Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 21(4): 667-676, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35086953

ABSTRACT

Prostate cancer remains a major cause of male mortality. Genetic alteration of the PI3K/AKT/mTOR pathway is one of the key events in tumor development and progression in prostate cancer, with inactivation of the PTEN tumor suppressor being very common in this cancer type. Extensive evaluation has been performed on the therapeutic potential of PI3K/AKT/mTOR inhibitors and the resistance mechanisms arising in patients with PTEN-mutant background. However, in patients with a PTEN wild-type phenotype, PI3K/AKT/mTOR inhibitors have not demonstrated efficacy, and this remains an area of clinical unmet need. In this study, we have investigated the response of PTEN wild-type prostate cancer cell lines to the dual PI3K/mTOR inhibitor DS-7423 alone or in combination with HER2 inhibitors or mGluR1 inhibitors. Upon treatment with the dual PI3K/mTOR inhibitor DS-7423, PTEN wild-type prostate cancer CWR22/22RV1 cells upregulate expression of the proteins PSMA, mGluR1, and the tyrosine kinase receptor HER2, while PTEN-mutant LNCaP cells upregulate androgen receptor and HER3. PSMA, mGluR1, and HER2 exert control over one another in a positive feedback loop that allows cells to overcome treatment with DS-7423. Concomitant targeting of PI3K/mTOR with either HER2 or mGluR1 inhibitors results in decreased cell survival and tumor growth in xenograft studies. Our results suggest a novel therapeutic possibility for patients with PTEN wild-type PI3K/AKT-mutant prostate cancer based in the combination of PI3K/mTOR blockade with HER2 or mGluR1 inhibitors.


Subject(s)
Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , Cell Line, Tumor , Cell Proliferation , Humans , MTOR Inhibitors , Male , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Metabotropic Glutamate , TOR Serine-Threonine Kinases/metabolism
2.
Front Mol Biosci ; 8: 661516, 2021.
Article in English | MEDLINE | ID: mdl-34568423

ABSTRACT

Tertiary lymphoid structures (TLSs) develop in non-lymphatic tissue in chronic inflammation and cancer. TLS can mature to lymph node (LN) like structures with germinal centers and associated vasculature. TLS neogenesis in cancer is highly varied and tissue dependent. The role of TLS in adaptive antitumor immunity is of great interest. However, data also show that TLS can play a role in cancer metastasis. The importance of lymphatics in cancer distant metastasis is clear yet the precise detail of how various immunosurveillance mechanisms interplay within TLS and/or draining LN is still under investigation. As part of the tumor lymphatics, TLS vasculature can provide alternative routes for the establishment of the pre-metastatic niche and cancer dissemination. The nature of the cytokine and chemokine signature at the heart of TLS induction can be key in determining the success of antitumor immunity or in promoting cancer invasiveness. Understanding the biochemical and biomechanical factors underlying TLS formation and the resulting impact on the primary tumor will be key in deciphering cancer metastasis and in the development of the next generation of cancer immunotherapeutics.

3.
Cell Rep ; 27(7): 1967-1978.e4, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091437

ABSTRACT

Lymphatic vasculature is crucial for metastasis in triple-negative breast cancer (TNBC); however, cellular and molecular drivers controlling lymphovascular metastasis are poorly understood. We define a macrophage-dependent signaling cascade that facilitates metastasis through lymphovascular remodeling. TNBC cells instigate mRNA changes in macrophages, resulting in ß4 integrin-dependent adhesion to the lymphovasculature. ß4 integrin retains macrophages proximal to lymphatic endothelial cells (LECs), where release of TGF-ß1 drives LEC contraction via RhoA activation. Macrophages promote gross architectural changes to lymphovasculature by increasing dilation, hyperpermeability, and disorganization. TGF-ß1 drives ß4 integrin clustering at the macrophage plasma membrane, further promoting macrophage adhesion and demonstrating the dual functionality of TGF-ß1 signaling in this context. ß4 integrin-expressing macrophages were identified in human breast tumors, and a combination of vascular-remodeling macrophage gene signature and TGF-ß signaling scores correlates with metastasis. We postulate that future clinical strategies for patients with TNBC should target crosstalk between ß4 integrin and TGF-ß1.


