Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Biochem Biophys Rep ; 17: 56-64, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30582008

ABSTRACT

The appropriate development and regulation of neuronal morphology are important to establish functional neuronal circuits and enable higher brain function of the central nervous system. R-Ras, a member of the Ras family of small GTPases, plays crucial roles in the regulation of axonal morphology, including outgrowth, branching, and guidance. GTP-bound activated R-Ras reorganizes actin filaments and microtubules through interactions with its downstream effectors, leading to the precise control of axonal morphology. However, little is known about the upstream regulatory mechanisms for R-Ras activation in neurons. In this study, we found that brain-derived neurotrophic factor (BDNF) has a positive effect on endogenous R-Ras activation and promotes R-Ras-mediated axonal growth. RNA interference knockdown and overexpression experiments revealed that RasGRF1, a guanine nucleotide exchange factor (GEF) for R-Ras, is involved in BDNF-induced R-Ras activation and the promotion of axonal growth. Phosphorylation of RasGRF1 by protein kinase A at Ser916/898 is needed for the full activation of its GEF activity and to facilitate Ras signaling. We observed that BDNF treatment markedly increased this phosphorylation. Our results suggest that BDNF is one of the critical extrinsic regulators for R-Ras activation, and that RasGRF1 is an intrinsic key mediator for BDNF-induced R-Ras activation and R-Ras-mediated axonal morphological regulation.

2.
Biochem Biophys Res Commun ; 499(4): 920-926, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29626472

ABSTRACT

EphA2, a member of the Eph family of receptor tyrosine kinases, has been reported to promote tumor malignancy through phosphorylation of serine 897 (S897). Here, we found that overexpression of wild-type EphA2 induced S897 phosphorylation through ERK activation without growth factors or cytokines and promoted glioblastoma cell proliferation. However, overexpression of a kinase-inactive mutant of EphA2 failed to induce ERK activation, S897 phosphorylation, and promotion of glioblastoma cell proliferation. These data suggest that when overexpressed, EphA2 induces ERK activation through its tyrosine kinase activity, leading to S897 phosphorylation and promotion of glioblastoma cell proliferation. Our findings provide a new insight into how EphA2 mediates glioblastoma progression.


Subject(s)
Glioblastoma/enzymology , Glioblastoma/pathology , Phosphoserine/metabolism , Receptor, EphA2/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Models, Biological , Phosphorylation
3.
J Biol Chem ; 292(48): 19721-19732, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29038291

ABSTRACT

Oncogenic signaling in cancer cells alters glucose uptake and utilization to supply sufficient energy and biosynthetic intermediates for survival and sustained proliferation. Oncogenic signaling also prevents oxidative stress and cell death caused by increased production of reactive oxygen species. However, elevated glucose metabolism in cancer cells, especially in glioblastoma, results in the cells becoming sensitive to glucose deprivation (i.e. in high glucose dependence), which rapidly induces cell death. However, the precise mechanism of this type of cell death remains unknown. Here, we report that glucose deprivation alone does not trigger glioblastoma cell death. We found that, for cell death to occur in glucose-deprived glioblastoma cells, cystine and glutamine also need to be present in culture media. We observed that cystine uptake through the cystine/glutamate antiporter xCT under glucose deprivation rapidly induces NADPH depletion, reactive oxygen species accumulation, and cell death. We conclude that although cystine uptake is crucial for production of antioxidant glutathione in cancer cells its transport through xCT also induces oxidative stress and cell death in glucose-deprived glioblastoma cells. Combining inhibitors targeting cancer-specific glucose metabolism with cystine and glutamine treatment may offer a therapeutic approach for glioblastoma tumors exhibiting high xCT expression.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Brain Neoplasms/pathology , Cell Death , Cystine/metabolism , Glioblastoma/pathology , Glucose/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , NADP/metabolism , Rats , Reactive Oxygen Species/metabolism
4.
PLoS One ; 11(7): e0159617, 2016.
Article in English | MEDLINE | ID: mdl-27437949

ABSTRACT

SGEF and Ephexin4 are members of the Ephexin subfamily of RhoGEFs that specifically activate the small GTPase RhoG. It is reported that Ephexin1 and Ephexin5, two well-characterized Ephexin subfamily RhoGEFs, are tyrosine-phosphorylated by Src, and that their phosphorylation affect their activities and functions. In this study, we show that SGEF, but not Ephexin4, is tyrosine-phosphorylated by Src. Tyrosine phosphorylation of SGEF suppresses its interaction with RhoG, the elevation of RhoG activity, and SGEF-mediated promotion of cell migration. We identified tyrosine 530 (Y530), which is located within the Dbl homology domain, as a major phosphorylation site of SGEF by Src, and Y530F mutation blocked the inhibitory effect of Src on SGEF. Taken together, these results suggest that the activity of SGEF is negatively regulated by tyrosine phosphorylation of the DH domain.


