Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 7300, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911937

ABSTRACT

Cancer stem cells (CSCs) play an important role during metastasis, but the dynamic behavior and induction mechanisms of CSCs are not well understood. Here, we employ high-resolution intravital microscopy using a CSC biosensor to directly observe CSCs in live mice with mammary tumors. CSCs display the slow-migratory, invadopod-rich phenotype that is the hallmark of disseminating tumor cells. CSCs are enriched near macrophages, particularly near macrophage-containing intravasation sites called Tumor Microenvironment of Metastasis (TMEM) doorways. Substantial enrichment of CSCs occurs on association with TMEM doorways, contributing to the finding that CSCs represent >60% of circulating tumor cells. Mechanistically, stemness is induced in non-stem cancer cells upon their direct contact with macrophages via Notch-Jagged signaling. In breast cancers from patients, the density of TMEM doorways correlates with the proportion of cancer cells expressing stem cell markers, indicating that in human breast cancer TMEM doorways are not only cancer cell intravasation portals but also CSC programming sites.


Subject(s)
Breast Neoplasms/immunology , Macrophages/immunology , Neoplastic Stem Cells/cytology , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Intravital Microscopy , Mice , Mice, SCID , Neoplasm Metastasis , Neoplastic Cells, Circulating/immunology , Neoplastic Stem Cells/immunology , Receptors, Notch/genetics , Receptors, Notch/immunology , Signal Transduction , Tumor Microenvironment/immunology
2.
Nat Chem Biol ; 16(9): 1034, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32694868

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nat Chem Biol ; 15(12): 1183-1190, 2019 12.
Article in English | MEDLINE | ID: mdl-31740825

ABSTRACT

Here we introduce Z-lock, an optogenetic approach for reversible, light-controlled steric inhibition of protein active sites. The light oxygen voltage (LOV) domain and Zdk, a small protein that binds LOV selectively in the dark, are appended to the protein of interest where they sterically block the active site. Irradiation causes LOV to change conformation and release Zdk, exposing the active site. Computer-assisted protein design was used to optimize linkers and Zdk-LOV affinity, for both effective binding in the dark, and effective light-induced release of the intramolecular interaction. Z-lock cofilin was shown to have actin severing ability in vitro, and in living cancer cells it produced protrusions and invadopodia. An active fragment of the tubulin acetylase αTAT was similarly modified and shown to acetylate tubulin on irradiation.


Subject(s)
Acetylesterase/chemistry , Actin Depolymerizing Factors/chemistry , Optogenetics , Tubulin/chemistry , Acetylation
4.
Biotechniques ; 66(3): 113-119, 2019 03.
Article in English | MEDLINE | ID: mdl-30869550

ABSTRACT

We systematically evaluated the performance and reliability of several widely used, commercially available actin-filament probes in a highly motile breast adenocarcinoma cell line to optimize the visualization of F-actin-rich dynamic lamellipodia. We evaluated four Phalloidin-fluorophores, two anti-actin antibodies, and three live-cell actin probes in five fixation conditions across three imaging platforms as a basis for the design of optimized protocols. Of the fluorescent phalloidin-dye conjugates tested, Alexa Fluor-488 Phalloidin ranked best in overall labeling of the actin cytoskeleton and maintenance of the fluorescence signal over time. Use of actin monoclonal antibodies revealed significant limitations under a variety of fixation-permeabilization conditions. Evaluation of commonly used live-cell probes provides evidence for actin filament bias, with TagRFP-Lifeact excluded from lamellipodia, but not mEGFP-Lifeact or F-tractin-EGFP.


Subject(s)
Actin Cytoskeleton/genetics , Actins/isolation & purification , Fluorescent Dyes/chemistry , Actin Cytoskeleton/chemistry , Actins/chemistry , Antibodies/chemistry , Antibodies/pharmacology , Fluorescent Dyes/pharmacology , Maleimides/chemistry , Maleimides/pharmacology , Phalloidine/chemistry , Phalloidine/pharmacology , Pseudopodia/chemistry , Pseudopodia/genetics
5.
Trends Cell Biol ; 27(8): 595-607, 2017 08.
Article in English | MEDLINE | ID: mdl-28412099

ABSTRACT

Invadopodia are a subset of invadosomes that are implicated in the integration of signals from the tumor microenvironment to support tumor cell invasion and dissemination. Recent progress has begun to define how tumor cells regulate the plasticity necessary for invadopodia to assemble and function efficiently in the different microenvironments encountered during dissemination in vivo. Exquisite mapping by many laboratories of the pathways involved in integrating diverse invadopodium initiation signals, from growth factors, to extracellular matrix (ECM) and cell-cell contact in the tumor microenvironment, has led to insight into the molecular basis of this plasticity. Here, we integrate this new information to discuss how the invadopodium is an important conductor that orchestrates tumor cell dissemination during metastasis.


Subject(s)
Cell Movement , Neoplasms/metabolism , Podosomes/metabolism , Tumor Microenvironment , Animals , Cell Communication , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Neoplasms/pathology
6.
Sci Rep ; 6: 37874, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901093

ABSTRACT

The process of intravasation involving transendothelial migration is a key step in metastatic spread. How the triple cell complex composed of a macrophage, Mena over-expressing tumor cell and endothelial cell, called the tumor microenvironment of metastasis (TMEM), facilitates tumor cell transendothelial migration is not completely understood. Previous work has shown that the physical contact between a macrophage and tumor cell results in the formation of invadopodia, actin-rich matrix degrading protrusions, important for tumor cell invasion and transendothelial migration and tumor cell dissemination. Herein, we show that the macrophage-induced invadopodium is formed through a Notch1/MenaINV signaling pathway in the tumor cell upon macrophage contact. This heterotypic tumor cell - macrophage interaction results in the upregulation of MenaINV through the activation of MENA transcription. Notch1 and MenaINV expression are required for tumor cell transendothelial migration, a necessary step during intravasation. Inhibition of the Notch signaling pathway blocked macrophage-induced invadopodium formation in vitro and the dissemination of tumor cells from the primary tumor in vivo. Our findings indicate a novel role for Notch1 signaling in the regulation of MenaINV expression and transendothelial migration and provide mechanistic information essential to the use of therapeutic inhibitors of metastasis.


Subject(s)
Macrophages/metabolism , Microfilament Proteins/metabolism , Podosomes/metabolism , Receptor, Notch1/metabolism , Transendothelial and Transepithelial Migration/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/pathology , Podosomes/physiology , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation/physiology
7.
Sci Rep ; 6: 36142, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824079

ABSTRACT

Invadopodia, actin-based protrusions of invasive carcinoma cells that focally activate extracellular matrix-degrading proteases, are essential for the migration and intravasation of tumor cells during dissemination from the primary tumor. We have previously shown that cortactin phosphorylation at tyrosine residues, in particular tyrosine 421, promotes actin polymerization at newly-forming invadopodia, promoting their maturation to matrix-degrading structures. However, the mechanism by which cells regulate the cortactin tyrosine phosphorylation-dephosphorylation cycle at invadopodia is unknown. Mena, an actin barbed-end capping protein antagonist, is expressed as various splice-isoforms. The MenaINV isoform is upregulated in migratory and invasive sub-populations of breast carcinoma cells, and is involved in tumor cell intravasation. Here we show that forced MenaINV expression increases invadopodium maturation to a far greater extent than equivalent expression of other Mena isoforms. MenaINV is recruited to invadopodium precursors just after their initial assembly at the plasma membrane, and promotes the phosphorylation of cortactin tyrosine 421 at invadopodia. In addition, we show that cortactin phosphorylation at tyrosine 421 is suppressed by the phosphatase PTP1B, and that PTP1B localization to the invadopodium is reduced by MenaINV expression. We conclude that MenaINV promotes invadopodium maturation by inhibiting normal dephosphorylation of cortactin at tyrosine 421 by the phosphatase PTP1B.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Cortactin/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Podosomes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cortactin/genetics , Female , Humans , Mice , Microfilament Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Phosphorylation/genetics , Podosomes/genetics , Podosomes/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
8.
Methods Mol Biol ; 1172: 115-23, 2014.
Article in English | MEDLINE | ID: mdl-24908299

ABSTRACT

Growth factor-dependent pairing and motility between tumor cells and tumor-associated macrophages on extracellular matrix (ECM) fibers of the tumor microenvironment have been shown to enhance intravasation and metastatic spread of breast carcinomas. We describe an in vitro motility assay that combines time-lapse wide-field microscopy and micro-patterned linear adhesive substrates to reconstitute the in vivo behavior between macrophages, tumor cells, and ECM fibers in orthotopic rodent tumor models observed by intravital imaging. Commercially available linear stripes of 650 nm dye-labeled fibronectin microlithographed onto glass cover slips are sequentially plated with fluorescently labeled MTLn3 tumor cells and bone marrow-derived macrophages and time-lapse imaged for up to 8 h. Incubation with pharmacological inhibitors during the assay can identify important paracrine or autocrine signaling pathways involved in the macrophage-tumor cell interaction. This high-resolution motility assay will lead to a more detailed description of immune cell-tumor cell behavior as well as interrogating additional cell types within the tumor microenvironment which use cytokine/growth factor paracrine signaling interactions to facilitate intravasation and metastasis.


Subject(s)
Biological Assay/methods , Cell Communication/drug effects , Epidermal Growth Factor/pharmacology , Macrophages/drug effects , Mammary Glands, Human/drug effects , Animals , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Collagen Type I/chemistry , Collagen Type I/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Female , Fibronectins/chemistry , Fibronectins/metabolism , Gefitinib , Humans , Macrophages/cytology , Macrophages/metabolism , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , Molecular Imaging , Quinazolines/pharmacology , Signal Transduction , Time-Lapse Imaging , Tyrphostins/pharmacology
9.
Nat Rev Mol Cell Biol ; 14(7): 405-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778968

ABSTRACT

Recently, a consensus has emerged that cofilin severing activity can generate free actin filament ends that are accessible for F-actin polymerization and depolymerization without changing the rate of G-actin association and dissociation at either filament end. The structural basis of actin filament severing by cofilin is now better understood. These results have been integrated with recently discovered mechanisms for cofilin activation in migrating cells, which led to new models for cofilin function that provide insights into how cofilin regulation determines the temporal and spatial control of cell behaviour.


Subject(s)
Actin Depolymerizing Factors/physiology , Cell Movement , Actin Depolymerizing Factors/chemistry , Actins/metabolism , Animals , Cell Surface Extensions/metabolism , Humans , Models, Molecular , Phosphorylation , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport
10.
Mol Biol Cell ; 24(11): 1661-75, S1-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23552693

ABSTRACT

ß1 integrin has been shown to promote metastasis in a number of tumor models, including breast, ovarian, pancreatic, and skin cancer; however, the mechanism by which it does so is poorly understood. Invasive membrane protrusions called invadopodia are believed to facilitate extracellular matrix degradation and intravasation during metastasis. Previous work showed that ß1 integrin localizes to invadopodia, but its role in regulating invadopodial function has not been well characterized. We find that ß1 integrin is required for the formation of mature, degradation-competent invadopodia in both two- and three-dimensional matrices but is dispensable for invadopodium precursor formation in metastatic human breast cancer cells. ß1 integrin is activated during invadopodium precursor maturation, and forced ß1 integrin activation enhances the rate of invadopodial matrix proteolysis. Furthermore, ß1 integrin interacts with the tyrosine kinase Arg and stimulates Arg-dependent phosphorylation of cortactin on tyrosine 421. Silencing ß1 integrin with small interfering RNA completely abrogates Arg-dependent cortactin phosphorylation and cofilin-dependent barbed-end formation at invadopodia, leading to a significant decrease in the number and stability of mature invadopodia. These results describe a fundamental role for ß1 integrin in controlling actin polymerization-dependent invadopodial maturation and matrix degradation in metastatic tumor cells.


Subject(s)
Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Integrin beta1/genetics , Protein-Tyrosine Kinases/genetics , Pseudopodia/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actins/genetics , Actins/metabolism , Cell Line, Tumor , Cell Movement , Cortactin/genetics , Cortactin/metabolism , Humans , Integrin beta1/metabolism , Phosphorylation , Protein Binding , Protein Multimerization , Protein-Tyrosine Kinases/metabolism , Pseudopodia/genetics , Pseudopodia/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tyrosine/metabolism
11.
Intravital ; 1(1): 77-85, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-24634804

ABSTRACT

In mammary tumors, intravital imaging techniques have uncovered an essential role for macrophages during tumor cell invasion and metastasis mediated by an epidermal growth factor (EGF) / colony stimulating factor-1 (CSF-1) paracrine loop. It was previously demonstrated that mammary tumors in mice derived from rat carcinoma cells (MTLn3) exhibited high velocity migration on extracellular matrix (ECM) fibers. These cells form paracrine loop-dependent linear assemblies of alternating host macrophages and tumor cells known as "streams." Here, we confirm by intravital imaging that similar streams form in close association with ECM fibers in a highly metastatic patient-derived orthotopic mammary tumor (TN1). To understand the in vivo cell motility behaviors observed in streams, an in vitro model of fibrillar tumor ECM utilizing adhesive 1D micropatterned substrates was developed. MTLn3 cells on 1D fibronectin or type I collagen substrates migrated with higher velocity than on 2D substrates and displayed enhanced lamellipodial protrusion and increased motility upon local interaction and pairing with bone marrow-derived macrophages (BMMs). Inhibitors of EGF or CSF-1 signaling disrupted this interaction and reduced tumor cell velocity and protrusion, validating the requirement for an intact paracrine loop. Both TN1 and MTLn3 cells in the presence of BMMs were capable of co-assembling into linear arrays of alternating tumor cells and BMMs that resembled streams in vivo, suggesting the stream assembly is cell autonomous and can be reconstituted on 1D substrates. Our results validate the use of 1D micropatterned substrates as a simple and defined approach to study fibrillar ECM-dependent cell pairing, migration and relay chemotaxis as a complementary tool to intravital imaging.

12.
Biochem J ; 411(2): 441-8, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18215145

ABSTRACT

Class IA PI3Ks (phosphoinositide 3-kinases) generate the secondary messenger PtdIns(3,4,5)P(3), which plays an important role in many cellular responses. The accumulation of PtdIns(3,4,5)P(3) in cell membranes is routinely measured using GFP (green fluorescent protein)-labelled PH (pleckstrin homology) domains. However, the kinetics of membrane PtdIns(3,4,5)P(3) synthesis and turnover as detected by PH domains have not been validated using an independent method. In the present study, we measured EGF (epidermal growth factor)-stimulated membrane PtdIns(3,4,5)P(3) production using a specific monoclonal anti-PtdIns(3,4,5)P(3) antibody, and compared the results with those obtained using PH-domain-dependent methods. Anti-PtdIns(3,4,5)P(3) staining rapidly accumulated at the leading edge of EGF-stimulated carcinoma cells. PtdIns(3,4,5)P(3) levels were maximal at 1 min, and returned to basal levels by 5 min. In contrast, membrane PtdIns(3,4,5)P(3) production, measured by the membrane translocation of an epitope-tagged (BTK)PH (PH domain of Bruton's tyrosine kinase), remained approx. 2-fold above basal level throughout 4-5 min of EGF stimulation. To determine the reason for this disparity, we measured the rate of PtdIns(3,4,5)P(3) hydrolysis by measuring the decay of the PtdIns(3,4,5)P(3) signal after LY294002 treatment of EGF-stimulated cells. LY294002 abolished anti-PtdIns(3,4,5)P(3) membrane staining within 10 s of treatment, suggesting that PtdIns(3,4,5)P(3) turnover occurs within seconds of synthesis. In contrast, (BTK)PH membrane recruitment, once initiated by EGF, was relatively insensitive to LY294002. These data suggest that sequestration of PtdIns(3,4,5)P(3) by PH domains may affect the apparent kinetics of PtdIns(3,4,5)P(3) accumulation and turnover; consistent with this hypothesis, we found that GRP-1 (general receptor for phosphoinositides 1) PH domains [which, like BTK, are specific for PtdIns(3,4,5)P(3)] inhibit PTEN (phosphatase and tensin homologue deleted on chromosome 10) dephosphorylation of PtdIns(3,4,5)P(3) in vitro. These data suggest that anti-PtdIns(3,4,5)P(3) antibodies are a useful tool to detect localized PtdIns(3,4,5)P(3), and illustrate the importance of using multiple approaches for the estimation of membrane phosphoinositides.


Subject(s)
Epidermal Growth Factor/pharmacology , Immunoassay/methods , Neoplasms/immunology , Neoplasms/metabolism , Phosphatidylinositol Phosphates/analysis , Phosphatidylinositol Phosphates/metabolism , Amino Acid Motifs , Animals , Cell Line, Tumor , Epitopes/immunology , Gastrointestinal Hormones/metabolism , Gene Expression Regulation , Kinetics , Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Rats , Sensitivity and Specificity
13.
J Cell Biol ; 179(6): 1247-59, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18086920

ABSTRACT

Lamellipodial protrusion and directional migration of carcinoma cells towards chemoattractants, such as epidermal growth factor (EGF), depend upon the spatial and temporal regulation of actin cytoskeleton by actin-binding proteins (ABPs). It is generally hypothesized that the activity of many ABPs are temporally and spatially regulated by PIP(2); however, this is mainly based on in vitro-binding and structural studies, and generally in vivo evidence is lacking. Here, we provide the first in vivo data that directly visualize the spatial and temporal regulation of cofilin by PIP(2) in living cells. We show that EGF induces a rapid loss of PIP(2) through PLC activity, resulting in a release and activation of a membrane-bound pool of cofilin. Upon release, we find that cofilin binds to and severs F-actin, which is coincident with actin polymerization and lamellipod formation. Moreover, our data provide evidence for how PLC is involved in the formation of protrusions in breast carcinoma cells during chemotaxis and metastasis towards EGF.


Subject(s)
Actin Depolymerizing Factors/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Epidermal Growth Factor/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Actin Depolymerizing Factors/analysis , Actins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Female , Hydrolysis , Phosphatidylinositol 4,5-Diphosphate/analysis , Protein Transport , Rats
14.
J Cell Sci ; 120(Pt 17): 3138-46, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17698922

ABSTRACT

Cell migration involves the localized extension of actin-rich protrusions, a process that requires Class I phosphoinositide 3-kinases (PI 3-kinases). Both Rac and Ras have been shown to regulate actin polymerization and activate PI 3-kinase. However, the coordination of Rac, Ras and PI 3-kinase activation during epidermal growth factor (EGF)-stimulated protrusion has not been analyzed. We examined PI 3-kinase-dependent protrusion in MTLn3 rat adenocarcinoma cells. EGF-stimulated phosphatidyl-inositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)] levels showed a rapid and persistent response, as PI 3-kinase activity remained elevated up to 3 minutes. The activation kinetics of Ras, but not Rac, coincided with those of leading-edge PtdIns(3,4,5)P(3) production. Small interfering RNA (siRNA) knockdown of K-Ras but not Rac1 abolished PtdIns(3,4,5)P(3) production at the leading edge and inhibited EGF-stimulated protrusion. However, Rac1 knockdown did inhibit cell migration, because of the inhibition of focal adhesion formation in Rac1 siRNA-treated cells. Our data show that in EGF-stimulated MTLn3 carcinoma cells, Ras is required for both PtdIns(3,4,5)P(3) production and lamellipod extension, whereas Rac1 is required for formation of adhesive structures. These data suggest an unappreciated role for Ras during protrusion, and a crucial role for Rac in the stabilization of protrusions required for cell motility.


Subject(s)
Cell Movement/physiology , Cell Surface Extensions/metabolism , Epidermal Growth Factor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , rac GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Adenocarcinoma , Animals , Cell Line, Tumor , Enzyme Activation , Phosphatidylinositol Phosphates/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rac GTP-Binding Proteins/genetics , ras Proteins/genetics
15.
J Cell Sci ; 119(Pt 14): 2871-81, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16803871

ABSTRACT

It has been demonstrated that the actin-severing activity of cofilin can be downregulated by LIM kinase (LIMK)-dependent phosphorylation at residue Ser3. Chemotactic stimulation in various cell types induces cofilin dephosphorylation, suggesting that cofilin activation in these cells occurs by a dephosphorylation mechanism. However, resting metastatic carcinoma cells have the majority of their cofilin in a dephosphorylated but largely inactive state. Stimulation with epidermal growth factor (EGF) induces an increase in cofilin activity after 60 seconds together with an increase in phosphorylated cofilin (p-cofilin), indicating that cofilin dephosphorylation is not coupled to cofilin activation in these cells. Suppression of LIMK function by inhibiting Rho-associated protein kinase (ROCK) or LIMK siRNA inhibited the EGF-induced cofilin phosphorylation but had no effect on cofilin activity or cofilin-dependent lamellipod protrusion induced by EGF. Correlation analysis revealed that cofilin, p-cofilin and LIMK are not colocalized, and changes in the location of these proteins upon stimulation with EGF indicate that they are not functionally coupled. Phospholipase C, which has been implicated in cofilin activation following stimulation with EGF, does not regulate p-cofilin levels following stimulation with EGF. Therefore, our results do not support a model for the initial activation of cofilin by dephosphorylation in response to chemoattractant stimulation in metastatic carcinoma cells.


Subject(s)
Actin Depolymerizing Factors/metabolism , Epidermal Growth Factor/pharmacology , Neoplasms/metabolism , Animals , Cytoskeleton/drug effects , Enzyme Activation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Lim Kinases , Neoplasms/pathology , Octoxynol/pharmacology , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Pseudopodia/drug effects , RNA, Small Interfering/genetics , Rats , Tumor Cells, Cultured , Type C Phospholipases/metabolism , rho-Associated Kinases
16.
J Biol Chem ; 280(7): 5836-42, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15579908

ABSTRACT

Activation of the epidermal growth factor (EGF) receptor can stimulate actin polymerization via the Arp2/3 complex using a number of signaling pathways, and specific stimulation conditions may control which pathways are activated. We have previously shown that localized stimulation of EGF receptor with EGF bound to beads results in localized actin polymerization and protrusion. Here we show that the actin polymerization is dependent upon activation of the Arp2/3 complex by neural Wiskott-Aldrich Syndrome protein (N-WASP) via Grb2 and Nck2. Suppression of Grb2 or Nck2 results in loss of localization of N-WASP at the activation site and reduced actin polymerization. Although cortactin has been found to synergize with N-WASP for Arp2/3-dependent actin polymerization in vitro, we find that cortactin can restrict N-WASP localization around EGF-bead-induced protrusions. In addition, cortactin-deficient cells have increased lamellipod dynamics but show reduced net translocation, suggesting that cortactin can contribute to cell polarity by controlling the extent of Arp2/3 activation by WASP family members and the stability of the F-actin network.


Subject(s)
Actins/chemistry , Actins/metabolism , Microfilament Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cattle , Cell Line , Cell Movement , Cortactin , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epidermal Growth Factor/pharmacology , GRB2 Adaptor Protein , Humans , Neurons/drug effects , Oncogene Proteins/metabolism , Protein Transport , Rats , Signal Transduction/drug effects , Wiskott-Aldrich Syndrome Protein , Wiskott-Aldrich Syndrome Protein Family
17.
J Biol Chem ; 278(12): 10831-41, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12522144

ABSTRACT

In response to chemoattractants neutrophils extend an actin-rich pseudopod, which imparts morphological polarity and is required for migration. Even when stimulated by an isotropic bath of chemoattractant, neutrophils exhibit persistent polarization and continued lamellipod formation at the front, suggesting that the cells establish an internal polarity. In this report, we show that perturbing lipid organization by depleting plasma membrane cholesterol levels reversibly inhibits cell polarization and migration. Among other receptor-mediated responses, beta(2) integrin up-regulation was unaffected, and initial calcium mobilization was only partially reduced by cholesterol depletion, indicating that this treatment did not abrogate initial receptor-mediated signal transduction. Interestingly, cholesterol depletion did not prevent initial activation of the GTPase Rac or an initial burst of actin polymerization, but rather it inhibited prolonged activation of Rac and sustained actin polymerization. Collectively, these findings support a model in which the plasma membrane is organized into domains that aid in amplifying the chemoattractant gradient and maintaining cell polarization.


Subject(s)
Membrane Lipids/chemistry , Neutrophils/cytology , beta-Cyclodextrins , Actins/metabolism , Calcium/metabolism , Cell Movement , Cell Polarity , Cholesterol/metabolism , Cyclodextrins/pharmacology , Humans , Hyaluronan Receptors/analysis , Membrane Lipids/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/chemistry , rac GTP-Binding Proteins/metabolism
18.
Mol Biol Cell ; 13(12): 4470-83, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12475966

ABSTRACT

The development of cell polarity in response to chemoattractant stimulation in human polymorphonuclear neutrophils (PMNs) is characterized by the rapid conversion from round to polarized morphology with a leading lamellipod at the front and a uropod at the rear. During PMN polarization, the microtubule (MT) array undergoes a dramatic reorientation toward the uropod that is maintained during motility and does not require large-scale MT disassembly or cell adhesion to the substratum. MTs are excluded from the leading lamella during polarization and motility, but treatment with a myosin light chain kinase inhibitor (ML-7) or the actin-disrupting drug cytochalasin D causes an expansion of the MT array and penetration of MTs into the lamellipod. Depolymerization of the MT array before stimulation caused 10% of the cells to lose their polarity by extending two opposing lateral lamellipodia. These multipolar cells showed altered localization of a leading lamella-specific marker, talin, and a uropod-specific marker, CD44. In summary, these results indicate that F-actin- and myosin II-dependent forces lead to the development and maintenance of MT asymmetry that may act to reinforce cell polarity during PMN migration.


Subject(s)
Microtubules/physiology , Neutrophils/cytology , Neutrophils/metabolism , Azepines/pharmacology , Cell Movement , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Fibronectins/metabolism , Humans , Hyaluronan Receptors/metabolism , Male , Microscopy, Fluorescence , Naphthalenes/pharmacology , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...