Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
PLoS One ; 10(10): e0140511, 2015.
Article in English | MEDLINE | ID: mdl-26509500

ABSTRACT

Focal adhesions are integrin-based structures that link the actin cytoskeleton and the extracellular matrix. They play an important role in various cellular functions such as cell signaling, cell motility and cell shape. To ensure and fine tune these different cellular functions, adhesions are regulated by a large number of proteins. The LIM domain protein zyxin localizes to focal adhesions where it participates in the regulation of the actin cytoskeleton. Because of its interactions with a variety of binding partners, zyxin has been proposed to act as a molecular scaffold. Here, we studied the interaction of zyxin with such a partner: Tes. Similar to zyxin, Tes harbors three highly conserved LIM domains of which the LIM1 domain directly interacts with zyxin. Using different zyxin variants in pull-down assays and ectopic recruitment experiments, we identified the Tes binding site in zyxin and showed that four highly conserved amino acids are crucial for its interaction with Tes. Based upon these findings, we used a zyxin mutant defective in Tes-binding to assess the functional consequences of abrogating the zyxin-Tes interaction in focal adhesions. Performing fluorescence recovery after photobleaching, we showed that zyxin recruits Tes to focal adhesions and modulates its turnover in these structures. However, we also provide evidence for zyxin-independent localization of Tes to focal adhesions. Zyxin increases focal adhesion numbers and reduces focal adhesion lifetimes, but does so independent of Tes. Quantitative analysis showed that the loss of interaction between zyxin and Tes affects the process of cell spreading. We conclude that zyxin influences focal adhesion dynamics, that it recruits Tes and that this interaction is functional in regulating cell spreading.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Interaction Mapping , Zyxin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Count , Cell Movement , Conserved Sequence , Cytoskeletal Proteins , Focal Adhesions/metabolism , Humans , Kinetics , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins , Structure-Activity Relationship , Zyxin/chemistry
2.
PLoS One ; 7(4): e35440, 2012.
Article in English | MEDLINE | ID: mdl-22514743

ABSTRACT

BACKGROUND: The majority of human cancer deaths are caused by metastasis. The metastatic dissemination is initiated by the breakdown of epithelial cell homeostasis. During this phenomenon, referred to as epithelial to mesenchymal transition (EMT), cells change their genetic and trancriptomic program leading to phenotypic and functional alterations. The challenge of understanding this dynamic process resides in unraveling regulatory networks involving master transcription factors (e.g. SNAI1/2, ZEB1/2 and TWIST1) and microRNAs. Here we investigated microRNAs regulated by SNAI1 and their potential role in the regulatory networks underlying epithelial plasticity. RESULTS: By a large-scale analysis on epithelial plasticity, we highlighted miR-203 and its molecular link with SNAI1 and the miR-200 family, key regulators of epithelial homeostasis. During SNAI1-induced EMT in MCF7 breast cancer cells, miR-203 and miR-200 family members were repressed in a timely correlated manner. Importantly, miR-203 repressed endogenous SNAI1, forming a double negative miR203/SNAI1 feedback loop. We integrated this novel miR203/SNAI1 with the known miR200/ZEB feedback loops to construct an a priori EMT core network. Dynamic simulations revealed stable epithelial and mesenchymal states, and underscored the crucial role of the miR203/SNAI1 feedback loop in state transitions underlying epithelial plasticity. CONCLUSION: By combining computational biology and experimental approaches, we propose a novel EMT core network integrating two fundamental negative feedback loops, miR203/SNAI1 and miR200/ZEB. Altogether our analysis implies that this novel EMT core network could function as a switch controlling epithelial cell plasticity during differentiation and cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , MicroRNAs/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
3.
BMC Bioinformatics ; 12: 67, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21375730

ABSTRACT

BACKGROUND: To understand biological processes and diseases, it is crucial to unravel the concerted interplay of transcription factors (TFs), microRNAs (miRNAs) and their targets within regulatory networks and fundamental sub-networks. An integrative computational resource generating a comprehensive view of these regulatory molecular interactions at a genome-wide scale would be of great interest to biologists, but is not available to date. RESULTS: To identify and analyze molecular interaction networks, we developed MIR@NT@N, an integrative approach based on a meta-regulation network model and a large-scale database. MIR@NT@N uses a graph-based approach to predict novel molecular actors across multiple regulatory processes (i.e. TFs acting on protein-coding or miRNA genes, or miRNAs acting on messenger RNAs). Exploiting these predictions, the user can generate networks and further analyze them to identify sub-networks, including motifs such as feedback and feedforward loops (FBL and FFL). In addition, networks can be built from lists of molecular actors with an a priori role in a given biological process to predict novel and unanticipated interactions. Analyses can be contextualized and filtered by integrating additional information such as microarray expression data. All results, including generated graphs, can be visualized, saved and exported into various formats. MIR@NT@N performances have been evaluated using published data and then applied to the regulatory program underlying epithelium to mesenchyme transition (EMT), an evolutionary-conserved process which is implicated in embryonic development and disease. CONCLUSIONS: MIR@NT@N is an effective computational approach to identify novel molecular regulations and to predict gene regulatory networks and sub-networks including conserved motifs within a given biological context. Taking advantage of the M@IA environment, MIR@NT@N is a user-friendly web resource freely available at http://mironton.uni.lu which will be updated on a regular basis.


Subject(s)
Databases, Genetic , Gene Regulatory Networks , MicroRNAs/genetics , Transcription Factors/genetics , Amino Acid Motifs/genetics , Computational Biology/methods , Gene Expression Regulation , Humans , Internet , MicroRNAs/metabolism , RNA, Messenger/genetics , Transcription Factors/metabolism
4.
PLoS One ; 5(2): e9210, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20169155

ABSTRACT

BACKGROUND: Initially detected in leukocytes and cancer cells derived from solid tissues, L-plastin/fimbrin belongs to a large family of actin crosslinkers and is considered as a marker for many cancers. Phosphorylation of L-plastin on residue Ser5 increases its F-actin binding activity and is required for L-plastin-mediated cell invasion. METHODOLOGY/PRINCIPAL FINDINGS: To study the kinetics of L-plastin and the impact of L-plastin Ser5 phosphorylation on L-plastin dynamics and actin turn-over in live cells, simian Vero cells were transfected with GFP-coupled WT-L-plastin, Ser5 substitution variants (S5/A, S5/E) or actin and analyzed by fluorescence recovery after photobleaching (FRAP). FRAP data were explored by mathematical modeling to estimate steady-state reaction parameters. We demonstrate that in Vero cell focal adhesions L-plastin undergoes rapid cycles of association/dissociation following a two-binding-state model. Phosphorylation of L-plastin increased its association rates by two-fold, whereas dissociation rates were unaffected. Importantly, L-plastin affected actin turn-over by decreasing the actin dissociation rate by four-fold, increasing thereby the amount of F-actin in the focal adhesions, all these effects being promoted by Ser5 phosphorylation. In MCF-7 breast carcinoma cells, phorbol 12-myristate 13-acetate (PMA) treatment induced L-plastin translocation to de novo actin polymerization sites in ruffling membranes and spike-like structures and highly increased its Ser5 phosphorylation. Both inhibition studies and siRNA knock-down of PKC isozymes pointed to the involvement of the novel PKC-delta isozyme in the PMA-elicited signaling pathway leading to L-plastin Ser5 phosphorylation. Furthermore, the L-plastin contribution to actin dynamics regulation was substantiated by its association with a protein complex comprising cortactin, which is known to be involved in this process. CONCLUSIONS/SIGNIFICANCE: Altogether these findings quantitatively demonstrate for the first time that L-plastin contributes to the fine-tuning of actin turn-over, an activity which is regulated by Ser5 phosphorylation promoting its high affinity binding to the cytoskeleton. In carcinoma cells, PKC-delta signaling pathways appear to link L-plastin phosphorylation to actin polymerization and invasion.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Algorithms , Amino Acid Substitution , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cortactin/metabolism , Fluorescence Recovery After Photobleaching , Focal Adhesions/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Models, Biological , Phosphorylation/drug effects , Protein Binding , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/genetics , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Vero Cells
5.
Eur Biophys J ; 39(4): 669-77, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19921173

ABSTRACT

A novel mathematical model of the actin dynamics in living cells under steady-state conditions has been developed for fluorescence recovery after photobleaching (FRAP) experiments. As opposed to other FRAP fitting models, which use the average lifetime of actins in filaments and the actin turnover rate as fitting parameters, our model operates with unbiased actin association/dissociation rate constants and accounts for the filament length. The mathematical formalism is based on a system of stochastic differential equations. The derived equations were validated on synthetic theoretical data generated by a stochastic simulation algorithm adapted for the simulation of FRAP experiments. Consistent with experimental findings, the results of this work showed that (1) fluorescence recovery is a function of the average filament length, (2) the F-actin turnover and the FRAP are accelerated in the presence of actin nucleating proteins, (3) the FRAP curves may exhibit both a linear and non-linear behaviour depending on the parameters of actin polymerisation, and (4) our model resulted in more accurate parameter estimations of actin dynamics as compared with other FRAP fitting models. Additionally, we provide a computational tool that integrates the model and that can be used for interpretation of FRAP data on actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/chemistry , Fluorescence Recovery After Photobleaching , Models, Biological , Actin Cytoskeleton/metabolism , Diffusion , Kinetics , Linear Models , Nonlinear Dynamics , Protein Multimerization , Protein Structure, Quaternary
6.
FASEB J ; 24(1): 105-18, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19726756
7.
J Cell Mol Med ; 14(6A): 1264-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19799649

ABSTRACT

We used a tumour necrosis factor (TNF)-alpha resistant breast adenocarcinoma MCF-7 cell line to investigate the involvement of the actin cytoskeleton in the mechanism of cell resistance to this cytokine. We found that TNF resistance correlates with the loss of cell epithelial properties and the gain of a mesenchymal phenotype, reminiscent of an epithelial-to-mesenchymal transition (EMT). Morphological changes were associated with a profound reorganization of the actin cytoskeleton and with a change in the repertoire of expressed actin cytoskeleton genes and EMT markers, as revealed by DNA microarray-based expression profiling. L-plastin, an F-actin cross-linking and stabilizing protein, was identified as one of the most significantly up-regulated genes in TNF-resistant cells. Knockdown of L-plastin in these cells revealed its crucial role in conferring TNF resistance. Importantly, overexpression of wild-type L-plastin in TNF-sensitive MCF-7 cells was sufficient to protect them against TNF-mediated cell death. Furthermore, we found that this effect is dependent on serine-5 phosphorylation of L-plastin and that non-conventional protein kinase C isoforms and the ceramide pathway may regulate its phosphorylation state. The protective role of L-plastin was not restricted to TNF-alpha resistant MCF-7 cells because a correlation between the expression of L-plastin and the resistance to TNF-alpha was observed in other breast cancer cell lines. Together, our study discloses a novel unexpected role of the actin bundling protein L-plastin as a cell protective protein against TNF-cytotoxicity.


Subject(s)
Actin Cytoskeleton/metabolism , Breast Neoplasms/metabolism , Cross-Linking Reagents/metabolism , Drug Resistance, Neoplasm/drug effects , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Actins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Ceramides/metabolism , Cytoskeleton/drug effects , Cytoskeleton/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/genetics , Humans , Mesoderm/drug effects , Mesoderm/pathology , Phenotype , Phosphorylation/drug effects , Protein Kinase C-delta/metabolism , Sphingomyelins/metabolism
8.
Biophys Chem ; 140(1-3): 24-34, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19101066

ABSTRACT

We report on an advanced universal Monte Carlo simulation model of actin polymerization processes offering a broad application panel. The model integrates major actin-related reactions, such as assembly of actin nuclei, association/dissociation of monomers to filament ends, ATP-hydrolysis via ADP-Pi formation and ADP-ATP exchange, filament branching, fragmentation and annealing or the effects of regulatory proteins. Importantly, these reactions are linked to information on the nucleotide state of actin subunits in filaments (ATP hydrolysis) and the distribution of actin filament lengths. The developed stochastic simulation modelling schemes were validated on: i) synthetic theoretical data generated by a deterministic model and ii) sets of our and published experimental data obtained from fluorescence pyrene-actin experiments. Build on an open-architecture principle, the designed model can be extended for predictive evaluation of the activities of other actin-interacting proteins and can be applied for the analysis of experimental pyrene actin-based or fluorescence microscopy data. We provide a user-friendly, free software package ActinSimChem that integrates the implemented simulation algorithms and that is made available to the scientific community for modelling in silico any specific actin-polymerization system.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Software , Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Computer Simulation , Escherichia coli/genetics , Fetal Proteins/chemistry , Fetal Proteins/metabolism , Formins , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Monte Carlo Method , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Rabbits
9.
Blood ; 113(2): 412-21, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18941112

ABSTRACT

Micro(mi)RNAs are small noncoding RNAs that orchestrate many key aspects of cell physiology and their deregulation is often linked to distinct diseases including cancer. Here, we studied the contribution of miRNAs in a well-characterized human myeloid leukemia, acute promyelocytic leukemia (APL), targeted by retinoic acid and trioxide arsenic therapy. We identified several miRNAs transcriptionally repressed by the APL-associated PML-RAR oncogene which are released after treatment with all-trans retinoic acid. These coregulated miRNAs were found to control, in a coordinated manner, crucial pathways linked to leukemogenesis, such as HOX proteins and cell adhesion molecules whose expressions are thereby repressed by the chemotherapy. Thus, APL appears linked to transcriptional perturbation of miRNA genes, and clinical protocols able to successfully eradicate cancer cells may do so by restoring miRNA expression. The identification of abnormal miRNA biogenesis in cancer may therefore provide novel biomarkers and therapeutic targets in myeloid leukemias.


Subject(s)
Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Leukemic , Leukemia, Promyelocytic, Acute/metabolism , MicroRNAs/biosynthesis , Oncogene Proteins, Fusion/metabolism , RNA, Neoplasm/biosynthesis , Transcription, Genetic , Antineoplastic Agents/therapeutic use , Arsenic/therapeutic use , Biomarkers, Tumor/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Gene Expression Regulation, Leukemic/drug effects , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , MicroRNAs/genetics , Oncogene Proteins, Fusion/genetics , RNA, Neoplasm/genetics , Transcription, Genetic/drug effects , Tretinoin/therapeutic use
10.
BMC Res Notes ; 1: 80, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18798985

ABSTRACT

BACKGROUND: Image analysis of microarrays and, in particular, spot quantification and spot quality control, is one of the most important steps in statistical analysis of microarray data. Recent methods of spot quality control are still in early age of development, often leading to underestimation of true positive microarray features and, consequently, to loss of important biological information. Therefore, improving and standardizing the statistical approaches of spot quality control are essential to facilitate the overall analysis of microarray data and subsequent extraction of biological information. FINDINGS: We evaluated the performance of two image analysis packages MAIA and GenePix (GP) using two complementary experimental approaches with a focus on the statistical analysis of spot quality factors. First, we developed control microarrays with a priori known fluorescence ratios to verify the accuracy and precision of the ratio estimation of signal intensities. Next, we developed advanced semi-automatic protocols of spot quality evaluation in MAIA and GP and compared their performance with available facilities of spot quantitative filtering in GP. We evaluated these algorithms for standardised spot quality analysis in a whole-genome microarray experiment assessing well-characterised transcriptional modifications induced by the transcription regulator SNAI1. Using a set of RT-PCR or qRT-PCR validated microarray data, we found that the semi-automatic protocol of spot quality control we developed with MAIA allowed recovering approximately 13% more spots and 38% more differentially expressed genes (at FDR = 5%) than GP with default spot filtering conditions. CONCLUSION: Careful control of spot quality characteristics with advanced spot quality evaluation can significantly increase the amount of confident and accurate data resulting in more meaningful biological conclusions.

11.
Hum Pathol ; 39(10): 1420-30, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18656242

ABSTRACT

The actin-binding protein gelsolin is involved in cell motility via the regulation of actin cytoskeleton, and its expression is modified in several human cancers. However, the potential implication of this protein in colorectal carcinogenesis is debated. By using immunohistochemistry, we studied gelsolin expression in 69 cases of colon adenocarcinomas and in 72 lesions representative of the different stages of colonic tumorigenesis. In addition, we performed Northern blot analysis of gelsolin messenger RNA in 12 paired samples of human colon cancer and normal corresponding mucosa. Gelsolin protein and messenger RNA expressions were severely down-regulated in all adenocarcinomas tested. Moreover, gelsolin protein was down-regulated in a large proportion of high-grade adenomas (14/16) before the acquisition of invasive properties but in only a small proportion of low grade adenomas and serrated adenomas (2/30) and in none of the 9 cases of nonneoplastic hyperplastic polyps tested. Our results therefore demonstrate that gelsolin down-regulation is an early and almost constant event in colon carcinogenesis and is associated with the transition from adenoma to carcinoma.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Gelsolin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adenoma/genetics , Adenoma/pathology , Aged , Cell Count , Colectomy , Colon/anatomy & histology , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation , Female , Gelsolin/genetics , Humans , Immunoenzyme Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Microsatellite Repeats , Middle Aged , Neoplasm Staging , RNA, Messenger/metabolism , RNA, Neoplasm/analysis
12.
BMC Genomics ; 8: 294, 2007 Aug 29.
Article in English | MEDLINE | ID: mdl-17727702

ABSTRACT

BACKGROUND: The actin cytoskeleton plays a crucial role in supporting and regulating numerous cellular processes. Mutations or alterations in the expression levels affecting the actin cytoskeleton system or related regulatory mechanisms are often associated with complex diseases such as cancer. Understanding how qualitative or quantitative changes in expression of the set of actin cytoskeleton genes are integrated to control actin dynamics and organisation is currently a challenge and should provide insights in identifying potential targets for drug discovery. Here we report the development of a dedicated microarray, the Actichip, containing 60-mer oligonucleotide probes for 327 genes selected for transcriptome analysis of the human actin cytoskeleton. RESULTS: Genomic data and sequence analysis features were retrieved from GenBank and stored in an integrative database called Actinome. From these data, probes were designed using a home-made program (CADO4MI) allowing sequence refinement and improved probe specificity by combining the complementary information recovered from the UniGene and RefSeq databases. Actichip performance was analysed by hybridisation with RNAs extracted from epithelial MCF-7 cells and human skeletal muscle. Using thoroughly standardised procedures, we obtained microarray images with excellent quality resulting in high data reproducibility. Actichip displayed a large dynamic range extending over three logs with a limit of sensitivity between one and ten copies of transcript per cell. The array allowed accurate detection of small changes in gene expression and reliable classification of samples based on the expression profiles of tissue-specific genes. When compared to two other oligonucleotide microarray platforms, Actichip showed similar sensitivity and concordant expression ratios. Moreover, Actichip was able to discriminate the highly similar actin isoforms whereas the two other platforms did not. CONCLUSION: Our data demonstrate that Actichip is a powerful alternative to commercial high density microarrays for cytoskeleton gene profiling in normal or pathological samples. Actichip is available upon request.


Subject(s)
Actins/metabolism , Computational Biology/methods , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Oligonucleotide Array Sequence Analysis/instrumentation , Oligonucleotide Array Sequence Analysis/methods , Actins/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cluster Analysis , Cytoskeleton/genetics , Data Interpretation, Statistical , Databases, Nucleic Acid , Equipment Design , Gene Expression , Humans , Microarray Analysis , Nucleic Acid Hybridization , Oligonucleotide Probes , Polymerase Chain Reaction , RNA/chemistry , Reproducibility of Results , Sensitivity and Specificity , Software
13.
Am J Hum Genet ; 80(1): 1-11, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17160889

ABSTRACT

Bardet-Biedl syndrome (BBS) is primarily an autosomal recessive ciliopathy characterized by progressive retinal degeneration, obesity, cognitive impairment, polydactyly, and kidney anomalies. The disorder is genetically heterogeneous, with 11 BBS genes identified to date, which account for ~70% of affected families. We have combined single-nucleotide-polymorphism array homozygosity mapping with in silico analysis to identify a new BBS gene, BBS12. Patients from two Gypsy families were homozygous and haploidentical in a 6-Mb region of chromosome 4q27. FLJ35630 was selected as a candidate gene, because it was predicted to encode a protein with similarity to members of the type II chaperonin superfamily, which includes BBS6 and BBS10. We found pathogenic mutations in both Gypsy families, as well as in 14 other families of various ethnic backgrounds, indicating that BBS12 accounts for approximately 5% of all BBS cases. BBS12 is vertebrate specific and, together with BBS6 and BBS10, defines a novel branch of the type II chaperonin superfamily. These three genes are characterized by unusually rapid evolution and are likely to perform ciliary functions specific to vertebrates that are important in the pathophysiology of the syndrome, and together they account for about one-third of the total BBS mutational load. Consistent with this notion, suppression of each family member in zebrafish yielded gastrulation-movement defects characteristic of other BBS morphants, whereas simultaneous suppression of all three members resulted in severely affected embryos, possibly hinting at partial functional redundancy within this protein family.


Subject(s)
Bardet-Biedl Syndrome/genetics , Chaperonins/genetics , Animals , Chaperonins/physiology , Chromosomes, Human, Pair 4/genetics , Embryo, Nonmammalian/abnormalities , Group II Chaperonins , Homozygote , Humans , Models, Molecular , Mutation , Oligonucleotide Array Sequence Analysis , Pedigree , Polymorphism, Single Nucleotide , Zebrafish/abnormalities , Zebrafish/embryology , Zebrafish/genetics
14.
J Cell Sci ; 119(Pt 9): 1947-60, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16636079

ABSTRACT

L-plastin, a malignant transformation-associated protein, is a member of a large family of actin filament cross-linkers. Here, we analysed how phosphorylation of L-plastin on Ser5 of the headpiece domain regulates its intracellular distribution and its interaction with F-actin in transfected cells and in in vitro assays. Phosphorylated wild-type L-plastin localised to the actin cytoskeleton in transfected Vero cells. Ser5Ala substitution reduced the capacity of L-plastin to localise with peripheral actin-rich membrane protrusions. Conversely, a Ser5Glu variant mimicking a constitutively phosphorylated state, accumulated in actin-rich regions and promoted the formation of F-actin microspikes in two cell lines. Similar to phosphorylated wild-type L-plastin, this variant remained associated with cellular F-actin in detergent-treated cells, whereas the Ser5Ala variant was almost completely extracted. When compared with non-phosphorylated protein, phosphorylated L-plastin and the Ser5Glu variant bound F-actin more efficiently in an in vitro assay. Importantly, expression of L-plastin elicited collagen invasion in HEK293T cells, in a manner dependent on Ser5 phosphorylation. Based on our findings, we propose that conversely to other calponin homology (CH)-domain family members, phosphorylation of L-plastin switches the protein from a low-activity to a high-activity state. Phosphorylated L-plastin might act as an integrator of signals controlling the assembly of the actin cytoskeleton and cell motility in a 3D-space.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Serine/metabolism , Actins/chemistry , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Cytoskeleton/metabolism , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding
15.
Mol Biol Cell ; 16(12): 5736-48, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16195354

ABSTRACT

Actin-related proteins (ARPs) are key players in cytoskeleton activities and nuclear functions. Two complexes, ARP2/3 and ARP1/11, also known as dynactin, are implicated in actin dynamics and in microtubule-based trafficking, respectively. ARP4 to ARP9 are components of many chromatin-modulating complexes. Conventional actins and ARPs codefine a large family of homologous proteins, the actin superfamily, with a tertiary structure known as the actin fold. Because ARPs and actin share high sequence conservation, clear family definition requires distinct features to easily and systematically identify each subfamily. In this study we performed an in depth sequence and comparative genomic analysis of ARP subfamilies. A high-quality multiple alignment of approximately 700 complete protein sequences homologous to actin, including 148 ARP sequences, allowed us to extend the ARP classification to new organisms. Sequence alignments revealed conserved residues, motifs, and inserted sequence signatures to define each ARP subfamily. These discriminative characteristics allowed us to develop ARPAnno (http://bips.u-strasbg.fr/ARPAnno), a new web server dedicated to the annotation of ARP sequences. Analyses of sequence conservation among actins and ARPs highlight part of the actin fold and suggest interactions between ARPs and actin-binding proteins. Finally, analysis of ARP distribution across eukaryotic phyla emphasizes the central importance of nuclear ARPs, particularly the multifunctional ARP4.


Subject(s)
Actins/genetics , Genome , Actins/chemistry , Actins/metabolism , Animals , Conserved Sequence , Internet , Mice , Models, Molecular , Mutagenesis, Insertional , Nucleic Acid Hybridization/methods , Phylogeny , Protein Conformation , Protein Folding , Protein Structure, Secondary , Sequence Deletion
16.
Traffic ; 6(4): 335-45, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15752138

ABSTRACT

T- and L-plastin are highly similar actin-bundling proteins implicated in the regulation of cell morphology, lamellipodium protrusion, bacterial invasion and tumor progression. We show that T-plastin localizes predominantly to the cytoplasm, whereas L-plastin distributes between nucleus and cytoplasm in HeLa or Cos cells. T-plastin shows nuclear accumulation upon incubation of cells with the CRM1 antagonist leptomycin B (LMB). We identified a Rev-like nuclear export sequence (NES) in T-plastin that is able to export an otherwise nuclear protein in an LMB-dependent manner. Deletion of the NES promotes nuclear accumulation of T-plastin. Mutation of residues L17, F21 or L26 in the T-plastin NES inhibits nuclear efflux. L-plastin harbors a less conserved NES and lacks the F21 T-plastin residue. Insertion of a Phe residue in the L-plastin NES specifically enhances its export activity. These findings explain why both isoforms exhibit specific distribution patterns in eukaryotic cells.


Subject(s)
Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chlorocebus aethiops , Fatty Acids, Unsaturated/pharmacology , Gene Products, rev/genetics , Gene Products, rev/metabolism , Humans , Leucine/genetics , Leucine/metabolism , Membrane Glycoproteins , Molecular Sequence Data , Phenotype , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphoproteins/genetics , Protein Isoforms/metabolism , Protein Sorting Signals/drug effects , Sequence Alignment
17.
J Cell Sci ; 118(Pt 6): 1255-65, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15741236

ABSTRACT

Increasing evidence suggests that actin cross-linking or bundling proteins might not only structure the cortical actin cytoskeleton but also control actin dynamics. Here, we analyse the effects of T-plastin/T-fimbrin, a representative member of an important actin-filament cross-linking protein by combining a quantitative biomimetic motility assay with biochemical and cell-based approaches. Beads coated with the VCA domain of the Wiskott/Aldrich-syndrome protein (WASP) recruit the actin-nucleating Arp2/3 complex, polymerize actin at their surface and undergo movement when placed in cell-free extracts. T-Plastin increased the velocity of VCA beads 1.5 times, stabilized actin comets and concomitantly displaced cofilin, an actin-depolymerizing protein. T-Plastin also decreased the F-actin disassembly rate and inhibited cofilin-mediated depolymerization of actin filaments in vitro. Importantly, a bundling-incompetent variant comprising the first actin-binding domain (ABD1) had similar effects. In cells, this domain induced the formation of long actin cables to which other actin-regulating proteins were recruited. Altogether, these results favor a mechanism in which binding of ABD1 controls actin turnover independently of cross-link formation. In vivo, this activity might contribute to the assembly and maintenance of the actin cytoskeleton of plasma-membrane protrusions.


Subject(s)
Actin Cytoskeleton/chemistry , Actins/metabolism , Cytoskeletal Proteins/metabolism , Phosphoproteins/chemistry , Actin Depolymerizing Factors , Actin-Related Protein 2 , Actin-Related Protein 3 , Actins/chemistry , Animals , Antibodies, Monoclonal/chemistry , Calcium/metabolism , Cell Membrane/metabolism , Cell-Free System , Chlorocebus aethiops , Cross-Linking Reagents/pharmacology , DNA/chemistry , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Membrane Glycoproteins , Microfilament Proteins/metabolism , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Neoplasm Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Time Factors , Transfection , Vero Cells , Wiskott-Aldrich Syndrome Protein
18.
Prog Cell Cycle Res ; 5: 511-25, 2003.
Article in English | MEDLINE | ID: mdl-14593746

ABSTRACT

Dynamic processes such as cell migration and division depend on the actin cytoskeleton, a dense meshwork of protein polymers capable of undergoing rapid cycles of assembly and disassembly, under the control of a large number of actin-associated proteins. In cancer cells, structural and functional perturbations of the actin cytoskeleton correlate with higher proliferation rates and uncontrolled movement. Therefore, small molecules that act on the actin cytoskeleton of tumour cells and thus inhibit cell division and movement, may be of high therapeutic value. The dynamic properties of the actin cytoskeleton and the mechanism of action of actin-targeting drugs will be described.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Antineoplastic Agents/pharmacology , Cytoskeleton/metabolism , Neoplasms/metabolism , Actin Cytoskeleton/drug effects , Actins/drug effects , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cytoskeleton/drug effects , Drug Evaluation, Preclinical/trends , Forecasting , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...