 2. Steady-state levels of wild-type and mutant Src proteins. (A)(Upper)
Src MEFs were transfected with pLNCX vector expressing c-Src, v-Src,
Y527F, E378G, Y416F, or K295R forms of Src, or pLNCX alone. Cells were
harvested after 48 hr, and 50
g of whole-cell lysate from each transfection
was separated by SDSPAGE, transferred to PVDF membrane, and probed with
mAb 327. (Lower) The same membrane reprobed with anti-actin antibody
C11. (B) Quantitation of the results shown in A by using densitometry.
Fig. 3. Pulse–chase analysis of wild-type and mutant Src proteins. Src
MEFs were transfected with pLNCX vector expressing c-Src, or the following
Src mutants: Y527F, E378G, Y416F, or K295R. Forty eight hours posttransfec-
tion, cells were pulse-labeled with 100
Ci
35
S MetCys for 45 min. Plates were
incubated in ‘‘chase’’ media with 100-fold excess methionine and then har-
vested at the indicated times. Lysates were immunoprecipitated by using mAb
327 and separated by SDSPAGE. Gels were quantitated with a PhosphorIm-
ager, and each time point was normalized to the 0-hr time point.
13740
www.pnas.org Harris et al.
could be detected in vivo. For these experiments c-Src or
c-SrcE378G were transfected into Cos-7 cells together with a
plasmid expressing Myc-tagged ubiquitin. Ubiquitinated forms
of c-SrcE378G were readily detected as a smear of higher
molecular weight bands in the cells coexpressing c-SrcE378G
and Myc-tagged ubiquitin (Fig. 5A, lanes 7 and 8). Although only
a faint ladder of multiubiquitinated Src was detected, the high
molecular weight smear is characteristic of multiubiquitinated
proteins and is similar to those found in previous reports using
Myc-tagged ubiquitin (35, 36). The addition of proteasome
inhibitor did not greatly stabilize the ubiquitinated forms of
c-SrcE378G, which is consistent with previous reports demon-
strating the overall stability of Myc-tagged ubiquitin protein
conjugates (37). We next examined the ubiquitination state of
endogenous Src in csk cells where Src is predominantly
active. After treatment with proteasome inhibitor for 14 hr, cell
lysates were immunoprecipitated with anti-Src antibodies and
then analyzed by immunoblotting with anti-ubiquitin antibodies.
As shown in Fig. 5B, we could detect the characteristic high
molecular weight smear indicative of ubiquitination. In longer
exposures of the transfection experiment (Fig. 5A) and in
experiments using csk cells we also were able to detect
evidence of high molecular bands indicative of ubiquitinated
c-Src (data not shown). Because the half-life of c-Src is 5–6 hr
(Fig. 3), the 14-hr proteasome inhibitor treatment period would
account for at least two half-lives of c-Src. This result suggests
that normal turnover of c-Src also may be regulated by ubiq-
uitination. Nonetheless, these results demonstrate the in vivo
ubiquitination of endogenous Src. Taken together with the
stabilization of Src observed with proteasome inhibitors, these
data confirm a role for ubiquitination in the regulation of Src
through targeted proteolysis of its activated forms (Fig. 6).
Discussion
Our laboratory recently has shown that activated forms of the Src
family member Blk are specifically degraded by ubiquitin-
mediated proteolysis (24). In the present study we have extended
this observation to Src itself and have examined whether this
mechanism of regulation is a property of other members of this
kinase family. Using csk cells, which lack the negative
regulator of Src, and mutant forms of Src as sources of active
kinase, we demonstrated a decrease in the steady-state levels of
the active forms of Src. Pulsechase experiments revealed
shorter half-lives for activated forms of c-Src, suggesting that the
decreased protein levels were the result of increased protein
turnover. In contrast, Src mutants fully or partially impaired in
their catalytic activity were quite stable and were present at
Fig. 4. Steady-state levels of Src in the presence of the proteasome inhibitor
Z-L
3
VS. (Upper) Src
MEFs were transfected with pLNCX vector expressing
c-Src, the active E378G mutant, or vector alone. Forty eight hours after
transfection, cells were treated with either 50
M Z-L
3
VS in DMSO or DMSO
alone as a negative control. Eight hours after treatment, cells were harvested,
separated by SDSPAGE, transferred to PVDF membrane, and probed with
mAb 327. The faint band in the LNCX lane is a background band. (Lower) The
same membrane reprobed with anti-actin antibody C11.
Fig. 5. In vivo ubiquitination of active c-Src. (A) Cos-7 cells were transiently transfected with pCMV4 vector alone (lane 1) or pCMV4 expressing c-Src (lane 2),
E378G (lane 3), Myc-tagged ubiquitin (lane 4) alone, or in the combinations indicated (lanes 5–8). For each transfection, 1 mg of lysate was immunoprecipitated
with polyclonal antibody (PAb) N16 (anti-Src), separated by SDSPAGE, transferred to PVDF membrane, and probed with mAb 9E10 (anti-myc). Cells used for lanes
6 and 8 were treated with 50
M Z-L
3
VS for 14 hr before harvesting.
*
indicates a background band. (B) csk cells were treated with either 50
M Z-L
3
VS in
DMSO or DMSO alone as a negative control. One milligram of lysate was immunoprecipitated with PAb N16 (anti-Src), separated by SDSPAGE, transferred to
PVDF membrane, and probed with mAb Ubi-1 (anti-ubiquitin).
Harris et al. PNAS
November 23, 1999
vol. 96
no. 24
13741
BIOCHEMISTRY
higher steady-state levels. Proteasome inhibitors stabilized only
the active forms of Src, implicating the proteasome machinery in
the proteolysis of the active forms of Src. We also demonstrated
the in vivo ubiquitination of kinase active Src, thus establishing
the involvement of ubiquitination in the regulation of Src.
Regulation of Src by ubiquitin-mediated degradation provides
a previously unrecognized mechanism for limiting Src activity. A
major switch for the activation and inactivation of Src involves
the phosphorylation and dephosphorylation of tyrosine residues
within the C terminus and kinase domains of the protein.
Regulation by reversible phosphorylation allows the kinase to
alternate between active and inactive states. The specific pro-
teolysis of the kinase-active forms of Src by ubiquitination as
demonstrated in this manuscript provides an alternative mech-
anism for restricting Src kinase activity. The irreversible nature
of ubiquitin-mediated proteolysis allows the cell to control and
attenuate the activity of a mitogenic protein.
Although ubiquitin-mediated degradation has not previously
been shown to be a mechanism for regulation of Src activity, the
correlation between increased activity and decreased protein
stability has been noted previously. Using NIH 3T3 cells stably
expressing various Src constructs, Kmiecik and Shalloway (20)
demonstrated that v-Src and c-SrcY527F have higher specific
kinase activities and significantly lower steady-state protein
levels than c-Src. In addition, other studies have shown that v-Src
has a short-half life that varies among different viral strains,
suggesting that the stability of v-Src depends on mutations
unique to each strain. Protein synthesis inhibitors greatly in-
creased the half-life of v-Src in these studies, indicating that
additional proteins are required for v-Src degradation (38, 39).
Unregulated, constitutive activation of Src induces cellular
transformation presumably through activation of mitogenic and
cellular adhesion pathways. In most cases, transformation by Src
occurs only when activated forms of the kinase are present;
however, there are data suggesting that elevated levels of c-Src
are transforming. Johnson et al. (40) found that c-Src transfected
into NIH 3T3 cells could induce low levels of foci. When these
transformed foci were examined, Src levels were consistently
higher than those found in transfected, nontransformed NIH
3T3 cells (average ratio of 5:1). Confirmation of the wild-type
status of the c-Src in these foci led these researchers to propose
that a level of c-Src above a certain threshold was oncogenic.
Defects in Src ubiquitination could cause Src protein levels to
exceed a ‘‘threshold’’ level and result in cellular transformation.
In fact, various human cancers, including breast cancer, exhibit
elevated Src levels despite the absence of any detectable Src
mutations (41). Elevated levels of Src in these cancers possibly
could result from mutations in genes that regulate Src levels,
possibly in those that participate in the proper targeting of Src
to the ubiquitin proteasome pathway.
Although we have established that the ubiquitinproteasome
pathway is involved in degrading activated Src, the specific
enzymes involved in the process have yet to be determined. The
E3 ubiquitin-protein ligases are the critical enzymes responsible
for substrate recognition by the ubiquitin machinery (for review
see ref. 42). E6AP was the first mammalian E3 enzyme to be
identified (26). In cooperation with the human papillomavirus
E6 protein, E6AP mediates the ubiquitination of p53 (25, 26).
E6AP is the prototype of a family of E3 proteins known as HECT
domain proteins (homology to E6AP carboxyl terminus), all of
which share similarities in their catalytic domains (43, 44). Based
on our previous results demonstrating a role for E6AP in
targeting Blk for degradation, E6AP, or perhaps a related HECT
family member, is a strong candidate for the E3 responsible for
recognizing activated forms of Src. In preliminary coimmuno-
precipitation experiments, we have detected an interaction
between E6AP and Src (K.F.H., E.M.C., and P.M.H., unpub-
lished results). Additional experiments, however, will need to be
performed to establish the specific ubiquitin protein ligase that
is involved in the degradation of Src.
In addition to identifying the E3 enzyme involved, it will be
important to determine the specific degradation signal(s) in the
active form of Src that is recognized by the ubiquitin machinery.
Activated Src adopts an open conformation in which tyrosine
416 becomes phosphorylated (2). It is possible that either the
change in conformation status or the phosphorylation of specific
residues such as tyrosine 416 serves as the degradation signal.
The stability of c-Src K295R argues for the latter possibility. This
mutant can adopt the open conformation, yet remains stable,
suggesting that conformation alone is not sufficient to target Src
for degradation. There also may be a requirement for interac-
tions with additional cellular proteins before recognition of Src
as a substrate for the ubiquitination machinery. These mecha-
nisms are not mutually exclusive and must be carefully explored.
The work presented in this report demonstrates the targeted
degradation of active forms of Src by the ubiquitinproteasome
pathway. These results reveal a previously unidentified cellular
mechanism for irreversibly limiting Src kinase activity. Further
investigation and elucidation of this pathway is necessary to gain
a general understanding of the cellular regulation of Src activity
and how this regulation may be disrupted in cancer cells.
We thank J. Brugge for plasmids expressing wild-type and mutant Src
kinases. We thank S. Thomas for anti-Src antibodies and src,
src, csk, and csk MEFs and H. Ploegh for Z-L
3
VS
proteasome inhibitor. We are grateful to J. Brugge, A. Hudson, L.
Decker, and W. Kao for a critical review of this manuscript. This work
was supported by a grant from the National Institutes of Health
(R01-CA64888). K.F.H. was supported by a postdoctoral fellowship from
the National Institutes of HealthNational Cancer Institute (award
number 1 F32 CA81727–01). S.K. was supported by a postdoctoral
fellowship from the American Cancer Society (award number PF-4309).
1. Thomas, S. M. & Brugge, J. S. (1997) Annu. Rev. Cell. Dev. Biol. 13, 513–609.
2. Brown, M. T. & Cooper, J. A. (1996) Biochim. Biophys. Acta 1287, 121–149.
3. Xu, W., Doshi, A., Lei, M., Eck, M. J. & Harrison, S. C. (1999) Mol. Cell 3,
629638.
4. Taylor, S. J. & Shalloway, D. (1993) Curr. Opin. Genet. Dev. 3, 26–34.
5. Xu, W., Harrison, S. C. & Eck, M. J. (1997) Nature (London) 385, 595–602.
6. Ralston, R. & Bishop, J. M. (1985) Proc. Natl. Acad. Sci. USA 82, 7845–7849.
7. Twamley-Stein, G. M., Pepperkok, R., Ansorge, W. & Courtneidge, S. A.
Fig. 6. Proposed model for the targeted degradation of active Src. Phos-
phorylation of Tyr-527 maintains Src in a ‘‘closed’’ inactive conformation.
Dephosphorylation of pTyr-527 and autophosphorylation of Tyr-416 releases
the kinase into an open, fully active state. The active form of Src is specifically
targeted for ubiquitin-mediated proteolysis. The E3 ubiquitin-protein ligase
that recognizes active Src has not yet been identified.
13742
www.pnas.org Harris et al.
(1993) Proc. Natl. Acad. Sci. USA 90, 7696–7700.
8. Roche, S., Koegl, M., Barone, M. V., Roussel, M. F. & Courtneidge, S. A.
(1995) Mol. Cell. Biol. 15, 1102–1109.
9. Barone, M. V. & Courtneidge, S. A. (1995) Nature (London) 378, 509–512.
10. Chackalaparampil, I. & Shalloway, D. (1988) Cell 52, 801–810.
11. Fumagalli, S., Totty, N. F., Hsuan, J. J. & Courtneidge, S. A. (1994) Nature
(London) 368, 871–874.
12. Roche, S., Fumagalli, S. & Courtneidge, S. A. (1995) Science 269, 1567–1569.
13. Klinghoffer, R. A., Sachsenmaier, C., Cooper, J. A. & Soriano, P. (1999) EMBO
J. 18, 2459–2471.
14. Juliano, R. L. & Haskill, S. (1993) J. Cell Biol. 120, 577–585.
15. Schwartz, M. A. (1993) Cancer Res. 53, 1503–1506.
16. Hanks, S. K. & Polte, T. R. (1997) BioEssays 19, 137–145.
17. Takeya, T. & Hanafusa, H. (1983) Cell 32, 881–890.
18. Hunter, T. (1987) Cell 49, 1–4.
19. Cartwright, C. A., Eckhart, W., Simon, S. & Kaplan, P. L. (1987) Cell 49, 83–91.
20. Kmiecik, T. E. & Shalloway, D. (1987) Cell 49, 65–73.
21. Piwnica-Worms, H., Saunders, K. B., Roberts, T. M., Smith, A. E. & Cheng,
S. H. (1987) Cell 49, 75–82.
22. Levy, J. B., Iba, H. & Hanafusa, H. (1986) Proc. Natl. Acad. Sci. USA 83,
42284232.
23. Bjorge, J. D., Bellagamba, C., Cheng, H. C., Tanaka, A., Wang, J. H. & Fujita,
D. J. (1995) J. Biol. Chem. 270, 24222–24228.
24. Oda, H., Kumar, S. & Howley, P. M. (1999) Proc. Nat. Acad. Sci. USA 96,
9557–9562.
25. Huibregtse, J. M., Scheffner, M. & Howley, P. M. (1993) Mol. Cell. Biol. 13,
775–784.
26. Scheffner, M., Huibregtse, J. M., Vierstra, R. D. & Howley, P. M. (1993) Cell
75, 495–505.
27. Snyder, M. A., Bishop, J. M., McGrath, J. P. & Levinson, A. D. (1985) Mol. Cell.
Biol. 5, 1772–1779.
28. Soriano, P., Montgomery, C., Geske, R. & Bradley, A. (1991) Cell 64, 693–702.
29. Nada, S., Yagi, T., Takeda, H., Tokunaga, T., Nakagawa, H., Ikawa, Y., Okada,
M. & Aizawa, S. (1993) Cell 73, 1125–1135.
30. Bogyo, M., McMaster, J. S., Gaczynska, M., Tortorella, D., Goldberg, A. L. &
Ploegh, H. (1997) Proc. Natl. Acad. Sci. USA 94, 66296634.
31. Imamoto, A. & Soriano, P. (1993) Cell 73, 1117–1124.
32. Nada, S., Okada, M., MacAuley, A., Cooper, J. A. & Nakagawa, H. (1991)
Nature (London) 351, 69–72.
33. Iba, H., Cross, F. R., Garber, E. A. & Hanafusa, H. (1985) Mol. Cell. Biol. 5,
1058–1066.
34. Hirai, H. & Varmus, H. E. (1990) Mol. Cell. Biol. 10, 1307–1318.
35. Ward, C. L., Omura, S. & Kopito, R. R. (1995) Cell 83, 121–127.
36. Hofmann, F., Martelli, F., Livingston, D. M. & Wang, Z. (1996) Genes Dev. 10,
2949–2959.
37. Ellison, M. J. & Hochstrasser, M. (1991) J. Biol. Chem. 266, 21150–21157.
38. Ziemiecki, A., Friis, R. R. & Bauer, H. (1982) Mol. Cell. Biol. 2, 355–360.
39. Sefton, B. M., Patschinsky, T., Berdot, C., Hunter, T. & Elliott, T. (1982)
J. Virol. 41, 813–820.
40. Johnson, P. J., Coussens, P. M., Danko, A. V. & Shalloway, D. (1985) Mol. Cell.
Biol. 5, 1073–1083.
41. Kolibaba, K. S. & Druker, B. J. (1997) Biochim. Biophys. Acta 1333, F217–F248.
42. Ciechanover, A. & Schwartz, A. L. (1994) FASEB J. 8, 182–191.
43. Huibregtse, J. M., Scheffner, M., Beaudenon, S. & Howley, P. (1995) Proc. Natl.
Acad. Sci. USA 92, 2563–2567.
44. Huibregtse, J. M., Scheffner, M. & Howley, P. M. (1994) in Cold Spring Harbor
Symposia on Quantitative Biology (Cold Spring Harbor Lab. Press, Plainview,
NY), Vol. 59, pp. 237–245.
Harris et al. PNAS
November 23, 1999
vol. 96
no. 24
13743
BIOCHEMISTRY
... SFKs, especially Src, can participate in the tumorigenesis process by activating STAT transcription factors [54]. A variety of human tumors have high levels of Src kinase activity expression-for example, breast, colorectal, and pancreatic cancers [55]. The increase in Src kinase activity in the tumor may be caused by tyrosine phosphatase-mediated dephosphorylation, increased Src levels, or upstream regulatory proteins. ...
Article
Full-text available
C-terminal Src kinase (CSK) is the major inhibitory kinase for Src family kinases (SFKs) through the phosphorylation of their C-tail tyrosine sites, and it regulates various types of cellular activity in association with SFK function. As a cytoplasmic protein, CSK needs be recruited to the plasma membrane to regulate SFKs’ activity. The regulatory mechanism behind CSK activity and its subcellular localization remains largely unclear. In this work, we developed a genetically encoded biosensor based on fluorescence resonance energy transfer (FRET) to visualize the CSK activity in live cells. The biosensor, with an optimized substrate peptide, confirmed the crucial Arg107 site in the CSK SH2 domain and displayed sensitivity and specificity to CSK activity, while showing minor responses to co-transfected Src and Fyn. FRET measurements showed that CSK had a relatively mild level of kinase activity in comparison to Src and Fyn in rat airway smooth muscle cells. The biosensor tagged with different submembrane-targeting signals detected CSK activity at both non-lipid raft and lipid raft microregions, while it showed a higher FRET level at non-lipid ones. Co-transfected receptor-type protein tyrosine phosphatase alpha (PTPα) had an inhibitory effect on the CSK FRET response. The biosensor did not detect obvious changes in CSK activity between metastatic cancer cells and normal ones. In conclusion, a novel FRET biosensor was generated to monitor CSK activity and demonstrated CSK activity existing in both non-lipid and lipid raft membrane microregions, being more present at non-lipid ones.
... SFKs, especially Src, can participate in the tumorigenesis process by activating STAT transcription factors [56]. A variety of human tumors have high levels of Src kinase activity expression, for example, breast, colorectal, and pancreatic cancers [57]. The increase in Src kinase activity in this tumor may be caused by tyrosine phosphatasemediated dephosphorylation of carboxyl terminal negative regulatory elements, or by increased protein levels and protein stability of Src, or by upregulation of upstream receptor tyrosine kinase activity and loss of key regulatory proteins. ...
Preprint
Full-text available
C-terminal Src kinase (CSK) is the major inhibitory kinase for Src family kinases (SFKs) through phosphorylation of their C-tail tyrosine site, and regulates various cellular activities in association with SFKs functions. As a cytoplasmic protein, CSK need be recruited to the plasma membrane to regulate SFKs activities. The regulatory mechanism for CSK activity and its subcellular localization remains largely unclear. In this work, we developed a genetically encoded biosensor based on fluorescence resonance energy transfer (FRET) to visualize CSK activity in live cells. The biosensor with optimized substrate peptide confirmed crucial Arg107 site in CSK SH2 domain, and displayed sensitivity and specificity to CSK activity while little responses to co-transfected Src and Fyn. FRET measurements showed CSK having relatively mild level of kinase activity in comparison to Src and Fyn ones in rat airway smooth muscle cells. The biosensor tagged with different submembrane-targeting signals detected CSK activity at both non-lipid raft and lipid raft microregions, while showed higher FRET level at non-lipid ones. Co-transfected receptor-type protein tyrosine phosphatase alpha (PTPα) had inhibitory effect on the CSK FRET response. The biosensor didn’t detect obvious changes of CSK activity between metastatic cancer cells and normal ones. In conclusion, a novel FRET biosensor was generated to monitor CSK activity, and demonstrated CSK activity existing in both non-lipid and lipid raft membrane microregions while more present at non-lipid ones.
... This is partially supported by results from our strain studies, where HUVECs treated with mechanical strain for 15 min displayed an increase in Src compared to samples treated with VEGF (Fig. 3f ). Previous literature has shown that activated Src is more unstable than inactive Src; therefore, active Src may be more quickly degraded [58]. Thus, the observed increase of Src protein levels in VEGFtreated cells could be due to Src inactivity and higher stability. ...
Article
Full-text available
Background Angiogenesis, or the growth of new vasculature from existing blood vessels, is widely considered a primary hallmark of cancer progression. When a tumor is small, diffusion is sufficient to receive essential nutrients; however, as the tumor grows, a vascular supply is needed to deliver oxygen and nutrients into the increasing mass. Several anti-angiogenic cancer therapies target VEGF and the receptor VEGFR-2, which are major promoters of blood vessel development. Unfortunately, many of these cancer treatments fail to completely stop angiogenesis in the tumor microenvironment (TME). Since these therapies focus on the biochemical activation of VEGFR-2 via VEGF ligand binding, we propose that mechanical cues, particularly those found in the TME, may be a source of VEGFR-2 activation that promotes growth of blood vessel networks even in the presence of VEGF and VEGFR-2 inhibitors. Results In this paper, we analyzed phosphorylation patterns of VEGFR-2, particularly at Y1054/Y1059 and Y1214, stimulated via either VEGF or biomechanical stimulation in the form of tensile strains. Our results show prolonged and enhanced activation at both Y1054/Y1059 and Y1214 residues when endothelial cells were stimulated with strain, VEGF, or a combination of both. We also analyzed Src expression, which is downstream of VEGFR-2 and can be activated through strain or the presence of VEGF. Finally, we used fibrin gels and microfluidic devices as 3D microtissue models to simulate the TME. We determined that regions of mechanical strain promoted increased vessel growth, even with VEGFR-2 inhibition through SU5416. Conclusions Overall, understanding both the effects that biomechanical and biochemical stimuli have on VEGFR-2 activation and angiogenesis is an important factor in developing effective anti-angiogenic therapies. This paper shows that VEGFR-2 can be mechanically activated through strain, which likely contributes to increased angiogenesis in the TME. These proof-of-concept studies show that small molecular inhibitors of VEGFR-2 do not fully prevent angiogenesis in 3D TME models when mechanical strains are introduced.
... The pancreatic islets express most SFKs similarly in Wistar and GK rats (undetectable level of Fyn), except for c-Src [84]. The level of c-Src pY416, which indicates Src activation, has been found to be higher in the islets of GK rats than in those of Wistar rats, and the levels of c-Src and carboxyl terminal Src kinase, a negative regulator of SFKs [85], have been found to be lower in the islets of GK rats than in those of Wistar rats [84], indicating that c-Src is activated under diabetic conditions and is degraded by ubiquitination [87] (Figure 2). Although the mechanism of c-Src activation under diabetic conditions is unclear, ROS themselves may regulate c-Src activity [88]. ...
Article
Full-text available
The dysfunction of pancreatic β-cells plays a central role in the onset and progression of type 2 diabetes mellitus (T2DM). Insulin secretory defects in β-cells are characterized by a selective impairment of glucose stimulation, and a reduction in glucose-induced ATP production, which is essential for insulin secretion. High glucose metabolism for insulin secretion generates reactive oxygen species (ROS) in mitochondria. In addition, the expression of antioxidant enzymes is very low in β-cells. Therefore, β-cells are easily exposed to oxidative stress. In islet studies using a nonobese T2DM animal model that exhibits selective impairment of glucose-induced insulin secretion (GSIS), quenching ROS generated by glucose stimulation and accumulated under glucose toxicity can improve impaired GSIS. Acute ROS generation and toxicity cause glucose metabolism disorders through different molecular mechanisms. Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor, is a master regulator of antioxidant defense and a potential therapeutic target in oxidative stress-related diseases, suggesting the possible involvement of Nrf2 in β-cell dysfunction caused by ROS. In this review, we describe the mechanisms of insulin secretory defects induced by oxidative stress in diabetic β-cells.
... Once signaling has terminated, SFKs can be inactivated by phosphorylation of Y537 or degraded by ubiquitination (26). Maintaining the balance between the inactivation of the kinases and their degradation is important to ensure a rapid response to extracellular signals while avoiding an excessive signaling that could lead to oncogenic processes. ...
Article
YES1 is a non-receptor tyrosine kinase that belongs to the SRC family of kinases (SFKs) and controls multiple cancer signaling pathways. YES1 is amplified and overexpressed in many tumor types, where it promotes cell proliferation, survival and invasiveness. Therefore, YES1 has been proposed as an emerging target in solid tumors. In addition, studies have shown that YES1 is a prognostic biomarker and a predictor of dasatinib activity. Several SFKs-targeting drugs have been developed and some of them have reached clinical trials. However, these drugs have encountered challenges to their utilization in the clinical practice in unselected patients due to toxicity and lack of efficacy. In the case of YES1, novel specific inhibitors have been developed and tested in preclinical models, with impressive antitumor effects. In this review, we summarize the structure and activation of YES1 and describe its role in cancer as a target and prognostic and companion biomarker. We also address the efficacy of SFKs inhibitors that are currently in clinical trials, highlighting the main hindrances for their clinical use. Current available information strongly suggests that inhibiting YES1 in tumors with high expression of this protein is a promising strategy against cancer.
... In contrast, phosphorylation of Y 416 leads to the release of the activation loop, resulting in increased SRC kinase activity (12,13). Phosphorylation of Y 416 also promotes SRC ubiquitination and subsequent degradation (14). ...
Article
Full-text available
Systemic sclerosis (SSc) is a fibrotic autoimmune disease characterized by pathogenic activation of fibroblasts enhanced by local oxidative stress. The tyrosine phosphatase PTP4A1 was identified as a critical promoter of TGF-β signaling in SSc. Oxidative stress is known to functionally inactivate tyrosine phosphatases. Here, we assessed whether oxidation of PTP4A1 modulates its profibrotic action and found that PTP4A1 forms a complex with the kinase SRC in scleroderma fibroblasts, but surprisingly, oxidative stress enhanced rather than reduced PTP4A1's association with SRC and its profibrotic action. Through structural assessment of the oxo-PTP4A1-SRC complex, we unraveled an unexpected mechanism whereby oxidation of a tyrosine phosphatase promotes its function through modification of its protein complex. Considering the importance of oxidative stress in the pathogenesis of SSc and fibrosis, our findings suggest routes for leveraging PTP4A1 oxidation as a potential strategy for developing antifibrotic agents.
... Moreover, the use of the PS341, a selective proteasome inhibitor, inhibited 21 PPARδ/β proteolysis, increasing the half-life of the DNA-bound receptor and therefore increasing its activity [96]. The 26S proteasome is a 2.4 MDa multifunctional ATP-dependent proteolytic complex, which degrades a large variety of cell proteins and is essential for many cellular regulatory mechanisms, that includes cell cycle progression, by the proteasomal degradation of cyclins and inhibitors of CDKs [164], transcriptional regulators (such as c-JUN, E2F-1, and β-catenin) [123], and kinases (such as SRC and protein kinase C (PKC)) [113,199], terminating specific signal transduction cascades. Furthermore, the ubiquitinproteasome pathway also plays an essential role in immune surveillance [141], muscle atrophy [220], regulation of metabolic pathways [108,226], acquisition of long-term memory [40], inflammatory response [211,241], and in the regulation of circadian rhythms [229] and tumor progression [123,239]. ...
Chapter
The peroxisome proliferator-activated receptors (PPAR) are transcription factors modulated by ligands and members of the nuclear receptor superfamily. There are three different human PPAR isotypes: PPARα, PPARδ/β, and PPARγ, which regulate the transcription of their target genes involved with energy metabolism, inflammatory process, and cellular differentiation in different human tissues. Because of these activities, PPARs are considered important targets for drugs to treat metabolic diseases, including diabetes, dyslipidemia, and obesity. Besides ligand modulation, PPARs activities can be modulated by posttranslational modifications (PTM), such as phosphorylation, SUMOylation, ubiquitination, acetylation, and O-GlcNAcylation. The understanding of PTMs modulation of PPARs function could contribute for the development of metabolic diseases treatment with more specificity and fewer side effects. Therefore, in this chapter, we present an overview of PTMs that modulate the activity of each PPAR isotype and strategies to modulate these PTMs and thus regulate PPARs action.
Article
Background: KRAS is a frequently mutated oncogene in human cancer. Clinical studies on the covalent inhibitors of the KRASG12C mutant have reported promising results. However, primary and acquired resistance may limit their clinical use. Methods: Sotorasib-resistant cell lines were established. We explored the signalling pathways activated in these resistant cell lines and their roles in sotorasib resistance. Results: The resistant cells exhibited increased cell-matrix adhesion with increased levels of stress fibres and focal adherens. p21-activated kinases (PAKs) were activated in resistant cells, which phosphorylate MEK at serine 298 of MEK and serine 338 of c-Raf to activate the mitogen-activated protein kinase pathway. The PAK inhibitors FRAX597 and FRAX486 in synergy with sotorasib reduced the viability of KRASG12C mutant cancer cells. Furthermore, the PI3K/AKT pathway was constitutively active in sotorasib-resistant cells. The overexpression of constitutively activated PI3K or the knockdown of PTEN resulted in resistance to sotorasib. PI3K inhibitor alpelisib was synergistic with sotorasib in compromising the viability of KRASG12C mutant cancer cells. Moreover, PI3K and PAK pathways formed a mutual positive regulatory loop that mediated sotorasib resistance. Conclusions: Our results indicate that the cell-matrix interaction-dependent activation of PAK mediates resistance to sotorasib through the activation of MAPK and PI3K pathways.
Article
Aims: Huntington's Disease (HD) is an autosomal-dominant neurodegenerative disorder with no effective therapies. Mutant huntingtin (mHTT), the main HD proteinaceous hallmark, has been linked to reactive oxygen species (ROS) formation and mitochondrial dysfunction, among other pathological mechanisms. Importantly, Src-related kinases, c-Src and Fyn, are activated by ROS and regulate mitochondrial activity. However, c-Src/Fyn involvement in HD is largely unexplored. Thus, in this study we aimed to explore changes in Src/Fyn proteins in HD models and their role in defining altered mitochondrial function and dynamics and redox regulation. Results: We show, for the first time, that c-Src/Fyn phosphorylation/activation and proteins levels are decreased in several human and mouse HD models mainly due to autophagy degradation, concomitantly with mHtt-expressing cells showing enhanced TFEB-mediated autophagy induction and autophagy flux. c-Src/Fyn co-localization with mitochondria is also reduced. Importantly, expression of constitutive active c-Src/Fyn to restore active SKF levels improves mitochondrial morphology and function, namely through improved mitochondrial transmembrane potential, mitochondrial basal respiration and ATP production, but did not affect mitophagy. Additionally, constitutive active c-Src/Fyn expression diminishes the levels of reactive species in cells expressing mHTT. Innovation: This work supports a relevant role for c-Src/Fyn proteins in controlling mitochondrial function and redox regulation in HD, revealing a potential HD therapeutic target. Conclusion: c-Src/Fyn restoration in HD improves mitochondrial morphology and function, precluding the rise in oxidant species and cell death.
Article
In most metazoans, early embryonic development is characterized by rapid division cycles that pause before gastrulation at the midblastula transition (MBT).¹ These cleavage divisions are accompanied by cytoskeletal rearrangements that ensure proper nuclear positioning. However, the molecular mechanisms controlling nuclear positioning are not fully elucidated. In Drosophila, early embryogenesis unfolds in a multinucleated syncytium. Nuclei rapidly move across the anterior-posterior (AP) axis at cell cycles 4–6 in a process driven by actomyosin contractility and cytoplasmic flows.²,³ In shackleton (shkl) mutants, this axial spreading is impaired.⁴ Here, we show that shkl mutants carry mutations in the cullin-5 (cul-5) gene. Live imaging experiments show that Cul-5 is downstream of the cell cycle but is required for cortical actomyosin contractility. The nuclear spreading phenotype of cul-5 mutants can be rescued by reducing Src activity, suggesting that a major target of cul-5 is Src kinase. cul-5 mutants display gradients of nuclear density across the AP axis that we exploit to study cell-cycle control as a function of the N/C ratio. We found that the N/C ratio is sensed collectively in neighborhoods of about 100 μm, and such collective sensing is required for a precise MBT, in which all the nuclei in the embryo pause their division cycle. Moreover, we found that the response to the N/C ratio is slightly graded along the AP axis. These two features can be linked to Cdk1 dynamics. Collectively, we reveal a new pathway controlling nuclear positioning and provide a dissection of how nuclear cycles respond to the N/C ratio.
Article
Full-text available
In the present work, a method based on an epitope-tagged ubiquitin derivative is described that allows for the unambiguous detection of ubiquitin-protein conjugates formed in vivo or in vitro. Expression in the yeast Saccharomyces cerevisiae of ubiquitin that has been tagged at its amino terminus with a peptide epitope results in the formation of tagged ubiquitin-protein conjugates that are detectable by immunoblotting with a monoclonal antibody that recognizes the tag. The expression of tagged ubiquitin has no adverse effect on vegetative growth and, moreover, can suppress the stress-hypersensitive phenotype of yeast lacking the polyubiquitin gene UBI4. We also show that tagged ubiquitin is correctly conjugated in vivo and in vitro to a short-lived test protein and can be covalently extended into the multimeric ubiquitin chain that is normally required for the degradation of this protein. Surprisingly, however, conjugation of tagged ubiquitin inhibits proteolysis. These and related results suggest that the amino-terminal region of ubiquitin is important in protease-substrate recognition and that the multiubiquitin chain is a dynamic transient structure. The potential of tagged ubiquitin for the identification and isolation of ubiquitin-protein conjugates and ubiquitin-related enzymes, and as a tool in mechanistic studies is discussed.
Article
The half-life of metabolically labeled pp60src of the Prague A strain of Rous sarcoma virus and of several transformation-defective, temperature-sensitive mutants was investigated by pulse-labeling infected cells with [35S]methionine, chasing for different times, and immunoprecipitating pp60src with tumor-bearing rabbit serum. These experiments showed that pp60src has a short half-life of approximately 60 min under normal physiological conditions and that the mutant pp60src proteins have similar half-lives to the wild type, irrespective of whether the cells are kept at the nonpermissive (42 degrees C) or permissive (35 degrees C) temperature. The half-life of the pp60src -associated kinase activity was determined by monitoring its decay by the immunoglobulin G heavy chain assay after the cells had been treated with several inhibitors of protein synthesis. In these experiments the kinase half-life was much longer than expected from the half-life of pp60src. The apparent contradiction between the half-lives of the kinase activity and the [35S]methionine-labeled pp60src protein could be resolved by the observation that treatment of cells with inhibitors of protein synthesis stabilized pp60src, resulting in a greatly extended half-life. Inhibitors of protein synthesis also extended the half-life of the gag precursor polypeptide, Pr76, suggesting that a host factor(s) may be required for the efficient intracellular processing of this polypeptide to the gag proteins.
Article
The E6 oncoproteins of the cancer-associated or high-risk human papillomaviruses (HPVs) target the cellular p53 protein. The association of E6 with p53 leads to the specific ubiquitination and degradation of p53 in vitro, suggesting a model by which E6 deregulates cell growth control by the elimination of the p53 tumor suppressor protein. Complex formation between E6 and p53 requires an additional cellular factor, designated E6-AP (E6-associated protein), which has a native and subunit molecular mass of approximately 100 kDa. Here we report the purification of E6-AP and the cloning of its corresponding cDNA, which contains a novel open reading frame encoding 865 amino acids. E6-AP, translated in vitro, has the following properties: (i) it associates with wild-type p53 in the presence of the HPV16 E6 protein and simultaneously stimulates the association of E6 with p53, (ii) it associates with the high-risk HPV16 and HPV18 E6 proteins in the absence of p53, and (iii) it induces the E6- and ubiquitin-dependent degradation of p53 in vitro.
Article
NIH 3T3 cells were transfected with plasmids containing Moloney murine leukemia virus long terminal repeats and either chicken c-src or v-src genes. In contrast with the effects observed after transfection with plasmids containing c-src and avian retrovirus or simian virus 40 promoter-enhancers (H. Hanafusa, H. Iba, T. Takeya, and F. R. Cross, p. 1-8, in G. F. Vande Woude, A. J. Levine, W. C. Topp, and J. D. Watson, ed., Cancer Cells, vol. 2, 1984; H. Iba, T. Takeya, F. R. Cross, T. Hanafusa, and H. Hanafusa, Proc. Natl. Acad. Sci. U.S.A. 81:4424-4428, 1984; R. C. Parker, R. Swanstrom, H. E. Varmus, and J. M. Bishop, p. 19-26, in G. F. Vande Woude et al., ed., Cancer Cells, vol. 2, 1984; R. C. Parker, H. E. Varmus, and J. M. Bishop, Cell 37:131-139, 1984; D. Shalloway, P. M. Coussens, and P. Yaciuk, p. 9-17, in G. F. Vande Woude et al., ed., Cancer Cells, vol. 2, 1984; D. Shalloway, P. M. Coussens, and P. Yaciuk, Proc. Natl. Acad. Sci. U.S.A. 81:7071-7075; and K. C. Wilhelmsen, W. G. Tarpley, and H. M. Temin, p. 303-308, in G. F. Vande Woude et al., ed., Cancer Cells, vol. 2, 1984), we found that both types of Moloney murine leukemia virus long terminal repeat-src expression plasmids induced focus formation, although c-src induced only 1% as many foci as v-src. The focus-selected c-src overexpressed cells had altered morphology and limited growth in soft agarose but were not tumorigenic in vivo. Cleveland digests, comparative in vitro kinase assays, secondary transfections, and immunoprecipitations indicated that focus formation was caused by rare transfection events that resulted in very high-level pp60c-src expression rather than by mutations of the transfected c-src genes. These results suggest that pp60v-src induced transformation is not a completely spurious activity which is unrelated to the function of pp60c-src but that it represents a perturbation of already existent molecular control processes involving pp60c-src.
Article
We constructed a mutant, called RSV-SF2, at the ATP-binding site of pp60v-src. In this mutant, lysine-295 is replaced with methionine. SF2 pp60v-src was found to have a half-life similar to that of wild-type pp60v-src and was localized in the membranous fraction of the cell. Rat cells expressing SF2 pp60v-src were morphologically untransformed and do not form tumors. The SF2 pp60v-src isolated from these cells lacked kinase activity with either specific immunoglobulin or other substrates, and expression of SF2 pp60v-src failed to cause an increase of total phosphotyrosine in the proteins of infected cells. Wild-type pp60v-src was phosphorylated on serine and tyrosine in infected cells, and the analogous phosphorylations could also be carried out in vitro. Phosphorylation of serine was catalyzed by a cyclic AMP-dependent protein kinase, and phosphorylation of tyrosine was perhaps catalyzed by pp60v-src itself. By contrast, SF2 pp60v-src could not be phosphorylated on serine or tyrosine either in infected cells or in vitro. These findings strengthen the belief that the phosphotransferase activity of pp60v-src is required for neoplastic transformation by the protein and suggest that the binding of ATP to pp60v-src elicits an allosteric change required for phosphorylation of serine in the protein.
Article
We have previously found that Rous sarcoma virus variants in which the viral src (v-src) gene is replaced by the cellular src (c-src) gene have no transforming activity. In this study, we analyzed the basis for the inability of the p60c-src overproduced by these variants to transform cells. Phosphorylations of tyrosine residues in total cell protein or in cellular 34K protein are known to be markedly enhanced upon infection with wild-type Rous sarcoma virus. We found that these tyrosine phosphorylations were only slightly increased in the c-src-containing virus-infected cells, whereas both levels were significantly increased by infection with wild-type Rous sarcoma virus, or transforming mutant viruses which are derived from c-src-containing viruses by spontaneous mutation. Phosphorylation at tyrosine 416 of p60 itself was also extremely low in overproduced p60c-src and high in p60s of transforming mutant viruses. In immunoprecipitates with monoclonal antibody, the overproduced p60c-src had much lower casein tyrosine kinase activity than did p60v-src. We previously showed that p60 myristylation and plasma membrane localization may be required for cell transformation. p60c-src was similar to transforming p60s in these properties. These results strongly suggest that the low level of tyrosine phosphorylation by overproduced p60c-src accounts for its inability to transform cells.
Article
The products of the viral and cellular src genes, p60v-src and p60c-src, appear to be composed of multiple functional domains. Highly conserved regions called src homology 2 and 3 (SH2 and SH3), comprising amino acid residues 88 to 250, are believed to modulate the protein-tyrosine kinase activity present in the carboxy-terminal halves of the src proteins. To explore the functions of these regions more fully, we have made 34 site-directed mutations in a transformation-competent c-src gene encoding phenylalanine in place of tyrosine 527 (Y527F c-src). Twenty of the new mutations change only one or two amino acids, and the remainder delete small or large portions of the SH2-SH3 region. These mutant alleles have been incorporated into a replication-competent Rous sarcoma virus vector to examine the biochemical and biological properties of the mutant proteins after infection of chicken embryo fibroblasts. Four classes of mutant proteins were observed: class 1, mutants with only slight differences from the parental gene products; class 2, mutant proteins with diminished transforming and specific kinase activities; class 3, mutant proteins with normal or enhanced specific kinase activity but impaired biological activity, often as a consequence of instability; and class 4, mutant proteins with augmented biological and catalytic activities. In general, there was a strong correlation between total kinase activity (or amounts of intracellular phosphotyrosine-containing proteins) and transforming activity. Deletion mutations and some point mutations affecting residues 109 to 156 inhibited kinase and transforming functions, whereas deletions affecting residues 187 to 226 generally had positive effects on one or both of those functions, confirming that SH2-SH3 has complex regulatory properties. Five mutations that augmented the transforming and kinase activities of Y527F c-src [F172P, R175L, delta(198-205), delta(206-226), and delta(176-226)] conferred transformation competence on an otherwise normal c-src gene, indicating that mutations in SH2 (like previously described lesions in SH3, the kinase domain, and a carboxy-terminal inhibitory domain) can activate c-src.
Article
E6-AP is a 100-kDa cellular protein that interacts with the E6 protein of the cancer-associated human papillomavirus types 16 and 18. The E6/E6-AP complex binds to and targets the p53 tumor-suppressor protein for ubiquitin-mediated proteolysis. E6-AP is an E3 ubiquitin-protein ligase which accepts ubiquitin from an E2 ubiquitin-conjugating enzyme in the form of a thioester and then directly transfers the ubiquitin to targeted substrates. The amino acid sequence of E6-AP shows similarity to a number of protein sequences over an approximately 350-aa region corresponding to the carboxyl termini of both E6-AP and the E6-AP-related proteins. Of particular note is a conserved cysteine residue within the last 32-34 aa, which in E6-AP is likely to be the site of ubiquitin thioester formation. Two of the E6-AP-related proteins, a rat 100-kDa protein and a yeast 95-kDa protein (RSP5), both of previously unknown function, are shown here to form thioesters with ubiquitin. Mutation of the conserved cysteine residue of these proteins destroys their ability to accept ubiquitin. These data strongly suggest that the rat 100-kDa protein and RSP5, as well as the other E6-AP-related proteins, belong to a class of functionally related E3 ubiquitin-protein ligases, defined by a domain homologous to the E6-AP carboxyl terminus (hect domain).
Article
The activity of the proto-oncogene encoded c-Src product is tightly regulated in vivo. In recent years, a model has emerged of how this regulation is achieved. In particular, protein kinases and phosphatases that are potential regulators of c-Src activity in the cell cycle have been identified and characterized.
Article
The protein-tyrosine kinase activity of the proto-oncogene product p60c-src is negatively regulated by the phosphorylation of a tyrosine residue close to the C terminus, tyrosine 527. The phosphorylation might be catalysed by a so-far-unidentified tyrosine kinase, distinct from p60c-src. Recently we purified a protein-tyrosine kinase that specifically phosphorylates tyrosine 527 of p60c-src from neonatal rat brain. We have now confirmed the specificity of this enzyme by using a mutant p60c-src that has a phenylalanine instead of tyrosine 527, and cloned a complementary DNA that encodes the enzyme. The enzyme is similar to kinases of the src family in that it has two conserved regions, Src-homology regions 2 and 3, upstream of a tyrosine kinase domain. The amino-acid identity of each region is no more than 47%, however, and the enzyme lacks phosphorylation sites corresponding to tyrosines 416 and 527 of p60c-src and has no myristylation signal. These results suggest that this protein-tyrosine kinase, which might negatively regulate p60c-src, represents a new type of tyrosine kinase.