Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cell ; 30(2): 214-26, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18439900

ABSTRACT

AMPK is a highly conserved sensor of cellular energy status that is activated under conditions of low intracellular ATP. AMPK responds to energy stress by suppressing cell growth and biosynthetic processes, in part through its inhibition of the rapamycin-sensitive mTOR (mTORC1) pathway. AMPK phosphorylation of the TSC2 tumor suppressor contributes to suppression of mTORC1; however, TSC2-deficient cells remain responsive to energy stress. Using a proteomic and bioinformatics approach, we sought to identify additional substrates of AMPK that mediate its effects on growth control. We report here that AMPK directly phosphorylates the mTOR binding partner raptor on two well-conserved serine residues, and this phosphorylation induces 14-3-3 binding to raptor. The phosphorylation of raptor by AMPK is required for the inhibition of mTORC1 and cell-cycle arrest induced by energy stress. These findings uncover a conserved effector of AMPK that mediates its role as a metabolic checkpoint coordinating cell growth with energy status.


Subject(s)
Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Transcription Factors/metabolism , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Apoptosis , Cell Cycle , Cell Line , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred Strains , Multienzyme Complexes/genetics , Multiprotein Complexes , Peptide Library , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteomics , Regulatory-Associated Protein of mTOR , Serine/metabolism , Substrate Specificity , TOR Serine-Threonine Kinases , Transcription Factors/antagonists & inhibitors
2.
J Exp Biol ; 211(Pt 1): 15-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18083727

ABSTRACT

Muscle LIM protein (MLP) is a cytoskeletal protein located at the Z-disc of sarcomeres. Mutations in the human MLP gene are associated with hypertrophic and dilated cardiomyopathy. MLP has been proposed to be a key player in the stretch-sensing response, but the molecular mechanisms underlying its function in normal and diseased cardiac muscle have not been established. A Drosophila homolog, Mlp84B, displays a similar subcellular localization at the Z-disc of sarcomeres throughout development and in the adult, suggesting Drosophila as a model to study MLP function. Here we employed genetic ablation and cardiac-specific RNA interference (RNAi) knockdown of mlp84B to investigate its role in heart function. We found that Mlp84B-deficient or heart-specific RNAi knockdown flies exhibit diastolic interval prolongation, heart rhythm abnormalities and a reduced lifespan, while showing no obvious structural phenotype. Our data demonstrate that Mlp84B is essential for normal cardiac function and establish the Drosophila model for the investigation of the mechanisms connecting defective cardiac Z-disc components to the development of cardiomyopathy.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Heart/physiology , Muscle Proteins/metabolism , Actinin/metabolism , Amino Acid Sequence , Animals , Diastole , Drosophila Proteins/chemistry , Drosophila Proteins/deficiency , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Heart/embryology , LIM Domain Proteins , Longevity , Molecular Sequence Data , Mortality , Muscle Proteins/chemistry , Muscle Proteins/deficiency , Myocardium/cytology , Myocardium/metabolism , Myofibrils/metabolism , Organ Specificity , Protein Transport , Sarcomeres/metabolism
3.
J Am Chem Soc ; 129(14): 4423-9, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-17367139

ABSTRACT

Plastocyanin is a small blue copper protein that shuttles electrons as part of the photosynthetic redox chain. Its redox behavior is changed at low pH as a result of protonation of the solvent-exposed copper-coordinating histidine. Protonation and subsequent redox inactivation could have a role in the down regulation of photosynthesis. As opposed to plastocyanin from other sources, in fern plastocyanin His90 protonation at low pH has been reported not to occur. Two possible reasons for that have been proposed: pi-pi stacking between Phe12 and His90 and lack of a hydrogen bond with the backbone oxygen of Gly36. We have produced this fern plastocyanin recombinantly and examined the properties of wild-type protein and mutants Phe12Leu, Gly36Pro, and the double mutant with NMR spectroscopy, X-ray crystallography, and cyclic voltammetry. The results demonstrate that, contrary to earlier reports, protonation of His90 in the wild-type protein does occur in solution with a pKa of 4.4 (+/-0.1). Neither the single mutants nor the double mutant exhibit a change in protonation behavior, indicating that the suggested interactions have no influence. The crystal structure at low pH of the Gly36Pro variant does not show His90 protonation, similar to what was found for the wild-type protein. The structure suggests that movement of the imidazole ring is hindered by crystal contacts. This study illustrates a significant difference between results obtained in solution by NMR and by crystallography.


Subject(s)
Dryopteris/chemistry , Histidine/analogs & derivatives , Organometallic Compounds/chemistry , Plastocyanin/chemistry , Protons , Amides/chemistry , Crystallography, X-Ray , Glycine/genetics , Glycine/metabolism , Histidine/chemistry , Hydrogen-Ion Concentration , Ligands , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Methods Mol Med ; 112: 175-82, 2005.
Article in English | MEDLINE | ID: mdl-16010016

ABSTRACT

Embryonic stem (ES) cells represent a source for cell-based regenerative therapies of heart failure. The pluripotency and the plasticity of ES cells allow them to be committed to a cardiac lineage following treatment with growth factors of the transforming growth factor (TGF)-beta superfamily. We describe a protocol designed to turn on expression of cardiac-specific genes in undifferentiated murine ES cells stimulated with BMP2 and/or TGF-beta. Cell commitment results in a significant improvement in spontaneous cardiac differentiation of ES cells both in vitro and in vivo.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Lineage , Cells, Cultured , Cricetinae , Myocardium/cytology , Polymerase Chain Reaction , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
5.
Hum Mol Genet ; 14(10): 1367-77, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15829506

ABSTRACT

Mutations of genes encoding contractile proteins are responsible for familial hypertrophic cardiomyopathies. Understanding the process of differentiation of cardiomyocytes carrying a mutated protein is a crucial step towards potential treatments of inherited cardiac disorders. Embryonic Stem (ES) cells which faithfully recapitulate in vitro the process of cardiac cell differentiation can be genetically modified to incorporate a mutation mimicking a cardiomyopathy. ES cell lines engineered to express a wild-type (MLC2vGFP) or a mutated form (R58QMLC2vGFP) of ventricular myosin light chain 2 (MLC2v) fused to GFP were differentiated into cardiomyocytes within embryoid bodies (EBs). Visualization of GFP combined with sarcomeric actinin immunofluorescence of EBs revealed that mutated MLC2v dramatically prevented myofibrillogenesis. Cardiomyocytes expressing wild-type MLC2v featured spontaneous Ca(2+) spiking, but not those harboring the mutation. Expression of cardiac transcription factors Mef2c, GATAs, myocardin and Nkx2.5 was not affected by cell expression of mutated MLC2v. A dramatic decrease in expression of mRNAs encoding alpha-actin, MLC2a and MLC2v was observed in R58QMLC2vGFP EBs. This event was attributed to a failure of Mef2c to translocate into the nucleus, a Ca(2+)-dependent process. Expression in mutated cells of a constitutively active Ca(2+)- and calmodulin-dependent kinase II or treating EBs with ionomycin fully restored translocation of Mef2c into the nucleus and expression of mRNAs encoding sarcomeric proteins partially rescued contractile activity of EBs. Alteration of Ca(2+) homeostasis in mutated cardioblasts affects the transcriptional program of cardiac cell differentiation leading to a defect in myofibrillogenesis, and, in turn, in contractility. Genetically modified ES cells provide a unique cell model to determine abnormalities in Ca(2+) homeostasis underlying progression of human cardiomyopathies.


Subject(s)
Calcium/physiology , Cardiomyopathies/metabolism , Homeostasis/physiology , Myocytes, Cardiac/metabolism , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Genes, Reporter , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism
6.
Mol Biol Cell ; 16(5): 2414-23, 2005 May.
Article in English | MEDLINE | ID: mdl-15758029

ABSTRACT

In the adult, the heart rate is driven by spontaneous and repetitive depolarizations of pacemaker cells to generate a firing of action potentials propagating along the conduction system and spreading into the ventricles. In the early embryo before E9.5, the pacemaker ionic channel responsible for the spontaneous depolarization of cells is not yet functional. Thus the mechanisms that initiate early heart rhythm during cardiogenesis are puzzling. In the absence of a functional pacemaker ionic channel, the oscillatory nature of inositol 1,4,5-trisphosphate (InsP3)-induced intracellular Ca2+ signaling could provide an alternative pacemaking mechanism. To test this hypothesis, we have engineered pacemaker cells from embryonic stem (ES) cells, a model that faithfully recapitulates early stages of heart development. We show that InsP3-dependent shuttle of free Ca2+ in and out of the endoplasmic reticulum is essential for a proper generation of pacemaker activity during early cardiogenesis and fetal life.


Subject(s)
Calcium Signaling/physiology , Fetal Heart/embryology , Fetal Heart/metabolism , Heart Conduction System/embryology , Heart Conduction System/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calreticulin/genetics , Calreticulin/metabolism , Clone Cells , DNA, Complementary/genetics , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Mice , Models, Cardiovascular , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
J Soc Biol ; 198(2): 145-51, 2004.
Article in French | MEDLINE | ID: mdl-15368965

ABSTRACT

Confocal microscopy offers important advantages compared to conventional epifluorescence microscopy. It works as an "optical microtome" leading to a accurate image resolution of a defined focal plane. Furthermore, the addition of a Nipkow disk on the confocal microscope greatly accelerates the image acquisition, up to 30 frames per second. Nevertheless, the software-assisted mathematical restoration of images acquired using a wide-field microscope allows to get images with a resolution similar to the one obtained in confocal microscopy. These imaging technologies allowed us to monitor on line cardiac differentiation of murine embryonic stem (ES) cells within 3D structures called embryoid bodies. The high rate acquisition of images using the confocal microscope equipped with a Nipkow disk allows to monitor calcium spiking in differentiating cardiomyocytes within embryoid bodies.


Subject(s)
Microscopy, Confocal/methods , Muscle Cells/cytology , Myocardium/cytology , Animals , Cell Differentiation , Humans , Stem Cells/cytology
8.
J Cell Biochem ; 93(4): 681-7, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15389971

ABSTRACT

Over the past decade, cell transplantation has been recognized as a mean of repairing infarcted myocardium. Both adult stem cells and differentiated cells have yielded encouraging results with regard to engraftment into postinfarction scars. However, these cells now feature serious restrictions. Asan alternative, embryonic stem (ES) cells are particularly attractive, because of their plasticity and the subsequent possibility to drive them towards a cardiomyogenic phenotype after exposure to appropriate growth factors. An additional theoretical advantage of ES cells is their expected immune privilege. In this article, we summarize the findings obtained in cell therapy using ES cells and discuss the molecular mechanisms of cardiac specification of the cells.


Subject(s)
Embryo, Mammalian/cytology , Myocardium/cytology , Pluripotent Stem Cells/physiology , Stem Cell Transplantation , Animals , Cell Differentiation , Cell Lineage , Humans , Regeneration
10.
Circ Res ; 92(4): 444-52, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12600892

ABSTRACT

Macromolecules are transported in and out of the nucleus through nuclear pores. It is poorly understood how these megadalton conduits support nucleocytoplasmic traffic during genetic reprogramming associated with cell commitment to a specific lineage. Murine embryonic stem cells were differentiated into cardiomyocytes within embryoid bodies, and contracting cells expressing myocardial-specific proteins were isolated from the mesodermal layer. Compared with postmitotic cardiac cells from heart muscle, these proliferative and differentiating stem cell-derived cardiomyocytes demonstrated a significantly lower density of nuclear pores. At nanoscale resolution, the pore channel was commonly unoccupied in heart muscle-isolated cardiac cells, yet a dense material, presumably the central transporter, protruded toward the cytosolic face of the nuclear pore complex in stem cell-derived cardiomyocytes. Stem cell-derived cardiac cells distributed the nuclear transport factor Ran in the nucleus, decreased the number of spare nuclear pore complexes from the cytosolic annulate lamellae reservoir, and expressed a set of nucleoporins, NUP214, NUP358, NUP153, and p62, involved in nuclear transport. Stem cell-derived cardiomyocytes secured transport of nuclear constitutive proteins, cardiogenic transcription factors, and cell cycle regulators, including the prototypic histone H1, myocyte enhancer binding factor 2, and p53. Thus, differentiating stem cell-derived cardiomyocytes undergo structural adaptation and mobilize nuclear transport regulators in support of nucleocytoplasmic communication during commitment to mature cardiac lineage.


Subject(s)
Myocytes, Cardiac/physiology , Nuclear Pore/physiology , Stem Cells/cytology , Animals , Biological Transport , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cell Lineage , Mice , Microfibrils/metabolism , Microfibrils/ultrastructure , Microscopy, Electron , Microscopy, Phase-Contrast , Myocytes, Cardiac/cytology , Myocytes, Cardiac/ultrastructure , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/metabolism , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Stem Cells/ultrastructure , Time Factors , ran GTP-Binding Protein/metabolism
11.
J Cell Biol ; 158(1): 103-13, 2002 Jul 08.
Article in English | MEDLINE | ID: mdl-12105184

ABSTRACT

Calreticulin (crt) is an ubiquitously expressed and multifunctional Ca(2+)-binding protein that regulates diverse vital cell functions, including Ca(2+) storage in the ER and protein folding. Calreticulin deficiency in mice is lethal in utero due to defects in heart development and function. Herein, we used crt(-/-) embryonic stem (ES) cells differentiated in vitro into cardiac cells to investigate the molecular mechanisms underlying heart failure of knockout embryos. After 8 d of differentiation, beating areas were prominent in ES-derived wild-type (wt) embryoid bodies (EBs), but not in ES-derived crt(-/-) EBs, despite normal expression levels of cardiac transcription factors. Crt(-/-) EBs exhibited a severe decrease in expression and a lack of phosphorylation of ventricular myosin light chain 2 (MLC2v), resulting in an impaired organization of myofibrils. Crt(-/-) phenotype could be recreated in wt cells by chelating extracellular or cytoplasmic Ca(2+) with EGTA or BAPTA, or by inhibiting Ca(2+)/calmodulin-dependent kinases (CaMKs). An imposed ionomycin-triggered cystolic-free Ca(2+) concentration ([Ca(2+)](c)) elevation restored the expression, phosphorylation, and insertion of MLC2v into sarcomeric structures and in turn the myofibrillogenesis. The transcription factor myocyte enhancer factor C2 failed to accumulate into nuclei of crt(-/-) cardiac cells in the absence of ionomycin-triggered [Ca(2+)](c) increase. We conclude that the absence of calreticulin interferes with myofibril formation. Most importantly, calreticulin deficiency revealed the importance of a Ca(2+)-dependent checkpoint critical for early events during cardiac myofibrillogenesis.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Heart/physiology , Ionomycin/pharmacology , Muscle Development/physiology , Myocardium/metabolism , Organic Chemicals , Ribonucleoproteins/metabolism , Animals , Benzothiazoles , Blotting, Western , Calreticulin , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Cytosol/metabolism , Diamines , Electrophoresis, Gel, Two-Dimensional , Fluorescent Dyes/pharmacology , Heart/embryology , Immunohistochemistry , Ionophores/metabolism , Ionophores/pharmacology , Mice , Phenotype , Phosphorylation , Quinolines , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
12.
Oncogene ; 21(18): 2901-7, 2002 Apr 25.
Article in English | MEDLINE | ID: mdl-11973651

ABSTRACT

We have previously shown that expression of active Rac1 and Cdc4Hs inhibits skeletal muscle cell differentiation. We show here, by bromodeoxyuridine incorporation and cyclin D1 expression, that the expression of active Rac1 and Cdc42Hs but not RhoA impairs cell cycle exit of L6 myoblasts cultured in differentiation medium. Furthermore, expression of activated forms of Rac1 and Cdc42Hs elicits the loss of cell contact inhibition and anchorage-dependent growth as measured by focus forming activity and growth in soft agar. RhoA was once again not found to have this effect. We found a constitutive Rac1 and Cdc42Hs activation in three human rhabdomyosarcoma-derived cell lines, one of the most common causes of solid tumours arising from muscle precursors during childhood. Finally, dominant negative forms of Rac1 and Cdc42Hs inhibit cell proliferation of the RD rhabdomyosarcoma cell line. These data suggest an important role for the small GTPases Rac1 and Cdc42Hs in the generation of skeletal muscle tumours.


Subject(s)
Cell Transformation, Neoplastic , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/physiology , Cell Division , Cell Line , Cell Line, Transformed , Humans , Muscle, Skeletal/cytology , Rhabdomyosarcoma , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...