Subject(s)
Integrin beta4/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/pathology , Macrophages/metabolism , Transforming Growth Factor beta1/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Integrin beta4/genetics , Lymphatic Metastasis , Lymphatic Vessels/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/genetics , Transforming Growth Factor beta1/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Kalinin
4.
Cell Rep ; 24(3): 630-641, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021161

ABSTRACT

The immunosuppressive transmembrane protein PD-L1 was shown to traffic via the multivesicular body (MVB) and to be released on exosomes. A high-content siRNA screen identified the endosomal sorting complexes required for transport (ESCRT)-associated protein ALIX as a regulator of both EGFR activity and PD-L1 surface presentation in basal-like breast cancer (BLBC) cells. ALIX depletion results in prolonged and enhanced stimulation-induced EGFR activity as well as defective PD-L1 trafficking through the MVB, reduced exosomal secretion, and its redistribution to the cell surface. Increased surface PD-L1 expression confers an EGFR-dependent immunosuppressive phenotype on ALIX-depleted cells. An inverse association between ALIX and PD-L1 expression was observed in human breast cancer tissues, while an immunocompetent mouse model of breast cancer revealed that ALIX-deficient tumors are larger and show an increased immunosuppressive environment. Our data suggest that ALIX modulates immunosuppression through regulation of PD-L1 and EGFR and may, therefore, present a diagnostic and therapeutic target for BLBC.


Subject(s)
B7-H1 Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , ErbB Receptors/metabolism , Immunosuppression Therapy , Animals , Biosensing Techniques , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cellular Microenvironment , Exosomes/metabolism , Exosomes/ultrastructure , Female , Fluorescence Resonance Energy Transfer , Humans , Ligands , Mice, Inbred BALB C
5.
Cancer Res ; 77(5): 1083-1096, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28082403

ABSTRACT

Cancer cells tend to metastasize first to tumor-draining lymph nodes, but the mechanisms mediating cancer cell invasion into the lymphatic vasculature remain little understood. Here, we show that in the human breast tumor microenvironment (TME), the presence of increased numbers of RORγt+ group 3 innate lymphoid cells (ILC3) correlates with an increased likelihood of lymph node metastasis. In a preclinical mouse model of breast cancer, CCL21-mediated recruitment of ILC3 to tumors stimulated the production of the CXCL13 by TME stromal cells, which in turn promoted ILC3-stromal interactions and production of the cancer cell motile factor RANKL. Depleting ILC3 or neutralizing CCL21, CXCL13, or RANKL was sufficient to decrease lymph node metastasis. Our findings establish a role for RORγt+ILC3 in promoting lymphatic metastasis by modulating the local chemokine milieu of cancer cells in the TME. Cancer Res; 77(5); 1083-96. ©2017 AACR.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Orphan Nuclear Receptors/immunology , Animals , Cell Line, Tumor , Chemokine CCL21/immunology , Chemokine CXCL13/immunology , Female , Humans , Immunity, Innate , Lymphatic Metastasis , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
6.
J Nucl Med ; 57(5): 765-70, 2016 05.
Article in English | MEDLINE | ID: mdl-26635342

ABSTRACT

UNLABELLED: Locoregional recurrence of breast cancer poses significant clinical problems because of frequent inoperability once the chest wall is involved. Early detection of recurrence by molecular imaging agents against therapeutically targetable receptors, such as c-Met, would be of potential benefit. The aim of this study was to assess (18)F-AH113804, a peptide-based molecular imaging agent with high affinity for human c-Met, for the detection of early-stage locoregional recurrence in a human basal-like breast cancer model, HCC1954. METHODS: HCC1954 tumor-bearing xenograft models were established, and (18)F-AH113804 was administered. Distribution of radioactivity was determined via PET at 60 min after radiotracer injection. PET and CT images were acquired 10 d after tumor inoculation, to establish baseline distribution and uptake, and then on selected days after surgical tumor resection. CT images and caliper were used to determine the tumor volume. Radiotracer uptake was assessed by (18)F-AH113804 PET imaging. c-Met expression was assessed by immunofluorescence imaging of tumor samples and correlated with (18)F-AH113804 PET imaging results. RESULTS: Baseline uptake of (18)F-AH113804, determined in tumor-bearing animals after 10 d, was approximately 2-fold higher in the tumor than in muscle tissue or the contralateral mammary fat pad. The tumor growth rate, determined from CT images, was comparable between the animals with recurrent tumors, with detection of tumors of low volume (<10 mm(3)) only possible by day 20 after tumor resection. (18)F-AH113804 PET detected local tumor recurrence as early as 6 d after surgery in the recurrent tumor-bearing animals and exhibited significantly higher (18)F-AH113804 uptake (in comparison to mammary fatty tissue), with a target-to-background (muscle) ratio of approximately 3:1 (P < 0.01). The c-Met expression of individual resected tumor samples, determined by immunofluorescence, correlated with the respective (18)F-AH113804 imaging signals (r = 0.82, P < 0.05). CONCLUSION: (18)F-AH113804 PET provides a new diagnostic tool for the detection of c-Met-expressing primary tumor and has potential utility for the detection of locoregional recurrence from an early stage.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Neoplasm Recurrence, Local , Positron-Emission Tomography , Proto-Oncogene Proteins c-met/metabolism , Animals , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Staging , Tomography, X-Ray Computed
7.
Biomed Opt Express ; 6(2): 277-96, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25780724

ABSTRACT

We demonstrate diffraction limited multiphoton imaging in a massively parallel, fully addressable time-resolved multi-beam multiphoton microscope capable of producing fluorescence lifetime images with sub-50ps temporal resolution. This imaging platform offers a significant improvement in acquisition speed over single-beam laser scanning FLIM by a factor of 64 without compromising in either the temporal or spatial resolutions of the system. We demonstrate FLIM acquisition at 500 ms with live cells expressing green fluorescent protein. The applicability of the technique to imaging protein-protein interactions in live cells is exemplified by observation of time-dependent FRET between the epidermal growth factor receptor (EGFR) and the adapter protein Grb2 following stimulation with the receptor ligand. Furthermore, ligand-dependent association of HER2-HER3 receptor tyrosine kinases was observed on a similar timescale and involved the internalisation and accumulation or receptor heterodimers within endosomes. These data demonstrate the broad applicability of this novel FLIM technique to the spatio-temporal dynamics of protein-protein interaction.

8.
J Cell Sci ; 128(2): 251-65, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25413351

ABSTRACT

Podosomes are integrin-containing adhesion structures commonly found in migrating leukocytes of the monocytic lineage. The actin cytoskeletal organisation of podosomes is based on a WASP- and Arp2/3-mediated mechanism. WASP also associates with a second protein, WIP (also known as WIPF1), and they co-localise in podosome cores. Here, we report for the first time that WIP can be phosphorylated on tyrosine residues and that tyrosine phosphorylation of WIP is a trigger for release of WASP from the WIP-WASP complex. Using a knockdown approach together with expression of WIP phosphomimics, we show that in the absence of WIP-WASP binding, cellular WASP is rapidly degraded, leading to disruption of podosomes and a failure of cells to degrade an underlying matrix. In the absence of tyrosine phosphorylation, the WIP-WASP complex remains intact and podosome lifetimes are extended. A screen of candidate kinases and inhibitor-based assays identified Bruton's tyrosine kinase (Btk) as a regulator of WIP tyrosine phosphorylation. We conclude that tyrosine phosphorylation of WIP is a crucial regulator of WASP stability and function as an actin-nucleation-promoting factor.


Subject(s)
Cytoskeletal Proteins/metabolism , Extracellular Matrix/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Cytoskeletal Proteins/genetics , Extracellular Matrix/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Phosphorylation/genetics , Podosomes/metabolism , Protein Binding , Protein-Tyrosine Kinases/genetics , Tyrosine/metabolism , Wiskott-Aldrich Syndrome Protein/genetics
9.
PLoS One ; 9(10): e110695, 2014.
Article in English | MEDLINE | ID: mdl-25360776

ABSTRACT

We present a novel imaging system combining total internal reflection fluorescence (TIRF) microscopy with measurement of steady-state acceptor fluorescence anisotropy in order to perform live cell Förster Resonance Energy Transfer (FRET) imaging at the plasma membrane. We compare directly the imaging performance of fluorescence anisotropy resolved TIRF with epifluorescence illumination. The use of high numerical aperture objective for TIRF required correction for induced depolarization factors. This arrangement enabled visualisation of conformational changes of a Raichu-Cdc42 FRET biosensor by measurement of intramolecular FRET between eGFP and mRFP1. Higher activity of the probe was found at the cell plasma membrane compared to intracellularly. Imaging fluorescence anisotropy in TIRF allowed clear differentiation of the Raichu-Cdc42 biosensor from negative control mutants. Finally, inhibition of Cdc42 was imaged dynamically in live cells, where we show temporal changes of the activity of the Raichu-Cdc42 biosensor.


