Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mov Disord ; 37(2): 365-374, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34820905

RESUMEN

BACKGROUND: The dystonias are a heterogeneous group of hyperkinetic disorders characterized by sustained or intermittent muscle contractions that cause abnormal movements and/or postures. Although more than 200 causal genes are known, many cases of primary dystonia have no clear genetic cause. OBJECTIVES: To identify the causal gene in a consanguineous family with three siblings affected by a complex persistent generalized dystonia, generalized epilepsy, and mild intellectual disability. METHODS: We performed exome sequencing in the parents and two affected siblings and characterized the expression of the identified gene by immunohistochemistry in control human and zebrafish brains. RESULTS: We identified a novel missense variant (c.142G>A (NM_032192); p.Glu48Lys) in the protein phosphatase 1 regulatory inhibitor subunit 1B gene (PPP1R1B) that was homozygous in all three siblings and heterozygous in the parents. This gene is also known as dopamine and cAMP-regulated neuronal phosphoprotein 32 (DARPP-32) and has been involved in the pathophysiology of abnormal movements. The uncovered variant is absent in public databases and modifies the conserved glutamate 48 localized close to the serine 45 phosphorylation site. The PPP1R1B protein was shown to be expressed in cells and regions involved in movement control, including projection neurons of the caudate-putamen, substantia nigra neuropil, and cerebellar Purkinje cells. The latter cells were also confirmed to be positive for PPP1R1B expression in the zebrafish brain. CONCLUSIONS: We report the association of a PPP1R1B/DARPP-32 variant with generalized dystonia in man. It might be relevant to include the sequencing of this new gene in the diagnosis of patients with otherwise unexplained movement disorders. © 2021 International Parkinson and Movement Disorder Society.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Distonía , Trastornos Distónicos , Animales , Trastornos Distónicos/genética , Homocigoto , Humanos , Pez Cebra
2.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32766723

RESUMEN

The Nck-associated protein 1-like (NCKAP1L) gene, alternatively called hematopoietic protein 1 (HEM-1), encodes a hematopoietic lineage-specific regulator of the actin cytoskeleton. Nckap1l-deficient mice have anomalies in lymphocyte development, phagocytosis, and neutrophil migration. Here we report, for the first time, NCKAP1L deficiency cases in humans. In two unrelated patients of Middle Eastern origin, recessive mutations in NCKAP1L abolishing protein expression led to immunodeficiency, lymphoproliferation, and hyperinflammation with features of hemophagocytic lymphohistiocytosis. Immunophenotyping showed an inverted CD4/CD8 ratio with a major shift of both CD4+ and CD8+ cells toward memory compartments, in line with combined RNA-seq/proteomics analyses revealing a T cell exhaustion signature. Consistent with the core function of NCKAP1L in the reorganization of the actin cytoskeleton, patients' T cells displayed impaired early activation, immune synapse morphology, and leading edge formation. Moreover, knockdown of nckap1l in zebrafish led to defects in neutrophil migration. Hence, NCKAP1L mutations lead to broad immune dysregulation in humans, which could be classified within actinopathies.


Asunto(s)
Síndromes de Inmunodeficiencia/complicaciones , Inflamación/complicaciones , Trastornos Linfoproliferativos/complicaciones , Proteínas de la Membrana/metabolismo , Actinas/metabolismo , Animales , Degranulación de la Célula , Proliferación Celular , Niño , Citotoxicidad Inmunológica , Familia , Femenino , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/inmunología , Sinapsis Inmunológicas/metabolismo , Lactante , Inflamación/inmunología , Inflamación/patología , Activación de Linfocitos/inmunología , Trastornos Linfoproliferativos/inmunología , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Mutación/genética , Linaje , Fenotipo , Síndrome , Pez Cebra
3.
Cells ; 9(3)2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32164332

