Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(3): e0301372, 2024.
Article in English | MEDLINE | ID: mdl-38547143

ABSTRACT

The importance of mitochondria in tissue homeostasis, stress responses and human diseases, combined to their ability to transition between various structural and functional states, makes them excellent organelles for monitoring cell health. There is therefore a need for technologies to accurately analyze and quantify changes in mitochondrial organization in a variety of cells and cellular contexts. Here we present an innovative computerized method that enables accurate, multiscale, fast and cost-effective analysis of mitochondrial shape and network architecture from confocal fluorescence images by providing more than thirty features. In order to facilitate interpretation of the quantitative results, we introduced two innovations: the use of Kiviat-graphs (herein named MitoSpider plots) to present highly multidimensional data and visualization of the various mito-cellular configurations in the form of morphospace diagrams (called MitoSigils). We tested our fully automated image analysis tool on rich datasets gathered from live normal human skin cells cultured under basal conditions or exposed to specific stress including UVB irradiation and pesticide exposure. We demonstrated the ability of our proprietary software (named MitoTouch) to sensitively discriminate between control and stressed dermal fibroblasts, and between normal fibroblasts and other cell types (including cancer tissue-derived fibroblasts and primary keratinocytes), showing that our automated analysis captures subtle differences in morphology. Based on this novel algorithm, we report the identification of a protective natural ingredient that mitigates the deleterious impact of hydrogen peroxide (H2O2) on mitochondrial organization. Hence we conceived a novel wet-plus-dry pipeline combining cell cultures, quantitative imaging and semiotic analysis for exhaustive analysis of mitochondrial morphology in living adherent cells. Our tool has potential for broader applications in other research areas such as cell biology and medicine, high-throughput drug screening as well as predictive and environmental toxicology.


Subject(s)
Hydrogen Peroxide , Mitochondria , Humans , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Software , Image Processing, Computer-Assisted/methods , Algorithms
2.
Comput Struct Biotechnol J ; 21: 5609-5619, 2023.
Article in English | MEDLINE | ID: mdl-38047232

ABSTRACT

Mitochondria are essential organelles that play crucial roles in cellular energy metabolism, calcium signaling and apoptosis. Their importance in tissue homeostasis and stress responses, combined to their ability to transition between various structural and functional states, make them excellent organelles for monitoring cellular health. Quantitative assessment of mitochondrial morphology can therefore provide valuable insights into environmentally-induced cell damage. High-content screening (HCS) provides a powerful tool for analyzing organelles and cellular substructures. We developed a fully automated and miniaturized HCS wet-plus-dry pipeline (MITOMATICS) exploiting mitochondrial morphology as a marker for monitoring cellular health or damage. MITOMATICS uses an in-house, proprietary software (MitoRadar) to enable fast, exhaustive and cost-effective analysis of mitochondrial morphology and its inherent diversity in live cells. We applied our pipeline and big data analytics software to assess the mitotoxicity of selected chemicals, using the mitochondrial uncoupler CCCP as an internal control. Six different pesticides (inhibiting complexes I, II and III of the mitochondrial respiratory chain) were tested as individual compounds and five other pesticides present locally in Occitanie (Southern France) were assessed in combination to determine acute mitotoxicity. Our results show that the assayed pesticides exhibit specific signatures when used as single compounds or chemical mixtures and that they function synergistically to impact mitochondrial architecture. Study of environment-induced mitochondrial damage has the potential to open new fields in mechanistic toxicology, currently underexplored by regulatory toxicology and exposome research. Such exploration could inform health policy guidelines and foster pharmacological intervention, water, air and soil pollution control and food safety.

3.
Biomolecules ; 10(11)2020 11 22.
Article in English | MEDLINE | ID: mdl-33266510

ABSTRACT

Protein phosphorylation is a post-translational modification essential for the control of the activity of most enzymes in the cell. This protein modification results from a fine-tuned balance between kinases and phosphatases. PP2A is one of the major serine/threonine phosphatases that is involved in the control of a myriad of different signaling cascades. This enzyme, often misregulated in cancer, is considered a tumor suppressor. In this review, we will focus on PP2A-B55, a particular holoenzyme of the family of the PP2A phosphatases whose specific role in cancer development and progression has only recently been highlighted. The discovery of the Greatwall (Gwl)/Arpp19-ENSA cascade, a new pathway specifically controlling PP2A-B55 activity, has been shown to be frequently altered in cancer. Herein, we will review the current knowledge about the mechanisms controlling the formation and the regulation of the activity of this phosphatase and its misregulation in cancer.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Protein Phosphatase 2/pharmacokinetics , Animals , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
5.
J Cell Biol ; 218(2): 541-558, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30626720

ABSTRACT

Greatwall (GWL) is an essential kinase that indirectly controls PP2A-B55, the phosphatase counterbalancing cyclin B/CDK1 activity during mitosis. In Xenopus laevis egg extracts, GWL-mediated phosphorylation of overexpressed ARPP19 and ENSA turns them into potent PP2A-B55 inhibitors. It has been shown that the GWL/ENSA/PP2A-B55 axis contributes to the control of DNA replication, but little is known about the role of ARPP19 in cell division. By using conditional knockout mouse models, we investigated the specific roles of ARPP19 and ENSA in cell division. We found that Arpp19, but not Ensa, is essential for mouse embryogenesis. Moreover, Arpp19 ablation dramatically decreased mouse embryonic fibroblast (MEF) viability by perturbing the temporal pattern of protein dephosphorylation during mitotic progression, possibly by a drop of PP2A-B55 activity inhibition. We show that these alterations are not prevented by ENSA, which is still expressed in Arpp19 Δ/Δ MEFs, suggesting that ARPP19 is essential for mitotic division. Strikingly, we demonstrate that unlike ARPP19, ENSA is not required for early embryonic development. Arpp19 knockout did not perturb the S phase, unlike Ensa gene ablation. We conclude that, during mouse embryogenesis, the Arpp19 and Ensa paralog genes display specific functions by differentially controlling cell cycle progression.


Subject(s)
Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mitosis/physiology , Phosphoproteins/metabolism , S Phase/physiology , Animals , Embryo, Mammalian/cytology , Embryonic Development/physiology , Fibroblasts/cytology , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Xenopus laevis
6.
Nat Commun ; 8(1): 206, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28785014

ABSTRACT

The Greatwall/Ensa/PP2A-B55 pathway is essential for controlling mitotic substrate phosphorylation and mitotic entry. Here, we investigate the effect of the knockdown of the Gwl substrate, Ensa, in human cells. Unexpectedly, Ensa knockdown promotes a dramatic extension of S phase associated with a lowered density of replication forks. Notably, Ensa depletion results in a decrease of Treslin levels, a pivotal protein for the firing of replication origins. Accordingly, the extended S phase in Ensa-depleted cells is completely rescued by the overexpression of Treslin. Our data herein reveal a new mechanism by which normal cells regulate S-phase duration by controlling the ubiquitin-proteasome degradation of Treslin in a Gwl/Ensa-dependent pathway.The Greatwall/Ensa/PP2A-B55 pathway controls mitotic substrate phosphorylation and mitotic entry. Here the authors show that cells regulate S phase duration by controlling the ubiquitin-proteasome degradation of Treslin in a Gwl/Ensa-dependent pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Peptides/metabolism , S Phase , Cell Cycle Proteins/genetics , Cell Division , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis , Peptides/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
7.
Sci Rep ; 7: 44001, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287092

ABSTRACT

In human eggs, aneuploidy increases with age and can result in infertility and genetic diseases. Studies in mouse oocytes suggest that reduced centromere cohesion and spindle assembly checkpoint (SAC) activity could be at the origin of chromosome missegregation. Little is known about these two features in humans. Here, we show that in human eggs, inter-kinetochore distances of bivalent chromosomes strongly increase with age. This results in the formation of univalent chromosomes during metaphase I (MI) and of single chromatids in metaphase II (MII). We also investigated SAC activity by checking the localization of BUB1 and BUBR1. We found that they localize at the kinetochore with a similar temporal timing than in mitotic cells and in a MPS1-dependent manner, suggesting that the SAC signalling pathway is active in human oocytes. Moreover, our data also suggest that this checkpoint is inactivated when centromere cohesion is lost in MI and consequently cannot inhibit premature sister chromatid separation. Finally, we show that the kinetochore localization of BUB1 and BUBR1 decreases with the age of the oocyte donors. This could contribute to oocyte aneuploidy.


Subject(s)
Aneuploidy , Kinetochores/metabolism , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Humans , Oocytes/cytology , Protein Transport
8.
Int J Dev Biol ; 60(7-8-9): 245-254, 2016.
Article in English | MEDLINE | ID: mdl-27759153

ABSTRACT

Entry into mitosis requires the coordinated activation of various protein kinases and phosphatases that together activate sequential signaling pathways allowing entry, progression and exit of mitosis. The limiting step is thought to be the activation of the mitotic Cdk1-cyclin B kinase. However, this model has recently evolved with new data showing that in addition to the Cdk1-cyclin B complex, Greatwall (Gwl) kinase is also required to enter into and maintain mitosis. This new concept proposes that entry into mitosis is now based on the combined activation of both kinases Cdk1-cyclin B and Gwl, the former promoting massive phosphorylation of mitotic substrates and the latter inhibiting PP2A-B55 phosphatase responsible for dephosphorylation of these substrates. Activated Gwl phosphorylates both Arpp19 and ENSA, which associate and inhibit PP2A-B55. This pathway seems relatively well conserved from yeast to humans, although some differences appear based on models or techniques used. While Gwl is activated by phosphorylation, its inactivation requires dephosphorylation of critical residues. Several phosphatases such as PP1, PP2A-B55 and FCP1 are required to control the dephosphorylation and inactivation of Gwl and a properly regulated mitotic exit. Gwl has also been reported to be involved in cancer processes and DNA damage recovery. These new findings support the idea that the Gwl-Arpp19/ENSA-PP2A-B55 pathway is essential to achieve an efficient division of cells and to maintain genomic stability.


Subject(s)
Meiosis/physiology , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Xenopus Proteins/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Phosphorylation , Xenopus laevis
9.
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
10.
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
11.
Chem Biol ; 16(6): 657-66, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19549603

ABSTRACT

RhoGEFs (guanine nucleotide exchange factors of the Rho GTPase family) are upstream regulators of cell adhesion and migration pathways, thus representing attractive yet relatively unexplored targets for the development of anti-invasive drugs. We screened for chemical inhibitors of TrioN, the N-terminal GEF domain of the multidomain Trio protein, and identified ITX3 as a nontoxic inhibitor. In transfected mammalian cells, ITX3 blocked TrioN-mediated dorsal membrane ruffling and Rac1 activation while having no effect on GEF337-, Tiam1-, or Vav2-mediated RhoA or Rac1 activation. ITX3 specifically inhibited endogenous TrioN activity, as evidenced by its ability to inhibit neurite outgrowth in nerve growth factor (NGF)-stimulated PC12 cells or C2C12 differentiation into myotubes. This study introduces a selective cell active inhibitor of the Trio/RhoG/Rac1 pathway and validates RhoGEFs as druggable targets.


Subject(s)
Benzimidazoles/pharmacology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Signal Transduction/drug effects , Thiazoles/pharmacology , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Benzimidazoles/chemistry , Cell Differentiation , Cell Line , Cell Movement , Guanine Nucleotide Exchange Factors/metabolism , Humans , Kinetics , Mice , Neurites/physiology , Nitrobenzenes/chemistry , Nitrobenzenes/pharmacology , PC12 Cells , Protein Structure, Tertiary , Rats , Thiazoles/chemistry
12.
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
13.
Cancer Res ; 68(16): 6559-68, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701479

ABSTRACT

Cadherins are transmembrane glycoproteins that mediate Ca(2+)-dependent homophilic cell-cell adhesion and play a crucial role in proliferation, differentiation, and cell transformation. The goal of this study was to understand why R-cadherin is found in rhabdomyosarcomas (RMS), tumors of skeletal muscle origin, whereas it is absent in normal myoblasts. We show that R-cadherin expression in C2C12 myoblasts causes inhibition of myogenesis induction and impairment of cell cycle exit when cells are cultured in differentiation medium. Furthermore, R-cadherin expression elicits myoblast transformation, as shown by anchorage-independent growth in soft agar in vivo tumor formation assays and increased cell motility. In contrast, inhibition of R-cadherin expression using RNA interference hinders growth of RD cell line in soft agar and its tumorigenicity in mice. The analysis of the nature of R-cadherin-mediated signals shows that R-cadherin-dependent adhesion increases Rac1 activity. Dominant-negative forms of Rac1 inhibit R-cadherin-mediated signaling and transformation. In addition, expression of R-cadherin results in perturbed function of endogenous N-cadherin and M-cadherin. Together, these data suggest that R-cadherin expression inhibits myogenesis and induces myoblast transformation through Rac1 activation. Therefore, the properties of R-cadherin make it an attractive target for therapeutic intervention in RMS.


Subject(s)
Cadherins/metabolism , Cell Transformation, Neoplastic , Muscle Development/physiology , Myoblasts/cytology , Myoblasts/metabolism , Rhabdomyosarcoma/pathology , rac1 GTP-Binding Protein/metabolism , Animals , Cadherins/genetics , Cell Adhesion , Cell Differentiation , Cell Movement , Cells, Cultured , Enzyme Activation , Gene Expression Regulation , Genes, Dominant , Humans , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Mice , Phosphorylation , Rhabdomyosarcoma/metabolism , Signal Transduction , Transfection , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics
14.
Biol Cell ; 99(9): 503-17, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17459003

ABSTRACT

BACKGROUND INFORMATION: N-cadherin, a member of the Ca(2+)-dependent cell-cell adhesion molecule family, plays an essential role in the induction of the skeletal muscle differentiation programme. However, the molecular mechanisms which govern the formation of N-cadherin-dependent cell-cell contacts in myoblasts remain unexplored. RESULTS: In the present study, we show that N-cadherin-dependent cell contact formation in myoblasts is defined by two stages. In the first phase, N-cadherin is highly mobile in the lamellipodia extensions between the contacting cells. The second stage corresponds to the formation of mature N-cadherin-dependent cell contacts, characterized by the immobilization of a pool of N-cadherin which appears to be clustered in the interdigitated membrane structures that are also membrane attachment sites for F-actin filaments. We also demonstrated that the formation of N-cadherin-dependent cell-cell contacts requires a co-ordinated and sequential activity of Rac1 and RhoA. Rac1 is involved in the first stage and facilitates N-cadherin-dependent cell-cell contact formation, but it is not absolutely required. Conversely, RhoA is necessary for N-cadherin-dependent cell contact formation, since, via ROCK (Rho-associated kinase) signalling and myosin 2 activation, it allows the stabilization of N-cadherin at the cell-cell contact sites. CONCLUSIONS: We have shown that Rac1 and RhoA have opposite effects on N-cadherin-dependent cell-cell contact formation in C2C12 myoblasts and act sequentially to allow its formation.


Subject(s)
Cadherins/metabolism , Myoblasts/metabolism , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/physiology , Cadherins/drug effects , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Humans
15.
Mol Biol Cell ; 18(5): 1734-43, 2007 May.
Article in English | MEDLINE | ID: mdl-17332503

ABSTRACT

Cadherins are transmembrane glycoproteins that mediate Ca(2+)-dependent homophilic cell-cell adhesion and play crucial role during skeletal myogenesis. M-cadherin is required for myoblast fusion into myotubes, but its mechanisms of action remain unknown. The goal of this study was to cast some light on the nature of the M-cadherin-mediated signals involved in myoblast fusion into myotubes. We found that the Rac1 GTPase activity is increased at the time of myoblast fusion and it is required for this process. Moreover, we showed that M-cadherin-dependent adhesion activates Rac1 and demonstrated the formation of a multiproteic complex containing M-cadherin, the Rho-GEF Trio, and Rac1 at the onset of myoblast fusion. Interestingly, Trio knockdown efficiently blocked both the increase in Rac1-GTP levels, observed after M-cadherin-dependent contact formation, and myoblast fusion. We conclude that M-cadherin-dependent adhesion can activate Rac1 via the Rho-GEF Trio at the time of myoblast fusion.


Subject(s)
Cadherins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Myoblasts, Skeletal/metabolism , Neuropeptides/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Base Sequence , Cadherins/antagonists & inhibitors , Cell Adhesion , Cell Fusion , Cell Line , Enzyme Activation , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Mice , Multiprotein Complexes , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , rac1 GTP-Binding Protein
16.
Mol Biol Cell ; 17(2): 749-59, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16291866

ABSTRACT

The Rho family of GTP-binding proteins plays critical roles during myogenesis induction. To elucidate their role later during myogenesis, we have analyzed RhoA function during myoblast fusion into myotubes. We find that RhoA activity is rapidly and transiently increased when cells are shifted into differentiation medium and then is decreased until myoblast fusion. RhoA activity must be down-regulated to allow fusion, because expression of a constitutively active form of RhoA (RhoAV14) inhibits this process. RhoAV14 perturbs the expression and localization of M-cadherin, a member of the Ca2+-dependent cell-cell adhesion molecule family that has an essential role in skeletal muscle cell differentiation. This mutant does not affect N-cadherin and other proteins involved in myoblast fusion, beta1-integrin and ADAM12. Active RhoA induces the entry of M-cadherin into a degradative pathway and thus decreases its stability in correlation with the monoubiquitination of M-cadherin. Moreover, p120 catenin association with M-cadherin is decreased in RhoAV14-expressing cells, which is partially reverted by the inhibition of the RhoA effector Rho-associated kinase ROCK. ROCK inhibition also restores M-cadherin accumulation at the cell-cell contact sites. We propose that the sustained activation of the RhoA pathway inhibits myoblast fusion through the regulation of p120 activity, which controls cadherin internalization and degradation.


Subject(s)
Cadherins/metabolism , Myoblasts/enzymology , rhoA GTP-Binding Protein/physiology , ADAM Proteins/metabolism , ADAM12 Protein , Animals , Cadherins/analysis , Catenins , Cell Adhesion Molecules/metabolism , Cell Fusion , Cell Line , Integrin beta1/metabolism , Lysosomes/metabolism , Mice , Models, Biological , Myoblasts/cytology , Myoblasts/metabolism , Phosphoproteins/metabolism , RNA Interference , rhoA GTP-Binding Protein/metabolism , Delta Catenin
17.
Eur J Biochem ; 271(15): 3255-64, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15265045

ABSTRACT

In living organisms, Ca2+ signalling is central to cell physiology. The Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) has been widely used as a probe to test the role of calcium in a large variety of cell functions. Here we show that in most cell types BAPTA has a potent actin and microtubule depolymerizing activity and that this activity is completely independent of Ca2+ chelation. Thus, the depolymerizing effect of BAPTA is shared by a derivative (D-BAPTA) showing a dramatically reduced calcium chelating activity. Because the extraordinary depolymerizing activity of BAPTA could be due to a general depletion of cell fuel molecules such as ATP, we tested the effects of BAPTA on cellular ATP levels and on mitochondrial function. We find that BAPTA depletes ATP pools and affects mitochondrial respiration in vitro as well as mitochondrial shape and distribution in cells. However, these effects are unrelated to the Ca2+ chelating properties of BAPTA and do not account for the depolymerizing effect of BAPTA on the cell cytoskeleton. We propose that D-BAPTA should be systematically introduced in calcium signalling experiments, as controls for the known and unknown calcium independent effects of BAPTA. Additionally, the concomitant depolymerizing effect of BAPTA on both tubulin and actin assemblies is intriguing and may lead to the identification of a new control mechanism for cytoskeleton assembly.


Subject(s)
Calcium/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Calcium/antagonists & inhibitors , Cell Line , Chelating Agents/pharmacology , Egtazic Acid/chemistry , Formaldehyde/metabolism , GTP Phosphohydrolases/metabolism , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Xenopus
18.
Oncogene ; 23(13): 2420-30, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-14691446

ABSTRACT

Cadherins are a family of transmembrane glycoproteins that mediate Ca(2+)-dependent homophilic cell-cell adhesion and play a crucial role in cell differentiation. E-cadherin-mediated cell-cell adhesion is lost during the development of most epithelial cancers. This study examines cadherin-dependent adhesion in cell lines derived from rhabdomyosarcoma (RMS), a highly malignant soft-tissue tumor committed to the myogenic lineage, but arrested prior to terminal differentiation. We analysed the expression of cadherins and associated catenins at the mRNA and protein levels as well as their localization in nine RMS-derived cell lines relative to normal myoblasts. We show a decrease in the expression of cadherins and catenins in all RMS-derived cell lines compared to control cells. This decrease in the expression of N- and M-cadherin was confirmed in RMS biopsies. In contrast, R-cadherin is found expressed in RMS, whereas it is normally absent in normal myoblasts. We show that a decrease of R-cadherin expression using RNA interference inhibits cell proliferation of the RD cell line. In addition to their diminished expression, cadherins and catenins do not localize to intercellular contacts in embryonal RMS (ERMS), whereas specific persistent localization is seen in alveolar RMS (ARMS)-derived cell lines. Thus, RMS exhibit defects in the expression of molecules of the cadherin family. Defects in the localization of these adhesion molecules at the sites of cell-cell contact are specifically observed in the ERMS subtype. In addition, our data suggest that R-cadherin is a specific diagnostic marker for RMS and is also an important factor of RMS cell proliferation.


Subject(s)
Cadherins/metabolism , Cell Division/physiology , Cell Transformation, Neoplastic/metabolism , Rhabdomyosarcoma/metabolism , Cadherins/genetics , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Desmoplakins , Humans , Rhabdomyosarcoma/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , alpha Catenin , beta Catenin , p120 GTPase Activating Protein/genetics , p120 GTPase Activating Protein/metabolism
19.
J Muscle Res Cell Motil ; 24(4-6): 309-13, 2003.
Article in English | MEDLINE | ID: mdl-14620744

ABSTRACT

The small GTPases of the Rho subfamily (RhoA, Rac1 and Cdc42) are signaling molecules involved in cytoskeleton remodeling and gene transcription. Their activities are important for many cellular processes, including myogenesis. Classical cadherin adhesion molecules are key determinants of cell recognition and tissus morphogenesis and act as adhesion-activated signaling receptors. Rho GTPases have emerged as key mediators of their activity. Not only signal transduction pathways link cadherins to Rho GTPases but also Rho GTPases to cadherins. We focus in this review on the role of cadherins and Rho GTPases in normal myogenesis as well as in pathological development of rhabdomyosarcoma.


Subject(s)
Cadherins/metabolism , Muscle, Skeletal/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cadherins/chemistry , Cell Adhesion/physiology , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , rho GTP-Binding Proteins/chemistry
20.
J Cell Biol ; 158(5): 953-65, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12213839

ABSTRACT

N-cadherin, a member of the Ca(2+)-dependent cell-cell adhesion molecule family, plays an essential role in skeletal muscle cell differentiation. We show that inhibition of N-cadherin-dependent adhesion impairs the upregulation of the two cyclin-dependent kinase inhibitors p21 and p27, the expression of the muscle-specific genes myogenin and troponin T, and C2C12 myoblast fusion. To determine the nature of N-cadherin-mediated signals involved in myogenesis, we investigated whether N-cadherin-dependent adhesion regulates the activity of Rac1, Cdc42Hs, and RhoA. N-cadherin-dependent adhesion decreases Rac1 and Cdc42Hs activity, and as a consequence, c-jun NH2-terminal kinase (JNK) MAPK activity but not that of the p38 MAPK pathway. On the other hand, N-cadherin-mediated adhesion increases RhoA activity and activates three skeletal muscle-specific promoters. Furthermore, RhoA activity is required for beta-catenin accumulation at cell-cell contact sites. We propose that cell-cell contacts formed via N-cadherin trigger signaling events that promote the commitment to myogenesis through the positive regulation of RhoA and negative regulation of Rac1, Cdc42Hs, and JNK activities.


Subject(s)
Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Trans-Activators/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Adhesion , Cell Differentiation , Gene Expression Regulation , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/enzymology , Promoter Regions, Genetic/genetics , Protein-Tyrosine Kinases/metabolism , Time Factors , beta Catenin , p38 Mitogen-Activated Protein Kinases , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...