Subject(s)
Cell Movement/genetics , Guanine Nucleotide Exchange Factors/genetics , rho GTP-Binding Proteins/genetics , src-Family Kinases/genetics , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Tyrosine/genetics , src Homology Domains/genetics
5.
Cell Signal ; 28(8): 937-45, 2016 08.
Article in English | MEDLINE | ID: mdl-27132626

ABSTRACT

EphA2, a member of the Eph receptor tyrosine kinases, is frequently overexpressed in a variety of malignancies, including glioblastoma, and its expression is correlated with poor prognosis. EphA2 acts as a tumor promoter through a ligand ephrin-independent mechanism, which requires phosphorylation of EphA2 on serine 897 (S897), leading to increased cell migration and invasion. In this study, we show that ligand-independent EphA2 signaling occurs downstream of the MEK/ERK/RSK pathway and mediates epidermal growth factor (EGF)-induced cell proliferation in glioblastoma cells. Suppression of EphA2 expression by long-term exposure to ligand ephrinA1 or EphA2-targeted shRNA inhibited EGF-induced cell proliferation. Stimulation of the cells with EGF induced EphA2 S897 phosphorylation, which was suppressed by MEK and RSK inhibitors, but not by phosphatidylinositol 3-kinase (PI3K) and Akt inhibitors. The RSK inhibitor or RSK2-targeted shRNA also suppressed EGF-induced cell proliferation. Furthermore, overexpression of wild-type EphA2 promoted cell proliferation without EGF stimulation, whereas overexpression of EphA2-S897A mutant suppressed EGF- or RSK2-induced proliferation. Taken together, these results suggest that EphA2 is a key downstream target of the MEK/ERK/RSK signaling pathway in the regulation of glioblastoma cell proliferation.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Glioblastoma/enzymology , Glioblastoma/pathology , MAP Kinase Signaling System , Receptor, EphA2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Signal Transduction/drug effects
6.
J Biochem ; 158(3): 245-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25922200

ABSTRACT

Eph/ephrin signalling plays essential roles in various tissue developments, such as axon guidance, angiogenesis and tissue separation. Interaction between Ephs and ephrins upon cell-cell contact results in forward (towards Eph-expressing cells) and reverse (towards ephrin-expressing cells) signalling. Although the molecular mechanisms downstream of Eph/ephrin forward signalling have been extensively studied, the functions and intracellular molecular mechanisms of Eph/ephrin reverse signalling are not fully understood. Rho GTPases are key regulators of the actin cytoskeleton to regulate cell morphology. In this study, we revealed that stimulation with the extracellular domain of EphB2 to activate Eph/ephrin reverse signalling induced axonal retraction in hippocampal neurons. The reduction of axonal length and branching by Eph/ephrin reverse signalling was blocked by inhibition of RhoA or Rho-associated coiled-coil-containing protein kinase (ROCK). These results suggest that Eph/ephrin reverse signalling negatively regulates axonal outgrowth and branching through RhoA/ROCK pathway in hippocampal neurons.


Subject(s)
Ephrins/metabolism , Neurons/metabolism , Receptor, EphB2/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Axons/metabolism , Ephrins/genetics , Hippocampus/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Receptor, EphB2/genetics , Signal Transduction , rho GTP-Binding Proteins/genetics
7.
J Cell Sci ; 128(10): 1912-21, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25908849

ABSTRACT

Expression of EphA2 is upregulated in various cancers that are derived from epithelial cells and correlates with the ability of a cancer cell to undergo migration and invasion. Here we have investigated the role of EphA2 in the epithelial morphogenesis of Madin-Darby canine kidney (MDCK) cells in three-dimensional culture. We show that EphA2 is phosphorylated on serine residue 897 through hepatocyte growth factor (HGF) stimulation using a phosphatidylinositol 3-kinase (PI3K)-Akt-dependent mechanism and that this phosphorylation is required for the formation of extensions, the first step of tubulogenesis, in MDCK cysts. By contrast, stimulation using the ligand ephrinA1 dephosphorylates EphA2 on serine residue 897 and suppresses the HGF-induced morphological change. Furthermore, activation of the small GTPase RhoG is involved in the HGF-induced formation of extensions downstream of EphA2. These observations suggest that a ligand-independent activity of EphA2 contributes to epithelial morphogenesis.


Subject(s)
Hepatocyte Growth Factor/pharmacology , Receptor, EphA2/metabolism , Serine/metabolism , Animals , Cell Movement/physiology , Dogs , Epithelial-Mesenchymal Transition , Madin Darby Canine Kidney Cells , Molecular Conformation , Phosphorylation
8.
Mol Biol Cell ; 26(10): 1957-70, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25808489

ABSTRACT

Precise wiring patterns of axons are among the remarkable features of neuronal circuit formation, and establishment of the proper neuronal network requires control of outgrowth, branching, and guidance of axons. R-Ras is a Ras-family small GTPase that has essential roles in multiple phases of axonal development. We recently identified afadin, an F-actin-binding protein, as an effector of R-Ras mediating axon branching through F-actin reorganization. Afadin comprises two isoforms--l-afadin, having the F-actin-binding domain, and s-afadin, lacking the F-actin-binding domain. Compared with l-afadin, s-afadin, the short splicing variant of l-afadin, contains RA domains but lacks the F-actin-binding domain. Neurons express both isoforms; however, the function of s-afadin in brain remains unknown. Here we identify s-afadin as an endogenous inhibitor of cortical axon branching. In contrast to the abundant and constant expression of l-afadin throughout neuronal development, the expression of s-afadin is relatively low when cortical axons branch actively. Ectopic expression and knockdown of s-afadin suppress and promote branching, respectively. s-Afadin blocks the R-Ras-mediated membrane translocation of l-afadin and axon branching by inhibiting the binding of l-afadin to R-Ras. Thus s-afadin acts as a dominant-negative isoform in R-Ras-afadin-regulated axon branching.


Subject(s)
Alternative Splicing , Axons/physiology , Cerebral Cortex/physiology , Microfilament Proteins/physiology , Animals , Axons/metabolism , Cerebral Cortex/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Microfilament Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , ras Proteins/metabolism
9.
Cell Signal ; 26(12): 2879-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239188

ABSTRACT

Anoikis is a specific type of apoptosis induced by detachment of epithelial cells from extracellular matrix, and acquiring resistance to anoikis is an important step that enables cancer cells to metastasize. EphA2, which is overexpressed in a variety of human cancers, is phosphorylated by Akt on serine 897 and mediates ligand ephrin-independent promotion of anoikis resistance through the RhoG activator Ephexin4. EphB6 is frequently silenced in invasive and metastatic cancers; however, its role in cancer progression is poorly understood. Here we show that EphB6 interacts with EphA2 and suppresses EphA2-mediated promotion of anoikis resistance in MCF7 breast cancer cells. On the other hand, knockdown of EphB6 promotes anoikis resistance. We further show that expression of EphB6 decreases serine 897 phosphorylation of EphA2 and suppresses EphA2-Ephexin4 interaction and the RhoG activation. These findings implicate EphB6 as a negative regulator of EphA2 oncogenic signaling.


Subject(s)
Anoikis , Receptor, EphA2/metabolism , Receptor, EphB6/metabolism , Signal Transduction , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Humans , Ligands , MCF-7 Cells , Phosphorylation , Phosphoserine/metabolism , Protein Structure, Tertiary , Receptor, EphA2/chemistry , Receptor, EphB6/chemistry , rho GTP-Binding Proteins/metabolism
10.
Cell Signal ; 26(5): 1082-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24508479

ABSTRACT

Dock4 is a member of the Dock180 family of proteins that mediates cancer cell migration through activation of Rac. However, the regulatory mechanism of Dock4 remains unclear. In this study, we show that the C-terminal proline-rich region of Dock4 is essential for the Dock4 mediated promotion of cell migration in MDA-MB-231 breast cancer cells. We found that a phosphoinositide-binding protein SH3YL1 interacted with the C-terminal proline-rich region of Dock4. Interaction of SH3YL1 with Dock4 promoted Dock4-mediated Rac1 activation and cell migration. Mutations in the phosphoinositide-binding domain disrupted the ability of SH3YL1 to promote Dock4-mediated cell migration. In addition, depletion of SH3YL1 in MDA-MB-231 cells suppressed cell migration. Taken together, these results provide evidence for a novel and functionally important interaction between Dock4 and SH3YL1 to promote cancer cell migration by regulating Rac1 activity.


Subject(s)
Carrier Proteins/metabolism , GTPase-Activating Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Membrane Proteins , Mice , Mutation , Protein Binding , Protein Structure, Tertiary , rac1 GTP-Binding Protein/metabolism
11.
FEBS Open Bio ; 3: 78-82, 2013.
Article in English | MEDLINE | ID: mdl-23772378

ABSTRACT

EphA2 is activated through phosphorylation on serine 897 (S897) by Akt to promote cancer cell motility and invasion, independently of stimulation by ephrin, its ligand. Here we show that S897 phosphorylation of EphA2 strengthens the interaction between EphA2 and Ephexin4, a guanine nucleotide exchange factor for the small GTPase RhoG. S897A mutation of EphA2 abolished the EphA2/Ephexin4-mediated RhoG activation, promotion of cell migration, and resistance to anoikis. Our results suggest that S897-phosphorylated EphA2 recruits Ephexin4 to promote cell migration and anoikis resistance, providing a molecular link between S897 phosphorylation of EphA2 and tumor progression.

12.
Mol Biol Cell ; 24(10): 1602-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23536706

ABSTRACT

In neuronal development, dendritic spine formation is important for the establishment of excitatory synaptic connectivity and functional neural circuits. Developmental deficiency in spine formation results in multiple neuropsychiatric disorders. Dock4, a guanine nucleotide exchange factor (GEF) for Rac, has been reported as a candidate genetic risk factor for autism, dyslexia, and schizophrenia. We previously showed that Dock4 is expressed in hippocampal neurons. However, the functions of Dock4 in hippocampal neurons and the underlying molecular mechanisms are poorly understood. Here we show that Dock4 is highly concentrated in dendritic spines and implicated in spine formation via interaction with the actin-binding protein cortactin. In cultured neurons, short hairpin RNA (shRNA)-mediated knockdown of Dock4 reduces dendritic spine density, which is rescued by coexpression of shRNA-resistant wild-type Dock4 but not by a GEF-deficient mutant of Dock4 or a truncated mutant lacking the cortactin-binding region. On the other hand, knockdown of cortactin suppresses Dock4-mediated spine formation. Taken together, the results show a novel and functionally important interaction between Dock4 and cortactin for regulating dendritic spine formation via activation of Rac.


Subject(s)
Cortactin/metabolism , Dendritic Spines/physiology , GTPase-Activating Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , HEK293 Cells , Hippocampus/cytology , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Sequence Deletion , rac GTP-Binding Proteins/metabolism
13.
J Neurosci ; 32(24): 8293-305, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22699910

ABSTRACT

Semaphorins have been identified as repulsive guidance molecules in the developing nervous system. We recently reported that the semaphorin 4D (Sema4D) receptor Plexin-B1 induces repulsion in axon and dendrites by functioning as a GTPase-activating protein (GAP) for R-Ras and M-Ras, respectively. In axons, Sema4D stimulation induces growth cone collapse, and downregulation of R-Ras activity by Plexin-B1-mediated GAP activity is required for the action. Axonal R-Ras GAP activity downregulates phosphatidylinositol 3-kinase signaling pathway, and thereby induces inactivation of a microtubule assembly promoter protein, CRMP-2. However, in contrast to the well studied roles of semaphorins and plexins in axonal guidance, signaling molecules linking M-Ras GAP to dendritic cytoskeleton remain obscure. Here we identified an Ena/VASP ligand, Lamellipodin (Lpd), as a novel effector of M-Ras in dendrites. Lpd was expressed in F-actin-rich distal dendritic processes and was required for both basal and M-Ras-mediated dendrite development. Subcellular fractionation showed M-Ras-dependent membrane translocation of Lpd, which was suppressed by Sema4D. Furthermore, the Ena/VASP-binding region within Lpd was required for dendrite development, and its membrane targeting was sufficient to overcome the Sema4D-mediated reduction of dendritic outgrowth and disappearance of F-actin from distal dendrites. Furthermore, in utero electroporation experiments also indicated that regulation of the M-Ras-Lpd system by the GAP activity of Plexin is involved in the normal development of cortical dendrites in vivo. Overall, our study sheds light on how repulsive guidance molecules regulate actin cytoskeleton in dendrites, revealing a novel mechanism that the M-Ras-Lpd system regulates actin-based dendrite remodeling by Sema/Plexin in rats or mice of either sex.