RESUMEN

The budding yeast Saccharomyces cerevisiae has an actin cytoskeleton that comprises a set of protein components analogous to those found in the actin cytoskeletons of higher eukaryotes. Furthermore, the actin cytoskeletons of S. cerevisiae and of higher eukaryotes have some similar physiological roles. The genetic tractability of budding yeast and the availability of a stable haploid cell type facilitates the application of molecular genetic approaches to assign functions to the various actin cytoskeleton components. This has provided information that is in general complementary to that provided by studies of the equivalent proteins of higher eukaryotes and hence has enabled a more complete view of the role of these proteins. Several human functional homologues of yeast actin effectors are implicated in diseases. A better understanding of the molecular mechanisms underpinning the functions of these proteins is critical to develop improved therapeutic strategies. In this article we chose as examples four evolutionarily conserved proteins that associate with the actin cytoskeleton: 1) yeast Hof1p/mammalian PSTPIP1, 2) yeast Rvs167p/mammalian BIN1, 3) yeast eEF1A/eEF1A1 and eEF1A2 and 4) yeast Yih1p/mammalian IMPACT. We compare the knowledge on the functions of these actin cytoskeleton-associated proteins that has arisen from studies of their homologues in yeast with information that has been obtained from in vivo studies using live animals or in vitro studies using cultured animal cell lines.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Levaduras/metabolismo , Humanos
4.
Front Genet ; 10: 504, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191616

RESUMEN

Amelogenesis imperfecta (AI) is a heterogeneous group of rare inherited diseases presenting with enamel defects. More than 30 genes have been reported to be involved in syndromic or non-syndromic AI and new genes are continuously discovered (Smith et al., 2017). Whole-exome sequencing was performed in a consanguineous family. The affected daughter presented with intra-uterine and postnatal growth retardation, skeletal dysplasia, macrocephaly, blue sclerae, and hypoplastic AI. We identified a homozygous missense mutation in exon 11 of SLC10A7 (NM_001300842.2: c.908C>T; p.Pro303Leu) segregating with the disease phenotype. We found that Slc10a7 transcripts were expressed in the epithelium of the developing mouse tooth, bones undergoing ossification, and in vertebrae. Our results revealed that SLC10A7 is overexpressed in patient fibroblasts. Patient cells display altered intracellular calcium localization suggesting that SLC10A7 regulates calcium trafficking. Mutations in this gene were previously reported to cause a similar syndromic phenotype, but with more severe skeletal defects (Ashikov et al., 2018;Dubail et al., 2018). Therefore, phenotypes resulting from a mutation in SLC10A7 can vary in severity. However, AI is the key feature indicative of SLC10A7 mutations in patients with skeletal dysplasia. Identifying this important phenotype will improve clinical diagnosis and patient management.

5.
J Neurol ; 266(2): 353-360, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30515627

RESUMEN

OBJECTIVE: To describe adult-onset limb-girdle-type muscular dystrophy caused by biallelic variants in the PYROXD1 gene, which has been recently linked to early-onset congenital myofibrillar myopathy. METHODS: Whole exome sequencing was performed for adult-onset neuromuscular disease patients with no molecular diagnosis. Patients with PYROXD1 variants underwent clinical characterization, lower limb muscle MRI, muscle biopsy and spirometry. A yeast complementation assay was used to determine the biochemical consequences of the genetic variants. RESULTS: We identified four patients with biallelic PYROXD1 variants. Three patients, who had symptom onset in their 20s or 30s, were homozygous for the previously described p.Asn155Ser. The fourth patient, with symptom onset at age 49, was compound heterozygous for p.Asn155Ser variant and previously unknown p.Tyr354Cys. All patients presented with a LGMD-type phenotype of symmetric muscle weakness and wasting. Symptoms started in proximal muscles of the lower limbs, and progressed slowly to involve also upper limbs in a proximal-predominant fashion. All patients remained ambulant past the age of 60. They had restrictive lung disease but no cardiac impairment. Muscle MRI showed strong involvement of anterolateral thigh muscles. Muscle biopsy displayed chronic myopathic changes. Yeast complementation assay demonstrated the p.Tyr354Cys mutation to impair PYROXD1 oxidoreductase ability. CONCLUSION: PYROXD1 variants can cause an adult-onset slowly progressive LGMD-type phenotype.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/fisiopatología , Oxidorreductasas/genética , Anciano , Femenino , Finlandia , Genes Recesivos , Humanos , Masculino , Distrofia Muscular de Cinturas/patología , Linaje , Secuenciación del Exoma
6.
J Clin Invest ; 127(12): 4477-4487, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29130937

