Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Commun Biol ; 4(1): 355, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742095

ABSTRACT

Mechanical stress induced by contractions constantly threatens the integrity of muscle Z-disc, a crucial force-bearing structure in striated muscle. The PDZ-LIM proteins have been proposed to function as adaptors in transducing mechanical signals to preserve the Z-disc structure, however the underlying mechanisms remain poorly understood. Here, we show that LDB3, a well-characterized striated muscle PDZ-LIM protein, modulates mechanical stress signaling through interactions with the mechanosensing domain in filamin C, its chaperone HSPA8, and PKCα in the Z-disc of skeletal muscle. Studies of Ldb3Ala165Val/+ mice indicate that the myopathy-associated LDB3 p.Ala165Val mutation triggers early aggregation of filamin C and its chaperones at muscle Z-disc before aggregation of the mutant protein. The mutation causes protein aggregation and eventually Z-disc myofibrillar disruption by impairing PKCα and TSC2-mTOR, two important signaling pathways regulating protein stability and disposal of damaged cytoskeletal components at a major mechanosensor hub in the Z-disc of skeletal muscle.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , LIM Domain Proteins/genetics , Mechanotransduction, Cellular , Muscle, Skeletal/enzymology , Myopathies, Structural, Congenital/enzymology , Point Mutation , Protein Kinase C-alpha/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Disease Models, Animal , Down-Regulation , Filamins/metabolism , HSC70 Heat-Shock Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/physiopathology , Protein Aggregates , Protein Aggregation, Pathological , Protein Kinase C-alpha/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
3.
EMBO Mol Med ; 10(2): 239-253, 2018 02.
Article in English | MEDLINE | ID: mdl-29246969

ABSTRACT

Rapid advances in allele-specific silencing by RNA interference established a strategy of choice to cure dominant inherited diseases by targeting mutant alleles. We used this strategy for autosomal-dominant centronuclear myopathy (CNM), a rare neuromuscular disorder without available treatment due to heterozygous mutations in the DNM2 gene encoding Dynamin 2. Allele-specific siRNA sequences were developed in order to specifically knock down the human and murine DNM2-mRNA harbouring the p.R465W mutation without affecting the wild-type allele. Functional restoration was achieved in muscle from a knock-in mouse model and in patient-derived fibroblasts, both expressing the most frequently encountered mutation in patients. Restoring either muscle force in a CNM mouse model or DNM2 function in patient-derived cells is an essential breakthrough towards future gene-based therapy for dominant centronuclear myopathy.


Subject(s)
Dynamin II/genetics , Genetic Therapy , Myopathies, Structural, Congenital , RNA, Small Interfering/therapeutic use , Alleles , Animals , Cells, Cultured , Humans , Mice , Mutation , Myopathies, Structural, Congenital/drug therapy , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/physiopathology
4.
Hum Mol Genet ; 26(19): 3736-3748, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28934386

ABSTRACT

Myotubularins (MTMs) are active or dead phosphoinositides phosphatases defining a large protein family conserved through evolution and implicated in different neuromuscular diseases. Loss-of-function mutations in MTM1 cause the severe congenital myopathy called myotubular myopathy (or X-linked centronuclear myopathy) while mutations in the MTM1-related protein MTMR2 cause a recessive Charcot-Marie-Tooth peripheral neuropathy. Here we aimed to determine the functional specificity and redundancy of MTM1 and MTMR2, and to assess their abilities to compensate for a potential therapeutic strategy. Using molecular investigations and heterologous expression of human MTMs in yeast cells and in Mtm1 knockout mice, we characterized several naturally occurring MTMR2 isoforms with different activities. We identified the N-terminal domain as responsible for functional differences between MTM1 and MTMR2. An N-terminal extension observed in MTMR2 is absent in MTM1, and only the short MTMR2 isoform lacking this N-terminal extension behaved similarly to MTM1 in yeast and mice. Moreover, adeno-associated virus-mediated exogenous expression of several MTMR2 isoforms ameliorates the myopathic phenotype owing to MTM1 loss, with increased muscle force, reduced myofiber atrophy, and reduction of the intracellular disorganization hallmarks associated with myotubular myopathy. Noteworthy, the short MTMR2 isoform provided a better rescue when compared with the long MTMR2 isoform. In conclusion, these results point to the molecular basis for MTMs functional specificity. They also provide the proof-of-concept that expression of the neuropathy-associated MTMR2 gene improves the MTM1-associated myopathy, thus identifying MTMR2 as a novel therapeutic target for myotubular myopathy.


Subject(s)
Myopathies, Structural, Congenital/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Humans , Male , Mice , Mice, Knockout , Mutation , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/metabolism , Phenotype , Protein Domains , Protein Isoforms , Protein Tyrosine Phosphatases, Non-Receptor/genetics
5.
J Neurol ; 264(8): 1791-1803, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28712002

ABSTRACT

Mutations in GFPT1 (glutamine-fructose-6-phosphate transaminase 1), a gene encoding an enzyme involved in glycosylation of ubiquitous proteins, cause a limb-girdle congenital myasthenic syndrome (LG-CMS) with tubular aggregates (TAs) characterized predominantly by affection of the proximal skeletal muscles and presence of highly organized and remodeled sarcoplasmic tubules in patients' muscle biopsies. We report here the first long-term clinical follow-up of 11 French individuals suffering from LG-CMS with TAs due to GFPT1 mutations, of which nine are new. Our retrospective clinical evaluation stresses an evolution toward a myopathic weakness that occurs concomitantly to ineffectiveness of usual CMS treatments. Analysis of neuromuscular biopsies from three unrelated individuals demonstrates that the maintenance of neuromuscular junctions (NMJs) is dramatically impaired with loss of post-synaptic junctional folds and evidence of denervation-reinnervation processes affecting the three main NMJ components. Moreover, molecular analyses of the human muscle biopsies confirm glycosylation defects of proteins with reduced O-glycosylation and show reduced sialylation of transmembrane proteins in extra-junctional area. Altogether, these results pave the way for understanding the etiology of this rare neuromuscular disorder that may be considered as a "tubular aggregates myopathy with synaptopathy".


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Neuromuscular Junction/pathology , Adolescent , Adult , Aged , Female , Follow-Up Studies , Glycosylation , Humans , Middle Aged , Muscle, Skeletal/enzymology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/enzymology , Myopathies, Structural, Congenital/drug therapy , Myopathies, Structural, Congenital/enzymology , Neuromuscular Junction/enzymology , Prospective Studies , Retrospective Studies , Young Adult
6.
Nature ; 529(7586): 408-12, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26760201

ABSTRACT

Phosphoinositides are a minor class of short-lived membrane phospholipids that serve crucial functions in cell physiology ranging from cell signalling and motility to their role as signposts of compartmental membrane identity. Phosphoinositide 4-phosphates such as phosphatidylinositol 4-phosphate (PI(4)P) and phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) are concentrated at the plasma membrane, on secretory organelles, and on lysosomes, whereas phosphoinositide 3-phosphates, most notably phosphatidylinositol 3-phosphate (PI(3)P), are a hallmark of the endosomal system. Directional membrane traffic between endosomal and secretory compartments, although inherently complex, therefore requires regulated phosphoinositide conversion. The molecular mechanism underlying this conversion of phosphoinositide identity during cargo exit from endosomes by exocytosis is unknown. Here we report that surface delivery of endosomal cargo requires hydrolysis of PI(3)P by the phosphatidylinositol 3-phosphatase MTM1, an enzyme whose loss of function leads to X-linked centronuclear myopathy (also called myotubular myopathy) in humans. Removal of endosomal PI(3)P by MTM1 is accompanied by phosphatidylinositol 4-kinase-2α (PI4K2α)-dependent generation of PI(4)P and recruitment of the exocyst tethering complex to enable membrane fusion. Our data establish a mechanism for phosphoinositide conversion from PI(3)P to PI(4)P at endosomes en route to the plasma membrane and suggest that defective phosphoinositide conversion at endosomes underlies X-linked centronuclear myopathy caused by mutation of MTM1 in humans.


Subject(s)
Endosomes/metabolism , Exocytosis , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols/metabolism , 1-Phosphatidylinositol 4-Kinase/metabolism , Biological Transport , Cell Line , Cell Membrane/metabolism , HeLa Cells , Humans , Hydrolysis , Membrane Fusion , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
7.
Biochim Biophys Acta ; 1851(6): 867-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25510381

ABSTRACT

More than twenty different genetic diseases have been described that are caused by mutations in phosphoinositide metabolizing enzymes, mostly in phosphoinositide phosphatases. Although generally ubiquitously expressed, mutations in these enzymes, which are mainly loss-of-function, result in tissue-restricted clinical manifestations through mechanisms that are not completely understood. Here we analyze selected disorders of phosphoinositide metabolism grouped according to the principle tissue affected: the nervous system, muscle, kidney, the osteoskeletal system, the eye, and the immune system. We will highlight what has been learnt so far from the study of these disorders about not only the cellular and molecular pathways that are involved or are governed by phosphoinositides, but also the many gaps that remain to be filled to gain a full understanding of the pathophysiological mechanisms underlying the clinical manifestations of this steadily growing class of diseases, most of which still remain orphan in terms of treatment. This article is part of a Special Issue entitled Phosphoinositides.


Subject(s)
Bone Diseases, Developmental/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Limb Deformities, Congenital/genetics , Mutation , Myopathies, Structural, Congenital/genetics , Phosphatidylinositols/metabolism , Animals , Bone Diseases, Developmental/enzymology , Bone Diseases, Developmental/pathology , Disease Models, Animal , Gene Expression , Hereditary Sensory and Motor Neuropathy/enzymology , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Limb Deformities, Congenital/enzymology , Limb Deformities, Congenital/pathology , Mice , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism
8.
Hum Mol Genet ; 22(8): 1525-38, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23307925

ABSTRACT

No effective treatment exists for patients with X-linked myotubular myopathy (XLMTM), a fatal congenital muscle disease caused by deficiency of the lipid phosphatase, myotubularin. The Mtm1δ4 and Mtm1 p.R69C mice model severely and moderately symptomatic XLMTM, respectively, due to differences in the degree of myotubularin deficiency. Contractile function of intact extensor digitorum longus (EDL) and soleus muscles from Mtm1δ4 mice, which produce no myotubularin, is markedly impaired. Contractile forces generated by chemically skinned single fiber preparations from Mtm1δ4 muscle were largely preserved, indicating that weakness was largely due to impaired excitation contraction coupling. Mtm1 p.R69C mice, which produce small amounts of myotubularin, showed impaired contractile function only in EDL muscles. Short-term replacement of myotubularin with a prototypical targeted protein replacement agent (3E10Fv-MTM1) in Mtm1δ4 mice improved contractile function and muscle pathology. These promising findings suggest that even low levels of myotubularin protein replacement can improve the muscle weakness and reverse the pathology that characterizes XLMTM.


Subject(s)
Enzyme Replacement Therapy , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/therapy , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Animals , Disease Models, Animal , Fatigue/metabolism , Fatigue/physiopathology , Female , Humans , Mice , Muscle Weakness/genetics , Muscle Weakness/therapy , Muscle, Skeletal/physiopathology , Muscles/enzymology , Muscles/metabolism , Muscles/pathology , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/genetics , Protein Tyrosine Phosphatases, Non-Receptor/biosynthesis , Protein Tyrosine Phosphatases, Non-Receptor/deficiency
9.
Subcell Biochem ; 58: 281-336, 2012.
Article in English | MEDLINE | ID: mdl-22403079

ABSTRACT

Two classes of lipid phosphatases selectively dephosphorylate the 3 position of the inositol ring of phosphoinositide signaling molecules: the PTEN and the Myotubularin families. PTEN dephosphorylates PtdIns(3,4,5)P(3), acting in direct opposition to the Class I PI3K enzymes in the regulation of cell growth, proliferation and polarity and is an important tumor suppressor. Although there are several PTEN-related proteins encoded by the human genome, none of these appear to fulfill the same functions. In contrast, the Myotubularins dephosphorylate both PtdIns(3)P and PtdIns(3,5)P(2), making them antagonists of the Class II and Class III PI 3-kinases and regulators of membrane traffic. Both phosphatase groups were originally identified through their causal mutation in human disease. Mutations in specific myotubularins result in myotubular myopathy and Charcot-Marie-Tooth peripheral neuropathy; and loss of PTEN function through mutation and other mechanisms is evident in as many as a third of all human tumors. This chapter will discuss these two classes of phosphatases, covering what is known about their biochemistry, their functions at the cellular and whole body level and their influence on human health.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Myopathies, Structural, Congenital/enzymology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Second Messenger Systems , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Gene Expression Regulation , Humans , Hydrolysis , Mutation , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Substrate Specificity
10.
J Vet Diagn Invest ; 23(1): 124-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21217042

ABSTRACT

Centronuclear myopathy (CNM) is an autosomal recessive hereditary disease affecting Labrador Retriever dogs. The disease is characterized by muscle lesions, typically encompassing reduction in the number and atrophy of type II fibers, and is caused by a short interspersed repeat element insertion in exon 2 of the protein tyrosine phosphatase-like member A. The actual allele frequency is unknown; a study was undertaken to ascertain it using a convenience-sample population composed of 217 Labrador Retrievers. In addition to 3 subjects already diagnosed with CNM, used as positive controls for polymerase chain reaction, only 2 unrelated dogs were heterozygous wild-type/mutation (wild-type/mut). Thus, the frequency of the CNM allele observed in the present study was 1.8% and 0.47% when including and excluding the 3 mut/mut homozygous cases, respectively. Based on the Hardy-Weinberg exact test (P  =  1.00), the genotype frequency without the CNM-affected dogs was in agreement with the Hardy-Weinberg equilibrium. Assuming the Hardy-Weinberg equilibrium law, the expected frequency of the homozygous mutated genotype was calculated to be approximately 0.00005, which corresponds to 1 case of CNM out of 20,000 dogs. In conclusion, the present study indicates that the CNM allele is present but rare in a convenience sample of Labrador Retrievers in Italy.


Subject(s)
Dog Diseases/genetics , Myopathies, Structural, Congenital/veterinary , Protein Tyrosine Phosphatases/genetics , Alleles , Animals , Chi-Square Distribution , DNA/chemistry , DNA/genetics , Dog Diseases/enzymology , Dog Diseases/epidemiology , Dog Diseases/pathology , Dogs , Female , Italy/epidemiology , Male , Muscle Fibers, Fast-Twitch/pathology , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/epidemiology , Myopathies, Structural, Congenital/genetics , Polymerase Chain Reaction/veterinary , Retrospective Studies
11.
Int J Biochem Cell Biol ; 42(5): 555-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20025992

ABSTRACT

Lipolysis in adipocytes, the hydrolysis of triacylglycerol (TAG) to release fatty acids (FAs) and glycerol for use by other organs, is a unique function of white adipose tissue. Lipolysis in adipocytes occurs at the surface of cytosolic lipid droplets, which have recently gained much attention as dynamic organelles integral to lipid metabolism. Desnutrin/ATGL is now established as a bona fide TAG hydrolase and mutations in human desnutrin/ATGL/PNPLA2, as well as in its activator, comparative gene identification 58, are associated with Neutral Lipid Storage Disease. Furthermore, recent identification of AdPLA as the major adipose phospholipase A(2), has led to the discovery of a dominant autocrine/paracrine regulation of lipolysis through PGE(2). Here, we review emerging concepts in the key players in lipolysis and the regulation of this process. We also examine recent findings in mouse models and humans with alterations/mutations in genes involved in lipolysis and discuss activation of lipolysis in adipocytes as a potential therapeutic target.


Subject(s)
Adipocytes, White/metabolism , Lipolysis/physiology , Adipocytes, White/drug effects , Adipocytes, White/enzymology , Animals , Humans , Lipidoses/enzymology , Lipidoses/genetics , Lipolysis/drug effects , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/genetics , Obesity/enzymology , Obesity/physiopathology , Obesity/prevention & control , Organelles/drug effects , Organelles/enzymology , Organelles/metabolism
12.
Curr Opin Struct Biol ; 15(6): 614-20, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16289848

ABSTRACT

The human neuromuscular diseases X-linked myotubular myopathy and Charcot-Marie-Tooth disease type 4B are caused by mutations in myotubularin family proteins. The myotubularins are a unique subfamily of protein tyrosine phosphatases that utilize inositol phospholipids, rather than phosphoproteins, as substrates. Recent structural studies, including the first crystal structure of a myotubularin family protein, have defined the structural features that are characteristic of the family and revealed the molecular basis of their unique substrate specificity. Interestingly, the myotubularin family contains a subgroup of proteins that are catalytically inactive. Recent biochemical studies have established that the inactive myotubularins function as adaptors for the active members and play an important regulatory role within the family.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Models, Molecular , Myopathies, Structural, Congenital/enzymology , Protein Tyrosine Phosphatases/metabolism , Charcot-Marie-Tooth Disease/genetics , Humans , Mutation , Myopathies, Structural, Congenital/genetics , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor
13.
Genes Dev ; 19(17): 2066-77, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140986

ABSTRACT

Myocyte enhancer factor 2 (MEF2) plays essential roles in transcriptional control of muscle development. However, signaling pathways acting downstream of MEF2 are largely unknown. Here, we performed a microarray analysis using Mef2c-null mouse embryos and identified a novel MEF2-regulated gene encoding a muscle-specific protein kinase, Srpk3, belonging to the serine arginine protein kinase (SRPK) family, which phosphorylates serine/arginine repeat-containing proteins. The Srpk3 gene is specifically expressed in the heart and skeletal muscle from embryogenesis to adulthood and is controlled by a muscle-specific enhancer directly regulated by MEF2. Srpk3-null mice display a new entity of type 2 fiber-specific myopathy with a marked increase in centrally placed nuclei; while transgenic mice overexpressing Srpk3 in skeletal muscle show severe myofiber degeneration and early lethality. We conclude that normal muscle growth and homeostasis require MEF2-dependent signaling by Srpk3.


Subject(s)
DNA-Binding Proteins/metabolism , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/genetics , Protein Serine-Threonine Kinases/deficiency , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , MEF2 Transcription Factors , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Muscle, Skeletal/embryology , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Myogenic Regulatory Factors , Myopathies, Structural, Congenital/etiology , Myopathies, Structural, Congenital/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
14.
Cell Mol Life Sci ; 60(10): 2084-99, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14618257

ABSTRACT

Phosphoinositides play a central role in the control of major eukaryotic cell signaling mechanisms. Accordingly, the list of phosphoinositide-metabolizing enzymes implicated in human diseases has considerably increased these last years. Here we will focus on myotubularin, the protein mutated in the X-linked myotubular myopathy (XLMTM) and the founding member of a family of 13 related proteins. Recent data demonstrate that myotubularin and several other members of the family are potent lipid phosphatases showing a marked specificity for phosphatidylinositol 3-phosphate [PtdIns(3)P]. This finding has raised considerable interest as PtdIns(3)P is implicated in vesicular trafficking and sorting through its binding to specific protein domains. The structure of myotubularin, the molecular mechanisms of its function and its implication in the etiology of XLMTM will be discussed, as well as the potential function and role of the other members of the family.


Subject(s)
Myopathies, Structural, Congenital/enzymology , Phosphatidylinositols/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Cytoplasmic Vesicles/metabolism , Disease Models, Animal , Mice , Mice, Knockout , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/physiopathology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor
15.
J Cell Sci ; 115(Pt 15): 3105-17, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12118066

ABSTRACT

Myotubularin, the phosphatase mutated in X-linked myotubular myopathy, was shown to dephosphorylate phosphatidylinositol 3-monophosphate (PtdIns3P) and was also reported to interact with nuclear transcriptional regulators from the trithorax family. We have characterized a panel of specific antibodies and investigated the subcellular localization of myotubularin. Myotubularin is not detected in the nucleus, and localizes mostly as a dense cytoplasmic network. Overexpression of myotubularin does not detectably affect vesicle trafficking in the mammalian cells investigated, in contrast to previous observations in yeast models. Both mutation of a key aspartate residue of myotubularin and dominant activation of Rac1 GTPase lead to the recruitment of myotubularin to specific plasma membrane domains. Localization to Rac1-induced ruffles is dependent on the presence of a domain highly conserved in the myotubularin family (that we named RID). We thus propose that myotubularin may dephosphorylate a subpool of PtdIns3P (or another related substrate) at the plasma membrane.


Subject(s)
Cell Surface Extensions/enzymology , Cytoplasm/enzymology , Eukaryotic Cells/enzymology , Myopathies, Structural, Congenital/enzymology , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases/deficiency , rac1 GTP-Binding Protein/metabolism , Animals , Antibodies , Cell Compartmentation/genetics , Cell Surface Extensions/ultrastructure , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Eukaryotic Cells/ultrastructure , Fluorescent Antibody Technique , HeLa Cells , Humans , Mice , Microscopy, Confocal , Mutation/genetics , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/physiopathology , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL