Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 13: 324, 2019.
Article in English | MEDLINE | ID: mdl-31379509

ABSTRACT

Proper axonal growth and guidance is essential for neuron differentiation and development. Abnormal neuronal development due to genetic or epigenetic influences can contribute to neurological and mental disorders such as Down syndrome, Rett syndrome, and autism. Identification of the molecular targets that promote proper neuronal growth and differentiation may restore structural and functional neuroplasticity, thus improving functional performance in neurodevelopmental disorders. Using differentiated human neuronal progenitor cells (NPCs) derived from induced pluripotent stem cells (iPSCs), the present study demonstrates that during early stage differentiation of human NPCs, neuron-targeted overexpression constitutively active Rac1 (Rac1CA) and constitutively active Cdc42 (Cdc42CA) enhance expression of P-Cav-1, T-Cav-1, and P-cofilin and increases axonal growth. Similarly, neuron-targeted over-expression of Cav-1 (termed SynCav1) increases axonal development by increasing both axon length and volume. Moreover, inhibition of Cav-1(Y14A) phosphorylation blunts Rac1/Cdc42-mediated both axonal growth and differentiation of human NPCs and SynCav1(Y14A)-treated NPCs exhibited blunted axonal growth. These results suggest that: (1) SynCav1-mediated dendritic and axonal growth in human NPCs is dependent upon P-Cav-1, (2) P-Cav-1 is necessary for proper axonal growth during early stages of neuronal differentiation, and (3) Rac1/Cdc42CA-mediated neuronal growth is in part dependent upon P-Cav-1. In conclusion, Cav-1 phosphorylation is essential for human neuronal axonal growth during early stages of neuronal differentiation.

2.
ACS Synth Biol ; 6(7): 1257-1262, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28365983

ABSTRACT

P21-activated kinases (PAKs) are important regulators of cell motility and morphology. It has been challenging to interrogate their functions because cells adapt to genetic manipulation of PAK, and because inhibitors act on multiple PAK isoforms. Here we describe genetically encoded PAK1 analogues that can be selectively activated by the membrane-permeable small molecule rapamycin. An engineered domain inserted away from the active site responds to rapamycin to allosterically control activity of the PAK1 isoform. To examine the mechanism of rapamycin-induced PAK1 activation, we used molecular dynamics with graph theory to predict amino acids involved in allosteric communication with the active site. This analysis revealed allosteric pathways that were exploited to generate kinase switches. Activation of PAK1 resulted in transient cell spreading in metastatic breast cancer cells, and long-term dendritic spine enlargement in mouse hippocampal CA1 neurons.


Subject(s)
Allosteric Regulation/physiology , p21-Activated Kinases/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , CA1 Region, Hippocampal/metabolism , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , In Vitro Techniques , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Sirolimus/pharmacology , p21-Activated Kinases/genetics
3.
Nat Chem Biol ; 12(10): 802-809, 2016 10.
Article in English | MEDLINE | ID: mdl-27501396

ABSTRACT

Guanine-nucleotide dissociation inhibitors (GDIs) are negative regulators of Rho family GTPases that sequester the GTPases away from the membrane. Here we ask how GDI-Cdc42 interaction regulates localized Cdc42 activation for cell motility. The sensitivity of cells to overexpression of Rho family pathway components led us to a new biosensor, GDI.Cdc42 FLARE, in which Cdc42 is modified with a fluorescence resonance energy transfer (FRET) 'binding antenna' that selectively reports Cdc42 binding to endogenous GDIs. Similar antennae could also report GDI-Rac1 and GDI-RhoA interaction. Through computational multiplexing and simultaneous imaging, we determined the spatiotemporal dynamics of GDI-Cdc42 interaction and Cdc42 activation during cell protrusion and retraction. This revealed remarkably tight coordination of GTPase release and activation on a time scale of 10 s, suggesting that GDI-Cdc42 interactions are a critical component of the spatiotemporal regulation of Cdc42 activity, and not merely a mechanism for global sequestration of an inactivated pool of signaling molecules.


Subject(s)
Fluorescence Resonance Energy Transfer , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/metabolism , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , Binding Sites , HEK293 Cells , Humans , Spatio-Temporal Analysis
4.
Proc Natl Acad Sci U S A ; 112(37): E5150-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324884

ABSTRACT

Cofilin, a critical player of actin dynamics, is spatially and temporally regulated to control the direction and force of membrane extension required for cell locomotion. In carcinoma cells, although the signaling pathways regulating cofilin activity to control cell direction have been established, the molecular machinery required to generate the force of the protrusion remains unclear. We show that the cofilin phosphatase chronophin (CIN) spatiotemporally regulates cofilin activity at the cell edge to generate persistent membrane extension. We show that CIN translocates to the leading edge in a PI3-kinase-, Rac1-, and cofilin-dependent manner after EGF stimulation to activate cofilin, promotes actin free barbed end formation, accelerates actin turnover, and enhances membrane protrusion. In addition, we establish that CIN is crucial for the balance of protrusion/retraction events during cell migration. Thus, CIN coordinates the leading edge dynamics by controlling active cofilin levels to promote MTLn3 cell protrusion.


Subject(s)
Cofilin 1/physiology , Gene Expression Regulation , Phosphoprotein Phosphatases/physiology , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Breast Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , ErbB Receptors/metabolism , Humans , Microfilament Proteins/physiology , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction
5.
J Cell Biol ; 208(7): 961-74, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25800056

ABSTRACT

We previously identified Waf1 Cip1 stabilizing protein 39 (WISp39) as a binding partner for heat shock protein 90 (Hsp90). We now report that WISp39 has an essential function in the control of directed cell migration, which requires WISp39 interaction with Hsp90. WISp39 knockdown (KD) resulted in the loss of directional motility of mammalian cells and profound changes in cell morphology, including the loss of a single leading edge. WISp39 binds Coronin 1B, known to regulate the Arp2/3 complex and Cofilin at the leading edge. WISp39 preferentially interacts with phosphorylated Coronin 1B, allowing it to complex with Slingshot phosphatase (SSH) to dephosphorylate and activate Cofilin. WISp39 also regulates Arp2/3 complex localization at the leading edge. WISp39 KD-induced morphological changes could be rescued by overexpression of Coronin 1B together with a constitutively active Cofilin mutant. We conclude that WISp39 associates with Hsp90, Coronin 1B, and SSH to regulate Cofilin activation and Arp2/3 complex localization at the leading edge.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Immunophilins/metabolism , Microfilament Proteins/metabolism , Actin Depolymerizing Factors/genetics , Cell Line, Tumor , Cell Movement/genetics , Enzyme Activation/genetics , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Immunophilins/genetics , Microfilament Proteins/biosynthesis , Phosphoprotein Phosphatases , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering , Tacrolimus Binding Proteins
6.
Mol Cell Biol ; 35(8): 1401-13, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25666508

ABSTRACT

Directional cell motility is essential for normal development and physiology, although how motile cells spatiotemporally activate signaling events remains largely unknown. Here, we have characterized an adhesion and signaling unit comprised of protein tyrosine phosphatase (PTP)-PEST and the extracellular matrix (ECM) adhesion receptor ß8 integrin that plays essential roles in directional cell motility. ß8 integrin and PTP-PEST form protein complexes at the leading edge of migrating cells and balance patterns of Rac1 and Cdc42 signaling by controlling the subcellular localization and phosphorylation status of Rho GDP dissociation inhibitor 1 (RhoGDI1). Translocation of Src-phosphorylated RhoGDI1 to the cell's leading edge promotes local activation of Rac1 and Cdc42, whereas dephosphorylation of RhoGDI1 by integrin-bound PTP-PEST promotes RhoGDI1 release from the membrane and sequestration of inactive Rac1/Cdc42 in the cytoplasm. Collectively, these data reveal a finely tuned regulatory mechanism for controlling signaling events at the leading edge of directionally migrating cells.


Subject(s)
Cell Movement , Integrin beta Chains/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Male , Mice , Phosphorylation , Protein Binding , Protein Interaction Maps , Ubiquitin-Protein Ligases/metabolism , rac1 GTP-Binding Protein/metabolism
7.
Cell Stem Cell ; 14(4): 523-34, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24702998

ABSTRACT

The creation of induced pluripotent stem cells (iPSCs) from somatic cells by ectopic expression of transcription factors has galvanized the fields of regenerative medicine and developmental biology. Here, we report a kinome-wide RNAi-based analysis to identify kinases that regulate somatic cell reprogramming to iPSCs. We prepared 3,686 small hairpin RNA (shRNA) lentiviruses targeting 734 kinase genes covering the entire mouse kinome and individually examined their effects on iPSC generation. We identified 59 kinases as barriers to iPSC generation and characterized seven of them further. We found that shRNA-mediated knockdown of the serine/threonine kinases TESK1 or LIMK2 promoted mesenchymal-to-epithelial transition, decreased COFILIN phosphorylation, and disrupted Actin filament structures during reprogramming of mouse embryonic fibroblasts. Similarly, knockdown of TESK1 in human fibroblasts also promoted reprogramming to iPSCs. Our study reveals the breadth of kinase networks regulating pluripotency and identifies a role for cytoskeletal remodeling in modulating the somatic cell reprogramming process.


Subject(s)
Cell Differentiation , Cellular Reprogramming/genetics , Cytoskeleton/metabolism , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Protein Serine-Threonine Kinases/genetics , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Humans , Induced Pluripotent Stem Cells/metabolism , Lim Kinases/antagonists & inhibitors , Lim Kinases/genetics , Lim Kinases/metabolism , Mice , Microscopy, Confocal , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Teratoma/metabolism , Teratoma/pathology
8.
Small GTPases ; 4(3): 174-9, 2013.
Article in English | MEDLINE | ID: mdl-23648943

ABSTRACT

Vesicle trafficking is crucial for delivery of membrane compartments as well as signaling molecules to specific sites on the plasma membrane for regulation of diverse processes such as cell division, migration, polarity establishment and secretion. Rho GTPases are well-studied signaling molecules that regulate actin cytoskeleton in response to variety of extracellular stimuli. Increasing amounts of evidence suggest that Rho proteins play a critical role in vesicle trafficking in both the exocytic and endocytic pathways; however, the molecular mechanism underlying the process remains largely unclear. We recently defined a mechanism of action for RhoA in membrane trafficking pathways through regulation of the octameric complex exocyst in a manuscript published in Developmental Cell. We have shown that microtubule-associated RhoA-activating factor GEF-H1 is involved in endocytic and excocytic vesicle trafficking. GEF-H1 activates RhoA in response to RalA GTPase, which in turn regulates the localization and the assembly of exocyst components and exocytosis. Our work defines a mechanism for RhoA activation in response to RalA signaling and during vesicle trafficking. These results provide a framework for understanding how RhoA/GEF-H1 regulates the coordination of actin and microtubule cytoskeleton modulation and vesicle trafficking during migration and cell division.


Subject(s)
Cytoskeleton/metabolism , Exosomes/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Actins/metabolism , Animals , Cell Movement , Exocytosis , Microtubules/metabolism , RNA Interference , Rats , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , ral GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Mol Biol Cell ; 24(3): 194-209, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23223568

ABSTRACT

Spatial control of RhoGTPase-inactivating GAP components remains largely enigmatic. We describe a brain-specific RhoGAP splice variant, BARGIN (BGIN), which comprises a combination of BAR, GAP, and partial CIN phosphatase domains spliced from adjacent SH3BP1 and CIN gene loci. Excision of BGIN exon 2 results in recoding of a 42-amino acid N-terminal stretch. The partial CIN domain is a poly-ubiquitin (poly-Ub)-binding module that facilitates BGIN distribution to membranous and detergent-insoluble fractions. Poly-Ub/BGIN interactions support BGIN-mediated inactivation of a membranous Rac1 population, which consequently inactivates membrane-localized Rac1 effector systems such as reactive oxygen species (ROS) generation by the Nox1 complex. Given that Ub aggregate pathology and proteotoxicity are central themes in various neurodegenerative disorders, we investigated whether BGIN/Rac1 signaling could be involved in neurodegenerative proteotoxicity. BGIN/Ub interactions are observed through colocalization in tangle aggregates in the Alzheimer's disease (AD) brain. Moreover, enhanced BGIN membrane distribution correlates with reduced Rac1 activity in AD brain tissue. Finally, BGIN contributes to Rac1 inhibition and ROS generation in an amyloid precursor protein (APP) proteotoxicity model. These results suggest that BGIN/poly-Ub interactions enhance BGIN membrane distribution and relay poly-Ub signals to enact Rac1 inactivation, and attenuation of Rac1 signaling is partially dependent on BGIN in a proteotoxic APP context.


Subject(s)
GTPase-Activating Proteins/genetics , Phosphoric Monoester Hydrolases/genetics , Polyubiquitin/metabolism , rac1 GTP-Binding Protein/metabolism , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Cell Membrane/enzymology , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Leupeptins/pharmacology , Molecular Sequence Data , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Proteasome Inhibitors/pharmacology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics
10.
PLoS One ; 7(8): e41342, 2012.
Article in English | MEDLINE | ID: mdl-22876286

ABSTRACT

Productive protrusions allowing motile cells to sense and migrate toward a chemotactic gradient of reactive oxygen species (ROS) require a tight control of the actin cytoskeleton. However, the mechanisms of how ROS affect cell protrusion and actin dynamics are not well elucidated yet. We show here that ROS induce the formation of a persistent protrusion. In migrating epithelial cells, protrusion of the leading edge requires the precise regulation of the lamellipodium and lamella F-actin networks. Using fluorescent speckle microscopy, we showed that, upon ROS stimulation, the F-actin retrograde flow is enhanced in the lamellipodium. This event coincides with an increase of cofilin activity, free barbed ends formation, Arp2/3 recruitment, and ERK activity at the cell edge. In addition, we observed an acceleration of the F-actin flow in the lamella of ROS-stimulated cells, which correlates with an enhancement of the cell contractility. Thus, this study demonstrates that ROS modulate both the lamellipodium and the lamella networks to control protrusion efficiency.


Subject(s)
Actins/physiology , Cell Surface Extensions/physiology , Reactive Oxygen Species/metabolism , Actin Depolymerizing Factors/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Surface Extensions/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/pharmacology , Nonmuscle Myosin Type IIA/metabolism , Protein Multimerization , Protein Transport , Tropomyosin/metabolism
11.
Dev Cell ; 23(2): 397-411, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22898781

ABSTRACT

The exocyst complex plays a critical role in targeting and tethering vesicles to specific sites of the plasma membrane. These events are crucial for polarized delivery of membrane components to the cell surface, which is critical for cell motility and division. Though Rho GTPases are involved in regulating actin dynamics and membrane trafficking, their role in exocyst-mediated vesicle targeting is not very clear. Herein, we present evidence that depletion of GEF-H1, a guanine nucleotide exchange factor for Rho proteins, affects vesicle trafficking. Interestingly, we found that GEF-H1 directly binds to exocyst component Sec5 in a Ral GTPase-dependent manner. This interaction promotes RhoA activation, which then regulates exocyst assembly/localization and exocytosis. Taken together, our work defines a mechanism for RhoA activation in response to RalA-Sec5 signaling and involvement of GEF-H1/RhoA pathway in the regulation of vesicle trafficking.


Subject(s)
Exocytosis , Guanine Nucleotide Exchange Factors/metabolism , Microtubules/metabolism , rhoA GTP-Binding Protein/metabolism , Biological Transport , Enzyme Activation , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Microscopy, Electron, Transmission , Protein Binding , Rho Guanine Nucleotide Exchange Factors , Signal Transduction
12.
J Cell Biol ; 193(7): 1289-303, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21708980

ABSTRACT

Cell motility requires the spatial and temporal coordination of forces in the actomyosin cytoskeleton with extracellular adhesion. The biochemical mechanism that coordinates filamentous actin (F-actin) assembly, myosin contractility, adhesion dynamics, and motility to maintain the balance between adhesion and contraction remains unknown. In this paper, we show that p21-activated kinases (Paks), downstream effectors of the small guanosine triphosphatases Rac and Cdc42, biochemically couple leading-edge actin dynamics to focal adhesion (FA) dynamics. Quantitative live cell microscopy assays revealed that the inhibition of Paks abolished F-actin flow in the lamella, displaced myosin IIA from the cell edge, and decreased FA turnover. We show that, by controlling the dynamics of these three systems, Paks regulate the protrusive activity and migration of epithelial cells. Furthermore, we found that expressing Pak1 was sufficient to overcome the inhibitory effects of excess adhesion strength on cell motility. These findings establish Paks as critical molecules coordinating cytoskeletal systems for efficient cell migration.


Subject(s)
Actins/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Nonmuscle Myosin Type IIA/metabolism , p21-Activated Kinases/physiology , Actins/genetics , Actins/ultrastructure , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Kinetics , Nonmuscle Myosin Type IIA/analysis , Paxillin/analysis , Paxillin/metabolism , Phenotype , Potoroidae
13.
Eur J Cell Biol ; 90(2-3): 164-71, 2011.
Article in English | MEDLINE | ID: mdl-20609497

ABSTRACT

NADPH oxidase (Nox) family enzymes are one of the main sources of cellular reactive oxygen species (ROS), which have been implicated in several physiological and pathophysiological processes. To date seven members of this family have been reported, including Nox1-5 and Duox1 and 2. With the exception of Nox2, the regulation of the Nox enzymes is still poorly understood. Nox1 is highly expressed in the colon, and requires two cytosolic regulators, the organizer subunit NoxO1 and the activator subunit NoxA1, as well as the binding of Rac1 GTPase, for its activity. Recently, we identified the c-Src substrate proteins Tks4 and Tks5 as functional members of a p47(phox)-related organizer superfamily. As a functional consequence of this interaction, Nox1 localizes to invadopodia, actin-rich membrane protrusions of cancer cells which facilitate pericellular proteolysis and invasive behavior. Here, we report that Tks4 and Tks5 directly bind to NoxA1. Moreover, the integrity of the N-terminal PRR of NoxA1 is essential for this direct interaction with the Tks proteins. When the PRR in NoxA1 is disrupted, Tks proteins cannot bind NoxA1 and lose their ability to support Nox1-dependent ROS generation. Consistent with this, Tks4 and Tks5 are unable to act as organizers for Nox2 because of their inability to interact with p67(phox), which lacks the N-terminal PRR, thus conferring a unique specificity to Tks4 and 5. Taken together, these results clarify the molecular basis for the interaction between NoxA1 and the Tks proteins and may provide new insights into the pharmacological design of a more effective anti-metastatic strategy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Sequence , Cell Line, Tumor , HEK293 Cells , Humans , Immunoprecipitation , Molecular Sequence Data , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Proline-Rich Protein Domains , Protein Binding , Sequence Alignment , Transfection
14.
Mol Biol Cell ; 21(23): 4287-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20943948

ABSTRACT

The NADPH oxidase family, consisting of Nox1-5 and Duox1-2, catalyzes the regulated formation of reactive oxygen species (ROS). Highly expressed in the colon, Nox1 needs the organizer subunit NoxO1 and the activator subunit NoxA1 for its activity. The tyrosine kinase c-Src is necessary for the formation of invadopodia, phosphotyrosine-rich structures which degrade the extracellular matrix (ECM). Many Src substrates are invadopodia components, including the novel Nox1 organizer Tks4 and Tks5 proteins. Nox1-dependent ROS generation is necessary for the maintenance of functional invadopodia in human colon cancer cells. However, the signals and the molecular machinery involved in the redox-dependent regulation of invadopodia formation remain unclear. Here, we show that the interaction of NoxA1 and Tks proteins is dependent on Src activity. Interestingly, the abolishment of Src-mediated phosphorylation of Tyr110 on NoxA1 and of Tyr508 on Tks4 blocks their binding and decreases Nox1-dependent ROS generation. The contemporary presence of Tks4 and NoxA1 unphosphorylable mutants blocks SrcYF-induced invadopodia formation and ECM degradation, while the overexpression of Tks4 and NoxA1 phosphomimetic mutants rescues this phenotype. Taken together, these results elucidate the role of c-Src activity on the formation of invadopodia and may provide insight into the mechanisms of tumor formation in colon cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Surface Extensions , Proto-Oncogene Proteins pp60(c-src)/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , Blotting, Western , Colonic Neoplasms , Extracellular Matrix/metabolism , Gene Expression , Genes, src , Humans , Immunoprecipitation , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation , Phosphotyrosine , Signal Transduction
15.
ACS Chem Biol ; 5(10): 981-93, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20715845

ABSTRACT

The NADPH oxidase (Nox) proteins catalyze the regulated formation of reactive oxygen species (ROS), which play key roles as signaling molecules in several physiological and pathophysiological processes. ROS generation by the Nox1 member of the Nox family is necessary for the formation of extracellular matrix (ECM)-degrading, actin-rich cellular structures known as invadopodia. Selective inhibition of Nox isoforms can provide reversible, mechanistic insights into these cellular processes in contrast to scavenging or inhibition of ROS production. Currently no specific Nox inhibitors have been described. Here, by high-throughput screening, we identify a subset of phenothiazines, 2-acetylphenothiazine (here referred to as ML171) (and its related 2-(trifluoromethyl)-phenothiazine) as nanomolar, cell-active, and specific Nox1 inhibitors that potently block Nox1-dependent ROS generation, with only marginal activity on other cellular ROS-producing enzymes and receptors including the other Nox isoforms. ML171 also blocks the ROS-dependent formation of ECM-degrading invadopodia in colon cancer cells. Such effects can be reversed by overexpression of Nox1 protein, which is suggestive of a selective mechanism of inhibition of Nox1 by this compound. These results elucidate the relevance of Nox1-dependent ROS generation in mechanisms of cancer invasion and define ML171 as a useful Nox1 chemical probe and potential therapeutic agent for inhibition of cancer cell invasion.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/pathology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Neoplasm Invasiveness/prevention & control , Phenothiazines/pharmacology , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , High-Throughput Screening Assays , Humans , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Phenothiazines/chemistry , Small Molecule Libraries/chemistry
16.
Cell Signal ; 21(12): 1974-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19765647

ABSTRACT

The regulation of Rho GTPase activities and expression is critical in the development and function of the kidney. Rho GTPase activities and cytosol-membrane cycling are regulated by Rho GDP Dissociation Inhibitor (RhoGDI), and RhoGDI knockout mice develop defects in kidney structure and function that lead to death due to renal failure. It is therefore important to understand the changes in RhoGDI-regulated Rho GTPase activities and cell morphology that lead to kidney failure in RhoGDI (-/-) mice. Here, we characterize a renal mesangial cell line derived from the RhoGDI (-/-) mouse in which we verify the absence of GDI proteins. In the absence of RhoGDI, we show an increase in the specific activity of Rac1, and to a lesser extent, RhoA and Cdc42 GTPases in these cells. This is accompanied by a compensatory decrease in the steady-state protein levels of Rho GTPases. Morphological analysis of RhoGDI (-/-) mesangial cells reveals a decrease in cell spreading and in focal contacts compared to wild-type cells. Finally, RhoGDI (-/-) mesangial cells show a decreased ability to proliferate and survive. These functional and structural changes are likely to contribute to the defects in renal architecture and function observed in the RhoGDI (-/-) mouse.


Subject(s)
Cell Proliferation , Guanine Nucleotide Dissociation Inhibitors/metabolism , Mesangial Cells/cytology , Animals , Cell Line , Cytoskeleton/ultrastructure , Gene Expression , Guanine Nucleotide Dissociation Inhibitors/genetics , Mesangial Cells/metabolism , Mice , Phenotype , rho GTP-Binding Proteins/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors
17.
FASEB J ; 22(6): 1737-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18198211

ABSTRACT

The signal transduction pathways involved in neuronal death are not well understood. Neuroglobin (Ngb), a recently discovered vertebrate globin expressed predominantly in the brain, shows increased expression in neurons in response to oxygen deprivation and protects neurons from ischemic and hypoxic death. The mechanism of this neuroprotection is unclear. We examined the surface distribution of raft membrane microdomains in cortical neuron cultures during hypoxia using the raft marker cholera toxin B (CTx-B) subunit. Mechanistically, we demonstrate that hypoxia induces rapid polarization of somal membranes and aggregation of microdomains with the subjacent mitochondrial network. This signaling complex is formed well before neurons commit to die, consistent with an early role in death signal transduction. Neurons from Ngb-overexpressing transgenic (Ngb-Tg) mice do not undergo microdomain polarization or mitochondrial aggregation in response to, and are resistant to death from hypoxia. We link the protective actions of Ngb to inhibition of Pak1 kinase activity and Rac1-GDP-dissociation inhibitor disassociation, and inhibition of actin assembly and death-signaling module polarization.


Subject(s)
Globins/physiology , Hypoxia/metabolism , Nerve Tissue Proteins/physiology , Neurons/pathology , Signal Transduction , Actins/antagonists & inhibitors , Animals , Cerebral Cortex , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Mice , Mice, Transgenic , Neuroglobin , p21-Activated Kinases/antagonists & inhibitors
18.
Dev Cell ; 13(5): 646-662, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17981134

ABSTRACT

Protrusion of the leading edge of migrating epithelial cells requires precise regulation of two actin filament (F-actin) networks, the lamellipodium and the lamella. Cofilin is a downstream target of Rho GTPase signaling that promotes F-actin cycling through its F-actin-nucleating, -severing, and -depolymerizing activity. However, its function in modulating lamellipodium and lamella dynamics, and the implications of these dynamics for protrusion efficiency, has been unclear. Using quantitative fluorescent speckle microscopy, immunofluorescence, and electron microscopy, we establish that the Rac1/Pak1/LIMK1 signaling pathway controls cofilin activity within the lamellipodium. Enhancement of cofilin activity accelerates F-actin turnover and retrograde flow, resulting in widening of the lamellipodium. This is accompanied by increased spatial overlap of the lamellipodium and lamella networks and reduced cell-edge protrusion efficiency. We propose that cofilin functions as a regulator of cell protrusion by modulating the spatial interaction of the lamellipodium and lamella in response to upstream signals.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/physiology , Epithelial Cells/physiology , Microfilament Proteins/physiology , Pseudopodia/physiology , p21-Activated Kinases/physiology , Cell Line , Cell Movement , Fluorescent Antibody Technique , Humans , Lim Kinases/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism
19.
Mol Biol Cell ; 17(11): 4760-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16943322

ABSTRACT

Rho GTPases (Rac, Rho, and Cdc42) play important roles in regulating cell function through their ability to coordinate the actin cytoskeleton, modulate the formation of signaling reactive oxidant species, and control gene transcription. Activation of Rho GTPase signaling pathways requires the regulated release of Rho GTPases from RhoGDI complexes, followed by their reuptake after membrane cycling. We show here that Src kinase binds and phosphorylates RhoGDI both in vitro and in vivo at Tyr156. Analysis of Rho GTPase-RhoGDI complexes using in vitro assays of complexation and in vivo by coimmunoprecipitation analysis indicates that Src-mediated phosphorylation of Tyr156 causes a dramatic decrease in the ability of RhoGDI to form a complex with RhoA, Rac1, or Cdc42. Phosphomimetic mutation of Tyr156-->Glu results in the constitutive association of RhoGDI(Y156E) with the plasma membrane and/or associated cortical actin. Substantial cortical localization of tyrosine-phosphorylated RhoGDI is also observed in fibroblasts expressing active Src, where it is most evident in podosomes and regions of membrane ruffling. Expression of membrane-localized RhoGDI(Y156E) mutant is associated with enhanced cell spreading and membrane ruffling. These results suggest that Src-mediated RhoGDI phosphorylation is a novel physiological mechanism for regulating Rho GTPase cytosol membrane-cycling and activity.


Subject(s)
Cell Membrane Structures/metabolism , Cytosol/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , rho GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Transformed , Fibroblasts/cytology , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Protein Transport , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors
20.
Methods Mol Biol ; 332: 269-79, 2006.
Article in English | MEDLINE | ID: mdl-16878699

ABSTRACT

The recognition that Rho guanosine triphosphatases (GTPases) (Rho, Rac, and Cdc42) play important regulatory roles in many areas of cell biology has made the ability to measure their activity in cells an important biological tool. Because Rho GTPases become activated by conversion from guanosine diphosphate-bound states to guanosine triphosphate (GTP)-bound forms, affinity-based methods to detect the formation of GTP-Rho GTPases have been developed and are widely used for the purpose of assessing Rho GTPase activities in biological studies.


Subject(s)
Biological Assay/methods , rho GTP-Binding Proteins/metabolism , Apoptosis Regulatory Proteins , Enzyme Activation , GTP-Binding Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , p21-Activated Kinases , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...