RESUMEN

Regulation of skeletal muscle development and organization is a complex process that is not fully understood. Here, we focused on amphiphysin 2 (BIN1, also known as bridging integrator-1) and dynamin 2 (DNM2), two ubiquitous proteins implicated in membrane remodeling and mutated in centronuclear myopathies (CNMs). We generated Bin1-/- Dnm2+/- mice to decipher the physiological interplay between BIN1 and DNM2. While Bin1-/- mice die perinatally from a skeletal muscle defect, Bin1-/- Dnm2+/- mice survived at least 18 months, and had normal muscle force and intracellular organization of muscle fibers, supporting BIN1 as a negative regulator of DNM2. We next characterized muscle-specific isoforms of BIN1 and DNM2. While BIN1 colocalized with and partially inhibited DNM2 activity during muscle maturation, BIN1 had no effect on the isoform of DNM2 found in adult muscle. Together, these results indicate that BIN1 and DNM2 regulate muscle development and organization, function through a common pathway, and define BIN1 as a negative regulator of DNM2 in vitro and in vivo during muscle maturation. Our data suggest that DNM2 modulation has potential as a therapeutic approach for patients with CNM and BIN1 defects. As BIN1 is implicated in cancers, arrhythmia, and late-onset Alzheimer disease, these findings may trigger research directions and therapeutic development for these common diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Dinamina II/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Dinamina II/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/patología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Proteínas del Tejido Nervioso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Supresoras de Tumor/genética
7.
Hum Mol Genet ; 26(19): 3736-3748, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28934386

RESUMEN

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.


Asunto(s)
Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Miopatías Estructurales Congénitas/enzimología , Miopatías Estructurales Congénitas/metabolismo , Fenotipo , Dominios Proteicos , Isoformas de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética
8.
Int J Mol Sci ; 18(3)2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294977

RESUMEN

Phosphoinositides are lipids involved in the vesicular transport of proteins and lipids between the different compartments of eukaryotic cells. They act by recruiting and/or activating effector proteins and thus are involved in regulating various cellular functions, such as vesicular budding, membrane fusion and cytoskeleton dynamics. Although detected in small concentrations in membranes, their role is essential to cell function, since imbalance in their concentrations is a hallmark of many cancers. Their synthesis involves phosphorylating/dephosphorylating positions D3, D4 and/or D5 of their inositol ring by specific lipid kinases and phosphatases. This process is tightly regulated and specific to the different intracellular membranes. Most enzymes involved in phosphoinositide synthesis are conserved between yeast and human, and their loss of function leads to severe diseases (cancer, myopathy, neuropathy and ciliopathy).


Asunto(s)
Membrana Celular/metabolismo , Metabolismo de los Lípidos , Fosfatidilinositoles/metabolismo , Transducción de Señal , Actinas/metabolismo , Animales , Autofagia , Transporte Biológico , Endocitosis , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Redes y Vías Metabólicas , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolípidos/metabolismo , Esteroles/metabolismo
9.
Adv Biol Regul ; 63: 49-58, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27666502

RESUMEN

Myotubularins define a large family of proteins conserved through evolution. Several members are mutated in different neuromuscular diseases including centronuclear myopathies and Charcot-Marie-Tooth (CMT) neuropathies, or are linked to a predisposition to obesity and cancer. While some members have phosphatase activity against the 3-phosphate of phosphoinositides, regulating the phosphorylation status of PtdIns3P and PtdIns(3,5)P2 implicated in membrane trafficking and autophagy, and producing PtdIns5P, others lack key residues in the catalytic site and are classified as dead-phosphatases. However, these dead phosphatases regulate phosphoinositide-dependent cellular pathways by binding to catalytically active myotubularins. Here we review previous studies on the molecular regulation and physiological roles of myotubularins. We also used the recent myotubularins three-dimensional structures to underline key residues that are mutated in neuromuscular diseases and required for enzymatic activity. In addition, through database mining and analysis, expression profile and specific isoforms of the different myotubularins are described in depth, as well as a revisited protein interaction network. Comparison of the interactome and expression data for each myotubularin highlights specific protein complexes and tissues where myotubularins should have a key regulatory role.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Miopatías Estructurales Congénitas/genética , Neoplasias/genética , Obesidad/genética , Proteínas Tirosina Fosfatasas no Receptoras/química , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Expresión Génica , Humanos , Modelos Moleculares , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Neoplasias/metabolismo , Neoplasias/patología , Obesidad/metabolismo , Obesidad/patología , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
10.
Nat Commun ; 7: 13586, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27882921

RESUMEN

Ciliopathies are a group of diseases that affect kidney and retina among other organs. Here, we identify a missense mutation in PIK3R4 (phosphoinositide 3-kinase regulatory subunit 4, named VPS15) in a family with a ciliopathy phenotype. Besides being required for trafficking and autophagy, we show that VPS15 regulates primary cilium length in human fibroblasts, as well as ciliary processes in zebrafish. Furthermore, we demonstrate its interaction with the golgin GM130 and its localization to the Golgi. The VPS15-R998Q patient mutation impairs Golgi trafficking functions in humanized yeast cells. Moreover, in VPS15-R998Q patient fibroblasts, the intraflagellar transport protein IFT20 is not localized to vesicles trafficking to the cilium but is restricted to the Golgi. Our findings suggest that at the Golgi, VPS15 and GM130 form a protein complex devoid of VPS34 to ensure the IFT20-dependent sorting and transport of membrane proteins from the cis-Golgi to the primary cilium.


Asunto(s)
Proteínas Portadoras/metabolismo , Cilios/metabolismo , Ciliopatías/genética , Aparato de Golgi/metabolismo , Proteína de Clasificación Vacuolar VPS15/genética , Anomalías Múltiples/genética , Adolescente , Animales , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Femenino , Fibroblastos/metabolismo , Deformidades Congénitas de la Mano/complicaciones , Deformidades Congénitas de la Mano/genética , Humanos , Discapacidades para el Aprendizaje/complicaciones , Discapacidades para el Aprendizaje/genética , Masculino , Mutación , Mutación Missense , Insuficiencia Renal/complicaciones , Insuficiencia Renal/genética , Retinitis Pigmentosa/complicaciones , Retinitis Pigmentosa/genética , Saccharomyces cerevisiae , Hermanos , Piel/citología , Adulto Joven , Pez Cebra
11.
Am J Hum Genet ; 99(5): 1086-1105, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27745833

RESUMEN

This study establishes PYROXD1 variants as a cause of early-onset myopathy and uses biospecimens and cell lines, yeast, and zebrafish models to elucidate the fundamental role of PYROXD1 in skeletal muscle. Exome sequencing identified recessive variants in PYROXD1 in nine probands from five families. Affected individuals presented in infancy or childhood with slowly progressive proximal and distal weakness, facial weakness, nasal speech, swallowing difficulties, and normal to moderately elevated creatine kinase. Distinctive histopathology showed abundant internalized nuclei, myofibrillar disorganization, desmin-positive inclusions, and thickened Z-bands. PYROXD1 is a nuclear-cytoplasmic pyridine nucleotide-disulphide reductase (PNDR). PNDRs are flavoproteins (FAD-binding) and catalyze pyridine-nucleotide-dependent (NAD/NADH) reduction of thiol residues in other proteins. Complementation experiments in yeast lacking glutathione reductase glr1 show that human PYROXD1 has reductase activity that is strongly impaired by the disease-associated missense mutations. Immunolocalization studies in human muscle and zebrafish myofibers demonstrate that PYROXD1 localizes to the nucleus and to striated sarcomeric compartments. Zebrafish with ryroxD1 knock-down recapitulate features of PYROXD1 myopathy with sarcomeric disorganization, myofibrillar aggregates, and marked swimming defect. We characterize variants in the oxidoreductase PYROXD1 as a cause of early-onset myopathy with distinctive histopathology and introduce altered redox regulation as a primary cause of congenital muscle disease.