Subject(s)
Cell Membrane/metabolism , Fluorescence Polarization/methods , Fluorescence Resonance Energy Transfer/methods , Molecular Imaging/methods , Biosensing Techniques , Humans , MCF-7 Cells , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
10.
Sci Signal ; 7(339): ra78, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25140053

ABSTRACT

The epidermal growth factor receptor (EGFR) is a member of the ErbB family that can promote the migration and proliferation of breast cancer cells. Therapies that target EGFR can promote the dimerization of EGFR with other ErbB receptors, which is associated with the development of drug resistance. Understanding how interactions among ErbB receptors alter EGFR biology could provide avenues for improving cancer therapy. We found that EGFR interacted directly with the CYT1 and CYT2 variants of ErbB4 and the membrane-anchored intracellular domain (mICD). The CYT2 variant, but not the CYT1 variant, protected EGFR from ligand-induced degradation by competing with EGFR for binding to a complex containing the E3 ubiquitin ligase c-Cbl and the adaptor Grb2. Cultured breast cancer cells overexpressing both EGFR and ErbB4 CYT2 mICD exhibited increased migration. With molecular modeling, we identified residues involved in stabilizing the EGFR dimer. Mutation of these residues in the dimer interface destabilized the complex in cells and abrogated growth factor-stimulated cell migration. An exon array analysis of 155 breast tumors revealed that the relative mRNA abundance of the ErbB4 CYT2 variant was increased in ER+ HER2- breast cancer patients, suggesting that our findings could be clinically relevant. We propose a mechanism whereby competition for binding to c-Cbl in an ErbB signaling heterodimer promotes migration in response to a growth factor gradient.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , ErbB Receptors/metabolism , Proteolysis , Receptor, ErbB-4/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Female , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Protein Structure, Tertiary , Protein Transport/genetics , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Receptor, ErbB-4/genetics
11.
PLoS One ; 9(2): e88251, 2014.
Article in English | MEDLINE | ID: mdl-24523880

ABSTRACT

Podosomes are highly dynamic actin-rich adhesive structures formed predominantly by cells of the monocytic lineage, which degrade the extracellular matrix. They consist of a core of F-actin and actin-regulating proteins, surrounded by a ring of adhesion-associated proteins such as vinculin. We have characterised the structure of podosomes in macrophages, particularly the structure of the ring, using three super-resolution fluorescence microscopy techniques: stimulated emission depletion microscopy, structured illumination microscopy and localisation microscopy. Rather than being round, as previously assumed, we found the vinculin ring to be created from relatively straight strands of vinculin, resulting in a distinctly polygonal shape. The strands bind preferentially at angles between 116° and 135°. Furthermore, adjacent vinculin strands are observed nucleating at the corners of the podosomes, suggesting a mechanism for podosome growth.


Subject(s)
Macrophages/cytology , Macrophages/metabolism , Microscopy, Fluorescence/methods , Microscopy/methods , Vinculin/chemistry , Actin Cytoskeleton , Actins/metabolism , Cell Adhesion , Cell Line , Cell Movement/physiology , Humans , Image Processing, Computer-Assisted , Talin/chemistry
12.
Blood ; 121(13): 2542-52, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23305739

ABSTRACT

Megakaryocytes give rise to platelets via extension of proplatelet arms, which are released through the vascular sinusoids into the bloodstream. Megakaryocytes and their precursors undergo varying interactions with the extracellular environment in the bone marrow during their maturation and positioning in the vascular niche. We demonstrate that podosomes are abundant in primary murine megakaryocytes adherent on multiple extracellular matrix substrates, including native basement membrane. Megakaryocyte podosome lifetime and density, but not podosome size, are dependent on the type of matrix, with podosome lifetime dramatically increased on collagen fibers compared with fibrinogen. Podosome stability and dynamics depend on actin cytoskeletal dynamics but not matrix metalloproteases. However, podosomes degrade matrix and appear to be important for megakaryocytes to extend protrusions across a native basement membrane. We thus demonstrate for the first time a fundamental requirement for podosomes in megakaryocyte process extension across a basement membrane, and our results suggest that podosomes may have a role in proplatelet arm extension or penetration of basement membrane.


Subject(s)
Basement Membrane/physiology , Cell Surface Extensions/physiology , Extracellular Matrix/metabolism , Megakaryocytes/physiology , Animals , Basement Membrane/metabolism , Blood Platelets/metabolism , Blood Platelets/physiology , Cell Surface Extensions/metabolism , Cells, Cultured , Fibrinogen/metabolism , HEK293 Cells , Humans , Infant, Newborn , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Type II/metabolism
13.
Haematologica ; 97(5): 687-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22133775

ABSTRACT

Podosomes are actin-based adhesions involved in migration of cells that have to cross tissue boundaries such as myeloid cells. The Wiskott Aldrich Syndrome Protein regulates de novo actin polymerization during podosome formation and it is cleaved by the protease calpain during podosome disassembly. The mechanisms that may induce the Wiskott Aldrich Syndrome Protein cleavage by calpain remain undetermined. We now report that in myeloid cells, tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein-tyrosine291 (Human)/tyrosine293 (mouse) not only enhances Wiskott Aldrich Syndrome Protein-mediated actin polymerization but also promotes its calpain-dependent degradation during podosome disassembly. We also show that activation of the Wiskott Aldrich Syndrome Protein leading to podosome formation occurs independently of tyrosine phosphorylation in spleen-derived dendritic cells. We conclude that tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein integrates dynamics of actin and cell adhesion proteins during podosome disassembly required for mobilization of myeloid cells during the immune response.


Subject(s)
Actin Cytoskeleton/physiology , Calpain/metabolism , Cell Membrane Structures/metabolism , Tyrosine/metabolism , Wiskott-Aldrich Syndrome Protein/physiology , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Fluorescent Antibody Technique , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism , Phosphorylation , Protein Binding
14.
Nat Methods ; 9(2): 195-200, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138825

ABSTRACT

We describe a localization microscopy analysis method that is able to extract results in live cells using standard fluorescent proteins and xenon arc lamp illumination. Our Bayesian analysis of the blinking and bleaching (3B analysis) method models the entire dataset simultaneously as being generated by a number of fluorophores that may or may not be emitting light at any given time. The resulting technique allows many overlapping fluorophores in each frame and unifies the analysis of the localization from blinking and bleaching events. By modeling the entire dataset, we were able to use each reappearance of a fluorophore to improve the localization accuracy. The high performance of this technique allowed us to reveal the nanoscale dynamics of podosome formation and dissociation throughout an entire cell with a resolution of 50 nm on a 4-s timescale.


Subject(s)
Bayes Theorem , Nanotechnology , Cell Line, Tumor , Humans
15.
Sci Signal ; 4(201): ra81, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22126964

ABSTRACT

Natural killer (NK) cells kill tumor cells and virally infected cells, and an effective NK cell response requires processes, such as motility, recognition, and directional secretion, that rely on cytoskeletal rearrangement. The Rho guanosine triphosphatase (GTPase) Cdc42 coordinates cytoskeletal reorganization downstream of many receptors. The Rho-related GTPase from plants 1 (ROP1) exhibits oscillatory activation behavior at the apical plasma membrane of growing pollen tubes; however, a similar oscillation in Rho GTPase activity has so far not been demonstrated in mammalian cells. We hypothesized that oscillations in Cdc42 activity might occur within NK cells as they interact with target cells. Through fluorescence lifetime imaging of a Cdc42 biosensor, we observed that in live NK cells forming immunological synapses with target cells, Cdc42 activity oscillated after exhibiting an initial increase. We used protein-protein interaction networks and structural databases to identify candidate proteins that controlled Cdc42 activity, leading to the design of a targeted short interfering RNA screen. The guanine nucleotide exchange factors RhoGEF6 and RhoGEF7 were necessary for Cdc42 activation within the NK cell immunological synapse. In addition, the kinase Akt and the p85α subunit of phosphoinositide 3-kinase (PI3K) were required for Cdc42 activation, the periodicity of the oscillation in Cdc42 activity, and the subsequent polarization of cytotoxic vesicles toward target cells. Given that PI3Ks are targets of tumor therapies, our findings suggest the need to monitor innate immune function during the course of targeted therapy against these enzymes.


Subject(s)
Immunological Synapses/immunology , Killer Cells, Natural/immunology , RNA, Small Interfering , cdc42 GTP-Binding Protein/immunology , Biological Clocks/genetics , Biological Clocks/immunology , Cell Line, Transformed , Cell- and Tissue-Based Therapy/methods , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/immunology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Cytoskeleton/genetics , Cytoskeleton/immunology , Cytoskeleton/metabolism , Enzyme Activation/genetics , Enzyme Activation/immunology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunity, Cellular/genetics , Immunity, Innate/genetics , Immunological Synapses/enzymology , Immunological Synapses/genetics , Killer Cells, Natural/enzymology , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Proto-Oncogene Proteins c-akt , Rho Guanine Nucleotide Exchange Factors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
16.
EMBO J ; 30(9): 1705-18, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21427700

ABSTRACT

Chemotactic migration of fibroblasts towards growth factors, such as during development and wound healing, requires precise spatial coordination of receptor signalling. However, the mechanisms regulating this remain poorly understood. Here, we demonstrate that ß1 integrins are required both for fibroblast chemotaxis towards platelet-derived growth factor (PDGF) and growth factor-induced dorsal ruffling. Mechanistically, we show that ß1 integrin stabilises and spatially regulates the actin nucleating endocytic protein neuronal Wiskott­Aldrich syndrome protein (N-WASP) to facilitate PDGF receptor traffic and directed motility. Furthermore, we show that in intact cells, PDGF binding leads to rapid activation of ß1 integrin within newly assembled actin-rich membrane ruffles. Active ß1 in turn controls assembly of N-WASP complexes with both Cdc42 and WASP-interacting protein (WIP), the latter of which acts to stabilise the N-WASP. Both of these protein complexes are required for PDGF internalisation and fibroblast chemotaxis downstream of ß1 integrins. This represents a novel mechanism by which integrins cooperate with growth factor receptors to promote localised signalling and directed cell motility.


Subject(s)
Chemotaxis/physiology , Fibroblasts/physiology , Integrin beta1/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Becaplermin , Blotting, Western , Endocytosis , Flow Cytometry , Fluorescence Resonance Energy Transfer , Gene Knockout Techniques , Immunoprecipitation , Lentivirus , Mice , NIH 3T3 Cells , Oligonucleotides/genetics , Plasmids/genetics , Platelet-Derived Growth Factor , Proto-Oncogene Proteins c-sis
17.
Eur J Cell Biol ; 90(2-3): 213-23, 2011.
Article in English | MEDLINE | ID: mdl-20952093

ABSTRACT

In immature dendritic cells (DCs) podosomes form and turn over behind the leading edge of migrating cells. The Arp2/3 complex activator Wiskott-Aldrich Syndrome Protein (WASP) localises to the actin core of forming podosomes together with WASP-Interacting Protein (WIP). A second weaker Arp2/3 activator, cortactin, is also found at podosomes where it has been proposed to participate in matrix metalloproteinase (MMP) secretion. We have previously shown that WIP(-/-) DCs are unable to make podosomes. WIP binds to cortactin and in this report we address whether WIP regulates cortactin-mediated MMP activity. Using DCs derived from splenic murine precursors, we found that wild-type cells were able to localise MMPs at podosomes where matrix degradation takes place. In contrast, WIP(-/-) DCs remain able to synthesise MMPs but do not degrade the extracellular matrix. Infection of WIP KO DCs with lentivirus expressing WIP restored both podosome formation and their ability to degrade the extracellular matrix, implicating WIP-induced podosomes as foci of functional MMP location. When WIP KO DCs were infected with a mutant form of WIP lacking the cortactin-binding domain (WIPΔ110-170) DCs were only able to elaborate disorganised podosomes that were unable to support MMP-mediated matrix degradation. Taken together, these results suggest a role for WIP not only in WASP-mediated actin polymerisation and podosome formation, but also in cortactin-mediated extracellular matrix degradation by MMPs.


Subject(s)
Carrier Proteins/metabolism , Cell Surface Extensions/metabolism , Cortactin/metabolism , Dendritic Cells/metabolism , Extracellular Matrix/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cytoskeletal Proteins , Dendritic Cells/enzymology , Enzyme Precursors/metabolism , Fibronectins/metabolism , Gelatinases/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Structure, Tertiary
18.
Eur J Cell Biol ; 90(2-3): 198-204, 2011.
Article in English | MEDLINE | ID: mdl-20609498

ABSTRACT

The integrin-dependent migration of myeloid cells requires tight coordination between actin-based cell membrane protrusion and integrin-mediated adhesion to form a stable leading edge. Under this mode of migration, polarised myeloid cells including dendritic cells, macrophages and osteoclasts develop podosomes that sustain the extending leading edge. Podosome integrity and dynamics vary in response to changes in the physical and biochemical properties of the cell environment. In the current article we discuss the role of various factors in initiation and stability of podosomes and the roles of the Wiskott Aldrich Syndrome Protein (WASP) in this process. We discuss recent data indicating that in a cellular context WASP is crucial not only for localised actin polymerisation at the leading edge and in podosome cores but also for coordination of integrin clustering and activation during podosome formation and disassembly.


Subject(s)
Cell Polarity/physiology , Cytoskeleton/physiology , Myeloid Cells/cytology , Wiskott-Aldrich Syndrome Protein/physiology , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Mice
19.
Mol Cell Biol ; 29(10): 2730-47, 2009 May.
Article in English | MEDLINE | ID: mdl-19273601

ABSTRACT

Cdc42 and Rac family GTPases are important regulators of morphology, motility, and polarity in a variety of mammalian cell types. However, comprehensive analysis of their roles in the morphological and behavioral aspects of chemotaxis within a single experimental system is still lacking. Here we demonstrate using a direct viewing chemotaxis assay that of all of the Cdc42/Rac1-related GTPases expressed in primary fibroblasts, Cdc42, Rac1, and RhoG are required for efficient migration towards platelet-derived growth factor (PDGF). During migration, Cdc42-, Rac1-, and RhoG-deficient cells show aberrant morphology characterized as cell elongation and cell body rounding, loss of lamellipodia, and formation of thick membrane extensions, respectively. Analysis of individual cell trajectories reveals that cell speed is significantly reduced, as well as persistence, but to a smaller degree, while the directional response to the gradient of PDGF is not affected. Combined knockdown of Cdc42, Rac1, and RhoG results in greater inhibition of cell speed than when each protein is knocked down alone, but the cells are still capable of migrating toward PDGF. We conclude that, Cdc42, Rac1, and RhoG function cooperatively during cell migration and that, while each GTPase is implicated in the control of morphology and cell speed, these and other Cdc42/Rac-related GTPases are not essential for the directional response toward PDGF.


Subject(s)
Cell Movement/physiology , Chemotaxis/physiology , Fibroblasts/physiology , Platelet-Derived Growth Factor/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Becaplermin , Biological Assay/instrumentation , Biological Assay/methods , Cell Shape , Cells, Cultured , Fibroblasts/cytology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-sis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , cdc42 GTP-Binding Protein/genetics , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
20.
J Cell Sci ; 121(Pt 20): 3403-12, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18827014

ABSTRACT

mDia1 belongs to the formin family of proteins that share FH1 and FH2 domains. Although formins play a critical role in the formation of many actin-based cellular structures, the physiological regulation of formin-mediated actin assembly within the cell is still unknown. Here we show that cells possess an acute actin polymer restoration mechanism involving mDia1. By using single-molecule live-cell imaging, we found that several treatments including low-dose G-actin-sequestering drugs and unpolymerizable actin mutants activate mDia1 to initiate fast directional movement. The FH2 region, the core domain for actin nucleation, is sufficient to respond to latrunculin B (LatB) to increase its actin nucleation frequency. Simulation analysis revealed an unexpected paradoxical effect of LatB that leads to a several fold increase in free G-actin along with an increase in total G-actin. These results indicate that in cells, the actin nucleation frequency of mDia1 is enhanced not only by Rho, but also strongly through increased catalytic efficiency of the FH2 domain. Consistently, frequent actin nucleation by mDia1 was found around sites of vigorous actin disassembly. Another major actin nucleator, the Arp2/3 complex, was not affected by the G-actin increase induced by LatB. Taken together, we propose that transient accumulation of G-actin works as a cue to promote mDia1-catalyzed actin nucleation to execute rapid reassembly of actin filaments.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Actin Cytoskeleton/genetics , Actin-Related Protein 2-3 Complex/genetics , Actins/genetics , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carrier Proteins/genetics , Cell Line , Formins , Mice , Protein Structure, Tertiary/physiology , Thiazolidines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...