Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cells ; 11(22)2022 11 11.
Article in English | MEDLINE | ID: mdl-36428994

ABSTRACT

Dock1, originally Dock180, was the first identified member of the Dock family of GTPase Exchange Factors. Early biochemical and genetic studies of Dock180 elucidated the functions and regulation of Dock180 and informed our understanding of all Dock family members. Dock180 activates Rac to stimulate actin polymerization in response to signals initiated by a variety of receptors. Dock180 dependent Rac activation is essential for processes such as apoptotic cell engulfment, myoblast fusion, and cell migration during development and homeostasis. Inappropriate Dock180 activity has been implicated in cancer invasion and metastasis and in the uptake of bacterial pathogens. Here, we give an overview of the history and current understanding of the activity, regulation, and impacts of Dock180.


Subject(s)
Actins , rac GTP-Binding Proteins , rac GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Cytoskeleton/metabolism , Microtubules/metabolism
2.
Soft Matter ; 15(36): 7203-7210, 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31475279

ABSTRACT

Cancer metastasis has been believed as a genetically programmed process that is commonly marked by biochemical signals. Here using extracellular matrix control of cellular mechanics, we establish that cellular force threshold can also mark in vitro metastatic phenotypic change and malignant transformation in HCT-8 cell colonies. We observe that for prolonged culture time the HCT-8 cell colonies disperse into individual malignant cells, and the metastatic-like dispersion depends on both cell-seeding gel stiffness and colony size. Cellular force microscopies show that gel stiffness and colony size are also two key parameters that modulate cellular forces, suggesting the correlations between the cellular forces and the metastatic phenotypic change. Using our recently developed biophysical model, we construct an extracellular traction phase diagram in the stiffness-size space, filled with experimental data on the colony behavior. From the phase diagram we identify a phase boundary as a traction force threshold above which the metastatic phenotypic transition occurs and below which the cell colonies remain cohesive. Our finding suggests that the traction threshold can be regarded as an effective mechano-marker for the onset of the metastatic-like dispersion and malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Cell Adhesion , Cell Line, Tumor , Colon/cytology , Extracellular Matrix/metabolism , Humans , Mechanotransduction, Cellular , Models, Biological , Phenotype , Stress, Mechanical
3.
Mol Biol Cell ; 30(11): 1249-1271, 2019 05 15.
Article in English | MEDLINE | ID: mdl-31084567

ABSTRACT

Detailed structural, biochemical, cell biological, and genetic studies of any gene/protein are required to develop models of its actions in cells. Studying a protein family in the aggregate yields additional information, as one can include analyses of their coevolution, acquisition or loss of functionalities, structural pliability, and the emergence of shared or variations in molecular mechanisms. An even richer understanding of cell biology can be achieved through evaluating functionally linked protein families. In this review, we summarize current knowledge of three protein families: the ARF GTPases, the guanine nucleotide exchange factors (ARF GEFs) that activate them, and the GTPase-activating proteins (ARF GAPs) that have the ability to both propagate and terminate signaling. However, despite decades of scrutiny, our understanding of how these essential proteins function in cells remains fragmentary. We believe that the inherent complexity of ARF signaling and its regulation by GEFs and GAPs will require the concerted effort of many laboratories working together, ideally within a consortium to optimally pool information and resources. The collaborative study of these three functionally connected families (≥70 mammalian genes) will yield transformative insights into regulation of cell signaling.


Subject(s)
GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Animals , Eukaryota/metabolism , Humans
4.
Small GTPases ; 9(3): 242-259, 2018 05 04.
Article in English | MEDLINE | ID: mdl-27562622

ABSTRACT

Hepatocyte growth factor (HGF) is a potent signaling factor that acts on epithelial cells, causing them to dissociate and scatter. This migration is coordinated by a number of small GTPases, such as ARF6 and Rac1. Active ARF6 is required for HGF-stimulated migration and intracellular levels of ARF6-GTP and Rac1-GTP increase following HGF treatment. During migration, cross talk between ARF6 and Rac1 occurs through formation of a multi-protein complex containing the ARF-GEF cytohesin-2, the scaffolding protein GRASP/Tamalin, and the Rac1-GEF Dock180. Previously, the role of ARF6 in this process was unclear. We have now found that ARF6 and ARF1 regulate trafficking of GRASP and Dock180 to the plasma membrane following HGF treatment. Trafficking of GRASP and Dock180 is impaired by blocking ARF6-mediated recycling pathways and is required for HGF-stimulated Rac1 activation. Finally, HGF treatment stimulates association of GRASP and Dock180. Inhibition of ARF6 trafficking pathways traps GRASP and Dock180 as a complex in the cell.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factors/metabolism , Carrier Proteins/metabolism , Hepatocyte Growth Factor/pharmacology , Membrane Proteins/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , ADP-Ribosylation Factor 1/deficiency , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Dogs , Endosomes/drug effects , Endosomes/metabolism , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Madin Darby Canine Kidney Cells , Protein Transport/drug effects
5.
Mol Biol Cell ; 26(23): 4265-79, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26378252

ABSTRACT

When expressed in epithelial cells, cytohesin-2/ARNO, a guanine nucleotide exchange factor (GEF) for ARF small GTPases, causes a robust migration response. Recent evidence suggests that cytohesin-2/ARNO acts downstream of small the GTPase R-Ras to promote spreading and migration. We hypothesized that cytohesin-2/ARNO could transmit R-Ras signals by regulating the recycling of R-Ras through ARF activation. We found that Eps15-homology domain 1 (EHD1), a protein that associates with the endocytic recycling compartment (ERC), colocalizes with active R-Ras in transiently expressed HeLa cells. In addition, we show that EHD1-positive recycling endosomes are a novel compartment for cytohesin-2/ARNO. Knockdown or expression of GEF-inactive (E156K) cytohesin-2/ARNO causes R-Ras to accumulate on recycling endosomes containing EHD1 and inhibits cell spreading. E156K-ARNO also causes a reduction in focal adhesion size and number. Finally, we demonstrate that R-Ras/ARNO signaling is required for recycling of α5-integrin and R-Ras to the plasma membrane. These data establish a role for cytohesin-2/ARNO as a regulator of R-Ras and integrin recycling and suggest that ARF-regulated trafficking of R-Ras is required for R-Ras-dependent effects on spreading and adhesion formation.


Subject(s)
GTPase-Activating Proteins/metabolism , Integrin alpha5/metabolism , Vesicular Transport Proteins/metabolism , ras Proteins/metabolism , Endosomes/metabolism , Epithelial Cells/metabolism , Focal Adhesions/metabolism , HeLa Cells , Humans , Signal Transduction
6.
Physiol Rep ; 3(6)2015 Jun.
Article in English | MEDLINE | ID: mdl-26116550

ABSTRACT

The lack of current treatment and preventable measures for acute kidney injury (AKI) in hospitalized patients results in an increased mortality rate of up to 80% and elevated health costs. Additionally, if not properly repaired, those who survive AKI may develop fibrosis and long-term kidney damage. The molecular aspects of kidney injury and repair are still uncertain. Hepatocyte growth factor (HGF) promotes recovery of the injured kidney by inducing survival and migration of tubular epithelial cells to repopulate bare tubule areas. HGF-stimulated kidney epithelial cell migration requires the activation of ADP-ribosylation factor 6 (Arf6) and Rac1 via the cytohesin family of Arf-guanine-nucleotide exchange factors (GEFs), in vitro. We used an ischemia and reperfusion injury (IRI) mouse model to analyze the effects of modulating this signaling pathway on kidney recovery. We treated IRI mice with either HGF, the cytohesin inhibitor SecinH3, or a combination of both. As previously reported, HGF treatment promoted rapid improvement of kidney function as evidenced by creatinine (Cre) and blood urea nitrogen (BUN) levels. In contrast, simultaneous treatment with SecinH3 and HGF blocks the ability of HGF to promote kidney recovery. Immunohistochemistry showed that HGF treatment promoted recovery of tubule structure, and had enhanced levels of active, GTP-bound Arf6 and GTP-Rac1. SecinH3 treatment, however, caused a dramatic decrease in GTP-Arf6 and GTP-Rac1 levels when compared to kidney sections from HGF-treated IRI mice. Additionally, SecinH3 counteracted the renal reparative effects of HGF. Our results support the conclusion that cytohesin function is required for HGF-stimulated renal IRI repair.

7.
PLoS One ; 8(11): e82084, 2013.
Article in English | MEDLINE | ID: mdl-24303080

ABSTRACT

Cell migration is regulated by a number of small GTPases, including members of the Arf family. Cytohesins, a family of Arf-activating proteins, have been extensively implicated in the regulation of Arfs during migration and cell shape change. Membrane association of both the Arf and its activating protein is a prerequisite for Arf activation. Therefore regulating the extent of cytohesin membrane association is a mechanism for controlling the initiation of cell movement. We have discovered a novel intramolecular interaction that controls the association of cytohesins with membranes. The presence of the coiled-coil domain reduces the association of cytohesin 2 with membranes. We demonstrate that this domain interacts with more C-terminal regions of the protein. This interaction is independent of another previously identified autoinhibitory conformation. A threonine residue (T276) in the cytohesin 2 PH domain is a target for phosphorylation by Akt. Mutation of this threonine to aspartic acid, to mimic phosphorylation, disrupts the binding of the coiled-coil domain to c-terminal regions and promotes membrane association of cytohesin 2. The presence of a second autoinhibitory interaction in the cytohesins suggests that these proteins can act a signal integrators that stimulate migration only after receive multiple pro-migratory signals.


Subject(s)
Cell Membrane/metabolism , GTPase-Activating Proteins/metabolism , Protein Interaction Domains and Motifs , Threonine/metabolism , Animals , Cell Line , GTPase-Activating Proteins/chemistry , Models, Biological , Phosphorylation , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism
8.
BMC Cell Biol ; 14: 9, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23441967

ABSTRACT

BACKGROUND: The transition of epithelial cells from their normal non-motile state to a motile one requires the coordinated action of a number of small GTPases. We have previously shown that epithelial cell migration is stimulated by the coordinated activation of Arf and Rac GTPases. This crosstalk depends upon the assembly of a multi-protein complex that contains the Arf-activating protein cytohesin 2/ARNO and the Rac activating protein Dock180. Two scaffolding proteins that bind directly to cytohesin 2 organize this complex. RESULTS: We now have found that Rac activation in response to hepatocyte growth factor (HGF) requires cytohesin 2 and Dock180. GRASP/Tamalin is one of the scaffolds that builds the complex containing cytohesin 2 and Dock180. We determine here that the Ala/Pro rich region of GRASP directly interacts with the SH3 domain of Dock180. By binding to both cytohesin 2/ARNO and Dock180, GRASP bridges the guanine nucleotide exchange factors (GEFs) that activate Arf and Rac, thereby promoting Arf-to-Rac signaling. Furthermore, we find that knockdown of GRASP impairs hepatocyte growth factor (HGF)-stimulated Rac activation and HGF-stimulated epithelial migration. CONCLUSIONS: GRASP binds directly both cytohesin 2 and Dock180 to coordinate their activities, and by doing so promotes crosstalk between Arf and Rac.


Subject(s)
Carrier Proteins/metabolism , GTP Phosphohydrolases/metabolism , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , rac GTP-Binding Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Movement/drug effects , Dogs , Enzyme Activation/drug effects , HEK293 Cells , Hepatocyte Growth Factor/pharmacology , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , src Homology Domains
9.
J Cell Sci ; 125(Pt 13): 3195-201, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22454518

ABSTRACT

Recycling of internalized integrins is a crucial step in adhesion remodeling and cell movement. Recently, we determined that the ADP-ribosylation factor-guanine nucleotide exchange factors (ARF-GEFs) cytohesin 2/ARNO and cytohesin 3/GRP1 have opposing effects on adhesion and stimulated ß1 integrin recycling even though they are very closely related proteins (80% sequence identity). We have now determined the sequence differences underlying the differential actions of cytohesin 2/ARNO and cytohesin 3/GRP1. We found that the ability of cytohesins to promote ß1 integrin recycling and adhesion depends upon the presence or absence of a key glycine residue in their pleckstrin homology (PH) domains. This glycine residue determines the phosphoinositide specificity and affinity of cytohesin PH domains. Switching the number of glycines in the PH domains of cytohesin 2 and cytohesin 3 is sufficient to reverse their effects on adhesion and spreading and to reverse their subcellular locations. Importantly, we also find that a mutant form of cytohesin 3/GRP1 that has three rather than two glycines in its PH domain rescues ß1 integrin recycling in cytohesin 2/ARNO knockdown cells. Conversely, a mutant form of cytohesin 2/ARNO with two glycines in its PH domain fails to rescue ß1 integrin recycling. Therefore, we conclude that phosphoinositide specificity is the sole functional difference that determines which cytohesin can promote integrin recycling.


Subject(s)
GTPase-Activating Proteins/metabolism , Integrin beta1/metabolism , Phosphatidylinositols/metabolism , Cell Adhesion , Endocytosis , Fibronectins/metabolism , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Glycine/metabolism , HeLa Cells , Humans , Integrin beta1/genetics , MCF-7 Cells , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Analysis, Protein , Substrate Specificity , Transfection
10.
Exp Cell Res ; 318(3): 228-37, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22085542

ABSTRACT

Epithelial cells are largely immotile under normal circumstances, but become motile during development, repair of tissue damage and during cancer metastasis. Numerous growth factors act to initiate epithelial cell movements. Hepatocyte growth factor (HGF) induces many epithelial cell lines to begin crawling. A number of small GTPases act downstream of HGF to alter cell shape and promote movement. Arf6 is one of these GTPases that can alter the cortical actin cytoskeleton and promote epithelial movement. Activation of Arf6 in MDCK cells by its guanine nucleotide exchange factor cytohesin 2/ARNO produces a scattering response strikingly reminiscent of the action of HGF. We have previously shown that IPCEF1, a scaffold that binds to cytohesin 2, is required for cytohesin-induced scattering. We report here that IPCEF1 is actually the C-terminal half of CNK3. CNKs are scaffolds involved in signal transduction downstream of Ras. We have found that both MDCK and CaCo-2 cells express a fused CNK3/IPCEF1 protein. Knockdown of this protein impairs HGF-induced Arf6 activation and migration in response to HGF treatment.


Subject(s)
ADP-Ribosylation Factors/metabolism , Carrier Proteins/physiology , Cell Movement/genetics , Hepatocyte Growth Factor/pharmacology , Membrane Proteins/physiology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , Caco-2 Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Dogs , Gene Expression Regulation/drug effects , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
11.
J Biol Chem ; 285(19): 14610-6, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20223830

ABSTRACT

ADP-ribosylation actor 6 (ARF6) regulates the endocytosis and recycling of a variety of proteins and also promotes peripheral actin rearrangements and cell motility. ARF6 is activated by a large number of guanine nucleotide exchange factors, which likely regulate ARF6 at different locations and during different processes. In this study we investigate the roles of the cytohesin ADP-ribosylation factor (ARF)-guanine nucleotide exchange factors during the recycling of integrin beta1. Intriguingly, we find that knockdown and overexpression of ARNO/cytohesin 2 and GRP1/cytohesin 3 have opposing effects on cell adhesion and spreading on fibronectin and on cell migration. We find that ARNO/cytohesin 2 is required for integrin beta1 recycling, whereas GRP1/cytohesin 3 is dispensable for this process. This is the first demonstration of unique roles for these proteins.


Subject(s)
GTPase-Activating Proteins/metabolism , Integrin beta1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Blotting, Western , Cell Adhesion , Cell Movement , Dogs , Endocytosis , Fibronectins/metabolism , Flow Cytometry , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Integrin beta1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Mol Biol Cell ; 21(4): 562-71, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20016009

ABSTRACT

ARFs are small GTPases that regulate vesicular trafficking, cell shape, and movement. ARFs are subject to extensive regulation by a large number of accessory proteins. The many different accessory proteins are likely specialized to regulate ARF signaling during particular processes. ARNO/cytohesin 2 is an ARF-activating protein that promotes cell migration and cell shape changes. We report here that protein-protein interactions mediated by the coiled-coil domain of ARNO are required for ARNO induced motility. ARNO lacking the coiled-coil domain does not promote migration and does not induce ARF-dependent Rac activation. We find that the coiled-coil domain promotes the assembly of a multiprotein complex containing both ARNO and the Rac-activating protein Dock180. Knockdown of either GRASP/Tamalin or IPCEF, two proteins known to bind to the coiled-coil of ARNO, prevents the association of ARNO and Dock180 and prevents ARNO-induced Rac activation. These data suggest that scaffold proteins can regulate ARF dependent processes by biasing ARF signaling toward particular outputs.


Subject(s)
ADP-Ribosylation Factors/metabolism , Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , ADP-Ribosylation Factors/genetics , Animals , Carrier Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Line , Dogs , Enzyme Activation , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Structure, Secondary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , rac GTP-Binding Proteins/genetics
13.
Biochem Biophys Res Commun ; 342(4): 1361-7, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16516854

ABSTRACT

Adipocytes are now known to secrete a range of adipokines that exhibit distinct biological functions. Here, we sought to understand the secretory pathways utilised by ACRP30 to the surface of adipocytes. We find that ACRP30 overlaps with adipsin in intracellular compartments distinct from Glut4, but nonetheless exhibits insulin-stimulated secretion from cells. Both adipsin and ACRP30 overlap with transferrin receptor-positive membranes, implying that the pathway of secretion involves the transferrin receptor-positive endosomal system. Consistent with this, we show that ablation of endosomes significantly inhibited the secretion of ACRP30, as did treatment of cells with Brefeldin A. In order to further probe the role of recycling endosomes on the secretion of ACRP30, we over-expressed a mutant form of Rab11, Rab11-S25N, in 3T3-L1 adipocytes and found that expression of this mutant significantly reduced basal and insulin-stimulated secretion. We also demonstrate that Arf6 also plays a role in the secretion of ACRP30. Collectively, these data implicate both Arf6 and Rab11 as crucial mediators of constitutive and insulin-stimulated secretion of ACRP30 and further suggest that recycling endosomes may play a central role in this process.


Subject(s)
Adipocytes/metabolism , Endosomes/metabolism , Protein Transport/physiology , Signal Transduction/physiology , rab GTP-Binding Proteins/metabolism , 3T3-L1 Cells , Adiponectin/biosynthesis , Animals , Mice
14.
J Biol Chem ; 281(19): 13300-13308, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16484220

ABSTRACT

ARNO is a guanine-nucleotide exchange protein for the ARF family of GTPases. Here we show that in polarized epithelial cells, ARNO is localized exclusively to the apical plasma membrane, where it regulates endocytosis. Expression of ARNO stimulates apical endocytosis of the polymeric immunoglobulin receptor, and coexpression of ARF6 with ARNO leads to a synergistic stimulation of apical endocytosis. Expression of a dominant negative ARF6 mutant, ARF6-T27N, antagonizes this stimulatory effect. Deletion of the N-terminal coiled-coil (CC) domain of ARNO causes the mutant ARNO to localize to both the apical and basolateral plasma membranes. Expression of the CC domain alone abolishes ARNO-induced apical endocytosis as well as co-localization of IgA-receptor complexes with ARNO and clathrin. These results suggest that the CC domain contributes to the specificity of apical localization of ARNO through association with components of the apical plasma membrane. We conclude that ARNO acts together with ARF6 to regulate apical endocytosis.


Subject(s)
ADP-Ribosylation Factors/metabolism , Endocytosis/physiology , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , Cell Line , Cell Membrane/metabolism , Dogs , Epithelial Cells/cytology , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Humans , Protein Structure, Tertiary , Protein Transport
15.
Curr Biol ; 15(19): 1749-54, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16213822

ABSTRACT

Cell motility requires extensions of the plasma membrane driven by reorganization of the actin cytoskeleton. Small GTPases, particularly the Rho family, are key regulators of this process. A second class of GTPases, the ADP-ribosylation factors (ARFs), have also been implicated in the regulation of the actin cytoskeleton and motility. ARF6 is intimately involved in the regulation of Rac activity; however, the mechanisms by which ARF activation leads to activation of Rac remain poorly understood. We have previously shown that expression of the ARF-GEF ARNO in MDCK cells induces robust activation of Rac, the formation of large lamellipodia, and the onset of motility. We report here that ARNO-dependent activation of Rac is mediated by a bipartite Rac GEF, the Dock180/Elmo complex. Both DOCK180 and Elmo colocalize extensively with ARNO in migrating MDCK cells. Importantly, both a catalytically inactive Dock180 mutant and an Elmo mutant that fails to couple to Dock180 block ARNO-induced Rac activation and motility. In contrast, a similar mutant of the Rac GEF beta-PIX fails to inhibit ARNO-induced Rac activation or motility. Together, these data suggest that ARNO and ARF6 coordinate with the Dock180/Elmo complex to promote Rac activation at the leading edge of migrating cells.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , ADP-Ribosylation Factor 6 , Animals , Cell Line , Dogs , GTPase-Activating Proteins/metabolism , Humans , Microscopy, Fluorescence
16.
Mol Cell Biol ; 24(22): 9752-62, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509780

ABSTRACT

TBC (Tre-2/Bub2/Cdc16) domains are predicted to encode GTPase-activating proteins (GAPs) for Rab family G proteins. While approximately 50 TBC proteins are predicted to exist in humans, little is known about their substrate specificity. Here we show that TRE17 (also called Tre-2 and USP6), a founding member of the TBC family, targets the Arf family GTPase Arf6, which regulates plasma membrane-endosome trafficking. Surprisingly, TRE17 does not function as a GAP for Arf6 but rather promotes its activation in vivo. TRE17 associates directly with Arf6 in its GDP- but not GTP-bound state. Mapping experiments pinpoint the site of interaction to the TBC domain of TRE17. Forced expression of TRE17 promotes the localization of Arf6 to the plasma membrane, leading to Arf6 activation, presumably due to facilitated access to membrane-associated guanine nucleotide exchange factors (GEFs). Furthermore, TRE17 cooperates with Arf6 GEFs to induce GTP loading of Arf6 in vivo. Finally, short interfering RNA-mediated loss of TRE17 leads to attenuated Arf6 activation. These studies identify TRE17 as a novel regulator of the Arf6-regulated plasma membrane recycling system and reveal an unexpected function for TBC domains.


Subject(s)
ADP-Ribosylation Factors/metabolism , Endopeptidases/metabolism , Oncogene Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Membrane/metabolism , DNA/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Endosomes/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Phenotype , Protein Structure, Tertiary , Proto-Oncogene Proteins , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , Ubiquitin Thiolesterase
17.
BMC Cell Biol ; 4: 13, 2003 09 11.
Article in English | MEDLINE | ID: mdl-12969509

ABSTRACT

BACKGROUND: Phospholipase D (PLD) is involved in many signaling pathways. In most systems, the activity of PLD is primarily regulated by the members of the ADP-Ribosylation Factor (ARF) family of GTPases, but the mechanism of activation of PLD and ARF by extracellular signals has not been fully established. Here we tested the hypothesis that ARF-guanine nucleotide exchange factors (ARF-GEFs) of the cytohesin/ARNO family mediate the activation of ARF and PLD by insulin. RESULTS: Wild type ARNO transiently transfected in HIRcB cells was translocated to the plasma membrane in an insulin-dependent manner and promoted the translocation of ARF to the membranes. ARNO mutants: DeltaCC-ARNO and CC-ARNO were partially translocated to the membranes while DeltaPH-ARNO and PH-ARNO could not be translocated to the membranes. Sec7 domain mutants of ARNO did not facilitate the ARF translocation. Overexpression of wild type ARNO significantly increased insulin-stimulated PLD activity, and mutations in the Sec7 and PH domains, or deletion of the PH or CC domains inhibited the effects of insulin. CONCLUSIONS: Small ARF-GEFs of the cytohesin/ARNO family mediate the activation of ARF and PLD by the insulin receptor.


Subject(s)
ADP-Ribosylation Factors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Insulin/pharmacology , Phospholipase D/metabolism , ADP-Ribosylation Factors/genetics , Animals , Cell Line , Cell Membrane/metabolism , Enzyme Activation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Mutation , Protein Binding , Protein Transport/drug effects , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Transfection
18.
J Biol Chem ; 277(43): 40185-8, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12218044

ABSTRACT

Studies of GTPase function often employ expression of dominant negative or constitutively active mutants. Dominant negative mutants cannot bind GTP and thus cannot be activated. Constitutively active mutants cannot hydrolyze GTP and therefore accumulate a large pool of GTP-bound GTPase. These mutations block the normal cycle of GTP binding, hydrolysis, and release. Therefore, although the GTPase-deficient mutants are in the active conformation, they do not fully imitate all the actions of the GTPase. This is particularly true for the ADP-ribosylation factors (ARFs), GTPases that regulate vesicular trafficking events. In Ras and Rho GTPases replacement of phenylalanine 28 with a leucine residue produces a "fast cycling" mutant that can undergo spontaneous GTP-GDP exchange and retains the ability to hydrolyze GTP. Unfortunately this phenylalanine residue is not conserved in the ARF family of GTPases. Here we report the design and characterization of a novel activated mutant of ARF6, ARF6 T157A. In vitro studies show that ARF6 T157A can spontaneously bind and release GTP more quickly than the wild-type protein suggesting that it is a fast cycling mutant. This mutant has enhanced activity in vivo and induces cortical actin rearrangements in HeLa cells and enhanced motility in Madin-Darby canine kidney cells.


Subject(s)
ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Animals , Cell Line , Dogs , GTP Phosphohydrolases/metabolism , Mutagenesis, Site-Directed
19.
Curr Biol ; 12(10): R360-2, 2002 May 14.
Article in English | MEDLINE | ID: mdl-12015138

ABSTRACT

Membrane traffic and actin cytoskeleton dynamics are intimately linked, and GTPases of the Rho and ARF families may work together to regulate both. Recent studies have identified a family of GTPase activating proteins (GAPs) that contain both ARF-GAP and Rho-GAP domains, providing the first direct link between these two signaling pathways.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing , GTPase-Activating Proteins/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , GTPase-Activating Proteins/chemistry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...