Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
BMC Cell Biol ; 18(1): 28, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851287

ABSTRACT

BACKGROUND: Eph signaling is known to induce contrasting cell behaviors such as promoting and inhibiting cell adhesion/spreading by altering F-actin organization and influencing integrin activities. We have previously demonstrated that EphA2 stimulation by ephrin-A1 promotes cell adhesion through interaction with integrins and integrin ligands in two monocyte/macrophage cell lines. Although mature mononuclear leukocytes express several members of the EphA/ephrin-A subclass, their expression has not been examined in monocytes undergoing during differentiation and maturation. RESULTS: Using RT-PCR, we have shown that EphA2, ephrin-A1, and ephrin-A2 expression was upregulated in murine bone marrow mononuclear cells during monocyte maturation. Moreover, EphA2 and EphA4 expression was induced, and ephrin-A4 expression was upregulated, in a human promyelocytic leukemia cell line, HL60, along with monocyte differentiation toward the classical CD14++CD16- monocyte subset. Using RT-PCR and flow cytometry, we have also shown that expression levels of αL, αM, αX, and ß2 integrin subunits were upregulated in HL60 cells along with monocyte differentiation while those of α4, α5, α6, and ß1 subunits were unchanged. Using a cell attachment stripe assay, we have shown that stimulation by EphA as well as ephrin-A, likely promoted adhesion to an integrin ligand-coated surface in HL60 monocytes. Moreover, EphA and ephrin-A stimulation likely promoted the formation of protrusions in HL60 monocytes. CONCLUSIONS: Notably, this study is the first analysis of EphA/ephrin-A expression during monocytic differentiation/maturation and of ephrin-A stimulation affecting monocyte adhesion to an integrin ligand-coated surface. Thus, we propose that monocyte adhesion via integrin activation and the formation of protrusions is likely promoted by stimulation of EphA as well as of ephrin-A.


Subject(s)
Cell Differentiation/physiology , Ephrins/genetics , Ephrins/metabolism , Monocytes , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Animals , Bone Marrow Cells/cytology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Surface Extensions/drug effects , Cell Surface Extensions/enzymology , Cell Surface Extensions/metabolism , Cells, Cultured , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-A1/pharmacology , HL-60 Cells , Humans , Integrins/genetics , Integrins/metabolism , Ligands , Male , Mice , Monocytes/cytology , Monocytes/enzymology , Monocytes/metabolism , Receptors, Eph Family/pharmacology , Signal Transduction/physiology , Up-Regulation/drug effects
2.
J Cell Sci ; 129(4): 743-56, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26769900

ABSTRACT

During angiogenesis, endothelial cells must coordinate matrix proteolysis with migration. Here, we tested whether the focal adhesion scaffold protein Hic-5 (also known as TGFB1I1) regulated endothelial sprouting in three dimensions. Hic-5 silencing reduced endothelial sprouting and lumen formation, and sprouting defects were rescued by the return of Hic-5 expression. Pro-angiogenic factors enhanced colocalization and complex formation between membrane type-1 matrix metalloproteinase (MT1-MMP, also known as MMP14) and Hic-5, but not between paxillin and MT1-MMP. The LIM2 and LIM3 domains of Hic-5 were necessary and sufficient for Hic-5 to form a complex with MT1-MMP. The degree of interaction between MT1-MMP and Hic-5 and the localization of the complex within detergent-resistant membrane fractions were enhanced during endothelial sprouting, and Hic-5 depletion lowered the surface levels of MT1-MMP. In addition, we observed that loss of Hic-5 partially reduced complex formation between MT1-MMP and focal adhesion kinase (FAK, also known as PTK2), suggesting that Hic-5 bridges MT1-MMP and FAK. Finally, Hic-5 LIM2-LIM3 deletion mutants reduced sprout initiation. Hic-5, MT1-MMP and FAK colocalized in angiogenic vessels during porcine pregnancy, supporting that this complex assembles during angiogenesis in vivo. Collectively, Hic-5 appears to enhance complex formation between MT1-MMP and FAK in activated endothelial cells, which likely coordinates matrix proteolysis and cell motility.


Subject(s)
Human Umbilical Vein Endothelial Cells/enzymology , Intracellular Signaling Peptides and Proteins/physiology , LIM Domain Proteins/physiology , Matrix Metalloproteinase 14/metabolism , Animals , Cell Movement , Cell Surface Extensions/enzymology , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Neovascularization, Physiologic , Pregnancy , Protein Interaction Domains and Motifs , Protein Transport , Sus scrofa
3.
J Cell Physiol ; 231(8): 1695-708, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26599499

ABSTRACT

Rab7 regulates the biogenesis of late endosomes, lysosomes, and autophagosomes. It has been proposed that a functional and physical interaction exists between Rab7 and Rac1 GTPases in CDH1 endocytosis and ruffled border formation. In FRT cells over-expressing Rab7, increased expression and activity of Rac1 was observed, whereas a reduction of Rab7 expression by RNAi resulted in reduced Rac1 activity, as measured by PAK1 phosphorylation. We found that CDH1 endocytosis was extremely reduced only in Rab7 over-expressing cells but was unchanged in Rab7 silenced cells. In Rab7 under or over-expressing cells, Rab7 and LC3B-II co-localized and co-localization in large circular structures occurred only in Rab7 over-expressing cells. These large circular structures occurred in about 10% of the cell population; some of them (61%) showed co-localization of Rab7 with cortactin and f-actin and were identified as circular dorsal ruffles (CDRs), the others as mature autophagosomes. We propose that the over-expression of Rab7 is sufficient to induce CDRs. Furthermore, in FRT cells, we found that the expression of the insoluble/active form of Rab7, rather than Rab5, or Rab8, was inducible by cAMP and that cAMP-stimulated FRT cells showed increased PAK1 phosphorylation and were no longer able to endocytose CDH1. Finally, we demonstrated that Rab7 over-expressing cells are able to endocytose exogenous thyroglobulin via pinocytosis/CDRs more efficiently than control cells. We propose that the major thyroglobulin endocytosis described in thyroid autonomous adenomas due to Rab7 increased expression, occurs via CDRs. J. Cell. Physiol. 231: 1695-1708, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cadherins/metabolism , Cell Surface Extensions/enzymology , Endocytosis , Thyroglobulin/metabolism , Thyroid Gland/enzymology , Vacuoles/enzymology , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Autophagy , Cell Line , Cell Surface Extensions/drug effects , Cortactin/metabolism , Cyclic AMP/metabolism , Endocytosis/drug effects , Microtubule-Associated Proteins/metabolism , Phosphorylation , Pinocytosis , RNA Interference , Rats, Inbred F344 , Second Messenger Systems , Thyroid Gland/cytology , Thyroid Gland/drug effects , Time Factors , Transfection , Vacuoles/drug effects , p21-Activated Kinases/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins , rac1 GTP-Binding Protein/metabolism
4.
Cell Signal ; 27(8): 1643-51, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25889896

ABSTRACT

Rho GTPases function as molecular switches that connect changes of the external environment to intracellular signaling pathways. They are active at various subcellular sites and require fast and tight regulation to fulfill their role as transducers of extracellular stimuli. New imaging technologies visualizing the active states of Rho proteins in living cells elucidated the necessity of precise spatiotemporal activation of the GTPases. The local regulation of Rho proteins is coordinated by the interaction with different guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) that turn on and off GTPase signaling to downstream effectors. GEFs and GAPs thus serve as critical signaling nodes that specify the amplitude and duration of a particular Rho signaling pathway. Despite their importance in Rho regulation, the molecular aspects underlying the spatiotemporal control of the regulators themselves are still largely elusive. In this review we will focus on the Deleted in Liver Cancer (DLC) family of RhoGAP proteins and summarize the evidence gathered over the past years revealing their different subcellular localizations that might account for isoform-specific functions. We will also highlight the importance of their tightly controlled expression in the context of neoplastic transformation.


Subject(s)
GTPase-Activating Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cell Surface Extensions/enzymology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytoplasmic Structures/enzymology , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Rho Guanine Nucleotide Exchange Factors/metabolism , Time Factors , Tumor Suppressor Proteins/genetics
5.
Pancreas ; 44(2): 331-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25426613

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of peroxiredoxin 1 (Prdx1) in the invasiveness of pancreatic ductal adenocarcinoma (PDAC) cells. METHODS: Immunohistochemistry was used to determine overexpression of Prdx1 in human PDAC tissues. Immunoprecipitation and immunocytochemistry were used to determine the interaction and intracellular distribution of Prdx1 and a member of the mitogen-activated protein kinase (MAPK) family protein, p38 MAPK, in PDAC cells. Finally, immunocytochemistry and Matrigel invasion assay were used to examine the effects of Prdx1 and p38 MAPK on the formation of cell protrusions and PDAC cell invasion. RESULTS: Prdx1 is overexpressed in human PDAC tissues. Peroxiredoxin 1 interacts with active forms of p38 MAPK, and complexes of Prdx1 and phosphorylated p38 MAPK localize at the leading edges of migrating PDAC cells. Suppression of Prdx1 decreases active p38 MAPK localized in cell protrusions and inhibits the invasiveness of PDAC cells. Consequently, suppression of Prdx1 inhibits membrane ruffling and protrusions. The p38 MAPK inhibitor SB203580 also decreases the formation of membrane protrusions and inhibits invasiveness. CONCLUSIONS: Prdx1 associates with the formation of membrane protrusions through modulation of the activity of p38 MAPK, which in turn promotes PDAC cell invasion.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Movement , Pancreatic Neoplasms/enzymology , Peroxiredoxins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Surface Extensions/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Peroxiredoxins/genetics , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Nat Cell Biol ; 16(6): 574-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24859002

ABSTRACT

Rho family GTPases control cell migration and participate in the regulation of cancer metastasis. Invadopodia, associated with invasive tumour cells, are crucial for cellular invasion and metastasis. To study Rac1 GTPase in invadopodia dynamics, we developed a genetically encoded, single-chain Rac1 fluorescence resonance energy (FRET) transfer biosensor. The biosensor shows Rac1 activity exclusion from the core of invadopodia, and higher activity when invadopodia disappear, suggesting that reduced Rac1 activity is necessary for their stability, and Rac1 activation is involved in disassembly. Photoactivating Rac1 at invadopodia confirmed this previously unknown Rac1 function. We describe here an invadopodia disassembly model, where a signalling axis involving TrioGEF, Rac1, Pak1, and phosphorylation of cortactin, causes invadopodia dissolution. This mechanism is critical for the proper turnover of invasive structures during tumour cell invasion, where a balance of proteolytic activity and locomotory protrusions must be carefully coordinated to achieve a maximally invasive phenotype.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement , Cell Surface Extensions/enzymology , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Biosensing Techniques , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Surface Extensions/pathology , Cortactin/metabolism , Extracellular Matrix/metabolism , Female , Fluorescence Resonance Energy Transfer , Guanine Nucleotide Exchange Factors/genetics , Humans , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Rats , Time Factors , Transfection , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
7.
Mol Biol Cell ; 25(13): 2061-70, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24807903

ABSTRACT

Movement through the extracellular matrix (ECM) requires cells to degrade ECM components, primarily through the action of matrix metalloproteinases (MMPs). Membrane type 1-matrix metalloproteinase (MT1-MMP) has an essential role in matrix degradation and cell invasion and localizes to subcellular degradative structures termed invadopodia. Trafficking of MT1-MMP to invadopodia is required for the function of these structures, and here we examine the role of N-ethylmaleimide-sensitive factor-activating protein receptor (SNARE)-mediated membrane traffic in the transport of MT1-MMP to invadopodia. During invadopodium formation in MDA-MB-231 human breast cancer cells, increased association of SNAP23, Syntaxin4, and vesicle-associated membrane protein 7 (VAMP7) is detected by coimmunoprecipitation. Blocking the function of these SNAREs perturbs invadopodium-based ECM degradation and cell invasion. Increased level of SNAP23-Syntaxin4-VAMP7 interaction correlates with decreased Syntaxin4 phosphorylation. These results reveal an important role for SNARE-regulated trafficking of MT1-MMP to invadopodia during cellular invasion of ECM.


Subject(s)
Cell Surface Extensions/enzymology , Matrix Metalloproteinase 14/metabolism , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , R-SNARE Proteins/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Protein Transport
9.
Cell Death Dis ; 4: e726, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868059

ABSTRACT

Lymphocytes form cell-cell connections by various mechanisms, including intercellular networks through actin-supported long-range plasma membrane (PM) extensions, termed tunneling nanotubes (TNTs). In this study, we tested in vitro whether TNTs form between human antigen-presenting B cells and T cells following cell contact and whether they enable the transfer of PM-associated proteins, such as green fluorescent protein (GFP)-tagged H-Ras (GFP-H-Ras). To address this question, we employed advanced techniques, including cell trapping by optical tweezers and live-cell imaging by 4D spinning-disk confocal microscopy. First, we showed that TNTs can form after optically trapped conjugated B and T cells are being pulled apart. Next, we determined by measuring fluorescence recovery after photobleaching that GFP-H-Ras diffuses freely in the membrane of TNTs that form spontaneously between B and T cells during coculturing. Importantly, by 4D time-lapse imaging, we showed that GFP-H-Ras-enriched PM patches accumulate at the junction between TNTs and the T-cell body and subsequently transfer to the T-cell surface. Furthermore, the PM patches adopted by T cells were enriched for another B-cell-derived transmembrane receptor, CD86. As predicted, the capacity of GFP-H-Ras to transfer between B and T cells, during coculturing, was dependent on its normal post-transcriptional lipidation and consequent PM anchorage. In summary, our data indicate that TNTs connecting B and T cells provide a hitherto undescribed route for the transfer of PM patches containing, for example, H-Ras from B to T cells.


Subject(s)
B-Lymphocytes/enzymology , Cell Surface Extensions/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , B-Lymphocytes/ultrastructure , Coculture Techniques , Diffusion , Green Fluorescent Proteins/metabolism , Humans , Jurkat Cells , Lipoylation , Microscopy, Confocal , Microscopy, Fluorescence , Nanotubes , Protein Prenylation , Protein Processing, Post-Translational , Protein Transport , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/ultrastructure , Time-Lapse Imaging
10.
J Dermatol Sci ; 70(3): 196-203, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23622765

ABSTRACT

BACKGROUND: Negative-pressure wound therapy (NPWT) is developed to facilitate wound healing at controlled subatmospheric pressures in modern medicine. Molecular mechanism for this therapy is still undefined. OBJECTIVE: This study highlights the localization and time-course of the cell division control protein 42 (Cdc42) in the cell membrane at ambient pressure (AP) and negative pressures of 75mmHg (NP75), 125mmHg (NP125) and 175mmHg (NP175). METHODS: The prepared cells were cultured in a negative pressure incubator with the same O2 and CO2 tensions at the four different pressures. The effective time, complete wound closure time, cell volume, cell viability, and the fluorescence of proliferating cell nuclear antigens (PCNA) and actins were evaluated in cells at different pressures. Wound-healing process and Cdc42 fluorescence were examined in cells with the knockdown of Cdc42. Cdc42 pathway proteins in cell membranes were analyzed after incubation at different pressures for 6 and 12h. RESULTS: The cells at NP125 had less wound closure time and obvious cell podia. Similar PCNA fluorescent intensity was observed in cells at different pressures. The Cdc42, neural Wiskott-Aldrich syndrome protein, and actin expression increased significantly (p<0.05) in plasma membranes of cells at NP125 for 12h. The knockdown of active Cdc42 resulted in the absence of Cdc42 expression at the cell leading edge. CONCLUSIONS: The activation and localization of Cdc42 pathway proteins in the cell membrane are involved in the cell podia formation in keratinocytes at NP125. NPWT may facilitate cell migration to accelerate wound healing.


Subject(s)
Cell Surface Extensions/enzymology , Keratinocytes/enzymology , Negative-Pressure Wound Therapy , Wound Healing , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Cell Culture Techniques/instrumentation , Cell Line , Cell Movement , Cell Surface Extensions/pathology , Cell Survival , Humans , Incubators , Keratinocytes/pathology , Polymerization , Pressure , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , Signal Transduction , Time Factors , Transfection , cdc42 GTP-Binding Protein/genetics
11.
Mol Cancer ; 12: 2, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23289900

ABSTRACT

BACKGROUND: ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a member of the ADAMTS family of metalloproteases. Here, we investigated mRNA and protein levels of ADAMTS-1 in normal and neoplastic tissues using qPCR, immunohistochemistry and immunoblot analyses, and we addressed the role of ADAMTS-1 in regulating migration, invasion and invadopodia formation in breast tumor cell lines. RESULTS: In a series of primary breast tumors, we observed variable levels of ADAMTS-1 mRNA expression but lower levels of ADAMTS-1 protein expression in human breast cancers as compared to normal tissue, with a striking decrease observed in high-malignancy cases (triple-negative for estrogen, progesterone and Her-2). This result prompted us to analyze the effect of ADAMTS-1 knockdown in breast cancer cells in vitro. MDA-MB-231 cells with depleted ADAMTS-1 expression demonstrated increased migration, invasion and invadopodia formation. The regulatory mechanisms underlying the effects of ADAMTS-1 may be related to VEGF, a growth factor involved in migration and invasion. MDA-MB-231 cells with depleted ADAMTS-1 showed increased VEGF concentrations in conditioned medium capable of inducing human endothelial cells (HUVEC) tubulogenesis. Furthermore, expression of the VEGF receptor (VEGFR2) was increased in MDA-MB-231 cells as compared to MCF7 cells. To further determine the relationship between ADAMTS-1 and VEGF regulating breast cancer cells, MDA-MB-231 cells with reduced expression of ADAMTS-1 were pretreated with a function-blocking antibody against VEGF and then tested in migration and invasion assays; both were partially rescued to control levels. CONCLUSIONS: ADAMTS-1 expression was decreased in human breast tumors, and ADAMTS-1 knockdown stimulated migration, invasion and invadopodia formation in breast cancer cells in vitro. Therefore, this series of experiments suggests that VEGF is involved in the effects mediated by ADAMTS-1 in breast cancer cells.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Cell Movement , Gene Expression , ADAM Proteins/genetics , ADAMTS1 Protein , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Cell Surface Extensions/enzymology , Female , Gene Knockdown Techniques , Humans , Lymphatic Metastasis , MCF-7 Cells , Middle Aged , Neoplasm Invasiveness , RNA, Small Interfering/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Time-Lapse Imaging , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
12.
Proc Natl Acad Sci U S A ; 110(3): 912-7, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23284172

ABSTRACT

RAC1 is a small, Ras-related GTPase that was recently reported to harbor a recurrent UV-induced signature mutation in melanoma, resulting in substitution of P29 to serine (RAC1(P29S)), ranking this the third most frequently occurring gain-of-function mutation in melanoma. Although the Ras family GTPases are mutated in about 30% of all cancers, mutations in the Rho family GTPases have rarely been observed. In this study, we demonstrate that unlike oncogenic Ras proteins, which are primarily activated by mutations that eliminate GTPase activity, the activated melanoma RAC1(P29S) protein maintains intrinsic GTP hydrolysis and is spontaneously activated by substantially increased inherent GDP/GTP nucleotide exchange. Determination and comparison of crystal structures for activated RAC1 GTPases suggest that RAC1(F28L)--a known spontaneously activated RAC1 mutant--and RAC1(P29S) are self-activated in distinct fashions. Moreover, the mechanism of RAC1(P29S) and RAC1(F28L) activation differs from the common oncogenic mutations found in Ras-like GTPases that abrogate GTP hydrolysis. The melanoma RAC1(P29S) gain-of-function point mutation therefore represents a previously undescribed class of cancer-related GTPase activity.


Subject(s)
Melanoma/enzymology , Melanoma/genetics , Mutation, Missense , Oncogenes , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Amino Acid Substitution , Animals , COS Cells , Cell Surface Extensions/enzymology , Chlorocebus aethiops , Crystallography, X-Ray , Enzyme Activation/genetics , Genetic Association Studies , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Kinetics , Mice , Microscopy, Fluorescence , Models, Molecular , NIH 3T3 Cells , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Static Electricity , rac1 GTP-Binding Protein/chemistry
13.
Eur J Cell Biol ; 91(11-12): 878-88, 2012.
Article in English | MEDLINE | ID: mdl-22823952

ABSTRACT

In the past decade, substantial progress has been made in understanding how Src family kinases regulate the formation and function of invadosomes. Invadosomes are organized actin-rich structures that contain an F-actin core surrounded by an adhesive ring and mediate invasive migration. Src kinases orchestrate, either directly or indirectly, each phase of the invadosome life cycle including invadosome assembly, maturation and matrix degradation and disassembly. Complex arrays of Src effector proteins are involved at different stages of invadosome maturation and their spatiotemporal activity must be tightly regulated to achieve effective invasive migration. In this review, we highlight some recent progress and the challenges of understanding how Src is regulated temporally and spatially to orchestrate the dynamics of invadosomes and mediate cell invasion.


Subject(s)
Cell Surface Extensions/enzymology , src-Family Kinases/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Movement , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Oxidation-Reduction , Signal Transduction
14.
Mol Cell Biol ; 32(8): 1374-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22331470

ABSTRACT

Our recent studies implicated key and distinct roles for the highly related RalA and RalB small GTPases (82% sequence identity) in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis and invasive and metastatic growth, respectively. How RalB may promote PDAC invasion and metastasis has not been determined. In light of known Ral effector functions in regulation of actin organization and secretion, we addressed a possible role for RalB in formation of invadopodia, actin-rich membrane protrusions that contribute to tissue invasion and matrix remodeling. We determined that a majority of KRAS mutant PDAC cell lines exhibited invadopodia and that expression of activated K-Ras is both necessary and sufficient for invadopodium formation. Invadopodium formation was not dependent on the canonical Raf-MEK-ERK effector pathway and was instead dependent on the Ral effector pathway. However, this process was more dependent on RalB than on RalA. Surprisingly, RalB-mediated invadopodium formation was dependent on RalBP1/RLIP76 but not Sec5 and Exo84 exocyst effector function. Unexpectedly, the requirement for RalBP1 was independent of its best known function as a GTPase-activating protein for Rho small GTPases. Instead, disruption of the ATPase function of RalBP1 impaired invadopodium formation. Our results identify a novel RalB-mediated biochemical and signaling mechanism for invadopodium formation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Surface Extensions/enzymology , GTPase-Activating Proteins/metabolism , ral GTP-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Surface Extensions/ultrastructure , Enzyme Activation , Humans , Neoplasm Invasiveness/ultrastructure , Pancreatic Neoplasms/pathology , Signal Transduction
15.
Free Radic Biol Med ; 52(2): 247-56, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22033009

ABSTRACT

Redox signaling contributes to the regulation of cancer cell proliferation, survival, and invasion and participates in the adaptation of cancer cells to their microenvironment. NADPH oxidases are important mediators of redox signaling in normal and cancer cells. Redox signal specificity in normal cells is in part achieved by targeting enzymes that generate reactive oxygen species to specific subcellular microdomains such as focal adhesions, dorsal ruffles, lipid rafts, or caveolae. In a similar fashion, redox signal specificity during cancer cell invasion can be regulated by targeting reactive oxygen generation to invasive microdomains such as invadopodia. Here we summarize recent advances in the understanding of the redox signaling processes that control the cancer cell proinvasive program by modulating cell adhesion, migration, and proteolysis as well as the interaction of cancer cells with the tumor microenvironment. We focus on redox signaling events mediated by invadopodia NADPH oxidase complexes and their contribution to cancer cell invasion.


Subject(s)
Membrane Microdomains/enzymology , Neoplasms/pathology , Signal Transduction , Animals , Cell Adhesion , Cell Movement , Cell Surface Extensions/enzymology , Cell Surface Extensions/metabolism , Humans , Membrane Microdomains/metabolism , NADPH Oxidases/metabolism , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Tumor Microenvironment
16.
Cell Signal ; 23(8): 1291-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21419846

ABSTRACT

Membrane ruffling is the formation of actin rich membrane protrusions, essential for cell motility. The exact mechanism of ruffling is not fully known. Using YFP and CFP fluorescent chimeras, we show for the first time a co-localization of Phospholipase D2 (PLD2) and Growth factor Receptor Bound protein-2 (Grb2) with actin-rich membrane protrusions of macrophages. Grb2 cooperates with PLD2 in enhancing membrane ruffling, whether in resting cells or in cells stimulated with the growth factor M-CSF, although in the latter an increase in dorsal ruffles was observed, consistent with receptor-ligand internalization. Cells transfected with PLD2 mutated in the PH domain (Y169F) or with Grb2 mutated in the SH2 site (R86K) negate this effect, indicating an association PLD2(Y169)-SH2-Grb2 that was confirmed by immunoprecipitation and Western blotting. The association results in enhanced PLD activity, but the lipase activity can only partially explain the formation of membrane ruffles in vivo. A third component involves the Rho-GTPase Rac2 and it is only when Rac2 is overexpressed along with PLD2 and Rac2 that a full biological effect, including actin polymerization in vivo, is obtained. The mechanism involved is, then, as follows: PLD enzymatic action, after having been increased due to the binding to Grb2-SH2 via Y169, cooperates with Rac2, and the three molecules stimulate actin polymerization and consequently, membrane ruffle formation. Since membrane ruffling precedes cell migration, the results herein provide a novel mechanism for control of membrane dynamics, crucial for the physiology of leukocytes.


Subject(s)
Cell Membrane/metabolism , GRB2 Adaptor Protein/metabolism , Phospholipase D/metabolism , rac GTP-Binding Proteins/metabolism , Actins/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Membrane/enzymology , Cell Movement , Cell Surface Extensions/enzymology , Cell Surface Extensions/metabolism , Chlorocebus aethiops , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mutation , Phospholipase D/chemistry , Phospholipase D/genetics , Protein Structure, Tertiary , RAC2 GTP-Binding Protein
17.
Cell Signal ; 23(8): 1225-34, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21342664

ABSTRACT

Podosomes are ventral adhesion structures prominent in cells of the myeloid lineage. A common aspect of these cells is that they are highly motile and must to traverse multiple tissue barriers in order to perform their functions. Recently podosomes have gathered attention from researchers as important cellular structures that can influence cell adhesion, motility and matrix remodeling. Adhesive and soluble ligands act via transmembrane receptors and propagate signals to the leukocyte cytoskeleton via small G proteins of the Rho family, tyrosine kinases and scaffold proteins and are able to induce podosome formation and rearrangements. Manipulation of the signals that regulate podosome formation and dynamics can therefore be a strategy to interfere with leukocyte functions in a multitude of pathological settings, such as infections, atherosclerosis and arthritis. Here, we review the major signaling molecules that act in the formation and regulation of podosomes.


Subject(s)
Cell Surface Extensions/metabolism , Leukocytes/metabolism , Signal Transduction , Actins/metabolism , Actins/physiology , Cell Adhesion/physiology , Cell Surface Extensions/enzymology , Leukocytes/immunology , Leukocytes/physiology , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , rho GTP-Binding Proteins/metabolism
18.
Arthritis Rheum ; 63(6): 1591-602, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21337539

ABSTRACT

OBJECTIVE: Invasive synovial fibroblasts are suggested to be the major effectors of cartilage and bone destruction, and this aggressive phenotype can lead to irreversible damage. In cancer cells, invasion across tissue boundaries and metastasis have recently been shown to depend on the capacity of the cells to breach the basement membrane, a process that was linked to the formation of the actin-rich cell protrusions called invadopodia. This study was undertaken to investigate whether arthritic synovial cells use invadopodia to invade and degrade cartilage components. METHODS: Fibroblast-like synoviocytes (FLS) from control rats or rats with collagen-induced arthritis (CIA) were cultured on fluorescent matrix in the presence of Src inhibitors or were transfected with wild-type or variants of Src kinases. The in vivo effect of Src inhibition on cartilage degradation and invasion was studied in a rat model of CIA. RESULTS: FLS from rats with CIA produced more invadopodia-like structures than did FLS from control rats, leading to increased extracellular matrix degradation. Furthermore, c-Src activation was increased in synovial cells from rats with CIA, and Src activity was found to mediate the formation of invadopodia. Pharmacologic blockade of Src activity by PP2 or intraarticular expression of a c-Src-specific short hairpin RNA in the CIA model reduced synovial membrane hyperplasia and cartilage degradation, an event linked to decreased invadopodia formation by synovial fibroblasts. CONCLUSION: This study demonstrates that inhibition of invadopodia formation in arthritic synovial cells leads to a direct effect on extracellular matrix degradation in vitro and in vivo, making invadopodia a relevant therapeutic target for interfering with this process.


Subject(s)
Arthritis, Experimental/enzymology , Cartilage/enzymology , Cell Surface Extensions/enzymology , Synovial Fluid/enzymology , src-Family Kinases/metabolism , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Cartilage/pathology , Cell Surface Extensions/pathology , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred Lew , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
19.
Eur J Cell Biol ; 90(2-3): 108-14, 2011.
Article in English | MEDLINE | ID: mdl-20605060

ABSTRACT

Invadopodia are actin-rich, adhesive protrusions that extend into and remodel the extracellular matrix. They are associated with high levels of pericellular proteolysis and correlate with the invasive capacity of a variety of tumour cells. Invadopodia have, thus, been proposed to recapitulate key events of the metastatic process. Although our understanding of the patho-physiology of invadopodia is still in its infancy, the molecular components and signalling pathways leading to their formation have received increasing attention. Recent studies have revealed that diverse membrane polarized secretory and endo/exocytic trafficking pathways converge at these structures for the delivery, in a temporally controlled and spatially confined manner, of key proteolytic enzymes. Here, we will focus our attention on MT1-MMP, a paradigmatic metalloprotease that is primarily responsible for the proteolytic activity of invadopodia. We propose that the biosynthetic/secretory pathway might be critical for the polarized delivery of MT1-MMP to invadopodia that form as "default response" whenever cells have to deal with extracellular matrix (ECM) of variable composition and stiffness. Conversely, "inducible" endo/exocytic trafficking routes might primarily control the delivery of MT1-MMP to invadopodia when cells need to respond in a fast and transient manner to soluble motogenic factors, rather than the insoluble ECM.


Subject(s)
Cell Membrane Structures/metabolism , Cell Surface Extensions/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 14/metabolism , Animals , Cell Membrane Structures/enzymology , Cell Surface Extensions/enzymology , Extracellular Matrix/enzymology , Humans , Protein Transport
20.
Eur J Cell Biol ; 90(2-3): 189-97, 2011.
Article in English | MEDLINE | ID: mdl-20573421

ABSTRACT

Myeloid cells form a first line of defense against infections. They migrate from the circulation to the infected tissues by adhering to and subsequently crossing the vascular wall. This process requires precise control and proper regulation of these interactions with the environment is therefore crucial. Podosomes are the most prominent adhesion structures in myeloid cells. Podosomes control both the adhesive and migratory properties of myeloid cells and the regulation of podosomes is key to the proper functioning of these cells. Here we discuss the regulation of podosomes by Rho GTPases, well known regulators of adhesion and migration, focusing on myeloid cells. In addition, the regulation of podosomes by GTPase regulators such as GEFs and GAPs, as well as the effects of some Rho GTPase effector pathways, will be discussed.


Subject(s)
Cell Surface Extensions/enzymology , Cytoskeleton/enzymology , Myeloid Cells/enzymology , rho GTP-Binding Proteins/metabolism , Animals , Humans , Myeloid Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL