Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(23): e2217971121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805272

ABSTRACT

Myogenesis is a multistep process that requires a spatiotemporal regulation of cell events resulting finally in myoblast fusion into multinucleated myotubes. Most major insights into the mechanisms underlying fusion seem to be conserved from insects to mammals and include the formation of podosome-like protrusions (PLPs) that exert a driving force toward the founder cell. However, the machinery that governs this process remains poorly understood. In this study, we demonstrate that MTM1 is the main enzyme responsible for the production of phosphatidylinositol 5-phosphate, which in turn fuels PI5P 4-kinase α to produce a minor and functional pool of phosphatidylinositol 4,5-bisphosphate that concentrates in PLPs containing the scaffolding protein Tks5, Dynamin-2, and the fusogenic protein Myomaker. Collectively, our data reveal a functional crosstalk between a PI-phosphatase and a PI-kinase in the regulation of PLP formation.


Subject(s)
Cell Fusion , Myoblasts , Phosphatidylinositol Phosphates , Podosomes , Animals , Phosphatidylinositol Phosphates/metabolism , Mice , Myoblasts/metabolism , Myoblasts/cytology , Podosomes/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Muscle Development/physiology
2.
Theranostics ; 13(15): 5435-5451, 2023.
Article in English | MEDLINE | ID: mdl-37908733

ABSTRACT

Doxorubicin (Dox) is an effective anticancer molecule, but its clinical efficacy is limited by strong cardiotoxic side effects. Lysosomal dysfunction has recently been proposed as a new mechanism of Dox-induced cardiomyopathy. However, to date, there is a paucity of therapeutic approaches capable of restoring lysosomal acidification and function in the heart. Methods: We designed novel poly(lactic-co-glycolic acid) (PLGA)-grafted silica nanoparticles (NPs) and investigated their therapeutic potential in the primary prevention of Dox cardiotoxicity in cardiomyocytes and mice. Results: We showed that NPs-PLGA internalized rapidly in cardiomyocytes and accumulated inside the lysosomes. Mechanistically, NPs-PLGA restored lysosomal acidification in the presence of doxorubicin or bafilomycin A1, thereby improving lysosomal function and autophagic flux. Importantly, NPs-PLGA mitigated Dox-related mitochondrial dysfunction and oxidative stress, two main mechanisms of cardiotoxicity. In vivo, inhalation of NPs-PLGA led to effective and rapid targeting of the myocardium, which prevented Dox-induced adverse remodeling and cardiac dysfunction in mice. Conclusion: Our findings demonstrate a pivotal role for lysosomal dysfunction in Dox-induced cardiomyopathy and highlight for the first time that pulmonary-driven NPs-PLGA administration is a promising strategy against anthracycline cardiotoxicity.


Subject(s)
Cardiomyopathies , Nanoparticles , Mice , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/prevention & control , Doxorubicin/pharmacology , Myocytes, Cardiac/metabolism , Cardiomyopathies/metabolism , Oxidative Stress , Lysosomes/metabolism
3.
Nat Cell Biol ; 25(7): 975-988, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37414850

ABSTRACT

Metabolic demands fluctuate rhythmically and rely on coordination between the circadian clock and nutrient-sensing signalling pathways, yet mechanisms of their interaction remain not fully understood. Surprisingly, we find that class 3 phosphatidylinositol-3-kinase (PI3K), known best for its essential role as a lipid kinase in endocytosis and lysosomal degradation by autophagy, has an overlooked nuclear function in gene transcription as a coactivator of the heterodimeric transcription factor and circadian driver Bmal1-Clock. Canonical pro-catabolic functions of class 3 PI3K in trafficking rely on the indispensable complex between the lipid kinase Vps34 and regulatory subunit Vps15. We demonstrate that although both subunits of class 3 PI3K interact with RNA polymerase II and co-localize with active transcription sites, exclusive loss of Vps15 in cells blunts the transcriptional activity of Bmal1-Clock. Thus, we establish non-redundancy between nuclear Vps34 and Vps15, reflected by the persistent nuclear pool of Vps15 in Vps34-depleted cells and the ability of Vps15 to coactivate Bmal1-Clock independently of its complex with Vps34. In physiology we find that Vps15 is required for metabolic rhythmicity in liver and, unexpectedly, it promotes pro-anabolic de novo purine nucleotide synthesis. We show that Vps15 activates the transcription of Ppat, a key enzyme for the production of inosine monophosphate, a central metabolic intermediate for purine synthesis. Finally, we demonstrate that in fasting, which represses clock transcriptional activity, Vps15 levels are decreased on the promoters of Bmal1 targets, Nr1d1 and Ppat. Our findings open avenues for establishing the complexity for nuclear class 3 PI3K signalling for temporal regulation of energy homeostasis.


Subject(s)
Circadian Clocks , Circadian Clocks/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Vacuolar Sorting Protein VPS15/genetics , Vacuolar Sorting Protein VPS15/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Purines , Lipids
4.
Biomolecules ; 13(4)2023 03 24.
Article in English | MEDLINE | ID: mdl-37189331

ABSTRACT

Phosphoinositides (PIs) play a crucial role in regulating intracellular signaling, actin cytoskeleton rearrangements, and membrane trafficking by binding to specific domains of effector proteins. They are primarily found in the membrane leaflets facing the cytosol. Our study demonstrates the presence of a pool of phosphatidylinositol 3-monophosphate (PI3P) in the outer leaflet of the plasma membrane of resting human and mouse platelets. This pool of PI3P is accessible to exogenous recombinant myotubularin 3-phosphatase and ABH phospholipase. Mouse platelets with loss of function of class III PI 3-kinase and class II PI 3-kinase α have a decreased level of external PI3P, suggesting a contribution of these kinases to this pool of PI3P. After injection in mouse, or incubation ex vivo in human blood, PI3P-binding proteins decorated the platelet surface as well as α-granules. Upon activation, these platelets were able to secrete the PI3P-binding proteins. These data sheds light on a previously unknown external pool of PI3P in the platelet plasma membrane that recognizes PI3P-binding proteins, leading to their uptake towards α-granules. This study raises questions about the potential function of this external PI3P in the communication of platelets with the extracellular environment, and its possible role in eliminating proteins from the plasma.


Subject(s)
Blood Platelets , Carrier Proteins , Mice , Humans , Animals , Blood Platelets/metabolism , Cell Membrane/metabolism , Carrier Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism
5.
EMBO Rep ; 22(6): e51299, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33880878

ABSTRACT

Endothelium protection is critical, because of the impact of vascular leakage and edema on pathological conditions such as brain ischemia. Whereas deficiency of class II phosphoinositide 3-kinase alpha (PI3KC2α) results in an increase in vascular permeability, we uncover a crucial role of the beta isoform (PI3KC2ß) in the loss of endothelial barrier integrity following injury. Here, we studied the role of PI3KC2ß in endothelial permeability and endosomal trafficking in vitro and in vivo in ischemic stroke. Mice with inactive PI3KC2ß showed protection against vascular permeability, edema, cerebral infarction, and deleterious inflammatory response. Loss of PI3KC2ß in human cerebral microvascular endothelial cells stabilized homotypic cell-cell junctions by increasing Rab11-dependent VE-cadherin recycling. These results identify PI3KC2ß as a potential new therapeutic target to prevent aggravating lesions following ischemic stroke.


Subject(s)
Endothelial Cells , Phosphatidylinositol 3-Kinases , Adherens Junctions/metabolism , Animals , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
6.
Methods Mol Biol ; 2251: 39-53, 2021.
Article in English | MEDLINE | ID: mdl-33481230

ABSTRACT

Our knowledge of the role and biology of the different phosphoinositides has greatly expanded over recent years. Reversible phosphorylation by specific kinases and phosphatases of positions 3, 4, and 5 on the inositol ring is a highly dynamic process playing a critical role in the regulation of the spatiotemporal recruitment and binding of effector proteins. The specific phosphoinositide kinases and phosphatases are key players in the control of many cellular functions, including proliferation, survival, intracellular trafficking, or cytoskeleton reorganization. Several of these enzymes are mutated in human diseases. The impact of the fatty acid composition of phosphoinositides in their function is much less understood. There is an important molecular diversity in the fatty acid side chains of PI. While stearic and arachidonic fatty acids are the major acyl species in PIP, PIP2, and PIP3, other fatty acid combinations are also found. The role of these different molecular species is still unknown, but it is important to quantify these different molecules and their potential changes during cell stimulation to better characterize this emerging field. Here, we describe a sensitive high-performance liquid chromatography-mass spectrometry method that we used for the first time to profile the changes in phosphoinositide molecular species (summed fatty acyl chain profiles) in human and mouse platelets under resting conditions and following stimulation. This method can be applied to other hematopoietic primary cells isolated from human or experimental animal models.


Subject(s)
Blood Platelets/metabolism , Phosphatidylinositols/analysis , Tandem Mass Spectrometry/methods , 1-Phosphatidylinositol 4-Kinase/metabolism , Animals , Biochemical Phenomena , Cell Line , Cells, Cultured , Chromatography, Liquid/methods , Fatty Acids/metabolism , Inositol/chemistry , Mice , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/analysis , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction/physiology
7.
Methods Mol Biol ; 2251: 177-184, 2021.
Article in English | MEDLINE | ID: mdl-33481239

ABSTRACT

Following their generation by lipid kinases and phosphatases, phosphoinositides regulate important biological processes such as cytoskeleton rearrangement, membrane remodeling/trafficking, and gene expression through the interaction of their phosphorylated inositol head group with a variety of protein domains such as PH, PX, and FYVE. Therefore, it is important to determine the specificity of phosphoinositides toward effector proteins to understand their impact on cellular physiology. Several methods have been developed to identify and characterize phosphoinositide effectors, and liposomes-based methods are preferred because the phosphoinositides are incorporated in a membrane, the composition of which can mimic cellular membranes. In this report, we describe the experimental setup for liposome flotation assay and a recently developed method called protein-lipid interaction by fluorescence (PLIF) for the characterization of phosphoinositide-binding specificities of proteins.


Subject(s)
Liposomes/analysis , Phosphatidylinositols/analysis , Protein Interaction Mapping/methods , Cell Membrane/metabolism , Humans , Liposomes/metabolism , Phosphatidylinositols/metabolism , Phosphorylation , Protein Binding/physiology , Protein Domains/physiology , Proteins/chemistry , Signal Transduction/physiology
8.
Biochem J ; 477(22): 4327-4342, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33242335

ABSTRACT

Our knowledge on the expression, regulation and roles of the different phosphoinositide 3-kinases (PI3Ks) in platelet signaling and functions has greatly expanded these last twenty years. Much progress has been made in understanding the roles and regulations of class I PI3Ks which produce the lipid second messenger phosphatidylinositol 3,4,5 trisphosphate (PtdIns(3,4,5)P3). Selective pharmacological inhibitors and genetic approaches have allowed researchers to generate an impressive amount of data on the role of class I PI3Kα, ß, δ and γ in platelet activation and in thrombosis. Furthermore, platelets do also express two class II PI3Ks (PI3KC2α and PI3KC2ß), thought to generate PtdIns(3,4)P2 and PtdIns3P, and the sole class III PI3K (Vps34), known to synthesize PtdIns3P. Recent studies have started to reveal the importance of PI3KC2α and Vps34 in megakaryocytes and platelets, opening new perspective in our comprehension of platelet biology and thrombosis. In this review, we will summarize previous and recent advances on platelet PI3Ks isoforms. The implication of these kinases and their lipid products in fundamental platelet biological processes and thrombosis will be discussed. Finally, the relevance of developing potential antithrombotic strategies by targeting PI3Ks will be examined.


Subject(s)
Blood Platelets/enzymology , Class II Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Thrombosis/enzymology , Thrombosis/therapy , Animals , Blood Platelets/pathology , Humans , Isoenzymes/metabolism , Phosphatidylinositol Phosphates/metabolism , Thrombosis/pathology
9.
Trends Mol Med ; 25(9): 760-774, 2019 09.
Article in English | MEDLINE | ID: mdl-31235369

ABSTRACT

For long-lived contractile cells, such as striated muscle cells, maintaining proteome integrity is a challenging task. These cells require hundreds of components that must be properly synthesized, folded, and incorporated into the basic contractile unit, the sarcomere. Muscle protein quality control in cells is mainly guaranteed by the ubiquitin-proteasome system (UPS), the lysosome-autophagy system, and various molecular chaperones. Recent studies establish the concept of dedicated UPS in the regulation of sarcomere assembly during development and in adult life to maintain the intricate and interwoven organization of protein complexes in muscle. Failure of sarcomere protein quality control often represents the basis of severe myopathies and cardiomyopathies in human, further highlighting its importance in producing and maintaining the contractile machinery of muscle cells in shape.


Subject(s)
Disease Susceptibility , Homeostasis , Muscle, Striated/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Autophagy , Humans , Molecular Chaperones/metabolism , Muscle, Striated/pathology , Protein Binding , Sarcomeres , Signal Transduction , Ubiquitination
10.
Nat Cell Biol ; 20(2): 198-210, 2018 02.
Article in English | MEDLINE | ID: mdl-29358706

ABSTRACT

The ubiquitin proteasome system and autophagy are major protein turnover mechanisms in muscle cells, which ensure stemness and muscle fibre maintenance. Muscle cells contain a high proportion of cytoskeletal proteins, which are prone to misfolding and aggregation; pathological processes that are observed in several neuromuscular diseases called proteinopathies. Despite advances in deciphering the mechanisms underlying misfolding and aggregation, little is known about how muscle cells manage cytoskeletal degradation. Here, we describe a process by which muscle cells degrade the misfolded intermediate filament proteins desmin and vimentin by the proteasome. This relies on the MTM1-UBQLN2 complex to recognize and guide these misfolded proteins to the proteasome and occurs prior to aggregate formation. Thus, our data highlight a safeguarding function of the MTM1-UBQLN2 complex that ensures cytoskeletal integrity to avoid proteotoxic aggregate formation.


Subject(s)
Autophagy/genetics , Cell Cycle Proteins/genetics , Intermediate Filament Proteins/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitins/genetics , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , Cell Cycle Proteins/chemistry , Cytoskeleton/genetics , Desmin/genetics , Humans , Intermediate Filament Proteins/chemistry , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Aggregates/genetics , Protein Folding , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Proteolysis , Ubiquitin/genetics , Ubiquitins/chemistry , Vimentin/genetics
12.
Sci Rep ; 6: 25140, 2016 04 29.
Article in English | MEDLINE | ID: mdl-27126782

ABSTRACT

MMP11 overexpression is a bad prognostic factor in various human carcinomas. Interestingly, this proteinase is not expressed in malignant cells themselves but is secreted by adjacent non-malignant mesenchymal/stromal cells, such as cancer associated fibroblasts (CAFs) and adipocytes (CAAs), which favors cancer cell survival and progression. As MMP11 negatively regulates adipogenesis in vitro, we hypothesized that it may play a role in whole body metabolism and energy homeostasis. We used an in vivo gain- (Mmp11-Tg mice) and loss- (Mmp11-/- mice) of-function approach to address the systemic function of MMP11. Strikingly, MMP11 overexpression protects against type 2 diabetes while Mmp11-/- mice exhibit hallmarks of metabolic syndrome. Moreover, Mmp11-Tg mice were protected from diet-induced obesity and display mitochondrial dysfunction, due to oxidative stress, and metabolic switch from oxidative phosphorylation to aerobic glycolysis. This Warburg-like effect observed in adipose tissues might provide a rationale for the deleterious impact of CAA-secreted MMP11, favouring tumor progression. MMP11 overexpression also leads to increased circulating IGF1 levels and the activation of the IGF1/AKT/FOXO1 cascade, an important metabolic signalling pathway. Our data reveal a major role for MMP11 in controlling energy metabolism, and provide new clues for understanding the relationship between metabolism, cancer progression and patient outcome.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Matrix Metalloproteinase 11/metabolism , Metabolic Syndrome , Animals , Energy Metabolism , Gene Expression , Gene Knockout Techniques , Glycolysis , Mice , Obesity/prevention & control , Oxidative Phosphorylation
13.
Dev Cell ; 35(2): 186-98, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26506308

ABSTRACT

Nucleus positioning is key for intracellular organization, cell differentiation, and organ development and is affected in many diseases, including myopathies due to alteration in amphiphysin-2 (BIN1). The actin and microtubule cytoskeletons are essential for nucleus positioning, but their crosstalk in this process is sparsely characterized. Here, we report that impairment of amphiphysin/BIN1 in Caenorhabditis elegans, mammalian cells, or muscles from patients with centronuclear myopathy alters nuclear position and shape. We show that AMPH-1/BIN1 binds to nesprin and actin, as well as to the microtubule-binding protein CLIP170 in both species. Expression of the microtubule-anchoring CAP-GLY domain of CLIP170 fused to the nuclear-envelope-anchoring KASH domain of nesprin rescues nuclear positioning defects of amph-1 mutants. Amphiphysins thus play a central role in linking the nuclear envelope with the actin and microtubule cytoskeletons. We propose that BIN1 has a direct and evolutionarily conserved role in nuclear positioning, altered in myopathies.


Subject(s)
Cell Nucleus/genetics , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Myopathies, Structural, Congenital/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Envelope/genetics , Nuclear Proteins/genetics , Actins/genetics , Animals , COS Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Shape/genetics , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , HEK293 Cells , Humans , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Microtubules/metabolism , Multiprotein Complexes , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism
14.
Cell Rep ; 11(10): 1564-76, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26051936

ABSTRACT

Desminopathies belong to a family of muscle disorders called myofibrillar myopathies that are caused by Desmin mutations and lead to protein aggregates in muscle fibers. To date, the initial pathological steps of desminopathies and the impact of desmin aggregates in the genesis of the disease are unclear. Using live, high-resolution microscopy, we show that Desmin loss of function and Desmin aggregates promote skeletal muscle defects and alter heart biomechanics. In addition, we show that the calcium dynamics associated with heart contraction are impaired and are associated with sarcoplasmic reticulum dilatation as well as abnormal subcellular distribution of Ryanodine receptors. Our results demonstrate that desminopathies are associated with perturbed excitation-contraction coupling machinery and that aggregates are more detrimental than Desmin loss of function. Additionally, we show that pharmacological inhibition of aggregate formation and Desmin knockdown revert these phenotypes. Our data suggest alternative therapeutic approaches and further our understanding of the molecular determinants modulating Desmin aggregate formation.


Subject(s)
Cardiomyopathies/genetics , Desmin/genetics , Desmin/metabolism , Heart/physiology , Muscle, Skeletal/physiology , Muscular Dystrophies/genetics , Animals , Biomechanical Phenomena , Cardiomyopathies/pathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Humans , Muscular Dystrophies/pathology , Mutation , Zebrafish
15.
Cell Tissue Res ; 360(3): 591-608, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25358400

ABSTRACT

Desmin is a muscle-specific type III intermediate filament essential for proper muscular structure and function. In human, mutations affecting desmin expression or promoting its aggregation lead to skeletal (desmin-related myopathies), or cardiac (desmin-related cardiomyopathy) phenotypes, or both. Patient muscles display intracellular accumulations of misfolded proteins and desmin-positive insoluble granulofilamentous aggregates, leading to a large spectrum of molecular alterations. Increasing evidence shows that desmin function is not limited to the structural and mechanical integrity of cells. This novel perception is strongly supported by the finding that diseases featuring desmin aggregates cannot be easily associated with mechanical defects, but rather involve desmin filaments in a broader spectrum of functions, such as in organelle positioning and integrity and in signaling. Here, we review desmin functions and related diseases affecting striated muscles. We detail emergent cellular functions of desmin based on reported phenotypes in patients and animal models. We discuss known desmin protein partners and propose an overview of the way that this molecular network could serve as a signal transduction platform necessary for proper muscle function.


Subject(s)
Desmin/chemistry , Desmin/metabolism , Muscular Diseases/metabolism , Animals , Desmin/genetics , Disease Models, Animal , Humans , Intermediate Filaments/metabolism , Models, Biological , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Organ Specificity
16.
Nat Commun ; 5: 5647, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25487648

ABSTRACT

Phosphoinositides play a central role in many physiological processes by assisting the recruitment of proteins to membranes through specific phosphoinositide-binding motifs. How this recruitment is coordinated in space and time is not well understood. Here we show that BIN1/M-Amphiphysin2, a protein involved in T-tubule biogenesis in muscle cells and frequently mutated in centronuclear myopathies, clusters PtdIns(4,5)P2 to recruit its downstream partner dynamin. By using several mutants associated with centronuclear myopathies, we find that the N-BAR and the SH3 domains of BIN1 control the kinetics and the accumulation of dynamin on membranes, respectively. We show that phosphoinositide clustering is a mechanism shared by other proteins that interact with PtdIns(4,5)P2, but do not contain a BAR domain. Our numerical simulations point out that clustering is a diffusion-driven process in which phosphoinositide molecules are not sequestered. We propose that this mechanism plays a key role in the recruitment of downstream phosphoinositide-binding proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Dynamins/chemistry , Nuclear Proteins/chemistry , Phosphatidylinositols/chemistry , Tumor Suppressor Proteins/chemistry , Amino Acid Motifs , Cell Membrane/chemistry , Endocytosis , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , HeLa Cells , Humans , Lipid Bilayers/chemistry , Liposomes/chemistry , Molecular Dynamics Simulation , Muscles/metabolism , Protein Binding , Protein Structure, Tertiary
17.
EMBO Mol Med ; 6(11): 1455-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25262827

ABSTRACT

Mutations in amphiphysin-2/BIN1, dynamin 2, and myotubularin are associated with centronuclear myopathy (CNM), a muscle disorder characterized by myofibers with atypical central nuclear positioning and abnormal triads. Mis-splicing of amphiphysin-2/BIN1 is also associated with myotonic dystrophy that shares histopathological hallmarks with CNM. How amphiphysin-2 orchestrates nuclear positioning and triad organization and how CNM-associated mutations lead to muscle dysfunction remains elusive. We find that N-WASP interacts with amphiphysin-2 in myofibers and that this interaction and N-WASP distribution are disrupted by amphiphysin-2 CNM mutations. We establish that N-WASP functions downstream of amphiphysin-2 to drive peripheral nuclear positioning and triad organization during myofiber formation. Peripheral nuclear positioning requires microtubule/Map7/Kif5b-dependent distribution of nuclei along the myofiber and is driven by actin and nesprins. In adult myofibers, N-WASP and amphiphysin-2 are only involved in the maintenance of triad organization but not in the maintenance of peripheral nuclear positioning. Importantly, we confirmed that N-WASP distribution is disrupted in CNM and myotonic dystrophy patients. Our results support a role for N-WASP in amphiphysin-2-dependent nuclear positioning and triad organization and in CNM and myotonic dystrophy pathophysiology.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Muscle, Skeletal/physiopathology , Myopathies, Structural, Congenital/physiopathology , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adult , Humans , Muscle Fibers, Skeletal/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics
18.
EMBO Rep ; 14(10): 907-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23917616

ABSTRACT

Myotubularin (MTM1) and amphiphysin 2 (BIN1) are two proteins mutated in different forms of centronuclear myopathy, but the functional and pathological relationship between these two proteins was unknown. Here, we identified MTM1 as a novel binding partner of BIN1, both in vitro and endogenously in skeletal muscle. Moreover, MTM1 enhances BIN1-mediated membrane tubulation, depending on binding and phosphoinositide phosphatase activity. BIN1 patient mutations induce a conformational change in BIN1 and alter its binding and regulation by MTM1. In conclusion, we identified the first molecular and functional link between MTM1 and BIN1, supporting a common pathological mechanism in different forms of centronuclear myopathy.


Subject(s)
Cell Membrane/metabolism , Myopathies, Structural, Congenital/metabolism , Nerve Tissue Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , COS Cells , Chlorocebus aethiops , Mice , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Nerve Tissue Proteins/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/genetics
19.
PLoS Genet ; 9(6): e1003583, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23818870

ABSTRACT

X-linked myotubular myopathy (XLMTM) is a congenital disorder caused by mutations of the myotubularin gene, MTM1. Myotubularin belongs to a large family of conserved lipid phosphatases that include both catalytically active and inactive myotubularin-related proteins (i.e., "MTMRs"). Biochemically, catalytically inactive MTMRs have been shown to form heteroligomers with active members within the myotubularin family through protein-protein interactions. However, the pathophysiological significance of catalytically inactive MTMRs remains unknown in muscle. By in vitro as well as in vivo studies, we have identified that catalytically inactive myotubularin-related protein 12 (MTMR12) binds to myotubularin in skeletal muscle. Knockdown of the mtmr12 gene in zebrafish resulted in skeletal muscle defects and impaired motor function. Analysis of mtmr12 morphant fish showed pathological changes with central nucleation, disorganized Triads, myofiber hypotrophy and whorled membrane structures similar to those seen in X-linked myotubular myopathy. Biochemical studies showed that deficiency of MTMR12 results in reduced levels of myotubularin protein in zebrafish and mammalian C2C12 cells. Loss of myotubularin also resulted in reduction of MTMR12 protein in C2C12 cells, mice and humans. Moreover, XLMTM mutations within the myotubularin interaction domain disrupted binding to MTMR12 in cell culture. Analysis of human XLMTM patient myotubes showed that mutations that disrupt the interaction between myotubularin and MTMR12 proteins result in reduction of both myotubularin and MTMR12. These studies strongly support the concept that interactions between myotubularin and MTMR12 are required for the stability of their functional protein complex in normal skeletal muscles. This work highlights an important physiological function of catalytically inactive phosphatases in the pathophysiology of myotubular myopathy and suggests a novel therapeutic approach through identification of drugs that could stabilize the myotubularin-MTMR12 complex and hence ameliorate this disorder.


Subject(s)
Myopathies, Structural, Congenital/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proteins/genetics , Zebrafish/genetics , Animals , Catalysis , Cell Line , Humans , Mice , Muscle, Skeletal , Muscles/metabolism , Muscles/physiopathology , Mutation , Myopathies, Structural, Congenital/physiopathology , Protein Stability , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proteins/chemistry , Proteins/metabolism
20.
J Cell Sci ; 126(Pt 8): 1806-19, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444364

ABSTRACT

The sarcoplasmic reticulum (SR) is a specialized form of endoplasmic reticulum (ER) in skeletal muscle and is essential for calcium homeostasis. The mechanisms involved in SR remodeling and maintenance of SR subdomains are elusive. In this study, we identified myotubularin (MTM1), a phosphoinositide phosphatase mutated in X-linked centronuclear myopathy (XLCNM, or myotubular myopathy), as a key regulator of phosphatidylinositol 3-monophosphate (PtdIns3P) levels at the SR. MTM1 is predominantly located at the SR cisternae of the muscle triads, and Mtm1-deficient mouse muscles and myoblasts from XLCNM patients exhibit abnormal SR/ER networks. In vivo modulation of MTM1 enzymatic activity in skeletal muscle using ectopic expression of wild-type or a dead-phosphatase MTM1 protein leads to differential SR remodeling. Active MTM1 is associated with flat membrane stacks, whereas dead-phosphatase MTM1 mutant promotes highly curved cubic membranes originating from the SR and enriched in PtdIns3P. Overexpression of a tandem FYVE domain with high affinity for PtdIns3P alters the shape of the SR cisternae at the triad. Our findings, supported by the parallel analysis of the Mtm1-null mouse and an in vivo study, reveal a direct function of MTM1 enzymatic activity in SR remodeling and a key role for PtdIns3P in promoting SR membrane curvature in skeletal muscle. We propose that alteration in SR remodeling is a primary cause of X-linked centronuclear myopathy. The tight regulation of PtdIns3P on specific membrane subdomains may be a general mechanism to control membrane curvature.


Subject(s)
Muscle, Skeletal/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Blotting, Western , Cell Line , Immunoprecipitation , Male , Mice , Microscopy, Electron, Transmission , Muscle, Skeletal/ultrastructure , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...