共查询到20条相似文献,搜索用时 15 毫秒
1.
Heterogeneity of Mitochondrial Protein Biogenesis during Primary
Leaf Development in Barley 总被引:4,自引:1,他引:4 下载免费PDF全文
The natural developmental gradient of light-grown primary leaves of barley (Hordeum vulgare L.) was used to analyze the biogenesis of mitochondrial proteins in relation to the age and physiological changes within the leaf. The data indicate that the protein composition of mitochondria changes markedly during leaf development. Three distinct patterns of protein development were noted: group A proteins, consisting of the E1 β-subunit of the pyruvate dehydrogenase complex, ORF156, ORF577, alternative oxidase, RPS12, cytochrome oxidase subunits II and III, malic enzyme, and the α- and β-subunits of F1-ATPase; group B proteins, consisting of the E1 α-subunit of the pyruvate dehydrogenase complex, isocitrate dehydrogenase, HSP70A, cpn60C, and cpn60B; and group C proteins, consisting of the four subunits of the glycine decarboxylase complex (P, H, T, and L proteins), fumarase, and formate dehydrogenase. All of the proteins increased in concentration from the basal meristem to the end of the elongation zone (20.0 mm from the leaf base), whereupon group A proteins decreased, group B proteins increased to a maximum at 50 mm from the leaf base, and group C proteins increased to a maximum at the leaf tip. This study provides evidence of a marked heterogeneity of mitochondrial protein composition, reflecting a changing function as leaf cells develop photosynthetic and photorespiratory capacity. 相似文献
2.
Mitotic Phosphorylation of Golgi Reassembly Stacking Protein 55
by Mitogen-activated Protein Kinase ERK2 下载免费PDF全文
Stephen A. Jesch Timothy S. Lewis Natalie G. Ahn Adam D. Linstedt 《Molecular biology of the cell》2001,12(6):1811-1817
The role of the mitogen-activated protein kinase kinase (MKK)/extracellular-activated protein kinase (ERK) pathway in mitotic Golgi disassembly is controversial, in part because Golgi-localized targets have not been identified. We observed that Golgi reassembly stacking protein 55 (GRASP55) was phosphorylated in mitotic cells and extracts, generating a mitosis-specific phospho-epitope recognized by the MPM2 mAb. This phosphorylation was prevented by mutation of ERK consensus sites in GRASP55. GRASP55 mitotic phosphorylation was significantly reduced, both in vitro and in vivo, by treatment with U0126, a potent and specific inhibitor of MKK and thus ERK activation. Furthermore, ERK2 directly phosphorylated GRASP55 on the same residues that generated the MPM2 phospho-epitope. These results are the first demonstration of GRASP55 mitotic phosphorylation and indicate that the MKK/ERK pathway directly phosphorylates the Golgi during mitosis. 相似文献
3.
Calcium-Dependent Protein Phosphorylation May Mediate the
Gibberellic Acid Response in Barley Aleurone 总被引:6,自引:0,他引:6 下载免费PDF全文
Peptide substrates of well-defined protein kinases were microinjected into aleurone protoplasts of barley (Hordeum vulgare L. cv Himalaya) to inhibit, and therefore identify, protein kinase-regulated events in the transduction of the gibberellin (GA) and abscisic acid signals. Syntide-2, a substrate designed for Ca2+- and calmodulin (CaM)-dependent kinases, selectively inhibited the GA response, leaving constitutive and abscisic acid-regulated events unaffected. Microinjection of syntide did not affect the GA-induced increase in cytosolic [Ca2+], suggesting that it inhibited GA action downstream of the Ca2+ signal. When photoaffinity-labeled syntide-2 was electroporated into protoplasts and cross-linked to interacting proteins in situ, it selectively labeled proteins of approximately 30 and 55 kD. A 54-kD, soluble syntide-2 phosphorylating protein kinase was detected in aleurone cells. This kinase was activated by Ca2+ and was CaM independent, but was inhibited by the CaM antagonist N-(6-aminohexyl)-5-chloro-1-naphthalene-sulfonamide (250 μm), suggesting that it was a CaM-domain protein kinase-like activity. These results suggest that syntide-2 inhibits the GA response of the aleurone via an interaction with this kinase, implicating the 54-kD kinase as a Ca2+-dependent regulator of the GA response in these cells. 相似文献
4.
Roger W. Hunter Carol MacKintosh Ingeborg Hers 《The Journal of biological chemistry》2009,284(18):12339-12348
The elevation of [cAMP]i is an important mechanism of
platelet inhibition and is regulated by the opposing activity of adenylyl
cyclase and phosphodiesterase (PDE). In this study, we demonstrate that a
variety of platelet agonists, including thrombin, significantly enhance the
activity of PDE3A in a phosphorylation-dependent manner. Stimulation of
platelets with the PAR-1 agonist SFLLRN resulted in rapid and transient
phosphorylation of PDE3A on Ser312, Ser428,
Ser438, Ser465, and Ser492, in parallel with
the PKC (protein kinase C) substrate, pleckstrin. Furthermore, phosphorylation
and activation of PDE3A required the activation of PKC, but not of PI3K/PKB,
mTOR/p70S6K, or ERK/RSK. Activation of PKC by phorbol esters also resulted in
phosphorylation of the same PDE3A sites in a PKC-dependent, PKB-independent
manner. This was further supported by the finding that IGF-1, which strongly
activates PI3K/PKB, but not PKC, did not regulate PDE3A. Platelet activation
also led to a PKC-dependent association between PDE3A and 14-3-3 proteins. In
contrast, cAMP-elevating agents such as PGE1 and forskolin-induced
phosphorylation of Ser312 and increased PDE3A activity, but did not
stimulate 14-3-3 binding. Finally, complete antagonism of
PGE1-evoked cAMP accumulation by thrombin required both
Gi and PKC activation. Together, these results demonstrate that
platelet activation stimulates PKC-dependent phosphorylation of PDE3A on
Ser312, Ser428, Ser438, Ser465,
and Ser492 leading to a subsequent increase in cAMP hydrolysis and
14-3-3 binding.Upon vascular injury, platelets adhere to the newly exposed subintimal
collagen and undergo activation leading to platelet spreading to cover the
damaged region and release of thrombogenic factors such as ADP and thromboxane
A2. In addition, platelets are activated by thrombin, which is
generated as a result of activation of the coagulation pathway, and stimulates
platelets by cleaving the protease-activated receptors
(PAR),2
PAR-1 and PAR-4. The final common pathway is the exposure of fibrinogen
binding sites on integrin αIIbβ3 resulting in
platelet aggregation and thrombus formation.Thrombin-mediated cleavage of PARs leads to activation of phospholipase C
β (PLC), hydrolysis of phosphatidylinositol (PI) 4,5-bisphosphate and a
subsequent increase in [Ca2+]i and activation
of protein kinase C (PKC). Protein kinase C contributes to platelet activation
both directly, through affinity regulation of the fibrinogen receptor,
integrin αIIbβ3
(1), and indirectly by
enhancing degranulation (2).
Thrombin also stimulates activation of PI 3-kinases and subsequent generation
of PI (3,
4,
5) trisphosphate and PI
(3,
4) bisphosphate
(3), which recruit protein
kinase B (PKB) to the plasma membrane where it becomes phosphorylated and
activated.Platelet activation is opposed by agents that raise intracellular
3′-5′-cyclic adenosine monophosphate
([cAMP]i). cAMP is a powerful inhibitory second messenger
that down-regulates platelet function by interfering with Ca2+
homeostasis, degranulation and integrin activation
(4). Synthesis of cAMP is
stimulated by mediators such as prostaglandin I2 (PGI2),
which bind to Gs-coupled receptors leading to activation of
adenylate cyclase (AC). This inhibitory pathway is opposed by thrombin, which
inhibits the elevation of cAMP indirectly via autocrine activation of the
Gi-coupled ADP receptor P2Y12. cAMP signaling is
terminated by hydrolysis to biologically inert 5′-AMP by
3′-phosphodiesterases. Platelets express two cAMP phosphodiesterase
isoforms, cGMP-stimulated PDE2 and cGMP-inhibited PDE3A. PDE3A is the most
abundant isoform in platelets and has a ∼250-fold lower
Km for cAMP than PDE2
(4). As a consequence of these
properties, PDE3A exerts a greater influence on cAMP homeostasis, particularly
at resting levels. The importance of PDE3A in platelet function is further
emphasized by the finding that the PDE3A inhibitors cilostamide and milrinone
raise basal cAMP levels and strongly inhibit thrombin-induced platelet
activation (5). Furthermore,
PDE3A-/- mice demonstrate increased resting levels of platelet cAMP
and are protected against a model of pulmonary thrombosis
(6). In contrast, the PDE2
inhibitor EHNA has no significant effect on cAMP levels and platelet
aggregation (7,
8). The activity of PDE3A is
therefore essential to maintain low equilibrium levels of cAMP and determine
the threshold for platelet activation
(7).Like its paralogue PDE3B, it has recently become clear that PDE3A activity
can be positively regulated by phosphorylation in platelets and human oocytes
(9,
10). There is some evidence
that PKB may be involved in this regulation, although the phosphorylation
sites are poorly characterized. In contrast, phosphorylation of PDE3A in HeLa
cells was stimulated by phorbol esters and blocked by inhibitors of PKC
(11). In this study, we aimed
to identify the signaling pathways and phosphorylation sites that are involved
in regulation of platelet PDE3A. Here, we show strong evidence that PKC, and
not PKB, is involved in agonist-stimulated PDE3A phosphorylation on
Ser312, Ser428, Ser438, Ser465,
and Ser492, leading to an increase in PDE3A activity, 14-3-3
binding and modulation of intracellular cAMP levels. 相似文献
5.
YongQiang Wang Mingxiang Liao Nicholas Hoe Poulomi Acharya Changhui Deng Andrew N. Krutchinsky Maria Almira Correia 《The Journal of biological chemistry》2009,284(9):5671-5684
Cytochromes P450 (P450s) incur phosphorylation. Although the precise role
of this post-translational modification is unclear, marking P450s for
degradation is plausible. Indeed, we have found that after structural
inactivation, CYP3A4, the major human liver P450, and its rat orthologs are
phosphorylated during their ubiquitin-dependent proteasomal degradation.
Peptide mapping coupled with mass spectrometric analyses of CYP3A4
phosphorylated in vitro by protein kinase C (PKC) previously
identified two target sites, Thr264 and Ser420. We now
document that liver cytosolic kinases additionally target Ser478 as
a major site. To determine whether such phosphorylation is relevant to in
vivo CYP3A4 degradation, wild type and CYP3A4 with single, double, or
triple Ala mutations of these residues were heterologously expressed in
Saccharomyces cerevisiae pep4Δ strains. We found that relative
to CYP3A4wt, its S478A mutant was significantly stabilized in these yeast, and
this was greatly to markedly enhanced for its S478A/T264A, S478A/S420A, and
S478A/T264A/S420A double and triple mutants. Similar relative
S478A/T264A/S420A mutant stabilization was also observed in HEK293T cells. To
determine whether phosphorylation enhances CYP3A4 degradation by enhancing its
ubiquitination, CYP3A4 ubiquitination was examined in an in vitro
UBC7/gp78-reconstituted system with and without cAMP-dependent protein kinase
A and PKC, two liver cytosolic kinases involved in CYP3A4 phosphorylation.
cAMP-dependent protein kinase A/PKC-mediated phosphorylation of CYP3A4wt but
not its S478A/T264A/S420A mutant enhanced its ubiquitination in this system.
Together, these findings indicate that phosphorylation of CYP3A4
Ser478, Thr264, and Ser420 residues by
cytosolic kinases is important both for its ubiquitination and proteasomal
degradation and suggest a direct link between P450 phosphorylation,
ubiquitination, and degradation.Hepatic cytochromes P450
(P450s)3 are integral
endoplasmic reticulum (ER)-anchored hemoproteins engaged in the oxidative
biotransformation of various endo- and xenobiotics. Of these, human CYP3A4 is
the most dominant liver enzyme, accounting for >30% of the hepatic
microsomal P450 complement, and responsible for the oxidative metabolism of
over 50% of clinically relevant drugs
(1). In common with all the
other ER-bound P450s, CYP3A4 is a monotopic protein with its N-terminal
≈33-residue domain embedded in the ER membrane with the bulk of its
structure in the cytosol. Our in vivo studies of the heterologously
expressed CYP3A4 in the yeast Saccharomyces cerevisiae as well as of
its rat liver CYP3A2/3A23 orthologs in primary hepatocytes have revealed that
human and rat liver CYPs 3A are turned over via ubiquitin (Ub)-dependent
proteasomal degradation (UPD)
(2–8).
Thus, CYPs 3A represent excellent prototypic substrates of ER-associated
degradation (ERAD), specifically of the ERAD-C pathway
(6–11).
Consistent with this CYP3A ERAD process, our studies of in vivo
and/or in vitro reconstituted systems have led us to conclude that
CYPs 3A are ubiquitinated by the UBC7/gp78 Ub-ligase complex and recruited by
the p97-Npl4-Ufd1 complex before their degradation by the 26 S proteasome
(4–8,
12). Because all these
processes are energy-dependent, it is not surprising that in vitro
reconstitution of CYP3A4 UPD requires ATP. However, inclusion of
γ-S-[32P]ATP in an in vitro reconstituted
CYP3A4 ubiquitination system catalyzed by rat liver cytosolic fraction II
(FII) resulted in CYP3A4 protein phosphorylation, i.e.
γ-[32P]phosphoryl transfer onto CYP3A4 target residues
(13,
14). This phosphorylation was
enhanced after cumene hydroperoxide (CuOOH)-mediated CYP3A4 inactivation. The
physiological role, if any, of this CYP3A4 post-translational modification is
unclear.CYP3A4 is not the only P450 that is phosphorylated. Since the in
vitro phosphorylation of a hepatic P450 (CYP2B4) by cAMP-dependent
protein kinase A (PKA) was first described
(15), various P450s,
particularly those belonging to the subfamily 2, were documented to be
phosphorylated in cell-free systems, hepatocyte incubations, and intact
animals
(16–32).
Common features of such P450 phosphorylation were the presence of a
cytosolically exposed PKA recognition sequence (RRXS) with the Ser
residue as the exclusive kinase target, and the ensuing loss of prosthetic
heme, conversion to the inactive P420 species, and consequent dramatic
functional inactivation
(15–20).
Studies in intact rats also identified CYPs 3A and 2C6 as kinase targets
(21). Although both these
P450s lack the hallmark PKA recognition sequence, apparently they possess
secondary PKA targeting sequences or are phosphorylated by other protein
kinases such as PKC. Indeed, in vitro studies revealed that P450s
were phosphorylated in an isoform-dependent manner by either PKA or PKC,
except for CYP2B1, which was heavily phosphorylated by both
(20). Over the years since
this particular post-translational P450 modification was recognized, it has
been assigned various functional roles
(17,
29–33).
Among these, as first proposed by Taniguchi et al.
(16) and later explored both
by Eliasson et al.
(23–26)
and us (13,
14), P450 phosphorylation
served as a marker for its degradation. Accordingly, the phosphorylation of
CYP2E1Ser129 and CYP3A1Ser393 by a microsomal
cAMP-dependent protein kinase has been proposed to predispose these P450s but
not the similarly phosphorylated CYP2B1 to proteolytic degradation by an
integral ER Mg2+-ATP-activated serine protease
(23–27).
However, heterologous expression of CYP2E1S129A/S129G site-directed mutants in
COS7 cells apparently had no effect on its relative stability thereby
revealing that if CYP2E1 phosphorylation is important for its degradation
(34,
35), then alternate Ser/Thr
residues (i.e. in plausible secondary PKA recognition sites,
Lys-Lys-Ser209-Lys and Lys-Lys-Ser449-Ala) may be
recruited.On the other hand, on the basis of rapid phosphorylation of
CuOOH-inactivated CYP3A4 that precedes its ubiquitination and 26 S proteasomal
degradation in an in vitro liver cytosolic FII-catalyzed system, we
have proposed that CYP3A4 phosphorylation was essential for targeting it to
proteins participating in its UPD/ERAD
(13). Indeed, several examples
of similar phosphorylation for targeting proteins to UPD exist, of which
IκBα phosphorylation is the most notable and perhaps the best
documented
(36–47;
see “Discussion”).Our in vitro studies with specific kinase inhibitors as probes
identified both PKC and PKA as the major FII kinases responsible for CYP3A4
phosphorylation (14). Indeed,
in vitro model studies of CYP3A4 with PKC as the kinase, coupled with
lysylendopeptidase C (Lys-C) digestion of the phosphorylated protein and
liquid chromatography-tandem mass spectrometric (LC-MS/MS) analyses of the
Lys-C digests, identified two PKC-phosphorylated CYP3A4 peptides
258ESRLEDpTQK266 and
414FLPERFpSK421 unambiguously phosphorylated at
Thr264 and Ser420
(14). These same residues were
also phosphorylated in corresponding studies with
PKA.4 Furthermore,
although both native and CuOOH-inactivated CYP3A4 were phosphorylated at
Thr264, Ser420 phosphorylation was particularly enhanced
after CuOOH-mediated CYP3A4 inactivation
(14). Corresponding studies of
CuOOH-inactivated CYP3A4 using rat liver cytosolic FII as the source of the
kinase(s), revealed 32P phosphorylation of both these peptides as
well as that of an additional CYP3A4 peptide
477LS(p)LGGLLQPEKPVVLK492. Unlike the unambiguous mass
spectrometric identification of Thr264 and Ser420 as the
phosphorylated CYP3A4 residues, the phosphorylation of Ser478, the
only plausible phosphorylatable residue in this 32P-labeled
peptide, was not similarly established. Nevertheless, the predominant
phosphorylation of Thr264 in native CYP3A4
(14), but of two additional
residues in the CuOOH-inactivated enzyme, is consistent with the
inactivation-induced structural unraveling of this enzyme with exposure of
otherwise concealed and/or kinase-inaccessible domains
(48). Such unraveling of
CYP3A4 protein stems from the irreversible modification of its active site by
fragments generated from CuOOH-mediated oxidative destruction of its
prosthetic heme (49). In this
study, using mass spectrometric analyses of Lys-C digests of
FII-phosphorylated CYP3A4, we have provided unambiguous evidence that in
addition to Thr264 and Ser420, Ser478 is
indeed phosphorylated. More importantly, through alanine-scanning mutagenesis
of these three residues, we now document that although neither the structural
conformation nor the catalytic function of this triple CYP3A4T264A/S420A/S478A
mutant is altered, its degradation after heterologous expression in S.
cerevisiae is significantly impaired. This is also true of
CYP3A4T264A/S420A/S478A mutant degradation in human embryonic kidney (HEK293T)
cells. Furthermore, using an in vitro reconstituted CYP3A4
ubiquitination system, catalyzed by human Ub-conjugating E2 enzyme UBC7 and
integral ER protein gp78 as the E3 Ub ligase
(12), we document that
PKA/PKC-mediated phosphorylation of the wild type CYP3A4 (CYP3A4wt)
considerably enhanced its UBC7/gp78-mediated ubiquitination. Together these
findings reveal the critical importance of CYP3A4 phosphorylation at these
residues for its UPD and suggest a direct link between phosphorylation and its
ubiquitination and degradation. 相似文献
6.
James Sinnett-Smith Rodrigo Jacamo Robert Kui YunZu M. Wang Steven H. Young Osvaldo Rey Richard T. Waldron Enrique Rozengurt 《The Journal of biological chemistry》2009,284(20):13434-13445
Rapid protein kinase D (PKD) activation and phosphorylation via protein
kinase C (PKC) have been extensively documented in many cell types cells
stimulated by multiple stimuli. In contrast, little is known about the role
and mechanism(s) of a recently identified sustained phase of PKD activation in
response to G protein-coupled receptor agonists. To elucidate the role of
biphasic PKD activation, we used Swiss 3T3 cells because PKD expression in
these cells potently enhanced duration of ERK activation and DNA synthesis in
response to Gq-coupled receptor agonists. Cell treatment with the
preferential PKC inhibitors GF109203X or Gö6983 profoundly inhibited PKD
activation induced by bombesin stimulation for <15 min but did not prevent
PKD catalytic activation induced by bombesin stimulation for longer times
(>60 min). The existence of sequential PKC-dependent and PKC-independent
PKD activation was demonstrated in 3T3 cells stimulated with various
concentrations of bombesin (0.3–10 nm) or with vasopressin, a
different Gq-coupled receptor agonist. To gain insight into the
mechanisms involved, we determined the phosphorylation state of the activation
loop residues Ser744 and Ser748. Transphosphorylation
targeted Ser744, whereas autophosphorylation was the predominant
mechanism for Ser748 in cells stimulated with Gq-coupled
receptor agonists. We next determined which phase of PKD activation is
responsible for promoting enhanced ERK activation and DNA synthesis in
response to Gq-coupled receptor agonists. We show, for the first
time, that the PKC-independent phase of PKD activation mediates prolonged ERK
signaling and progression to DNA synthesis in response to bombesin or
vasopressin through a pathway that requires epidermal growth factor
receptor-tyrosine kinase activity. Thus, our results identify a novel
mechanism of Gq-coupled receptor-induced mitogenesis mediated by
sustained PKD activation through a PKC-independent pathway.The understanding of the mechanisms that control cell proliferation
requires the identification of the molecular pathways that govern the
transition of quiescent cells into the S phase of the cell cycle. In this
context the activation and phosphorylation of protein kinase D
(PKD),4 the founding
member of a new protein kinase family within the
Ca2+/calmodulin-dependent protein kinase (CAMK) group and separate
from the previously identified PKCs (for review, see Ref.
1), are attracting intense
attention. In unstimulated cells, PKD is in a state of low catalytic (kinase)
activity maintained by autoinhibition mediated by the N-terminal domain, a
region containing a repeat of cysteinerich zinc finger-like motifs and a
pleckstrin homology (PH) domain
(1–4).
Physiological activation of PKD within cells occurs via a
phosphorylation-dependent mechanism first identified in our laboratory
(5–7).
In response to cellular stimuli
(1), including phorbol esters,
growth factors (e.g. PDGF), and G protein-coupled receptor (GPCR)
agonists (6,
8–16)
that signal through Gq, G12, Gi, and Rho
(11,
15–19),
PKD is converted into a form with high catalytic activity, as shown by in
vitro kinase assays performed in the absence of lipid co-activators
(5,
20).During these studies multiple lines of evidence indicated that PKC activity
is necessary for rapid PKD activation within intact cells. For example, rapid
PKD activation was selectively and potently blocked by cell treatment with
preferential PKC inhibitors (e.g. GF109203X or Gö6983) that do
not directly inhibit PKD catalytic activity
(5,
20), implying that PKD
activation in intact cells is mediated directly or indirectly through PKCs.
Many reports demonstrated the operation of a rapid PKC/PKD signaling cascade
induced by multiple GPCR agonists and other receptor ligands in a range of
cell types (for review, see Ref.
1). Our previous studies
identified Ser744 and Ser748 in the PKD activation loop
(also referred as activation segment or T-loop) as phosphorylation sites
critical for PKC-mediated PKD activation
(1,
4,
7,
17,
21). Collectively, these
findings demonstrated the existence of a rapidly activated PKC-PKD protein
kinase cascade(s). In a recent study we found that the rapid PKC-dependent PKD
activation was followed by a late, PKC-independent phase of catalytic
activation and phosphorylation induced by stimulation of the bombesin
Gq-coupled receptor ectopically expressed in COS-7 cells
(22). This study raised the
possibility that PKD mediates rapid biological responses downstream of PKCs,
whereas, in striking contrast, PKD could mediate long term responses through
PKC-independent pathways. Despite its potential importance for defining the
role of PKC and PKD in signal transduction, this hypothesis has not been
tested in any cell type.Accumulating evidence demonstrates that PKD plays an important role in
several cellular processes and activities, including signal transduction
(14,
23–25),
chromatin organization (26),
Golgi function (27,
28), gene expression
(29–31),
immune regulation (26), and
cell survival, adhesion, motility, differentiation, DNA synthesis, and
proliferation (for review, see Ref.
1). In Swiss 3T3 fibroblasts, a
cell line used extensively as a model system to elucidate mechanisms of
mitogenic signaling
(32–34),
PKD expression potently enhances ERK activation, DNA synthesis, and cell
proliferation induced by Gq-coupled receptor agonists
(8,
14). Here, we used this model
system to elucidate the role and mechanism(s) of biphasic PKD activation.
First, we show that the Gq-coupled receptor agonists bombesin and
vasopressin, in contrast to phorbol esters, specifically induce PKD activation
through early PKC-dependent and late PKC-independent mechanisms in Swiss 3T3
cells. Subsequently, we demonstrate for the first time that the
PKC-independent phase of PKD activation is responsible for promoting ERK
signaling and progression to DNA synthesis through an epidermal growth factor
receptor (EGFR)-dependent pathway. Thus, our results identify a novel
mechanism of Gq-coupled receptor-induced mitogenesis mediated by
sustained PKD activation through a PKC-independent pathway. 相似文献
7.
Marlise I. Klein Jin Xiao Bingwen Lu Claire M. Delahunty John R. Yates III Hyun Koo 《PloS one》2012,7(9)
Biofilms formed on tooth surfaces are comprised of mixed microbiota enmeshed in an extracellular matrix. Oral biofilms are constantly exposed to environmental changes, which influence the microbial composition, matrix formation and expression of virulence. Streptococcus mutans and sucrose are key modulators associated with the evolution of virulent-cariogenic biofilms. In this study, we used a high-throughput quantitative proteomics approach to examine how S. mutans produces relevant proteins that facilitate its establishment and optimal survival during mixed-species biofilms development induced by sucrose. Biofilms of S. mutans, alone or mixed with Actinomyces naeslundii and Streptococcus oralis, were initially formed onto saliva-coated hydroxyapatite surface under carbohydrate-limiting condition. Sucrose (1%, w/v) was then introduced to cause environmental changes, and to induce biofilm accumulation. Multidimensional protein identification technology (MudPIT) approach detected up to 60% of proteins encoded by S. mutans within biofilms. Specific proteins associated with exopolysaccharide matrix assembly, metabolic and stress adaptation processes were highly abundant as the biofilm transit from earlier to later developmental stages following sucrose introduction. Our results indicate that S. mutans within a mixed-species biofilm community increases the expression of specific genes associated with glucan synthesis and remodeling (gtfBC, dexA) and glucan-binding (gbpB) during this transition (P<0.05). Furthermore, S. mutans up-regulates specific adaptation mechanisms to cope with acidic environments (F1F0-ATPase system, fatty acid biosynthesis, branched chain amino acids metabolism), and molecular chaperones (GroEL). Interestingly, the protein levels and gene expression are in general augmented when S. mutans form mixed-species biofilms (vs. single-species biofilms) demonstrating fundamental differences in the matrix assembly, survival and biofilm maintenance in the presence of other organisms. Our data provide insights about how S. mutans optimizes its metabolism and adapts/survives within the mixed-species community in response to a dynamically changing environment. This reflects the intricate physiological processes linked to expression of virulence by this bacterium within complex biofilms. 相似文献
8.
Yonghua Liu Youhua Wang Ying Chen Xiaohong Li Jiao Yang Yang Liu Aiguo Shen 《The Journal of biological chemistry》2015,290(22):13888-13894
Axon loss is a destructive consequence of a wide range of neurological diseases without a clearly defined mechanism. Recent data demonstrate that SCG10 is a novel axonal maintenance factor and that rapid SCG10 loss after injury requires JNK activity; how JNK induces degradation of SCG10 is not well known. Here we showed that SCG10 was a binding partner of Spy1, a Speedy/RINGO family protein, which participated in cellular response to sciatic nerve injury. During the early stage of axonal injury, Spy1 expression was inversely correlated with SCG10. Spy1 mediated SCG10 phosphorylation and degradation partly in a JNK-dependent manner. Inhibition of Spy1 attenuated SCG10 phosphorylation and delayed injury-induced axonal degeneration. Taken together, these data suggest that Spy1 is an important regulator of SCG10 and can be targeted in future axo-protective therapeutics. 相似文献
9.
Yan Li Yiwei Cheng Tianyu Zhu Hao Zhang Wen Li Yueshuai Guo Yaling Qi Xu Chen Jun Zhang Jiahao Sha Zuomin Zhou Hui Zhu Xuejiang Guo 《Proteomics》2019,19(11)
The characteristic tadpole shape of sperm is formed from round spermatids via spermiogenesis, a process which results in dramatic morphological changes in the final stage of spermatogenesis in the testis. Protein phosphorylation, as one of the most important post‐translational modifications, can regulate spermiogenesis; however, the phosphorylation events taking place during this process have not been systematically analyzed. In order to better understand the role of phosphorylation in spermiogenesis, large‐scale phosphoproteome profiling is performed using IMAC and TiO2 enrichment. In total, 13 835 phosphorylation sites, in 4196 phosphoproteins, are identified in purified mouse spermatids undergoing spermiogenesis in two biological replicates. Overall, 735 testis‐specific proteins are identified to be phosphorylated, and are expressed at high levels during spermiogenesis. Gene ontology analysis shows enrichment of the identified phosphoproteins in terms of histone modification, cilium organization, centrosome and the adherens junction. Further characterization of the kinase‐substrate phosphorylation network demonstrates enrichment of phosphorylation substrates related to the regulation of spermiogenesis. This global protein phosphorylation landscape of spermiogenesis shows wide phosphoregulation across a diverse range of processes during spermiogenesis and can help to further characterize the process of sperm generation. All MS data are available via ProteomeXchange with the identifier PXD011890. 相似文献
10.
Michel Varrin-Doyer Peggy Vincent Sylvie Cavagna Nathalie Auvergnon Nelly Noraz Véronique Rogemond Jér?me Honnorat Mahnaz Moradi-Améli Pascale Giraudon 《The Journal of biological chemistry》2009,284(19):13265-13276
In the central nervous system, collapsin response mediator protein 2 (CRMP2) is a transducer protein that supports the semaphorin-induced guidance of axons toward their cognate target. However, we previously showed that CRMP2 is also expressed in immune cells and plays a crucial role in T lymphocyte migration. Here we further investigated the molecular mechanisms underlying CRMP2 function in chemokine-directed T-cell motility. Examining Jurkat T-cells treated with the chemokine CXCL12, we found that 1) CXCL12 induces a dynamic re-localization of CRMP2 to uropod, the flexible structure of migrating lymphocyte, and increases its binding to the cytoskeletal protein vimentin; 2) CXCL12 decreases phosphorylation of the glycogen synthase kinase-3β-targeted residues CRMP2-Thr-509/514; and 3) tyrosine Tyr-479 is a new phosphorylation CRMP2 residue and a target for the Src-family kinase Yes. Moreover, phospho-Tyr-479 increased under CXCL12 signaling while phospho-Thr-509/514 decreased. The functional importance of this tyrosine phosphorylation was demonstrated by Y479F mutation that strongly reduced CXCL12-mediated T-cell polarization and motility as tested in a transmigration model and on neural tissue. We propose that differential phosphorylation by glycogen synthase kinase-3β and Yes modulates the contribution of CRMP2 to cytoskeletal reorganization during chemokine-directed T-cell migration. In addition to providing a novel mechanism for T lymphocyte motility, our findings reveal CRMP2 as a transducer of chemokine signaling.T lymphocyte migration is the basis of major immune functions such as responses to infection and inflammation, as well as normal recirculation through the lymphoid organs. Indeed, the role of T-cells depends strongly on their ability to travel between organs via the blood and lymph and to move rapidly within these tissues, by extravasation (1). This latter function is dependent on extracellular signals, among which chemokines play a major role.Chemokines form a superfamily of small proteins that orchestrate lymphocyte polarization and migration (2). These proteins exert their functions by binding specific seven-transmembrane-domain G-protein-coupled receptors on the T-cell surface (3). T-lymphocytes exposed to chemokines, in a soluble or surface-bound gradient, develop a polarized shape, extending at the front, an F-actin-rich lamellipodium, which constitutes the leading edge, and a trailing edge or uropod in which both the microtubule and vimentin networks are retracted during migration. Although F-actin has the well known function of producing the mechanical forces required to generate movement (4), the role of microtubules and vimentin in T-cell migration requires further investigation.Cytoskeletal remodeling is of key importance in migrating cells (5) and is one of the functions carried out by the chemokine stromal cell-derived factor-1α, also named CXCL12. In association with its cognate receptor CXCR4, CXCL12 is a potent chemoattractant for mature T-cells and monocytes (6). Following ligand recognition and binding, CXCR4 signaling starts with the activation of G proteins, followed by various signaling cascade effectors, including MAP2 kinases, phosphoinositide 3-kinase, and phospholipase Cγ (7). Although this intracellular signaling cascade has not been completely elucidated, the Src family non-receptor tyrosine kinase Lck and the Syk kinase ZAP-70 have emerged as the main candidates for delivering the input signal following CXCR4 activation (8). Thus, tyrosine kinase activity appears as a central step in CXCR4-dependent chemotaxis.While searching for molecules involved in T-cell motility, we recently identified collapsin response mediator protein 2 (CRMP2) (9), a protein first described in the context of neuronal growth cone advance (10, 11). We demonstrated that CRMP2 regulated both T-cell polarization and spontaneous/chemokine-induced migration of T-lymphocytes. Moreover, CRMP2 was found at the uropod of motile T-cells and has the ability to bind cytoskeletal elements, including vimentin. A correlation between CRMP2 expression levels and cell migratory rates toward a chemokine gradient, including CXCL12, was demonstrated by overexpression and knockdown experiments in T-cells (9). In addition, we recently reported that, in mouse model of neuroinflammation, elevated CRMP2 expression in T lymphocytes correlated with their elevated migratory rates and their ability to target the central nervous system (12). The importance of CXCL12 in the central nervous system and its implication in the pathogenesis of central nervous system disorders, including neuroinflammatory diseases, are well documented (review in Ref. 13). Thus, the aim of the present study was to determine whether and how CRMP2 participates in the transduction pathway induced by CXCL12 on T lymphocytes. 相似文献
11.
12.
Amanda J. Bell Timothy J. Satchwell Kate J. Heesom Bethan R. Hawley Sabine Kupzig Matthew Hazell Rosey Mushens Andrew Herman Ashley M. Toye 《PloS one》2013,8(4)
Enucleation is the step in erythroid terminal differentiation when the nucleus is expelled from developing erythroblasts creating reticulocytes and free nuclei surrounded by plasma membrane. We have studied protein sorting during human erythroblast enucleation using fluorescence activated cell sorting (FACS) to obtain pure populations of reticulocytes and nuclei produced by in vitro culture. Nano LC mass spectrometry was first used to determine the protein distribution profile obtained from the purified reticulocyte and extruded nuclei populations. In general cytoskeletal proteins and erythroid membrane proteins were preferentially restricted to the reticulocyte alongside key endocytic machinery and cytosolic proteins. The bulk of nuclear and ER proteins were lost with the nucleus. In contrast to the localization reported in mice, several key erythroid membrane proteins were detected in the membrane surrounding extruded nuclei, including band 3 and GPC. This distribution of key erythroid membrane and cytoskeletal proteins was confirmed using western blotting. Protein partitioning during enucleation was investigated by confocal microscopy with partitioning of cytoskeletal and membrane proteins to the reticulocyte observed to occur at a late stage of this process when the nucleus is under greatest constriction and almost completely extruded. Importantly, band 3 and CD44 were shown not to restrict specifically to the reticulocyte plasma membrane. This highlights enucleation as a stage at which excess erythroid membrane proteins are discarded in human erythroblast differentiation. Given the striking restriction of cytoskeleton proteins and the fact that membrane proteins located in macromolecular membrane complexes (e.g. GPA, Rh and RhAG) are segregated to the reticulocyte, we propose that the membrane proteins lost with the nucleus represent an excess mobile population of either individual proteins or protein complexes. 相似文献
13.
14.
Adriana A. Paulucci-Holthauzen Leoncio A. Vergara Larry J. Bellot David Canton John D. Scott Kathleen L. O'Connor 《The Journal of biological chemistry》2009,284(9):5956-5967
Protein kinase A (PKA) has been suggested to be spatially regulated in
migrating cells due to its ability to control signaling events that are
critical for polarized actin cytoskeletal dynamics. Here, using the
fluorescence resonance energy transfer-based A-kinase activity reporter
(AKAR1), we find that PKA activity gradients form with the strongest activity
at the leading edge and are restricted to the basal surface in migrating
cells. The existence of these gradients was confirmed using
immunocytochemistry using phospho-PKA substrate antibodies. This observation
holds true for carcinoma cells migrating randomly on laminin-1 or stimulated
to migrate on collagen I with lysophosphatidic acid. Phosphodiesterase
inhibition allows the formation of PKA activity gradients; however, these
gradients are no longer polarized. PKA activity gradients are not detected
when a non-phosphorylatable mutant of AKAR1 is used, if PKA activity is
inhibited with H-89 or protein kinase inhibitor, or when PKA anchoring is
perturbed. We further find that a specific A-kinase anchoring protein,
AKAP-Lbc, is a major contributor to the formation of these gradients. In
summary, our data show that PKA activity gradients are generated at the
leading edge of migrating cells and provide additional insight into the
mechanisms of PKA regulation of cell motility.Cell motility is controlled by a complex network of signals that are
initiated by binding to the extracellular matrix. Understanding the
biochemical mechanisms that control cell migration is necessary for better
comprehension of processes like wound healing, embryonic development, and
angiogenesis as well as cancer metastasis
(1).
PKA3 is an important
regulator of cell signaling and various biological functions
(2-4).
Previous studies have shown that cell motility is delicately controlled by
synthesis and breakdown of cAMP through its effects on PKA. PKA regulates key
signaling events that are critical for actin cytoskeletal remodeling and cell
polarization during migration, including control of the activation states of
RhoA, Rac, cdc42, Pak, and c-Abl. For example, PKA is known to inhibit the
activation of RhoA, whereas it is required for the activation of Rac1, two
proteins that are spatially regulated during cell migration. Therefore, it has
been suggested that PKA activity in migrating cells is spatially regulated
(5-9).
The mounting evidence for the formation of cAMP/PKA gradients and their
influence over directed cell motility is compelling. To conclusively determine
that PKA activity gradients exist, the visualization of these gradients in
single cells is needed to determine the nature of gradients and the mechanisms
governing how they are formed.The compartmental action of cAMP was suggested over three decades ago
(10,
11) and has hence been shown
to mediate the precise spatiotemporal control of its effectors
(12-15).
Tight control of cAMP levels is governed by the coordinated actions of cyclic
nucleotide phosphodiesterases (PDEs) and adenylyl cyclases. Gradients of cAMP
and, thus, PKA activity are expected to exist in a cell. This idea is based,
most simplistically, on the fact that cAMP is generated by membrane-bound
adenylyl cyclases and broken down by cytosolic PDEs; that is, the two arms of
cAMP metabolism are spatially separated. Further compartmentalization of PKA
activity also occurs as a result of the anchoring of PKA and cAMP-specific
PDEs to A-kinase anchoring proteins (AKAPs), which has been demonstrated in a
variety of cell types (16,
17). The anchoring of PKA
occurs typically through the binding of the type II regulatory (RII) subunits
to AKAPs where the relative levels of PDE activity and cAMP generated regulate
the regional activity of PKA. PKA anchoring, in addition to cAMP synthesis and
degradation, is believed to control spatial signaling of PKA
(14,
15). Until recently, we have
lacked both the model systems and technology to adequately study the
possibility that cAMP/PKA activity gradients exist. We and others
(5-8)
have established that polarization and migration of cells are dependent on
cAMP synthesis and breakdown. Here, we sought to demonstrate the existence of
cAMP/PKA gradients in single migrating cells using the fluorescence resonance
energy transfer (FRET)-based PKA biosensor A-kinase activity reporter (AKAR1)
and determine how signaling components that regulate PKA activity, including
cAMP synthesis, PDEs, and PKA anchoring, affect the formation of these
gradients. 相似文献
15.
Capillaries in vertebrate brain have unique permeability properties that make up the blood-brain barrier (BBB). Although it is known that capillaries are innervated by nerve endings of intracerebral origin and that brain capillary function is likely acutely regulated by neuronal inputs, the possible mechanisms of neuronal regulation of capillary function are at present unknown. One possible mode of regulation is via the phosphorylation of brain capillary proteins. The present studies characterize, for the first time, the major phosphoproteins in the bovine brain capillary using both intact bovine brain capillaries and plasma membrane fractions from bovine brain capillaries. The patterns of endogenous phosphorylation of capillary proteins are compared to similar patterns obtained with synaptosomal (P2) fractions from bovine brain. The major findings of this study are: (a) The activity of protein phosphorylation in brain capillaries is localized almost exclusively to the capillary plasma membrane, and is nearly comparable to the activity of protein phosphorylation in synaptosomal membranes. (b) A major phosphoprotein doublet in the capillary fraction comigrates on a sodium dodecyl sulfate gel with a major phosphoprotein doublet of approximate molecular weight of 80K in the synaptosomal fraction, and the latter is presumed to be synapsin I; in dephosphorylation assays the synaptosomal 80K phosphoprotein doublet is not subject to measurable dephosphorylation, whereas the capillary 80K doublet is subject to rapid dephosphorylation, and is essentially completely dephosphorylated within 5 s at 0 degrees C. (c) A prominent triplet of phosphoproteins with molecular weight of 50-55K is present in the capillary fraction, and is not present in the synaptosomal fraction; thus, this 50-55K triplet of phosphoproteins appears specific for brain capillaries.(ABSTRACT TRUNCATED AT 250 WORDS) 相似文献
16.
Sarah B. Scruggs Aaron C. Hinken Ariyaporn Thawornkaiwong Jeffrey Robbins Lori A. Walker Pieter P. de Tombe David L. Geenen Peter M. Buttrick R. John Solaro 《The Journal of biological chemistry》2009,284(8):5097-5106
There is little direct evidence on the role of myosin regulatory light
chain phosphorylation in ejecting hearts. In studies reported here we
determined the effects of regulatory light chain (RLC) phosphorylation on
in situ cardiac systolic mechanics and in vitro myofibrillar
mechanics. We compared data obtained from control nontransgenic mice (NTG)
with a transgenic mouse model expressing a cardiac specific
nonphosphorylatable RLC (TG-RLC(P-). We also determined whether the depression
in RLC phosphorylation affected phosphorylation of other sarcomeric proteins.
TG-RLC(P-) demonstrated decreases in base-line load-independent measures of
contractility and power and an increase in ejection duration together with a
depression in phosphorylation of myosin-binding protein-C (MyBP-C) and
troponin I (TnI). Although TG-RLC(P-) displayed a significantly reduced
response to β1-adrenergic stimulation, MyBP-C and TnI were
phosphorylated to a similar level in TG-RLC(P-) and NTG, suggesting
cAMP-dependent protein kinase signaling to these proteins was not disrupted. A
major finding was that NTG controls were significantly phosphorylated at RLC
serine 15 following β1-adrenergic stimulation, a mechanism
prevented in TG-RLC(P-), thus providing a biochemical difference in
β1-adrenergic responsiveness at the level of the sarcomere.
Our measurements of Ca2+ tension and Ca2+-ATPase rate
relations in detergent-extracted fiber bundles from LV trabeculae demonstrated
a relative decrease in maximum Ca2+-activated tension and tension
cost in TG-RLC(P-) fibers, with no change in Ca2+ sensitivity. Our
data indicate that RLC phosphorylation is critical for normal ejection and
response to β1-adrenergic stimulation. Our data also indicate
that the lack of RLC phosphorylation promotes compensatory changes in MyBP-C
and TnI phosphorylation, which when normalized do not restore function.Phosphorylation of sarcomeric proteins tunes the intensity and dynamics of
cardiac contraction and relaxation independent of membrane Ca2+
fluxes to meet physiologic demands
(1,
2). We focus here on
ventricular myosin regulatory light chain, which is phosphorylated in
vivo
(3–5)
but whose functional role in control of cardiac dynamics has remained unclear.
The identification of
RLC2 mutations linked
to familial hypertrophic cardiomyopathy
(6) underscores the importance
of understanding its action as a regulator of contraction. Functionally,
in vitro cardiac RLC phosphorylation by MLCK produces a sensitizing
shift in the force-Ca2+ relation in skinned fibers
(7–11).
Moreover, studies show that RLC phosphorylation manifests as a gradient across
the wall of the heart, which may be important for both normalizing wall stress
and for generation of torsion about the long axis of the ejecting heart
(12–14).
Yet there remains a lack of understanding of the in situ functional
effects of RLC phosphorylation and whether phosphorylation of RLC influences
other sarcomeric sites as substrates for kinases and phosphatases.Understanding the precise mechanisms by which phosphorylation of RLC
affects function of ejecting ventricles is particularly important, because
mechanisms downstream of Ca2+ fluxes at the level of the sarcomere
appear to dominate ejection and to sustain ventricular elastance
(15). Myosin motors are
important in this, and RLC is well positioned at the S1-S2 junction to
modulate myosin heavy chain directly by fine-tuning lever arm motion and
indirectly by interacting with the essential light chain, the thick filament
backbone, and MyBP-C (16,
17). Accordingly, the
hypothesis underlying this study was that ablation of N-terminal RLC
phosphorylation would elicit a depression in ventricular ejection and
compensatory changes in phosphorylation of sarcomeric proteins neighboring
RLC.To understand the role of RLC phosphorylation in the ejection phase of the
cardiac cycle, we determined in situ pressure-volume functions in
ejecting, auxotonically loaded ventricles expressing either wild type RLC
(NTG) or a nonphosphorylatable RLC (TG-RLC(P-))
(10). Our experiments provide
novel data demonstrating the importance of RLC phosphorylation in systolic
pump function and provide new insights into how a lack of phosphorylation of
RLC induces a redistribution of charge among myofilament proteins.
Furthermore, our data demonstrate an enigmatic blunting of TG-RLC(P-)
functional response to β1-adrenergic simulation despite a
normal TnI and MyBP-C phosphorylation profile. RLC serine 15 phosphorylation
increased significantly in NTG controls but was not permitted in TG-RLC(P-)
(RLC S14/15/19/A), suggesting that a change in RLC phosphorylation following
β1-adrenergic simulation may be critical for eliciting a
normal response. 相似文献
17.
18.
19.
20.
Muhammad Saeed Florian Schwarze Adele Loidl Joachim Meraner Markus Lechner Peter Loidl 《PloS one》2012,7(9)
The retinoblastoma protein (pRb) and the related proteins Rb2/p130 and 107 represent the “pocket protein” family of cell cycle regulators. A key function of these proteins is the cell cycle dependent modulation of E2F-regulated genes. The biological activity of these proteins is controlled by acetylation and phosphorylation in a cell cycle dependent manner. In this study we attempted to investigate the interdependence of acetylation and phosphorylation of Rb2/p130 in vitro. After having identified the acetyltransferase p300 among several acetyltransferases to be associated with Rb2/p130 during S-phase in NIH3T3 cells in vivo, we used this enzyme and the CDK4 protein kinase for in vitro modification of a variety of full length Rb2/p130 and truncated versions with mutations in the acetylatable lysine residues 1079, 128 and 130. Mutation of these residues results in the complete loss of Rb2/p130 acetylation. Replacement of lysines by arginines strongly inhibits phosphorylation of Rb2/p130 by CDK4; the inhibitory effect of replacement by glutamines is less pronounced. Preacetylation of Rb2/p130 strongly enhances CDK4-catalyzed phosphorylation, whereas deacetylation completely abolishes in vitro phosphorylation. In contrast, phosphorylation completely inhibits acetylation of Rb2/p130 by p300. These results suggest a mutual interdependence of modifications in a way that acetylation primes Rb2/p130 for phosphorylation and only dephosphorylated Rb2/p130 can be subject to acetylation. Human papillomavirus 16-E7 protein, which increases acetylation of Rb2/p130 by p300 strongly reduces phosphorylation of this protein by CDK4. This suggests that the balance between phosphorylation and acetylation of Rb2/p130 is essential for its biological function in cell cycle control. 相似文献