Subject(s)
Antigens, CD/physiology , Carrier Proteins/physiology , Dendrites/physiology , GTPase-Activating Proteins/metabolism , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Semaphorins/physiology , ras Proteins/physiology , Actins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cytoskeleton/metabolism , Dendrites/metabolism , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , Primary Cell Culture , Protein Transport/physiology , Rats , Signal Transduction/physiology , ras Proteins/metabolism
14.
Mol Biol Cell ; 23(14): 2793-804, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22593211

ABSTRACT

Regulation of axon growth, guidance, and branching is essential for constructing a correct neuronal network. R-Ras, a Ras-family small GTPase, has essential roles in axon formation and guidance. During axon formation, R-Ras activates a series of phosphatidylinositol 3-kinase signaling, inducing activation of a microtubule-assembly promoter-collapsin response mediator protein-2. However, signaling molecules linking R-Ras to actin cytoskeleton-regulating axonal morphology remain obscure. Here we identify afadin, an actin-binding protein harboring Ras association (RA) domains, as an effector of R-Ras inducing axon branching through F-actin reorganization. We observe endogenous interaction of afadin with R-Ras in cortical neurons during the stage of axonal development. Ectopic expression of afadin increases axon branch number, and the RA domains and the carboxyl-terminal F-actin binding domain are required for this action. RNA interference knockdown experiments reveal that knockdown of endogenous afadin suppressed both basal and R-Ras-mediated axon branching in cultured cortical neurons. Subcellular localization analysis shows that active R-Ras-induced translocation of afadin and its RA domains is responsible for afadin localizing to the membrane and inducing neurite development in Neuro2a cells. Overall, our findings demonstrate a novel signaling pathway downstream of R-Ras that controls axon branching.


Subject(s)
Axons/physiology , Cerebral Cortex/metabolism , Microfilament Proteins/metabolism , ras Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Cell Differentiation , Cell Line , Cell Proliferation , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Microfilament Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Phosphatidylinositol 3-Kinase/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Signal Transduction
15.
Mol Biol Cell ; 23(8): 1593-604, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22357615

ABSTRACT

The Rnd proteins Rnd1, Rnd2, and Rnd3/RhoE are well known as key regulators of the actin cytoskeleton in various cell types, but they comprise a distinct subgroup of the Rho family in that they are GTP bound and constitutively active. Functional differences of the Rnd proteins in RhoA inhibition signaling have been reported in various cell types. Rnd1 and Rnd3 antagonize RhoA signaling by activating p190 RhoGAP, whereas Rnd2 does not. However, all the members of the Rnd family have been reported to bind directly to p190 RhoGAP and equally induce activation of p190 RhoGAP in vitro, and there is no evidence that accounts for the functional difference of the Rnd proteins in RhoA inhibition signaling. Here we report the role of the N-terminal region in signaling. Rnd1 and Rnd3, but not Rnd2, have a KERRA (Lys-Glu-Arg-Arg-Ala) sequence of amino acids in their N-terminus, which functions as the lipid raft-targeting determinant. The sequence mediates the lipid raft targeting of p190 RhoGAP correlated with its activation. Overall, our results demonstrate a novel regulatory mechanism by which differential membrane targeting governs activities of Rnd proteins to function as RhoA antagonists.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Membrane Microdomains/physiology , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , HEK293 Cells , HeLa Cells , Humans , Mice , Protein Binding , Repressor Proteins/antagonists & inhibitors , Signal Transduction , rho GTP-Binding Proteins/chemistry
16.
J Cell Physiol ; 227(2): 618-29, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21448919

ABSTRACT

Bradykinin (BK) plays a major role in producing peripheral sensitization in response to peripheral inflammation and in pain transmission in the central nerve system (CNS). Because BK activates protein kinase C (PKC) through phospholipase C (PLC)-ß and myristoylated alanine-rich C kinase substrate (MARCKS) has been found to be a substrate of PKC, we explored the possibility that BK could induce MARCKS phosphorylation and regulate its function. BK stimulation induced transient MARCKS phosphorylation on Ser159 with a peak at 1 min in human neuroblastoma SH-SY5Y cells. By contrast, PKC activation by the phorbol ester phorbol 12,13-dibutyrate (PDBu) elicited MARCKS phosphorylation which lasted more than 10 min. Western blotting analyses and glutathione S-transferase (GST) pull-down analyses showed that the phosphorylation by BK was the result of activation of the PKC-dependent RhoA/Rho-associated coiled-coil kinase (ROCK) pathway. Protein phosphatase (PP) 2A inhibitors calyculin A and fostriecin inhibited the dephosphorylation of MARCKS after BK-induced phosphorylation. Moreover, immunoprecipitation analyses showed that PP2A interacts with MARCKS. These results indicated that PP2A is the dominant PP of MARCKS after BK stimulation. We established SH-SY5Y cell lines expressing wild-type MARCKS and unphosphorylatable MARCKS, and cell morphology changes after cell stimulation were studied. PDBu induced lamellipodia formation on the neuroblastoma cell line SH-SY5Y and the morphology was sustained, whereas BK induced neurite outgrowth of the cells via lamellipodia-like actin accumulation that depended on transient MARCKS phosphorylation. Thus these findings show a novel BK signal cascade-that is, BK promotes neurite outgrowth through transient MARCKS phosphorylation involving the PKC-dependent RhoA/ROCK pathway and PP2A in a neuroblastoma cell line.


Subject(s)
Bradykinin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurites/physiology , Actins/metabolism , Cell Line, Tumor , Gene Expression Regulation/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Myristoylated Alanine-Rich C Kinase Substrate , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/metabolism , Phosphorylation/physiology , Pseudopodia/physiology , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Signal Transduction
17.
J Biol Chem ; 286(37): 32672-83, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21768103

ABSTRACT

Pombe Cdc15 homology proteins, characterized by Fer/CIP4 homology Bin-Amphiphysin-Rvs/extended Fer/CIP4 homology (F-BAR/EFC) domains with membrane invaginating property, play critical roles in a variety of membrane reorganization processes. Among them, Rapostlin/formin-binding protein 17 (FBP17) has attracted increasing attention as a critical coordinator of endocytosis. Here we found that Rapostlin was expressed in the developing rat brain, including the hippocampus, in late developmental stages when accelerated dendritic spine formation and maturation occur. In primary cultured rat hippocampal neurons, knockdown of Rapostlin by shRNA or overexpression of Rapostlin-QQ, an F-BAR domain mutant of Rapostlin that has no ability to induce membrane invagination, led to a significant decrease in spine density. Expression of shRNA-resistant wild-type Rapostlin effectively restored spine density in Rapostlin knockdown neurons, whereas expression of Rapostlin deletion mutants lacking the protein kinase C-related kinase homology region 1 (HR1) or Src homology 3 (SH3) domain did not. In addition, knockdown of Rapostlin or overexpression of Rapostlin-QQ reduced the uptake of transferrin in hippocampal neurons. Knockdown of Rnd2, which binds to the HR1 domain of Rapostlin, also reduced spine density and the transferrin uptake. These results suggest that Rapostlin and Rnd2 cooperatively regulate spine density. Indeed, Rnd2 enhanced the Rapostlin-induced tubular membrane invagination. We conclude that the F-BAR protein Rapostlin, whose activity is regulated by Rnd2, plays a key role in spine formation through the regulation of membrane dynamics.


Subject(s)
Cell Membrane Structures/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Membrane Structures/genetics , Dendritic Spines/genetics , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Humans , Mutation , Nerve Tissue Proteins/genetics , Rats , Rats, Wistar , Transferrin/genetics , Transferrin/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , src Homology Domains
18.
Exp Cell Res ; 317(12): 1701-13, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21621533

ABSTRACT

Disruption of cell-extracellular matrix interaction causes epithelial cells to undergo apoptosis called anoikis, and resistance to anoikis has been suggested to be a critical step for cancer cells to metastasize. EphA2 is frequently overexpressed in a variety of human cancers, and recent studies have found that overexpression of EphA2 contributes to malignant cellular behavior, including resistance to anoikis, in several different types of cancer cells. Here we show that Ephexin4, a guanine nucleotide exchange factor for the small GTPase RhoG that interacts with EphA2, plays an important role in the regulation of anoikis. Knockdown of Ephexin4 promoted anoikis in HeLa cells, and experiments using a knockdown-rescue approach showed that activation of RhoG, phosphatidylinositol 3-kinase (PI3K), and Akt was required for the Ephexin4-mediated suppression of anoikis. Indeed, Ephexin4 knockdown caused a decrease in RhoG activity and Akt phosphorylation in HeLa cells cultured in suspension. In addition, Ephexin4 was involved in the EphA2-mediated suppression of anoikis. Taken together, these results suggest that Ephexin4 mediates resistance to anoikis through activation of RhoG and PI3K downstream of EphA2.


Subject(s)
Anoikis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA2/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured , Dogs , Female , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Receptor, EphA2/genetics , Signal Transduction , rho GTP-Binding Proteins/genetics
19.
J Cell Biol ; 190(3): 461-77, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20679435

ABSTRACT

EphA2, a member of the Eph receptor family, is frequently overexpressed in a variety of human cancers, including breast cancers, and promotes cancer cell motility and invasion independently of its ligand ephrin stimulation. In this study, we identify Ephexin4 as a guanine nucleotide exchange factor (GEF) for RhoG that interacts with EphA2 in breast cancer cells, and knockdown and rescue experiments show that Ephexin4 acts downstream of EphA2 to promote ligand-independent breast cancer cell migration and invasion toward epidermal growth factor through activation of RhoG. The activation of RhoG recruits its effector ELMO2 and a Rac GEF Dock4 to form a complex with EphA2 at the tips of cortactin-rich protrusions in migrating breast cancer cells. In addition, the Dock4-mediated Rac activation is required for breast cancer cell migration. Our findings reveal a novel link between EphA2 and Rac activation that contributes to the cell motility and invasiveness of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Receptor, EphA2/metabolism , rho GTP-Binding Proteins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Guanine Nucleotide Exchange Factors/chemistry , HeLa Cells , Humans
20.
J Biol Chem ; 285(36): 28200-9, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20610402

ABSTRACT

Plexins are receptors for axonal guidance molecules semaphorins. We recently reported that the semaphorin 4D (Sema4D) receptor, Plexin-B1, suppresses PI3K signaling through the R-Ras GTPase-activating protein (GAP) activity, inducing growth cone collapse. Phosphatidylinositol 3-phosphate level is critically regulated by PI3K and PTEN (phosphatase and tensin homologue deleted chromosome ten). Here we examined the involvement of PTEN in the Plexin-B1-induced repulsive response. Phosphorylation of PTEN at Ser-380 is known to suppress its phosphatase activity. Sema4D induced the dephosphorylation of PTEN at Ser-380 and stimulated PTEN phosphatase activity in hippocampal neurons. Knockdown of endogenous PTEN suppressed the Sema4D-induced growth cone collapse. Phosphorylation mimic PTEN mutant suppressed the Sema4D-induced growth cone collapse, whereas phosphorylation-resistant PTEN mutant by itself induced growth cone collapse. Plexin-B1-induced PTEN dephosphorylation through R-Ras GAP activity and R-Ras GAP activity was by itself sufficient for PTEN dephosphorylation and activation. We also suggested that the Sema4D-induced PTEN dephosphorylation and growth cone collapse were mediated by the inhibition of casein kinase 2 alpha activity. Thus, we propose that Sema4D/Plexin-B1 promotes the dephosphorylation and activation of PTEN through the R-Ras GAP activity, inducing growth cone collapse.


Subject(s)
Antigens, CD/metabolism , Growth Cones/metabolism , Hippocampus/cytology , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , COS Cells , Casein Kinase II/metabolism , Chlorocebus aethiops , Enzyme Activation , Humans , Phosphorylation , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL