Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 549, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724689

ABSTRACT

Amphiphysin 2 (BIN1) is a membrane and actin remodeling protein mutated in congenital and adult centronuclear myopathies. Here, we report an unexpected function of this N-BAR domain protein BIN1 in filopodia formation. We demonstrated that BIN1 expression is necessary and sufficient to induce filopodia formation. BIN1 is present at the base of forming filopodia and all along filopodia, where it colocalizes with F-actin. We identify that BIN1-mediated filopodia formation requires IRSp53, which allows its localization at negatively-curved membrane topologies. Our results show that BIN1 bundles actin in vitro. Finally, we identify that BIN1 regulates the membrane-to-cortex architecture and functions as a molecular platform to recruit actin-binding proteins, dynamin and ezrin, to promote filopodia formation.


Subject(s)
Actins , Adaptor Proteins, Signal Transducing , Nerve Tissue Proteins , Pseudopodia , Tumor Suppressor Proteins , Pseudopodia/metabolism , Actins/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Cell Membrane/metabolism , Mice , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
2.
J Cell Sci ; 135(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35394045

ABSTRACT

Altered endocytosis and vesicular trafficking are major players during tumorigenesis. Flotillin overexpression, a feature observed in many invasive tumors and identified as a marker of poor prognosis, induces a deregulated endocytic and trafficking pathway called upregulated flotillin-induced trafficking (UFIT). Here, we found that in non-tumoral mammary epithelial cells, induction of the UFIT pathway promotes epithelial-to-mesenchymal transition (EMT) and accelerates the endocytosis of several transmembrane receptors, including AXL, in flotillin-positive late endosomes. AXL overexpression, frequently observed in cancer cells, is linked to EMT and metastasis formation. In flotillin-overexpressing non-tumoral mammary epithelial cells and in invasive breast carcinoma cells, we found that the UFIT pathway-mediated AXL endocytosis allows its stabilization and depends on sphingosine kinase 2, a lipid kinase recruited in flotillin-rich plasma membrane domains and endosomes. Thus, the deregulation of vesicular trafficking following flotillin upregulation, and through sphingosine kinase 2, emerges as a new mechanism of AXL overexpression and EMT-inducing signaling pathway activation.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Membrane Proteins , Phosphotransferases (Alcohol Group Acceptor) , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Cell Line, Tumor , Female , Humans , Membrane Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
3.
J Biol Chem ; 295(39): 13474-13487, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32690605

ABSTRACT

Yes-associated protein (YAP) signaling has emerged as a crucial pathway in several normal and pathological processes. Although the main upstream effectors that regulate its activity have been extensively studied, the role of the endosomal system has been far less characterized. Here, we identified the late endosomal/lysosomal adaptor MAPK and mTOR activator (LAMTOR) complex as an important regulator of YAP signaling in a preosteoblast cell line. We found that p18/LAMTOR1-mediated peripheral positioning of late endosomes allows delivery of SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) to the plasma membrane and promotes activation of an SRC-dependent signaling cascade that controls YAP nuclear shuttling. Moreover, ß1 integrin engagement and mechano-sensitive cues, such as external stiffness and related cell contractility, controlled LAMTOR targeting to the cell periphery and thereby late endosome recycling and had a major impact on YAP signaling. Our findings identify the late endosome recycling pathway as a key mechanism that controls YAP activity and explains YAP mechano-sensitivity.


Subject(s)
Cell Cycle Proteins/metabolism , Endosomes/metabolism , Integrin beta1/metabolism , Transcription Factors/metabolism , src-Family Kinases/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Line , HEK293 Cells , Humans , Mice , Mice, Knockout , Proto-Oncogene Mas , Signal Transduction , Transcription Factors/deficiency , src-Family Kinases/deficiency
4.
Cancer Metastasis Rev ; 39(2): 361-374, 2020 06.
Article in English | MEDLINE | ID: mdl-32297092

ABSTRACT

Flotillins 1 and 2 are two ubiquitous, highly conserved homologous proteins that assemble to form heterotetramers at the cytoplasmic face of the plasma membrane in cholesterol- and sphingolipid-enriched domains. Flotillin heterotetramers can assemble into large oligomers to form molecular scaffolds that regulate the clustering of at the plasma membrane and activity of several receptors. Moreover, flotillins are upregulated in many invasive carcinomas and also in sarcoma, and this is associated with poor prognosis and metastasis formation. When upregulated, flotillins promote plasma membrane invagination and induce an endocytic pathway that allows the targeting of cargo proteins in the late endosomal compartment in which flotillins accumulate. These late endosomes are not degradative, and participate in the recycling and secretion of protein cargos. The cargos of this Upregulated Flotillin-Induced Trafficking (UFIT) pathway include molecules involved in signaling, adhesion, and extracellular matrix remodeling, thus favoring the acquisition of an invasive cellular behavior leading to metastasis formation. Thus, flotillin presence from the plasma membrane to the late endosomal compartment influences the activity, and even modifies the trafficking and fate of key protein cargos, favoring the development of diseases, for instance tumors. This review summarizes the current knowledge on flotillins and their role in cancer development focusing on their function in cellular membrane remodeling and vesicular trafficking regulation.


Subject(s)
Membrane Proteins/metabolism , Neoplasms/metabolism , Animals , Carcinogenesis , Cell Membrane/metabolism , Humans , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Membrane Proteins/biosynthesis , Neoplasms/pathology
5.
J Cell Sci ; 132(21)2019 11 08.
Article in English | MEDLINE | ID: mdl-31604795

ABSTRACT

Directional collective cell migration (DCCM) is crucial for morphogenesis and cancer metastasis. P-cadherin (also known as CDH3), which is a cell-cell adhesion protein expressed in carcinoma and aggressive sarcoma cells and associated with poor prognosis, is a major DCCM regulator. However, it is unclear how P-cadherin-mediated mechanical coupling between migrating cells influences force transmission to the extracellular matrix (ECM). Here, we found that decorin, a small proteoglycan that binds to and organizes collagen fibers, is specifically expressed and secreted upon P-cadherin, but not E- and R-cadherin (also known as CDH1 and CDH4, respectively) expression. Through cell biological and biophysical approaches, we demonstrated that decorin is required for P-cadherin-mediated DCCM and collagen fiber orientation in the migration direction in 2D and 3D matrices. Moreover, P-cadherin, through decorin-mediated collagen fiber reorientation, promotes the activation of ß1 integrin and of the ß-Pix (ARHGEF7)/CDC42 axis, which increases traction forces, allowing DCCM. Our results identify a novel P-cadherin-mediated mechanism to promote DCCM through ECM remodeling and ECM-guided cell migration.


Subject(s)
Cadherins/metabolism , Cell Movement/physiology , Collagen/metabolism , Decorin/metabolism , Cell Adhesion/physiology , Extracellular Matrix/metabolism , Humans , Mechanical Phenomena , cdc42 GTP-Binding Protein/metabolism
6.
J Cell Sci ; 131(17)2018 09 05.
Article in English | MEDLINE | ID: mdl-30111578

ABSTRACT

Tumor cell invasion and metastasis formation are the major cause of death in cancer patients. These processes rely on extracellular matrix (ECM) degradation mediated by organelles termed invadopodia, to which the transmembrane matrix metalloproteinase MT1-MMP (also known as MMP14) is delivered from its reservoir, the RAB7-containing endolysosomes. How MT1-MMP is targeted to endolysosomes remains to be elucidated. Flotillin-1 and -2 are upregulated in many invasive cancers. Here, we show that flotillin upregulation triggers a general mechanism, common to carcinoma and sarcoma, which promotes RAB5-dependent MT1-MMP endocytosis and its delivery to RAB7-positive endolysosomal reservoirs. Conversely, flotillin knockdown in invasive cancer cells greatly reduces MT1-MMP accumulation in endolysosomes, its subsequent exocytosis at invadopodia, ECM degradation and cell invasion. Our results demonstrate that flotillin upregulation is necessary and sufficient to promote epithelial and mesenchymal cancer cell invasion and ECM degradation by controlling MT1-MMP endocytosis and delivery to the endolysosomal recycling compartment.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Matrix Metalloproteinase 14/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Cell Line, Tumor , Endocytosis , Endosomes/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Lysosomes/genetics , Matrix Metalloproteinase 14/genetics , Membrane Proteins/genetics , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/pathology , Podosomes/genetics , Podosomes/metabolism , Protein Transport , Up-Regulation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
7.
Biol Cell ; 109(5): 210-221, 2017 May.
Article in English | MEDLINE | ID: mdl-28225561

ABSTRACT

Zebrafish gastrulation and particularly epiboly that involves coordinated movements of several cell layers is a dynamic process for which regulators remain to be identified. We show here that Flotillin 1 and 2, ubiquitous and highly conserved proteins, are required for epiboly. Flotillins knockdown compromised embryo survival, strongly delayed epiboly and impaired deep cell radial intercalation and directed collective migration without affecting enveloping layer cell movement. At the molecular level, we identified that Flotillins are required for the formation of E-cadherin-mediated cell-cell junctions. These results provide the first in vivo evidence that Flotillins regulate E-cadherin-mediated cell-cell junctions to allow epiboly progression.


Subject(s)
Cadherins/metabolism , Cell Movement , Membrane Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Adhesion , Cell Communication , Gene Knockdown Techniques , beta Catenin/metabolism
8.
Mol Biol Cell ; 27(17): 2653-61, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27413008

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of skeletal muscle origin in children and adolescents. Among RMS subtypes, alveolar rhabdomyosarcoma (ARMS), which is characterized by the presence of the PAX3-FOXO1A or PAX7-FOXO1A chimeric oncogenic transcription factor, is associated with poor prognosis and a strong risk of metastasis compared with the embryonal subtype (ERMS). To identify molecular pathways involved in ARMS aggressiveness, we first characterized the migratory behavior of cell lines derived from ARMS and ERMS biopsies using a three-dimensional spheroid cell invasion assay. ARMS cells were more invasive than ERMS cells and adopted an ellipsoidal morphology to efficiently invade the extracellular matrix. Moreover, the invasive potential of ARMS cells depended on ROCK activity, which is regulated by the GTPase RhoE. Specifically, RhoE expression was low in ARMS biopsies, and its overexpression in ARMS cells reduced their invasion potential. Conversely, ARHGAP25, a GTPase-activating protein for Rac, was up-regulated in ARMS biopsies. Moreover, we found that ARHGAP25 inhibits Rac activity downstream of ROCKII and is required for ARMS cell invasion. Our results indicate that the RhoE/ROCK/ARHGAP25 signaling pathway promotes ARMS invasive potential and identify these proteins as potential therapeutic targets for ARMS treatment.


Subject(s)
GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Cell Differentiation , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Muscle, Skeletal/metabolism , Oncogene Proteins, Fusion/metabolism , Rhabdomyosarcoma , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Signal Transduction/genetics , rac GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
9.
Small GTPases ; 7(3): 156-63, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27152729

ABSTRACT

This commentary addresses the role of P-cadherin in collective cell migration (CCM), a cooperative and coordinated migration mode, used by cells during normal and pathological migration processes. We discuss how cadherin-mediated cell-cell junctions (CCJs) play a critical role in CCM through their ability to regulate Rho GTPase-dependent pathways and how this leads to the generation and orientation of mechanical forces. We will also highlight the key function of P-cadherin (a poor prognostic marker in several tumors) in promoting collective cell movement in epithelial and mesenchymal cells.


Subject(s)
Cadherins/metabolism , Cell Movement , rho GTP-Binding Proteins/metabolism , Animals , Biomechanical Phenomena , Humans , Intercellular Junctions/metabolism
10.
J Cell Biol ; 212(2): 199-217, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26783302

ABSTRACT

Collective cell migration (CCM) is essential for organism development, wound healing, and metastatic transition, the primary cause of cancer-related death, and it involves cell-cell adhesion molecules of the cadherin family. Increased P-cadherin expression levels are correlated with tumor aggressiveness in carcinoma and aggressive sarcoma; however, how P-cadherin promotes tumor malignancy remains unknown. Here, using integrated cell biology and biophysical approaches, we determined that P-cadherin specifically induces polarization and CCM through an increase in the strength and anisotropy of mechanical forces. We show that this mechanical regulation is mediated by the P-cadherin/ß-PIX/Cdc42 axis; P-cadherin specifically activates Cdc42 through ß-PIX, which is specifically recruited at cell-cell contacts upon CCM. This mechanism of cell polarization and migration is absent in cells expressing E- or R-cadherin. Thus, we identify a specific role of P-cadherin through ß-PIX-mediated Cdc42 activation in the regulation of cell polarity and force anisotropy that drives CCM.


Subject(s)
Cadherins/metabolism , Cell Movement , cdc42 GTP-Binding Protein/metabolism , Animals , Biomechanical Phenomena , Cell Polarity , Cells, Cultured , Mice , Myoblasts/cytology , Myoblasts/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism
11.
ACS Biomater Sci Eng ; 2(3): 415-425, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-33429544

ABSTRACT

Controlling the different steps of cell differentiation in vitro using bioactive surfaces may be useful in view of future cell therapies. Substrates presenting peptides, which are minimal fragments of extracellular matrix (ECM) proteins may be used for this purpose. In this work, we used polyelectrolyte multilayer films presenting two peptides derived from different muscle ECM proteins to target syndecan or/and integrin receptors. We showed that the presence of laminin-derived peptide to target syndecan-1 promotes lamellipodia formation, increases migration speed, directionality, and cell proliferation but impaired myotube formation. The cellular effects of L2synd are under the control of Rac1 and Cdc42 activities and involved ß1 integrin in contrast to RGD-containing peptide, which enabled adhesion via ß3 integrins and muscle cell differentiation. Our results show that peptides grafted onto multilayered films can guide the proliferation/differentiation balance and reveal crosstalk between different adhesion receptors.

12.
J Cell Biol ; 207(4): 517-33, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25422375

ABSTRACT

Accumulation of type I collagen fibrils in tumors is associated with an increased risk of metastasis. Invadosomes are F-actin structures able to degrade the extracellular matrix. We previously found that collagen I fibrils induced the formation of peculiar linear invadosomes in an unexpected integrin-independent manner. Here, we show that Discoidin Domain Receptor 1 (DDR1), a collagen receptor overexpressed in cancer, colocalizes with linear invadosomes in tumor cells and is required for their formation and matrix degradation ability. Unexpectedly, DDR1 kinase activity is not required for invadosome formation or activity, nor is Src tyrosine kinase. We show that the RhoGTPase Cdc42 is activated on collagen in a DDR1-dependent manner. Cdc42 and its specific guanine nucleotide-exchange factor (GEF), Tuba, localize to linear invadosomes, and both are required for linear invadosome formation. Finally, DDR1 depletion blocked cell invasion in a collagen gel. Altogether, our data uncover an important role for DDR1, acting through Tuba and Cdc42, in proteolysis-based cell invasion in a collagen-rich environment.


Subject(s)
Collagen Type I/metabolism , Cytoskeletal Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , cdc42 GTP-Binding Protein/metabolism , Actin Cytoskeleton , Actins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Line, Tumor , Collagenases/metabolism , Dipeptides/pharmacology , Discoidin Domain Receptor 1 , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Neoplasm Invasiveness/genetics , RNA Interference , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/genetics
13.
J Cell Sci ; 127(Pt 24): 5139-47, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25413346

ABSTRACT

Flotillin 1 and 2 are ubiquitous and highly conserved proteins. They were initially discovered in 1997 as being associated with specific caveolin-independent cholesterol- and glycosphingolipid-enriched membrane microdomains and as being expressed during axon regeneration. Flotillins have a role in a large number of physiopathological processes, mainly through their function in membrane receptor clustering and in the regulation of clathrin-independent endocytosis. In this Commentary, we summarize the research performed so far on the role of flotillins in cell-cell adhesion. Recent studies have demonstrated that flotillins directly regulate the formation of cadherin complexes. Indeed, flotillin microdomains are required for the dynamic association and stabilization of cadherins at cell-cell junctions and also for cadherin signaling. Moreover, because flotillins regulate endocytosis and also the actin cytoskeleton, they could have an indirect role in the assembly and stabilization of cadherin complexes. Because it has also recently been shown that flotillins are overexpressed during neurodegenerative diseases and in human cancers, where their upregulation is associated with metastasis formation and poor prognosis, understanding to what extent flotillin upregulation participates in the development of such pathologies is thus of particular interest, as well as how, at the molecular level, it might affect cell adhesion processes.


Subject(s)
Cell Physiological Phenomena , Disease , Membrane Proteins/metabolism , Cadherins/metabolism , Cell Adhesion , Humans , Membrane Proteins/chemistry , Models, Biological
14.
J Cell Sci ; 126(Pt 22): 5293-304, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24046456

ABSTRACT

Cadherins are essential in many fundamental processes and assemble at regions of cell-cell contact in large macromolecular complexes named adherens junctions. We have identified flotillin 1 and 2 as new partners of the cadherin complexes. We show that flotillins are localised at cell-cell junctions (CCJs) in a cadherin-dependent manner. Flotillins and cadherins are constitutively associated at the plasma membrane and their colocalisation at CCJ increases with CCJ maturation. Using three-dimensional structured illumination super-resolution microscopy, we found that cadherin and flotillin complexes are associated with F-actin bundles at CCJs. The knockdown of flotillins dramatically affected N- and E-cadherin recruitment at CCJs in mesenchymal and epithelial cell types and perturbed CCJ integrity and functionality. Moreover, we determined that flotillins are required for cadherin association with GM1-containing plasma membrane microdomains. This allows p120 catenin binding to the cadherin complex and its stabilization at CCJs. Altogether, these data demonstrate that flotillin microdomains are required for cadherin stabilization at CCJs and for the formation of functional CCJs.


Subject(s)
Cadherins/metabolism , Intercellular Junctions/genetics , Membrane Proteins/metabolism , Cadherins/genetics , Catenins/metabolism , Cell Membrane/metabolism , Gene Knockdown Techniques , HCT116 Cells , Humans , Intercellular Junctions/metabolism , MCF-7 Cells , Membrane Proteins/genetics , Protein Structure, Tertiary , Sphingolipid Activator Proteins/metabolism , Delta Catenin
15.
Mol Biol Cell ; 24(3): 234-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23197472

ABSTRACT

Cadherins are homophilic cell-cell adhesion molecules implicated in many fundamental processes, such as morphogenesis, cell growth, and differentiation. They accumulate at cell-cell contact sites and assemble into large macromolecular complexes named adherens junctions (AJs). Cadherin targeting and function are regulated by various cellular processes, many players of which remain to be uncovered. Here we identify the small GTPase Rab35 as a new regulator of cadherin trafficking and stabilization at cell-cell contacts in C2C12 myoblasts and HeLa cells. We find that Rab35 accumulates at cell-cell contacts in a cadherin-dependent manner. Knockdown of Rab35 or expression of a dominant-negative form of Rab35 impaired N- and M-cadherin recruitment to cell-cell contacts, their stabilization at the plasma membrane, and association with p120 catenin and led to their accumulation in transferrin-, clathrin-, and AP-2-positive intracellular vesicles. We also find that Rab35 function is required for PIP5KIγ accumulation at cell-cell contacts and phosphatidyl inositol 4,5-bisphosphate production, which is involved in cadherin stabilization at contact sites. Finally, we show that Rab35 regulates myoblast fusion, a major cellular process under the control of cadherin-dependent signaling. Taken together, these results reveal that Rab35 regulates cadherin-dependent AJ formation and myoblast fusion.


Subject(s)
Adherens Junctions/metabolism , Myoblasts/physiology , rab GTP-Binding Proteins/physiology , Animals , Cadherins/metabolism , Cell Differentiation , Cell Fusion , Gene Knockdown Techniques , HeLa Cells , Humans , Mice , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Stability , Protein Transport , Signal Transduction
16.
Acta Biomater ; 9(5): 6468-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23261924

ABSTRACT

Skeletal muscle tissue engineering holds promise for the replacement of muscle damaged by injury and for the treatment of muscle diseases. Although arginylglycylaspartic acid (RGD) substrates have been widely explored in tissue engineering, there have been no studies aimed at investigating the combined effects of RGD nanoscale presentation and matrix stiffness on myogenesis. In the present work we use polyelectrolyte multilayer films made of poly(L-lysine) (PLL) and poly(L-glutamic) acid (PGA) as substrates of tunable stiffness that can be functionalized by a RGD adhesive peptide to investigate important events in myogenesis, including adhesion, migration, proliferation and differentiation. C2C12 myoblasts were used as cellular models. RGD presentation on soft films and increasing film stiffness could both induce cell adhesion, but the integrins involved in adhesion were different in the case of soft and stiff films. Soft films with RGD peptide appeared to be the most appropriate substrate for myogenic differentiation, while the stiff PLL/PGA films induced significant cell migration and proliferation and inhibited myogenic differentiation. ROCK kinase was found to be involved in the myoblast response to the different films. Indeed, its inhibition was sufficient to rescue differentiation on stiff films, but no significant changes were observed on stiff films with the RGD peptide. These results suggest that different signaling pathways may be activated depending on the mechanical and biochemical properties of multilayer films. This study emphasizes the advantage of soft PLL/PGA films presenting the RGD peptide in terms of myogenic differentiation. This soft RGD-presenting film may be further used as a coating of various polymeric scaffolds for muscle tissue engineering.


Subject(s)
Cell Differentiation , Electrolytes/chemistry , Muscle, Skeletal/cytology , Oligopeptides/chemistry , Amino Acid Sequence , Humans , Molecular Sequence Data , Osteoblasts/cytology
17.
Mol Biol Cell ; 21(14): 2412-24, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20505075

ABSTRACT

Myoblast fusion is an essential step during myoblast differentiation that remains poorly understood. M-cadherin-dependent pathways that signal through Rac1 GTPase activation via the Rho-guanine nucleotide exchange factor (GEF) Trio are important for myoblast fusion. The ADP-ribosylation factor (ARF)6 GTPase has been shown to bind to Trio and to regulate Rac1 activity. Moreover, Loner/GEP(100)/BRAG2, a GEF of ARF6, has been involved in mammalian and Drosophila myoblast fusion, but the specific role of ARF6 has been not fully analyzed. Here, we show that ARF6 activity is increased at the time of myoblast fusion and is required for its implementation in mouse C2C12 myoblasts. Specifically, at the onset of myoblast fusion, ARF6 is associated with the multiproteic complex that contains M-cadherin, Trio, and Rac1 and accumulates at sites of myoblast fusion. ARF6 silencing inhibits the association of Trio and Rac1 with M-cadherin. Moreover, we demonstrate that ARF6 regulates myoblast fusion through phospholipase D (PLD) activation and phosphatidylinositol 4,5-bis-phosphate production. Together, these data indicate that ARF6 is a critical regulator of C2C12 myoblast fusion and participates in the regulation of PLD activities that trigger both phospholipids production and actin cytoskeleton reorganization at fusion sites.


Subject(s)
ADP-Ribosylation Factors/metabolism , Myoblasts/enzymology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase D/metabolism , Signal Transduction , ADP-Ribosylation Factor 6 , Animals , Cadherins/metabolism , Cell Fusion , Cell Line , Enzyme Activation , Gene Knockdown Techniques , Gene Silencing , Guanine Nucleotide Exchange Factors/metabolism , Mice , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myoblasts/cytology , Myoblasts/ultrastructure , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Regeneration , rac1 GTP-Binding Protein/metabolism
18.
J Biol Chem ; 284(34): 23137-45, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19546217

ABSTRACT

p120 catenin is a major regulator of cadherin stability at cell-cell contacts and a modulator of Rho GTPase activities. In C2C12 myoblasts, N-cadherin is stabilized at cell contacts through its association with cholesterol-rich membrane domains or lipid rafts (LR) and acts as an adhesion-activated receptor that activates RhoA, an event required for myogenesis induction. Here, we report that association of p120 catenin with N-cadherin at cell contacts occurs specifically in LR. We demonstrate that interaction of p120 catenin with N-cadherin is required for N-cadherin association with LR and for its stabilization at cell contacts. LR disruption inhibits myogenesis induction and N-cadherin-dependent RhoA activation as does the perturbation of the N-cadherin-p120 catenin complex after p120 catenin knockdown. Finally, we observe an N-cadherin-dependent accumulation of RhoA at phosphatidylinositol 4,5-bisphosphate-enriched cell contacts which is lost after LR disruption. Thus, a functional N-cadherin-catenin complex occurs in cholesterol-rich membrane microdomains which allows the recruitment of RhoA and the regulation of its activity during myogenesis induction.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cholesterol/chemistry , Muscle Development/physiology , Phosphoproteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Catenins , Cell Adhesion Molecules/genetics , Cell Line , Fluorescence Recovery After Photobleaching , Immunohistochemistry , Immunoprecipitation , Intercellular Junctions/metabolism , Mice , Muscle Development/genetics , Phosphoproteins/genetics , Protein Binding , Delta Catenin
19.
J Biol Chem ; 284(28): 18808-15, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19429675

ABSTRACT

In the pituitary gland, activated protein kinase C (PKC) isoforms accumulate either selectively at the cell-cell contact (alpha and epsilon) or at the entire plasma membrane (beta1 and delta). The molecular mechanisms underlying these various subcellular locations are not known. Here, we demonstrate the existence within PKCepsilon of a cell-cell contact targeting sequence (3CTS) that, upon stimulation, is capable of targeting PKCdelta, chimerin-alpha1, and the PKCepsilon C1 domain to the cell-cell contact. We show that this selective targeting of PKCepsilon is lost upon overexpression of 3CTS fused to a (R-Ahx-R)(4) (where Ahx is 6-aminohexanoic acid) vectorization peptide, reflecting a dominant-negative effect of the overexpressed 3CTS on targeting selectivity. 3CTS contains a putative amphipathic alpha-helix, a 14-3-3-binding site, and the Glu-374 amino acid, involved in targeting selectivity. We show that the integrity of the alpha-helix is important for translocation but that 14-3-3 is not involved in targeting selectivity. However, PKCepsilon translocation is increased when PKCepsilon/14-3-3 interaction is abolished, suggesting that phorbol 12-myristate 13-acetate activation may initiate two sets of PKCepsilon functions, those depending on 14-3-3 and those depending on translocation to cell-cell contacts. Thus, 3CTS is involved in the modulation of translocation via its 14-3-3-binding site, in cytoplasmic desequestration via the alpha-helix, and in selective PKCepsilon targeting at the cell-cell contact via Glu-374.


Subject(s)
Protein Kinase C-epsilon/physiology , 14-3-3 Proteins/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Binding Sites , Cell Communication , Glutamic Acid/chemistry , Humans , Molecular Sequence Data , Pituitary Gland/metabolism , Protein Conformation , Protein Kinase C-epsilon/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Tetradecanoylphorbol Acetate/chemistry
20.
Biochem Biophys Res Commun ; 384(3): 322-8, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19409875

ABSTRACT

Up-regulation of utrophin, a homolog of dystrophin, is known to ameliorate the dystrophic phenotype in animal models of Duchenne muscular dystrophy. We have previously demonstrated that the active form of RhoA (RhoAV14) increases the expression of utrophin and its localization at the plasma membrane in cultured myoblasts. In this paper, we ask whether RhoAV14 can up-regulate utrophin also in mice. A plasmid encoding for RhoAV14 was injected into skeletal muscles followed by electroporation. Muscles expressing RhoAV14 were analyzed by Western-immunoblotting, real time PCR amplification and immunohistochemistry. We found that RhoAV14 increased utrophin protein expression and distribution specifically at the plasma membrane in muscle fibers without any effect on utrophin transcription. Utrophin up-regulation, uncoupled from that of its mRNA, has been previously observed in pathological processes and in normal regenerating conditions.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Utrophin/metabolism , rho GTP-Binding Proteins/physiology , Animals , Cell Membrane/metabolism , Electroporation , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Duchenne/therapy , Transfection , Up-Regulation , Utrophin/genetics , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...