Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Res ; 76(16): 4720-4727, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325650

ABSTRACT

Other than an association with HPV infection, little is known about the genetic alterations determining the development of penile cancer. Although penile cancer is rare in the developed world, it presents a significant burden in developing countries. Here, we report the findings of whole-exome sequencing (WES) to determine the somatic mutational landscape of penile cancer. WES was performed on penile cancer and matched germline DNA from 27 patients undergoing surgical resection. Targeted resequencing of candidate genes was performed in an independent 70 patient cohort. Mutation data were also integrated with DNA methylation and copy-number information from the same patients. We identified an HPV-associated APOBEC mutation signature and an NpCpG signature in HPV-negative disease. We also identified recurrent mutations in the novel penile cancer tumor suppressor genes CSN1(GPS1) and FAT1 Expression of CSN1 mutants in cells resulted in colocalization with AGO2 in cytoplasmic P-bodies, ultimately leading to the loss of miRNA-mediated gene silencing, which may contribute to disease etiology. Our findings represent the first comprehensive analysis of somatic alterations in penile cancer, highlighting the complex landscape of alterations in this malignancy. Cancer Res; 76(16); 4720-7. ©2016 AACR.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Intracellular Signaling Peptides and Proteins/genetics , Penile Neoplasms/genetics , COP9 Signalosome Complex , DNA Copy Number Variations , DNA Methylation , DNA Mutational Analysis , Fluorescent Antibody Technique , Humans , Male , Mutation , Polymerase Chain Reaction
2.
PLoS One ; 10(5): e0126478, 2015.
Article in English | MEDLINE | ID: mdl-25945928

ABSTRACT

After birth, stem cells in the subventricular zone (SVZ) generate neuroblasts that migrate along the rostral migratory stream (RMS) to become interneurons in the olfactory bulb (OB). This migration is crucial for the proper integration of newborn neurons in a pre-existing synaptic network and is believed to play a key role in infant human brain development. Many regulators of neuroblast migration have been identified; however, still very little is known about the intracellular molecular mechanisms controlling this process. Here, we have investigated the function of drebrin, an actin-binding protein highly expressed in the RMS of the postnatal mammalian brain. Neuroblast migration was monitored both in culture and in brain slices obtained from electroporated mice by time-lapse spinning disk confocal microscopy. Depletion of drebrin using distinct RNAi approaches in early postnatal mice affects neuroblast morphology and impairs neuroblast migration and orientation in vitro and in vivo. Overexpression of drebrin also impairs migration along the RMS and affects the distribution of neuroblasts at their final destination, the OB. Drebrin phosphorylation on Ser142 by Cyclin-dependent kinase 5 (Cdk5) has been recently shown to regulate F-actin-microtubule coupling in neuronal growth cones. We also investigated the functional significance of this phosphorylation in RMS neuroblasts using in vivo postnatal electroporation of phosphomimetic (S142D) or non-phosphorylatable (S142A) drebrin in the SVZ of mouse pups. Preventing or mimicking phosphorylation of S142 in vivo caused similar effects on neuroblast dynamics, leading to aberrant neuroblast branching. We conclude that drebrin is necessary for efficient migration of SVZ-derived neuroblasts and propose that regulated phosphorylation of drebrin on S142 maintains leading process stability for polarized migration along the RMS, thus ensuring proper neurogenesis.


Subject(s)
Lateral Ventricles/cytology , Neurons/metabolism , Neuropeptides/metabolism , Olfactory Bulb/cytology , Actins/metabolism , Animals , Cell Movement , Cyclin-Dependent Kinase 5/metabolism , Electroporation , Female , Humans , Lateral Ventricles/growth & development , Male , Mice , Microtubules/metabolism , Neurogenesis/physiology , Neurons/cytology , Neuropeptides/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
3.
J Cell Biol ; 202(5): 793-806, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23979715

ABSTRACT

Drebrin is an actin filament (F-actin)-binding protein with crucial roles in neuritogenesis and synaptic plasticity. Drebrin couples dynamic microtubules to F-actin in growth cone filopodia via binding to the microtubule-binding +TIP protein EB3 and organizes F-actin in dendritic spines. Precisely how drebrin interacts with F-actin and how this is regulated is unknown. We used cellular and in vitro assays with a library of drebrin deletion constructs to map F-actin binding sites. We discovered two domains in the N-terminal half of drebrin-a coiled-coil domain and a helical domain-that independently bound to F-actin and cooperatively bundled F-actin. However, this activity was repressed by an intramolecular interaction relieved by Cdk5 phosphorylation of serine 142 located in the coiled-coil domain. Phospho-mimetic and phospho-dead mutants of serine 142 interfered with neuritogenesis and coupling of microtubules to F-actin in growth cone filopodia. These findings show that drebrin contains a cryptic F-actin-bundling activity regulated by phosphorylation and provide a mechanistic model for microtubule-F-actin coupling.


Subject(s)
Actins/metabolism , Cyclin-Dependent Kinase 5/metabolism , Neuropeptides/metabolism , Animals , COS Cells , Chlorocebus aethiops , Growth Cones/metabolism , Humans , Microtubules/metabolism , Models, Biological , Mutant Proteins/metabolism , Neurogenesis , Neuropeptides/chemistry , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Structure, Tertiary , Pseudopodia/metabolism , Rabbits , Rats , Stress Fibers/metabolism , Stress Fibers/ultrastructure , Structure-Activity Relationship
4.
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
5.
J Cell Sci ; 123(Pt 21): 3629-38, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20971702

ABSTRACT

Adhesion is fundamental to the survival and function of many different cell types, and regulates basic events such as mitosis, cell survival and migration, in both embryonic and adult organisms. Cell-matrix adhesion also regulates the dynamic interplay between cells and surrounding tissues during processes such as immune cell recruitment, wound healing and cancer cell metastasis. The study of cell adhesion has gained momentum in recent years, in large part because of the emergence of imaging techniques that have facilitated detailed analysis of the molecular composition and dynamics of the structures involved. In this Commentary, we discuss the recent application of different imaging techniques to study cell-matrix adhesions, emphasising common strategies used for the analysis of adhesion dynamics both in cells in culture and in whole organisms.


Subject(s)
Cell Adhesion , Microscopy/methods , Animals , Fluorescence Resonance Energy Transfer , Focal Adhesions/metabolism , Humans , Microscopy/trends
6.
J Cell Biol ; 189(2): 369-83, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20404115

ABSTRACT

Integrins are fundamental to the control of protrusion and motility in adherent cells. However, the mechanisms by which specific members of this receptor family cooperate in signaling to cytoskeletal and adhesion dynamics are poorly understood. Here, we show that the loss of beta3 integrin in fibroblasts results in enhanced focal adhesion turnover and migration speed but impaired directional motility on both 2D and 3D matrices. These motility defects are coupled with an increased rate of actin-based protrusion. Analysis of downstream signaling events reveals that loss of beta3 integrin results in a loss of protein kinase A-dependent phosphorylation of the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP). Dephosphorylated VASP in beta3-null cells is preferentially associated with Rap1-GTP-interacting adaptor molecule (RIAM) both in vitro and in vivo, which leads to enhanced formation of a VASP-RIAM complex at focal adhesions and subsequent increased binding of talin to beta1 integrin. These data demonstrate a novel mechanism by which alphavbeta3 integrin acts to locally suppress beta1 integrin activation and regulate protrusion, adhesion dynamics, and persistent migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Integrin alphaVbeta3/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Actins/genetics , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion Molecules/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Focal Adhesions/metabolism , Humans , Integrin alphaVbeta3/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Phosphoproteins/genetics , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Talin/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
7.
Methods Mol Biol ; 591: 311-23, 2010.
Article in English | MEDLINE | ID: mdl-19957138

ABSTRACT

Cell surface receptors are crucial in the regulation of a wide variety of signalling responses to extracellular stimuli such as soluble growth factors or matrix proteins. To respond effectively to rapidly changing environmental cues, many receptors are rapidly endo- or exo-cytosed to either subcellular or membrane compartments or they recruit specific intracellular binding partners. Recent advances in microscopy techniques have made it possible to study receptor behaviour in live cells to gain a better understanding of dynamics, binding partners and sub-cellular localisation. Here we describe several common currently used techniques to study receptor behaviour in living cells.


Subject(s)
Molecular Imaging/methods , Receptors, Cell Surface/metabolism , Cell Survival , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/metabolism , Humans , Kinetics , Microscopy, Confocal , Photobleaching , Protein Multimerization , Protein Structure, Quaternary , Protein Transport , Receptors, Cell Surface/chemistry
8.
Int J Biochem Cell Biol ; 40(11): 2397-409, 2008.
Article in English | MEDLINE | ID: mdl-18485788

ABSTRACT

Adhesion to the extracellular matrix (ECM) is a fundamental requirement for survival, differentiation and migration of numerous cell types during both embryonic development and adult homeostasis. Different types of adhesion structures have been classified in different cell types or tissue environments. The best studied of these are focal adhesions which are found on a wide variety of cell types and will be the main focus of this review. Many years of research into the control of adhesion has yielded a wealth of information regarding the complexity of protein composition of these critical points of cell:ECM contact. Moreover, it has emerged that adhesions are not only highly ordered, but also dynamic structures under tight spatial control at the subcellular level to enable localised responses to extracellular cues. However, it is only in the last decade that the relative dynamics of these adhesion proteins have been closely studied. Here we provide an overview of the imaging strategies that have been developed and implemented to study the intricacies and hierarchy of protein turnover within focal adhesions. The caveats of employing these imaging techniques, as well as future directions will also be discussed.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Focal Adhesions/metabolism , Extracellular Matrix/metabolism , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , Focal Adhesions/chemistry , Imaging, Three-Dimensional , Microscopy/methods
SELECTION OF CITATIONS
SEARCH DETAIL