Asunto(s)
Núcleo Celular/genética , Miopatías Distales/genética , Variación Genética , Miopatías Estructurales Congénitas/genética , Oxidorreductasas/genética , Secuencia de Aminoácidos , Animales , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Estudios de Cohortes , Creatina Quinasa/genética , Creatina Quinasa/metabolismo , Citoplasma/metabolismo , Miopatías Distales/patología , Proteína 4 Similar a ELAV/genética , Proteína 4 Similar a ELAV/metabolismo , Femenino , Flavoproteínas/metabolismo , Eliminación de Gen , Estudio de Asociación del Genoma Completo , Glutatión Reductasa/genética , Glutatión Reductasa/metabolismo , Células HEK293 , Humanos , Masculino , Músculo Esquelético/patología , Mutación Missense , Miopatías Estructurales Congénitas/patología , Oxidorreductasas/metabolismo , Linaje , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Pez Cebra/genética
12.
Biol Aujourdhui ; 209(1): 97-109, 2015.
Artículo en Francés | MEDLINE | ID: mdl-26115715

RESUMEN

Phosphoinositides (PPIn) are lipids involved in the vesicular transport of proteins between the different intracellular compartments. They act by recruiting and/or activating effector proteins and are thus involved in crucial cellular functions including vesicle budding, fusion and dynamics of membranes and regulation of the cytoskeleton. Although they are present in low concentrations in membranes, their activity is essential for cell survival and needs to be tightly controlled. Therefore, phosphatases and kinases specific of the various cellular membranes can phosphorylate/dephosphorylate their inositol ring on the positions D3, D4 and/or D5. The differential phosphorylation determines the intracellular localisation and the activity of the PPIn. Indeed, non-phosphorylated phosphatidylinositol (PtdIns) is the basic component of the PPIn and can be found in all eukaryotic cells at the cytoplasmic face of the ER, the Golgi, mitochondria and microsomes. It can get phosphorylated on position D4 to obtain PtdIns4P, a PPIn enriched in the Golgi compartment and involved in the maintenance of this organelle as well as anterograde and retrograde transport to and from the Golgi. PtdIns phosphorylation on position D3 results in PtdIns3P that is required for endosomal transport and multivesicular body (MVB) formation and sorting. These monophosphorylated PtdIns can be further phosphorylated to produce bisphophorylated PtdIns. Thus, PtdIns(4,5)P2, mainly produced by PtdIns4P phosphorylation, is enriched in the plasma membrane and involved in the regulation of actin cytoskeleton and endocytosis. PtdIns(3,5)P2, mainly produced by PtdIns3P phosphorylation, is enriched in late endosomes, MVBs and the lysosome/vacuole and plays a role in endosome to vacuole transport. PtdIns(3,4)P2 is absent in yeast, cells and mainly produced by PtdIns4P phosphorylation in human cells; PtdIns(3,4)P2 is localised in the plasma membrane and plays an important role as a second messenger by recruiting specific protein kinases (Akt and PDK1). Finally the triple phosphorylated PPIn, PtdIns(3,4,5)P3 also absent in yeast, is produced by the phosphorylation of PtdIns(3,4)P2 and localized at the plasma membrane of human cells where it binds proteins via their PH domain. Interaction partners include members of the Arf (ADP-ribosylation factors) family, PDK1 (Phosphoinositide Dependent Kinase 1) and Akt. Therefore this last PPIn is essential for the control of cell proliferation and its deregulation leads to the development of numerous cancers. In conclusion, the regulation of PPIn phosphorylation/dephosphorylation is complex and needs to be very precisely regulated. Indeed phosphatases and kinases allow the maintenance of the equilibrium between the different forms. PPIn play a crucial role in numerous cellular functions and a loss in their synthesis or regulation results in severe genetic diseases.


Asunto(s)
Espacio Intracelular/metabolismo , Fosfatidilinositoles/fisiología , Vesículas Transportadoras/fisiología , Transporte Biológico , Membrana Celular/química , Membrana Celular/enzimología , Endocitosis , Retículo Endoplásmico/química , Endosomas , Aparato de Golgi/química , Humanos , Inositol/metabolismo , Espacio Intracelular/química , Microsomas/química , Mitocondrias/química , Fosfatos de Fosfatidilinositol/fisiología , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Fosfotransferasas/metabolismo , Sistemas de Mensajero Secundario , Vacuolas , Proteínas de Transporte Vesicular
13.
PLoS Genet ; 8(10): e1002965, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071445

RESUMEN

Myotubularin MTM1 is a phosphoinositide (PPIn) 3-phosphatase mutated in X-linked centronuclear myopathy (XLCNM; myotubular myopathy). We investigated the involvement of MTM1 enzymatic activity on XLCNM phenotypes. Exogenous expression of human MTM1 in yeast resulted in vacuolar enlargement, as a consequence of its phosphatase activity. Expression of mutants from patients with different clinical progression and determination of PtdIns3P and PtdIns5P cellular levels confirmed the link between vacuolar morphology and MTM1 phosphatase activity, and showed that some disease mutants retain phosphatase activity. Viral gene transfer of phosphatase-dead myotubularin mutants (MTM1(C375S) and MTM1(S376N)) significantly improved most histological signs of XLCNM displayed by a Mtm1-null mouse, at similar levels as wild-type MTM1. Moreover, the MTM1(C375S) mutant improved muscle performance and restored the localization of nuclei, triad alignment, and the desmin intermediate filament network, while it did not normalize PtdIns3P levels, supporting phosphatase-independent roles of MTM1 in maintaining normal muscle performance and organelle positioning in skeletal muscle. Among the different XLCNM signs investigated, we identified only triad shape and fiber size distribution as being partially dependent on MTM1 phosphatase activity. In conclusion, this work uncovers MTM1 roles in the structural organization of muscle fibers that are independent of its enzymatic activity. This underlines that removal of enzymes should be used with care to conclude on the physiological importance of their activity.


Asunto(s)
Miopatías Estructurales Congénitas/genética , Fenotipo , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Desmina/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/genética , Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Fuerza Muscular/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Mutación , Miopatías Estructurales Congénitas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
14.
Dev Cell ; 5(3): 499-511, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12967568

RESUMEN

PtdIns(3,5)P(2) is required for cargo-selective sorting to the vacuolar lumen via the multivesicular body (MVB). Here we show that Ent3p, a yeast epsin N-terminal homology (ENTH) domain-containing protein, is a specific PtdIns(3,5)P(2) effector localized to endosomes. The ENTH domain of Ent3p is essential for its PtdIns(3,5)P(2) binding activity and for its membrane interaction in vitro and in vivo. Ent3p is required for protein sorting into the MVB but not for the internalization step of endocytosis. Ent3p is associated with clathrin and is necessary for normal actin cytoskeleton organization. Our results show that Ent3p is required for protein sorting into intralumenal vesicles of the MVB through PtdIns(3,5)P(2) binding via its ENTH domain.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Fúngicas , Neuropéptidos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Señales de Clasificación de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología , Vacuolas/metabolismo , Proteínas de Transporte Vesicular , Actinas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Transferasas Alquil y Aril/metabolismo , Western Blotting , Clatrina/metabolismo , Endocitosis/fisiología , Endosomas/metabolismo , Proteínas Fluorescentes Verdes , Técnicas In Vitro , Cinética , Proteínas Luminiscentes/metabolismo , Oxigenasas de Función Mixta/metabolismo , Complejos Multienzimáticos/metabolismo , Mutación , Pruebas de Precipitina , Transporte de Proteínas , Compuestos de Piridinio/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Alineación de Secuencia/métodos , Temperatura , Factores de Tiempo , Levaduras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA