首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
Phosphatidylinositol-4,5-bisphosphate (PI4,5P2) is a critical regulator of cell migration, but the roles of the type I phosphatidylinositol-4-phosphate 5-kinases (PIPKIs), which synthesize PI4,5P2, have yet to be fully defined in this process. In this study, we report that one kinase, PIPKI-α, is a novel upstream regulator of Rac1 that links activated integrins to the regulation of cell migration. We show that PIPKI-α controls integrin-induced translocation of Rac1 to the plasma membrane and thereby regulates Rac1 activation. Strikingly, this function is not shared with other PIPKI isoforms, is independent of catalytic activity, and requires physical interaction of PIPKI-α with the Rac1 polybasic domain. Consistent with its role in Rac1 activation, depletion of PIPKI-α causes pronounced defects in membrane ruffling, actin organization, and focal adhesion formation, and ultimately affects the directional persistence of migration. Thus, our study defines the role of PIPKI-α in cell migration and describes a new mechanism for the spatial regulation of Rac1 activity that is critical for cell migration.  相似文献   

2.
Star-PAP is a nuclear non-canonical poly(A) polymerase (PAP) that shows specificity toward mRNA targets. Star-PAP activity is stimulated by lipid messenger phosphatidyl inositol 4,5 bisphoshate (PI4,5P2) and is regulated by the associated Type I phosphatidylinositol-4-phosphate 5-kinase that synthesizes PI4,5P2 as well as protein kinases. These associated kinases act as coactivators of Star-PAP that regulates its activity and specificity toward mRNAs, yet the mechanism of control of these interactions are not defined. We identified a phosphorylated residue (serine 6, S6) on Star-PAP in the zinc finger region, the domain required for PIPKIα interaction. We show that S6 is phosphorylated by CKIα within the nucleus which is required for Star-PAP nuclear retention and interaction with PIPKIα. Unlike the CKIα mediated phosphorylation at the catalytic domain, Star-PAP S6 phosphorylation is insensitive to oxidative stress suggesting a signal mediated regulation of CKIα activity. S6 phosphorylation together with coactivator PIPKIα controlled select subset of Star-PAP target messages by regulating Star-PAP-mRNA association. Our results establish a novel role for phosphorylation in determining Star-PAP target mRNA specificity and regulation of 3′-end processing.  相似文献   

3.
Cell migration is a fundamental cellular process required for embryonic development to wound healing and also plays a key role in tumor metastasis and atherosclerosis. Migration is regulated at multiple strata, from cytoskeletal reorganization to vesicle trafficking. In migrating cells, signaling pathways are integrated with vesicle trafficking machineries in a highly coordinated fashion to accomplish the recruitment and trafficking of the trans-membrane proteins toward the leading edge. Different signaling molecules regulate cell migration in different physio-pathological contexts, among them, phosphatidylinositol-4,5-biphosphate (PIP2) is an integral component of the plasma membrane and pleiotropic lipid signaling molecule modulating diverse biological processes, including actin cytoskeletal dynamics and vesicle trafficking required for cell migration. In this commentary, we provide a brief overview of our current understandings on the phosphoinositide signaling and its implication in regulation of cell polarity and vesicle trafficking in migrating cells. In addition, we highlight the coordinated role of PIPKIγi2, a focal adhesion-targeted enzyme that synthesizes PIP2, and the exocyst complex, a PIP2-effector, in the trafficking of E-cadherin in epithelial cells and integrins in migrating cancer cells.  相似文献   

4.
A fundamental property of tumor cells is to defy anoikis, cell death caused by a lack of cell-matrix interaction, and grow in an anchorage-independent manner. How tumor cells organize signaling molecules at the plasma membrane to sustain oncogenic signals in the absence of cell-matrix interactions remains poorly understood. Here, we describe a role for phosphatidylinositol 4-phosphate 5-kinase (PIPK) Iγi2 in controlling anchorage-independent growth of tumor cells in coordination with the proto-oncogene Src. PIPKIγi2 regulated Src activation downstream of growth factor receptors and integrins. PIPKIγi2 directly interacted with the C-terminal tail of Src and regulated its subcellular localization in concert with talin, a cytoskeletal protein targeted to focal adhesions. Co-expression of PIPKIγi2 and Src synergistically induced the anchorage-independent growth of nonmalignant cells. This study uncovers a novel mechanism where a phosphoinositide-synthesizing enzyme, PIPKIγi2, functions with the proto-oncogene Src, to regulate oncogenic signaling.  相似文献   

