scholarly journals The MEK1 Scaffolding Protein MP1 Regulates Cell Spreading by Integrating PAK1 and Rho Signals

2005 ◽  
Vol 25 (12) ◽  
pp. 5119-5133 ◽  
Author(s):  
Ashok Pullikuth ◽  
Evangeline McKinnon ◽  
Hans-Joerg Schaeffer ◽  
Andrew D. Catling

ABSTRACT How the extracellular signal-regulated kinase (ERK) cascade regulates diverse cellular functions, including cell proliferation, survival, and motility, in a context-dependent manner remains poorly understood. Compelling evidence indicates that scaffolding molecules function in yeast to channel specific signals through common components to appropriate targets. Although a number of putative ERK scaffolding proteins have been identified in mammalian systems, none has been linked to a specific biological response. Here we show that the putative scaffold protein MEK partner 1 (MP1) and its partner p14 regulate PAK1-dependent ERK activation during adhesion and cell spreading but are not required for ERK activation by platelet-derived growth factor. MP1 associates with active but not inactive PAK1 and controls PAK1 phosphorylation of MEK1. Our data further show that MP1, p14, and MEK1 serve to inhibit Rho/Rho kinase functions necessary for the turnover of adhesion structures and cell spreading and reveal a signal-channeling function for a MEK1/ERK scaffold in orchestrating cytoskeletal rearrangements important for cell motility.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1895-1895
Author(s):  
Zhenyun Yang ◽  
Sarah Sitarski ◽  
Tirajeh Saadatzadeh ◽  
Fuqin Yin ◽  
Rebecca J. Chan

Abstract Abstract 1895 Poster Board I-918 Juvenile myelomoncytic leukemia (JMML) is a lethal childhood disease characterized by the in vitro phenotype of hematopoitic progenitor hypersensitivity to granulocyte-macrophage-colony-stimulating factor (GM-CSF). At the molecular level, Ras hyperactivation is implicated based on the majority of JMML patients bearing either loss-of-function NF1 mutations or gain-of-function RAS or PTPN11 mutations. We demonstrated previously that the Shp2 gain-of-function mutants Shp2E76K and Shp2D61Y induce constitutively elevated and sustained activation of Erk. Signal transduction among Raf1/MEK/Erk kinases is mediated through direct phosphorylation, but scaffolding proteins also play an important role in regulating the location, strength, and duration of Raf1/MEK/Erk signaling. One of the best-defined scaffolding proteins that positively facilitates the Raf1/MEK/Erk cascade is Kinase Suppressor of Ras (Ksr). In its inactivated state, Ksr is phosphorylated and constitutively associated with MEK. In response to growth factor stimulation or Ras activation, Ksr is dephosphorylated (serine 392), translocates to cell membrane, recruits Raf1 and Erk, and, thus, promotes Erk activation. We hypothesized that Ksr contributes to the hyperproliferation and GM-CSF hypersensitivity of mutant Shp2-expressing cells. Upon examination of phosphorylated Ksr levels, we observed lower GM-CSF-stimulated phospho- Ksr levels in the Shp2D61Y- and Shp2E76K-expressing macrophage progenitors compared to cells expressing empty vector or WT Shp2. Consistently, in co-immunoprecipitation assays, we found that upon GM-CSF stimulation, macrophage progenitors expressing Shp2D61Y or Shp2E76K demonstrated an increased physical association between phospho-Erk and Ksr, suggesting that Ksr promotes enhanced Erk activation in mutant Shp2-expressing cells and may contribute functionally to GM-CSF hypersensitivity of mutant Shp2-expressing cells. To examine this hypothesis, we subjected retrovirally transduced WT and Ksr1-/- bone marrow low density mononuclear cells (LDMNCs) to 3H-thymidine incorporation assays and found that GM-CSF-stimulated proliferation of Ksr1-/- cells expressing Shp2E76K was significantly reduced, but not entirely normalized to the level of WT Shp2-expressing cells. In contrast, the proliferation of Ksr1-/- cells expressing Shp2D61Y was unchanged compared to WT cells expressing Shp2D61Y. To examine the effect of genetic disruption of Ksr1 on GM-CSF-stimulated activation of Erk and Akt, western blot analysis was performed using retrovirally transduced WT and Ksr1-/- bone marrow LDMNCs, as described above. Activation of phospho-Erk was similarly reduced in both Shp2E76K- and Shp2D61Y-expressing cells upon genetic disruption of Ksr1 both at baseline and in response to GM-CSF. However, in contrast, Akt activation was increased, rather than decreased, in both Ksr1-/- Shp2E76K- and Shp2D61Y-expressing cells, suggesting that a compensatory mechanism in the absence of Ksr leads to enhanced signaling through the phospho-inositol-3-kinase (PI3K) pathway in mutant Shp2-expressing cells. Taken together, these findings suggest that the E76K mutant is dependent on Ksr-mediated Erk activation for GM-CSF-stimulated hyperactivation and that the compensatory upregulation of Akt activation in the absence of Ksr may contribute to the incomplete correction of GM-CSF hypersensitivity. Regarding the D61Y mutant, although Erk activation is reduced in the absence of Ksr, the lack of GM-CSF hypersensitivity correction suggests that the Shp2D61Y-expressing cells are more sensitive to the compensatory upregulation of Akt activation. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 34 (5) ◽  
pp. 833-836 ◽  
Author(s):  
D.B. Sacks

Signal transduction networks allow cells to recognize and respond to changes in the extracellular environment. All eukaryotic cells have MAPK (mitogen-activated protein kinase) pathways that participate in diverse cellular functions, including differentiation, survival, transformation and movement. Five distinct groups of MAPKs have been characterized in mammals, the most extensively studied of which is the Ras/Raf/MEK [MAPK/ERK (extracellular-signal-regulated kinase) kinase]/ERK cascade. Numerous stimuli, including growth factors and phorbol esters, activate MEK/ERK signalling. How disparate extracellular signals are translated by MEK/ERK into different cellular functions remains obscure. Originally identified in yeast, scaffold proteins are now recognized to contribute to the specificity of MEK/ERK pathways in mammalian cells. These scaffolds include KSR (kinase suppressor of Ras), β-arrestin, MEK partner-1, Sef and IQGAP1. Scaffolds organize multiprotein signalling complexes. This targets MEK/ERK to specific substrates and facilitates communication with other pathways, thereby mediating diverse functions. The adaptor proteins regulate the kinetics, amplitude and localization of MEK/ERK signalling, providing an efficient mechanism that enables an individual extracellular stimulus to promote a specific biological response.


2015 ◽  
Vol 43 (2) ◽  
pp. 199-204 ◽  
Author(s):  
Masaki Fukata ◽  
Atsushi Sekiya ◽  
Tatsuro Murakami ◽  
Norihiko Yokoi ◽  
Yuko Fukata

Precise regulation of protein assembly at specialized membrane domains is essential for diverse cellular functions including synaptic transmission. However, it is incompletely understood how protein clustering at the plasma membrane is initiated, maintained and controlled. Protein palmitoylation, a common post-translational modification, regulates protein targeting to the plasma membrane. Such modified proteins are enriched in these specialized membrane domains. In this review, we focus on palmitoylation of PSD-95, which is a major postsynaptic scaffolding protein and makes discrete postsynaptic nanodomains in a palmitoylation-dependent manner and discuss a determinant role of local palmitoylation cycles in creating highly localized hotspots at the membrane where specific proteins concentrate to organize functional domains.


2006 ◽  
Vol 26 (9) ◽  
pp. 3432-3445 ◽  
Author(s):  
Mark G. H. Scott ◽  
Vincenzo Pierotti ◽  
Hélène Storez ◽  
Erika Lindberg ◽  
Alain Thuret ◽  
...  

ABSTRACT β-Arrestins (βarr) are multifunctional adaptor proteins that can act as scaffolds for G protein-coupled receptor activation of mitogen-activated protein kinases (MAPK). Here, we identify the actin-binding and scaffolding protein filamin A (FLNA) as a βarr-binding partner using Son of sevenless recruitment system screening, a classical yeast two-hybrid system, coimmunoprecipitation analyses, and direct binding in vitro. In FLNA, the βarr-binding site involves tandem repeat 22 in the carboxyl terminus. βarr binds FLNA through both its N- and C-terminal domains, indicating the presence of multiple binding sites. We demonstrate that βarr and FLNA act cooperatively to activate the MAPK extracellular signal-regulated kinase (ERK) downstream of activated muscarinic M1 (M1MR) and angiotensin II type 1a (AT1AR) receptors and provide experimental evidence indicating that this phenomenon is due to the facilitation of βarr-ERK2 complex formation by FLNA. In Hep2 cells, stimulation of M1MR or AT1AR results in the colocalization of receptor, βarr, FLNA, and active ERK in membrane ruffles. Reduction of endogenous levels of βarr or FLNA and a catalytically inactive dominant negative MEK1, which prevents ERK activation, inhibit membrane ruffle formation, indicating the functional requirement for βarr, FLNA, and active ERK in this process. Our results indicate that βarr and FLNA cooperate to regulate ERK activation and actin cytoskeleton reorganization.


2010 ◽  
Vol 104 (11) ◽  
pp. 875-885 ◽  
Author(s):  
Yi Wu ◽  
Yuchuan Liu ◽  
Robert Colman

SummaryWe have shown that cleaved high-molecular-weight kininogen inhibits endoththelial cell tube and vacuole formation in a concentration-dependent manner and this correlates with its recognised anti-angiogenic activity. The antibody against the urokinase plasminogen activator receptor (uPAR) mimicked the inhibitory effect of cleaved kininogen (HKa) on apoptosis (HKa: 30% and uPAR antibody: 26%) and tube formation. In tumour angiogenesis, cancer cells release angiogenic stimulators, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), thus stimulating the transformation of endogenous pro-uPA to uPA. The proteolytic enzyme urokinase plasminogen activator (uPA) then binds to its receptor in a complex with its inhibitor PAI-1, which results in the internalisation of this complex, and activates extracellular signal-regulated kinase (ERK). Recycling of the uPAR regulates the migration of endothelial cells (ECs). ERK activation stimulates migration and proliferation and suppresses apoptosis of ECs. HKa disrupted the uPA-uPAR complex, inhibited ERK activation, and blocked the internalization of uPAR, eventually resulting in cell death and cell motility arrest. Both are critical steps in angiogenesis.


2002 ◽  
Vol 283 (1) ◽  
pp. C251-C260 ◽  
Author(s):  
Philippe Robin ◽  
Isaline Boulven ◽  
Christine Desmyter ◽  
Simone Harbon ◽  
Denis Leiber

In this study, we analyzed in rat myometrial cells the signaling pathways involved in the endothelin (ET)-1-induced extracellular signal-regulated kinase (ERK) activation required for the induction of DNA synthesis. We found that inhibition of protein kinase C (PKC) by Ro-31–8220 abolished ERK activation. Inhibition of phospholipase C (PLC) by U-73122 or of phosphoinositide (PI) 3-kinase by wortmannin partially reduced ERK activation. A similar partial inhibition was observed after treatment with pertussis toxin or PKC downregulation by phorbol ester treatment. The effect of wortmannin was additive with that produced by PKC downregulation but not with that due to pertussis toxin. These results suggest that both diacylglycerol-sensitive PKC, activated by PLC products, and diacylglycerol-insensitive PKC, possibly activated by a Gi-PI 3-kinase-dependent process, are involved in ET-1-induced ERK activation. These two pathways were found to be activated mainly through the ETA receptor subtype. ET-1 and phorbol ester stimulated Src activity in a PKC-dependent manner, both responses being abolished in the presence of Ro-31–8220. Inhibition of Src kinases by PP1 abrogated phorbol ester- and ET-1-induced ERK activation. Finally, ET-1 activated Ras in a PP1- and Ro-31–8220-sensitive manner. Altogether, our results indicate that ET-1 induces ERK activation in rat myometrial cells through the sequential stimulation of PKC, Src, and Ras.


1997 ◽  
Vol 272 (3) ◽  
pp. L558-L565 ◽  
Author(s):  
A. Y. Karpova ◽  
M. K. Abe ◽  
J. Li ◽  
P. T. Liu ◽  
J. M. Rhee ◽  
...  

We tested whether activation of mitogen-activated protein kinase/ extracellular signal-regulated kinase kinase-1 (MEK1) is required and sufficient for extracellular signal-regulated kinase (ERK) activation in airway smooth muscle cells. First, we transiently cotransfected bovine tracheal myocytes with an epitope-tagged ERK2 and a dominant-negative or a constitutively active form of the gene encoding MEK1 and assessed ERK2 activation by in vitro phosphorylation assay. Expression of the dominant-negative MEK1 inhibited platelet-derived growth factor (PDGF)-induced ERK2 activation, whereas expression of the constitutively active MEK1 induced ERK2 activation, suggesting that MEK1 is required and sufficient for ERK activation in these cells. Next, we assessed the effect of PD-98059, a synthetic MEK inhibitor, on PDGF-induced MEK1 and ERK activation. PD-98059 (10 microM) inhibited MEK1 and ERK activation, confirming that MEK1 is required for ERK activation in bovine tracheal myocytes. PD-98059 had no effect on Src or Raf-1 activity, evidence that PD-98059 is a specific inhibitor of MEK in this system. Finally, PD-98059 reduced PDGF-induced [(3)H]thymidine incorporation in a concentration-dependent manner, suggesting that catalytic activation of MEK1 and ERKs is required for DNA synthesis. We conclude that MEK1 is required for PDGF-induced ERK activation in bovine tracheal myocytes and that MEK1 and ERKs are required for PDGF-induced DNA synthesis in these cells.


2015 ◽  
Vol 209 (6) ◽  
pp. 895-912 ◽  
Author(s):  
Mutsuki Amano ◽  
Tomonari Hamaguchi ◽  
Md. Hasanuzzaman Shohag ◽  
Kei Kozawa ◽  
Katsuhiro Kato ◽  
...  

Protein kinases play pivotal roles in numerous cellular functions; however, the specific substrates of each protein kinase have not been fully elucidated. We have developed a novel method called kinase-interacting substrate screening (KISS). Using this method, 356 phosphorylation sites of 140 proteins were identified as candidate substrates for Rho-associated kinase (Rho-kinase/ROCK2), including known substrates. The KISS method was also applied to additional kinases, including PKA, MAPK1, CDK5, CaMK1, PAK7, PKN, LYN, and FYN, and a lot of candidate substrates and their phosphorylation sites were determined, most of which have not been reported previously. Among the candidate substrates for Rho-kinase, several functional clusters were identified, including the polarity-associated proteins, such as Scrib. We found that Scrib plays a crucial role in the regulation of subcellular contractility by assembling into a ternary complex with Rho-kinase and Shroom2 in a phosphorylation-dependent manner. We propose that the KISS method is a comprehensive and useful substrate screen for various kinases.


2000 ◽  
Vol 11 (9) ◽  
pp. 2863-2872 ◽  
Author(s):  
Joe W. Ramos ◽  
Paul E. Hughes ◽  
Mark W. Renshaw ◽  
Martin A. Schwartz ◽  
Etienne Formstecher ◽  
...  

PEA-15 is a small, death effector-domain (DED)–containing protein that was recently demonstrated to inhibit tumor necrosis factor-α–induced apoptosis and to reverse the inhibition of integrin activation due to H-Ras. This led us to investigate the involvement of PEA-15 in Ras signaling. Surprisingly, PEA-15 activates the extracellular signal receptor-activated kinase (ERK) mitogen-activated protein kinase pathway in a Ras-dependent manner. PEA-15 expression in Chinese hamster ovary cells resulted in an increased mitogen-activated protein kinase kinase and ERK activity. Furthermore, PEA-15 expression leads to an increase in Ras guanosine 5′-triphosphate loading. PEA-15 bypasses the anchorage dependence of ERK activation. Finally, the effects of PEA-15 on integrin signaling are separate from those on ERK activation. Heretofore, all known DEDs functioned in the regulation of apoptosis. In contrast, the DED of PEA-15 is essential for its capacity to activate ERK. The ability of PEA-15 to simultaneously inhibit apoptosis and potentiate Ras-to-Erk signaling may be of importance for oncogenic processes.


2021 ◽  
Author(s):  
Tanya A. Baldwin ◽  
Yong Li ◽  
Autumn Marsden ◽  
Roland F.R. Schindler ◽  
Musi Zhang ◽  
...  

The establishment of macromolecular complexes by scaffolding proteins such as A-kinase anchoring proteins is key to the local production of cAMP by anchored adenylyl cyclase (AC) and the subsequent cAMP signaling necessary for many cardiac functions. We have identified herein a novel AC scaffold, the Popeye domain-containing (POPDC) protein. Unlike other AC scaffolding proteins, POPDC1 binds cAMP with high affinity. The POPDC family of proteins are important for cardiac pacemaking and conduction, due in part to their cAMP-dependent binding and regulation of TREK-1 potassium channels. TREK-1 binds the AC9:POPDC1 complex and co-purifies in a POPDC1-dependent manner with AC9-associated activity in heart. Although the interaction of AC9 and POPDC1 is cAMP independent, TREK-1 association with AC9 and POPDC1 is reduced in an isoproterenol-dependent manner, requiring an intact cAMP binding Popeye domain and AC activity within the complex. We show that deletion of Adcy9 (AC9) gives rise to bradycardia at rest and stress-induced heart rate variability. The phenotype for deletion of Adcy9 is milder than previously observed upon loss of Popdc1, but similar to loss of Kcnk2 (TREK-1). Thus, POPDC1 represents a novel scaffolding protein for AC9 to regulate heart rate control.


Sign in / Sign up

Export Citation Format

Share Document