5.
We previously demonstrated that RhoA-dependent signaling regulates transforming growth factor-β1 (TGF-β1)-induced cytoskeletal reorganization in the human retinal pigment epithelial cell line ARPE-19. Smad pathways have also been shown to mediate TGF-β1 activity. Here, we examined what regulates Rho GTPase activity and tested whether Smad signaling cross-talks with Rho pathways during TGF-β1-induced actin rearrangement. Using small interfering RNAs, we found that NET1, the guanine nucleotide exchange factor of RhoA, is critical for TGF-β1-induced cytoskeletal reorganization, N-cadherin expression, and RhoA activation. In ARPE-19 cells lacking NET1, TGF-β1-induced stress fibers and N-cadherin expression were not observed. Interestingly, in dominant-negative Smad3-expressing or constitutively active Smad7 cells, TGF-β1 failed to induce NET1 mRNA and protein expression. Consistent with these results, both dominant-negative Smad3 and constitutively active Smad7 blocked the cytoplasmic localization of NET1 and inhibited interactions between NET1 and RhoA. Finally, we found that NET1 is a direct gene target of TGF-β1 via Smad3. Taken together, our results demonstrate that Smad3 regulates RhoA activation and cytoskeletal reorganization by controlling NET1 in TGF-β1-induced ARPE-19 cells. These data define a new role for Smad3 as a modulator of RhoA activation in the regulation of TGF-β1-induced epithelial-mesenchymal transitions.  相似文献   

6.
Integrin signaling plays critical roles in cell adhesion, spreading, and migration, and it is generally accepted that to regulate these integrin functions accurately, localized actin remodeling is required. However, the molecular mechanisms that control the targeting of actin regulation molecules to the proper sites are unknown. We previously demonstrated that integrin-mediated cell spreading and migration on fibronectin are dependent on the localized activation of phospholipase D (PLD). However, the mechanism underlying PLD activation by integrin is largely unknown. Here we demonstrate that protein kinase Cδ (PKCδ) is required for integrin-mediated PLD signaling. After integrin stimulation, PKCδ is activated and translocated to the edges of lamellipodia, where it colocalizes with PLD2. The abrogation of PKCδ activity inhibited integrin-induced PLD activation and cell spreading. Finally, we show that Thr566 of PLD2 is directly phosphorylated by PKCδ and that PLD2 mutation in this region prevents PLD2 activation, PLD2 translocation to the edge of lamellipodia, Rac translocation, and cell spreading after integrin activation. Together, these results suggest that PKCδ is a primary regulator of integrin-mediated PLD activation via the direct phosphorylation of PLD, which is essential for directing integrin-induced cell spreading.Integrin-mediated cell adhesion, spreading, and migration, which are essential for cellular differentiation, proliferation, survival, chemotaxis, and wound healing, require cell polarization with an environmental stimulus (32). To regulate these integrin-mediated functions accurately, coordinated and spatial control of localized cytoskeletal rearrangement is required. The key downstream signaling molecules of integrin-mediated actin cytoskeletal rearrangements include small GTPases of the Rho family, such as Rho, Cdc42, and Rac (57, 58). Recently it was suggested that integrin indirectly regulates the recruitment of small G proteins and their localized activation at a specific plasma membrane region called the cholesterol-enriched membrane microdomain. Furthermore, the membrane targeting of these molecules appears to be required for the activation of downstream effectors that induce actin reorganization (8, 9, 48). However, in the absence of integrin signaling, despite the GTP loading status, activated Rac and Cdc42 remain in the cytosol and cannot activate downstream effectors, such as p21-activated kinase (PAK) (8). This regulation of the localization of small GTPases to a specific site is supported by the observation that Rac1 is localized and activated at the leading edges of migrating cells, while Cdc42 is also activated in cellular protrusions and in the peripheral region (33, 51). The differentially localized activation of small GTPases results in coordinated spatially confined signaling leading to cytoskeletal rearrangement, which is critical for the regulation of integrin-mediated cell spreading and directional cell migration.The hydrolysis of phosphatidylcholine by phospholipase D1 (PLD1) and PLD2 generates the messenger lipid phosphatidic acid (PA) in response to a variety of signals, which include hormones, neurotransmitters, and growth factors (17). It has been shown that PA affects actin cytoskeletal rearrangement and hence lamellipodium extension and integrin-mediated cell spreading as well as migration. PLD activity has been found in detergent-insoluble membrane fractions in which a wide variety of cytoskeletal proteins, such as F-actin, α-actinin, vinculin, paxillin, and talin, were enriched (34). Furthermore, the stimulation of PLD with physiologic and pharmacologic agonists results in its association with actin filaments (34). In addition, actin polymerization and stress fiber formation are tightly coupled to the activation of PLD (14). The formation of lamellipodium structures and membrane ruffles is blocked by PLD inhibition (53, 60), and PLD activity is critical for epithelial cell, leukocyte, and neutrophil adhesion and migration (41, 43, 52). Furthermore, we have previously shown that the activity of PLD is upregulated, and that the activated PLD is translocated to lamellipodia, after integrin activation (3). The PLD product PA acts as a lipid anchor for the membrane translocation of Rac, and this PA-mediated localized activation of Rac is critical for integrin-mediated cell spreading and migration through Rac downstream signaling activation and actin cytoskeleton rearrangement (3). However, the mechanisms that regulate the activation and localization of PLD, which induce the localized downstream activation of integrin signaling, have not been elucidated.Members of the protein kinase C (PKC) family of serine-threonine kinases are known to play important roles in the transduction of signals from the activation of integrin to cell adhesion and spreading, as well as in cell migration via actin reorganization (11, 25, 61, 66). Several studies have shown that the activities of several PKC isozymes are modulated and are crucially required for integrin-mediated cell spreading and migration. The PKCα, -δ, and -ɛ isotypes were activated and then translocated from the cytosol to the membrane after integrin activation, and inhibition of these PKC isozymes prevented cell spreading (10, 47, 66). In addition, the activation of PKCα, -δ, and -ɛ rescued the spreading of α5 integrin-deficient cells on fibronectin (10), and PKCβΙ mediated platelet cell spreading and migration on fibrinogen (2). It has also been demonstrated that PKCθ activity is involved in endothelial cell migration (65). These results suggest that the kinase activities of diverse members of PKC are involved in the integrin-mediated signaling pathway leading to the actin cytoskeletal rearrangement required for cell spreading and migration. Several PKC substrates are known to influence the actin cytoskeleton directly (42). However, the natures of the isoform-specific functions of PKC members and of their specific downstream effectors for actin cytoskeletal rearrangement induction by integrin signaling remain to be elucidated.In this study, we found that PKCδ is an upstream modulator of localized PLD activation in the integrin signaling pathway. We demonstrate for the first time that PKCδ activity (not PKCα or PKCɛ activity) is critical for integrin-mediated PLD activation, and we found that PLD2 is phosphorylated at Thr566 by PKCδ in the integrin signaling pathway. Furthermore, we show that this phosphorylation is critical for integrin-mediated targeting of PLD to membrane ruffles, Rac translocation to the membrane, and lamellipodium formation during cell spreading. These findings strongly suggest a bridge between PKCδ and the signaling of actin cytoskeletal rearrangement by the integrin signaling pathway via PLD activation, and they provide a novel molecular mechanism for localized PLD activation via PKCδ phosphorylation, which is critical for the actin cytoskeletal rearrangements required for integrin-mediated cell spreading.  相似文献   

7.
Directional cell movement in response to external chemical gradients requires establishment of front–rear asymmetry, which distinguishes an up-gradient protrusive leading edge, where Rac-induced F-actin polymerization takes place, and a down-gradient retractile tail (uropod in leukocytes), where RhoA-mediated actomyosin contraction occurs. The signals that govern this spatial and functional asymmetry are not entirely understood. We show that the human type I phosphatidylinositol 4-phosphate 5-kinase isoform β (PIPKIβ) has a role in organizing signaling at the cell rear. We found that PIPKIβ polarized at the uropod of neutrophil-differentiated HL60 cells. PIPKIβ localization was independent of its lipid kinase activity, but required the 83 C-terminal amino acids, which are not homologous to other PIPKI isoforms. The PIPKIβ C terminus interacted with EBP50 (4.1-ezrin-radixin-moesin (ERM)-binding phosphoprotein 50), which enabled further interactions with ERM proteins and the Rho-GDP dissociation inhibitor (RhoGDI). Knockdown of PIPKIβ with siRNA inhibited cell polarization and impaired cell directionality during dHL60 chemotaxis, suggesting a role for PIPKIβ in these processes.  相似文献   

8.
Phosphatidylinositol phosphate kinases (PIPKs) are lipid kinases that generate phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a critical lipid signaling molecule that regulates diverse cellular functions, including the activities of membrane channels and transporters. IRBIT (IP3 R-binding protein released with inositol 1,4,5-trisphosphate) is a multifunctional protein that regulates diverse target proteins. Here, we report that IRBIT forms signaling complexes with members of the PIPK family. IRBIT bound to all PIPK isoforms in heterologous expression systems and specifically interacted with PIPK type Iα (PIPKIα) and type IIα (PIPKIIα) in mouse cerebellum. Site-directed mutagenesis revealed that two conserved catalytic aspartate residues of PIPKIα and PIPKIIα are involved in the interaction with IRBIT. Furthermore, phosphatidylinositol 4-phosphate, Mg2+, and/or ATP interfered with the interaction, suggesting that IRBIT interacts with catalytic cores of PIPKs. Mutations of phosphorylation sites in the serine-rich region of IRBIT affected the selectivity of its interaction with PIPKIα and PIPKIIα. The structural flexibility of the serine-rich region, located in the intrinsically disordered protein region, is assumed to underlie the mechanism of this interaction. Furthermore, in vitro binding experiments and immunocytochemistry suggest that IRBIT and PIPKIα interact with the Na+/HCO3 cotransporter NBCe1-B. These results suggest that IRBIT forms signaling complexes with PIPKIα and NBCe1-B, whose activity is regulated by PI(4,5)P2.  相似文献   

9.
Phospholipase D activity has been extensively implicated in the regulation of the actin cytoskeleton. Through this regulation the enzyme controls a number of physiological functions such as cell migration and adhesion and, it also is implicated in the regulation of membrane trafficking. The two phospholipase Ds are closely implicated with the control of the ARF and Rho families of small GTPases. In this article it is proposed that PLD2 plays the role of ‘master regulator’ and in an ill-defined manner regulates Rho function, PLD1 activity is downstream of this activation, however the generated phosphatidic acid controls changes in cytoskeletal organisation through its regulation of phosphatidylinositol-4-phosphate-5-kinase activity.  相似文献   

10.
Phospholipase Ds (PLDs) are regulated enzymes that generate phosphatidic acid (PA), a putative second messenger implicated in the regulation of vesicular trafficking and cytoskeletal reorganization. Mast cells, when stimulated with antigen, show a dramatic alteration in their cytoskeleton and also release their secretory granules by exocytosis. Butan-1-ol, which diverts the production of PA generated by PLD to the corresponding phosphatidylalcohol, was found to inhibit membrane ruffling when added together with antigen or when added after antigen. Inhibition by butan-1-ol was completely reversible because removal of butan-1-ol restored membrane ruffling. Measurements of PLD activation by antigen indicate a requirement for continual PA production during membrane ruffling, which was maintained for at least 30 min. PLD1 and PLD2 are both expressed in mast cells and green fluorescent protein-tagged proteins were used to identify PLD2 localizing to membrane ruffles of antigen-stimulated mast cells together with endogenous ADP ribosylation factor 6 (ARF6). In contrast, green fluorescent protein-PLD1 localized to intracellular vesicles and remained in this location after stimulation with antigen. Membrane ruffling was independent of exocytosis of secretory granules because phorbol 12-myristate 13-acetate increased membrane ruffling in the absence of exocytosis. Antigen or phorbol 12-myristate 13-acetate stimulation increased both PLD1 and PLD2 activity when expressed individually in RBL-2H3 cells. Although basal activity of PLD2-overexpressing cells is very high, membrane ruffling was still dependent on antigen stimulation. In permeabilized cells, antigen-stimulated phosphatidylinositol(4,5)bisphosphate synthesis was dependent on both ARF6 and PA generated from PLD. We conclude that both activation of ARF6 by antigen and a continual PLD2 activity are essential for local phosphatidylinositol(4,5)bisphosphate generation that regulates dynamic actin cytoskeletal rearrangements.  相似文献   

11.
Phosphatidic acid (PA) has been increasingly recognized as an important signaling lipid regulating cell growth and proliferation, membrane trafficking, and cytoskeletal reorganization. Recent studies indicate that the signaling PA generated from phospholipase D (PLD) and diacylglycerol kinase (DGK) plays critical roles in regulating the activity of some members of Ras superfamily of small guanosine triphosphatases (GTPases), such as Ras, Rac and Arf. Change of PA levels regulates the activity of small GTPases by modulating membrane localization and activity of small GTPase regulatory proteins, guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). In addition, PA also targets some small GTPases to membranes by direct binding. This review summarizes the roles of PLD and DGK in regulating the activity of several Ras superfamily members and cellular processes they control. Some future directions and the implication of PA regulation of Ras small GTPases in pathology are also discussed.  相似文献   

12.
Cell migration requires the regulated disassembly of focal adhesions, but the underlying mechanisms remain poorly defined. We have previously shown that focal adhesion disassembly requires the dynamin 2- and clathrin-dependent endocytosis of ligand-activated β1 integrins. Here, we identify type I phosphatidylinositol phosphate kinase beta (PIPKIβ), an enzyme that generates phosphatidylinositol-4,5-bisphosphate (PI4,5P2), as a key regulator of this process. We found that knockdown of PIPKIβ by RNA interference blocks the internalization of active β1 integrins and impairs focal adhesion turnover and cell migration. These defects are caused by the failure to target the endocytic machinery, including clathrin adaptors and dynamin 2, to focal adhesion sites. As a consequence, depletion of PIPKIβ blocks clathrin assembly at adhesion plaques and prevents complex formation between dynamin 2 and focal adhesion kinase (FAK), a critical step in focal adhesion turnover. Together, our findings identify PIPKIβ as a novel regulator of focal adhesion disassembly and suggest that PIPKIβ spatially regulates integrin endocytosis at adhesion sites to control cell migration.Cell migration is a highly dynamic process that depends on the ability of a cell to adhere to and deadhere from the extracellular matrix in a coordinated manner. Adhesion is mediated through focal adhesion sites, which assemble in response to activation and clustering of integrin receptors and comprise signaling and scaffolding proteins, such as focal adhesion kinase (FAK), talin, vinculin, paxillin, and zyxin (9, 55). These complexes anchor the extracellular matrix to the actin cytoskeleton and also serve as signaling platforms (9, 55). The formation of adhesive complexes is essential for the stabilization of membrane protrusions and to provide the tensile forces for migration (9, 55). However, rapid cell movement requires that focal adhesions not only be continuously formed, but also disassembled (9, 56). The coordinated control of cell adhesion, and release thereof, is therefore a critical regulatory function for migrating cells. However, while much has been learned about the mechanisms underlying focal adhesion assembly, comparatively little is known about how the turnover of adhesion sites is regulated, despite the importance of this process for cell migration.Recently, the protease calpain, FAK, and phosphatases and kinases that control the activity of FAK, as well as microtubules and the large GTPase dynamin 2, have been identified as regulators of focal adhesion disassembly (9, 10, 21, 22). In particular, a pathway has been defined in which microtubule targeting of focal adhesions leads to their disassembly (21). A critical step in this process is the formation of a protein complex between FAK and dynamin 2, a key regulator of endocytosis (21). Dynamin 2, together with components of the clathrin machinery, then mediates the turnover of focal adhesions by promoting the internalization of β1 integrins (14, 41). Notably, dynamin 2 and clathrin adaptors become enriched at focal adhesion sites prior to their disassembly (14, 21). Therefore, mechanisms that control the recruitment of the endocytic machinery to focal adhesion sites must exist. However, how this process is regulated during focal adhesion turnover remains unknown.Phosphatidylinositol-4,5-bisphosphate (PI4,5P2) has recently emerged as an important regulator of focal adhesion dynamics (38, 47, 51, 57). In addition to serving as the precursor to other second messengers, PI4,5P2 directly binds and modulates many focal adhesion components, including talin, vinculin, and α-actinin, that regulate adhesion assembly and their linkage to the actin cytoskeleton (38, 47, 51, 57). Adhesion to the extracellular matrix stimulates the synthesis of PI4,5P2, and the general paradigm has been that the resulting local increase in PI4,5P2 levels promotes focal adhesion assembly (23, 38, 40). Intriguingly, emerging evidence suggests that PI4,5P2 also promotes the disassembly of focal adhesions (13, 46). This finding implies that PI4,5P2 levels at adhesion sites must be tightly regulated, both spatially and temporally, to elicit its specific, yet inverse, effects on focal adhesion dynamics.The generation of PI4,5P2 at specific subcellular sites is modulated in part by the selective targeting and activation of specific type I phosphatidylinositol phosphate kinases (PIPKI), which synthesize PI4,5P2 (38). Three related PIPKI isoforms, designated PIPKIα, PIPKIβ, and PIPKIγ, and multiple splice variants are present in mammalian cells (27, 28, 39, 49). Recent studies have shown that the increase of PI4,5P2 synthesis leading to focal adhesion assembly is mediated through the specific recruitment of PIPKIγ661, a splice variant of PIPKIγ, to focal adhesions (18, 37). However, whether PIPKIγ661 or another member of the PIPKI family is responsible for synthesizing the PI4,5P2 pool regulating focal adhesion disassembly is currently unknown, and the molecular mechanisms whereby PI4,5P2 regulates this process are not well defined.Coincidently, PI4,5P2 is also an important organizer of clathrin assembly at the plasma membrane (17). In this study, we therefore set out to determine whether PI4,5P2 promotes focal adhesion disassembly through its effects on endocytosis and to identify the PIPKI isoform involved in generating this pool of PI4,5P2. We show that knockdown of one specific PIPKI isoform, PIPKIβ, blocks adhesion turnover leading to the inhibition of cell migration. We further show that PIPKIβ is necessary for the uptake of activated β1 integrins and provide evidence that PI4,5P2 produced by PIPKIβ orchestrates the recruitment of components of the endocytic machinery to adhesion sites. Together, these studies define the role of PI4,5P2 in the regulation of focal adhesion disassembly and identify PIPKIβ as the enzyme synthesizing this pool of PI4,5P2.  相似文献   

13.
Reorganization of the actin cytoskeleton is an early cellular response to a variety of extracellular signals. Dissection of pathways leading to actin rearrangement has focused largely on those initiated by growth factor receptors or integrins, although stimulation of G protein-coupled receptors also leads to cytoskeletal changes. In transfected Cos-7SH cells, activation of the chemoattractant formyl peptide receptor induces cortical actin polymerization and a decrease in the number of central actin bundles. In this report, we show that cytoskeletal reorganization can be transduced by G protein βγ heterodimers (Gβγ), phosphoinositide 3-kinase γ (PI3-Kγ), a guanosine exchange factor (GEF) for Rac, and Rac. Expression of inactive variants of either PI3-Kγ, the Rac GEF Vav, or Rac blocked the actin rearrangement. Neither wortmannin nor LY294002, pharmacologic inhibitors of PI3-K, could inhibit the actin rearrangement induced by a constitutively active Rac. The inhibition of cytoskeletal reorganization by the dominant negative Vav variants could be rescued by coexpression of a constitutively active form of Rac. In contrast, a Vav variant with its pleckstrin homology (PH) domain missing constitutively induced JNK activation and led to cytoskeletal reorganization, even without stimulation by PI3-Kγ. This suggests that the PH domain of Vav controls the guanosine exchange activity of Vav, perhaps by a mechanism regulated by D3 phosphoinositides generated by PI3-K. Taken together, these findings delineate a pathway leading from activation of a G protein-coupled receptor to actin reorganization which sequentially involves Gβγ, PI3-Kγ, a Rac GEF, and Rac.  相似文献   

14.
Neuronal nicotinic acetylcholine receptors (nAChRs) are widely expressed throughout the central and peripheral nervous system and are localized at synaptic and extrasynaptic sites of the cell membrane. However, the mechanisms regulating the localization of nicotinic receptors in distinct domains of the cell membrane are not well understood. N-cadherin is a cell adhesion molecule that mediates homotypic binding between apposed cell membranes and regulates the actin cytoskeleton through protein interactions with the cytoplasmic domain. At synaptic contacts, N-cadherin is commonly localized adjacent to the active zone and the postsynaptic density, suggesting that N-cadherin contributes to the assembly of the synaptic complex. To examine whether N-cadherin homotypic binding regulates the cell surface localization of nicotinic receptors, this study used heterologous expression of N-cadherin and α3β4 nAChR subunits C-terminally fused to a myc-tag epitope in Chinese hamster ovary cells. Expression levels of α3β4 nAChRs at cell-cell contacts and at contact-free cell membrane were analyzed by confocal microscopy. α3β4 nAChRs were found distributed over the entire surface of contacting cells lacking N-cadherin. In contrast, N-cadherin-mediated cell-cell contacts were devoid of α3β4 nAChRs. Cell-cell contacts mediated by N-cadherin-deleted proteins lacking the β-catenin binding region or the entire cytoplasmic domain showed control levels of α3β4 nAChRs expression. Inhibition of actin polymerization with latrunculin A and cytochalasin D did not affect α3β4 nAChRs localization within N-cadherin-mediated cell-cell contacts. However, treatment with the Rho associated kinase inhibitor Y27632 resulted in a significant increase in α3β4 nAChR levels within N-cadherin-mediated cell-cell contacts. Analysis of α3β4 nAChRs localization in polarized Caco-2 cells showed specific expression on the apical cell membrane and colocalization with apical F-actin and the actin nucleator Arp3. These results indicate that actomyosin contractility downstream of N-cadherin homotypic binding regulates the cell surface localization of α3β4 nAChRs presumably through interactions with a particular pool of F-actin.  相似文献   

15.
The signalling pathway leading, for example, to actin cytoskeletal reorganisation, secretion or superoxide generation involves phospholipase D (PLD)-catalysed hydrolysis of phosphatidylcholine to generate phosphatidic acid, which appears to mediate the messenger functions of this pathway. Two PLD genes (PLD1 and PLD2) with similar domain structures have been doned and progress has been made in identifying the protein regulators of PLD1 activation, for example Arf and Rho family members. The activities of both PLD isoforms are dependent on phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and our sequence analysis suggested the presence of a pleckstrin homology (PH) domain in PLD1, although its absence has also been daimed. Investigation of the inositide dependence showed that a bis-phosphorylated lipid with a vicinal pair of phosphates was required for PLD1 activity. Furthermore, PLD1 bound specifically and with high affinity to lipid surfaces containing PI(4,5)P2 independently of the substrate phosphatidylcholine, suggesting a key role for the PH domain in PLD function. Importantly, a glutathione-S-transferase (GST) fusion protein comprising GST and the PH domain of PLD1 (GST-PLD1-PH) also bound specifically to supported lipid monolayers containing PI(4,5)P2. Point mutations within the PLD1 PH domain inhibited enzyme activity, whereas deletion of the domain both inhibited enzyme activity and disrupted normal PLD1 localisation. Thus, the functional PH domain regulates PLD by mediating its interaction with polyphosphoinositide-containing membranes; this might also induce a conformational change, thereby regulating catalytic activity.  相似文献   

16.
Macrophages and dendritic cells continuously survey their environment in search of foreign particles and soluble antigens. Such surveillance involves the ongoing extension of actin-rich protrusions and the consequent formation of phagosomes and macropinosomes. The signals inducing this constitutive cytoskeletal remodeling have not been defined. We report that, unlike nonphagocytic cells, macrophages and immature dendritic cells have elevated levels of phosphatidic acid (PA) in their plasma membrane. The plasmalemmal PA is synthesized by phosphorylation of diacylglycerol, which is in turn generated by a G protein–stimulated phospholipase C. Inhibition of diacylglycerol kinase activity results in the detachment of T-cell lymphoma invasion and metastasis–inducing protein 1 (TIAM1)—a Rac guanine exchange factor—from the plasma membrane, thereby depressing Rac activity and abolishing the constitutive ruffling and macropinocytosis that characterize macrophages and immature dendritic cells. Accumulation of PA and binding of TIAM1 to the membrane require the activity of phosphatidylinositol-4,5-bisphosphate 3-kinase. Thus a distinctive, constitutive pathway of PA biosynthesis promotes the actin remodeling required for immune surveillance.  相似文献   

17.
The small GTPase, ADP-ribosylation factor-6 (ARF6), has been implicated in regulating membrane traffic and remodeling cortical F-actin. Using real-time video analysis of actin assembly in living cells, we investigated the function and mechanism of ARF6 in control of actin assembly. Expression of an activated form of ARF6 that mimicks the GTP-bound form of the GTPase induced actin assembly resulting in the movement of vesicle-like particles, some of which contain markers for pinosomes. Activated ARF6 also stimulated actin assembly at foci on the ventral surface of the cell and stimulated fluid phase pinocytosis. Particle motility induced by ARF6 involved Arp2/3 complex, tyrosine kinase activity, phospholipase D (PLD) and D3-phosphoinositides, but not phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). We conclude that ARF6 regulates actin assembly for pinosome motility and at foci on the ventral cell surface.  相似文献   

18.
Phosphatidic acid (PA) is a lipid second messenger located at the intersection of several lipid metabolism and cell signaling events including membrane trafficking, survival, and proliferation. Generation of signaling PA has long been primarily attributed to the activation of phospholipase D (PLD). PLD catalyzes the hydrolysis of phosphatidylcholine into PA. A variety of both receptor-tyrosine kinase and G-protein-coupled receptor stimulations have been shown to lead to PLD activation and PA generation. This study focuses on profiling the PA pool upon P2Y6 receptor signaling manipulation to determine the major PA producing enzymes. Here we show that PLD, although highly active, is not responsible for the majority of stable PA being produced upon UDP stimulation of the P2Y6 receptor and that PA levels are tightly regulated. By following PA flux in the cell we show that PLD is involved in an initial increase in PA upon receptor stimulation; however, when PLD is blocked, the cell compensates by increasing PA production from other sources. We further delineate the P2Y6 signaling pathway showing that phospholipase Cβ3 (PLCβ3), PLCδ1, DGKζ and PLD are all downstream of receptor activation. We also show that DGKζ is a novel negative regulator of PLD activity in this system that occurs through an inhibitory mechanism with PKCα. These results further define the downstream events resulting in PA production in the P2Y6 receptor signaling pathway.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号