Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mov Disord ; 37(2): 365-374, 2022 02.
Article in English | MEDLINE | ID: mdl-34820905

ABSTRACT

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.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dystonia , Dystonic Disorders , Animals , Dystonic Disorders/genetics , Homozygote , Humans , Zebrafish
2.
Biol Open ; 10(3)2021 03 22.
Article in English | MEDLINE | ID: mdl-33753324

ABSTRACT

COPI (coatomer complex I) coated vesicles are involved in Golgi-to-ER and intra-Golgi trafficking pathways, and mediate retrieval of ER resident proteins. Functions and components of the COPI-mediated trafficking pathways, beyond the canonical set of Sec/Arf proteins, are constantly increasing in number and complexity. In mammalian cells, GORAB, SCYL1 and SCYL3 proteins regulate Golgi morphology and protein glycosylation in concert with the COPI machinery. Here, we show that Cex1, homologous to the mammalian SCYL proteins, is a component of the yeast COPI machinery, by interacting with Sec27, Sec28 and Sec33 (Ret1/Cop1) proteins of the COPI coat. Cex1 was initially reported to mediate channeling of aminoacylated tRNA outside of the nucleus. Our data show that Cex1 localizes at membrane compartments, on structures positive for the Sec33 α-COP subunit. Moreover, the Wbp1 protein required for N-glycosylation and interacting via its di-lysine motif with the Sec27 ß'-COP subunit is mis-targeted in cex1Δ deletion mutant cells. Our data point to the possibility of developing Cex1 yeast-based models to study neurodegenerative disorders linked to pathogenic mutations of its human homologue SCYL1.


Subject(s)
Coat Protein Complex I/metabolism , Fungal Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/metabolism , Chromatography, Liquid , Coat Protein Complex I/genetics , Endoplasmic Reticulum/metabolism , Fungal Proteins/genetics , Gene Deletion , Golgi Apparatus/metabolism , Intracellular Space , Mass Spectrometry , Mutation , Nucleocytoplasmic Transport Proteins/genetics , Protein Binding , Protein Transport , Proteomics/methods , RNA-Binding Proteins/genetics
3.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32766723

ABSTRACT

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.


Subject(s)
Immunologic Deficiency Syndromes/complications , Inflammation/complications , Lymphoproliferative Disorders/complications , Membrane Proteins/metabolism , Actins/metabolism , Animals , Cell Degranulation , Cell Proliferation , Child , Cytotoxicity, Immunologic , Family , Female , Homozygote , Humans , Immunologic Deficiency Syndromes/immunology , Immunological Synapses/metabolism , Infant , Inflammation/immunology , Inflammation/pathology , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/immunology , Male , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mutation/genetics , Pedigree , Phenotype , Syndrome , Zebrafish
4.
Elife ; 92020 07 13.
Article in English | MEDLINE | ID: mdl-32657755

ABSTRACT

A single nuclear gene can be translated into a dual localized protein that distributes between the cytosol and mitochondria. Accumulating evidences show that mitoproteomes contain lots of these dual localized proteins termed echoforms. Unraveling the existence of mitochondrial echoforms using current GFP (Green Fluorescent Protein) fusion microscopy approaches is extremely difficult because the GFP signal of the cytosolic echoform will almost inevitably mask that of the mitochondrial echoform. We therefore engineered a yeast strain expressing a new type of Split-GFP that we termed Bi-Genomic Mitochondrial-Split-GFP (BiG Mito-Split-GFP). Because one moiety of the GFP is translated from the mitochondrial machinery while the other is fused to the nuclear-encoded protein of interest translated in the cytosol, the self-reassembly of this Bi-Genomic-encoded Split-GFP is confined to mitochondria. We could authenticate the mitochondrial importability of any protein or echoform from yeast, but also from other organisms such as the human Argonaute 2 mitochondrial echoform.


Subject(s)
Fungal Proteins/metabolism , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae/physiology , Cytosol/metabolism , Green Fluorescent Proteins/metabolism , Mitochondria/physiology , Protein Transport
5.
EMBO Mol Med ; 12(7): e11861, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32500975

ABSTRACT

The ubiquitin-proteasome system degrades ubiquitin-modified proteins to maintain protein homeostasis and to control signalling. Whole-genome sequencing of patients with severe deafness and early-onset cataracts as part of a neurological, sensorial and cutaneous novel syndrome identified a unique deep intronic homozygous variant in the PSMC3 gene, encoding the proteasome ATPase subunit Rpt5, which lead to the transcription of a cryptic exon. The proteasome content and activity in patient's fibroblasts was however unaffected. Nevertheless, patient's cells exhibited impaired protein homeostasis characterized by accumulation of ubiquitinated proteins suggesting severe proteotoxic stress. Indeed, the TCF11/Nrf1 transcriptional pathway allowing proteasome recovery after proteasome inhibition is permanently activated in the patient's fibroblasts. Upon chemical proteasome inhibition, this pathway was however impaired in patient's cells, which were unable to compensate for proteotoxic stress although a higher proteasome content and activity. Zebrafish modelling for knockout in PSMC3 remarkably reproduced the human phenotype with inner ear development anomalies as well as cataracts, suggesting that Rpt5 plays a major role in inner ear, lens and central nervous system development.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Cataract/genetics , Deafness/genetics , Mutation , Proteasome Endopeptidase Complex/genetics , Proteolysis , Stress, Physiological , Zebrafish Proteins/genetics , Adolescent , Animals , Cataract/pathology , Child , Child, Preschool , Consanguinity , Deafness/physiopathology , Female , Humans , Infant , Male , Nuclear Respiratory Factor 1/genetics , Pedigree , Phenotype , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Stress, Physiological/drug effects , Stress, Physiological/genetics , Syndrome , Ubiquitin/metabolism , Zebrafish/genetics
6.
Eur J Hum Genet ; 28(10): 1403-1413, 2020 10.
Article in English | MEDLINE | ID: mdl-32467598

ABSTRACT

Cause of complex dyskinesia remains elusive in some patients. A homozygous missense variant leading to drastic decrease of PDE2A enzymatic activity was reported in one patient with childhood-onset choreodystonia preceded by paroxysmal dyskinesia and associated with cognitive impairment and interictal EEG abnormalities. Here, we report three new cases with biallelic PDE2A variants identified by trio whole-exome sequencing. Mitochondria network was analyzed after Mitotracker™ Red staining in control and mutated primary fibroblasts. Analysis of retrospective video of patients' movement disorder and refinement of phenotype was carried out. We identified a homozygous gain of stop codon variant c.1180C>T; p.(Gln394*) in PDE2A in siblings and compound heterozygous variants in young adult: a missense c.446C>T; p.(Pro149Leu) and splice-site variant c.1922+5G>A predicted and shown to produce an out of frame transcript lacking exon 22. All three patients had cognitive impairment or developmental delay. The phenotype of the two oldest patients, aged 9 and 26, was characterized by childhood-onset refractory paroxysmal dyskinesia initially misdiagnosed as epilepsy due to interictal EEG abnormalities. The youngest patient showed a proven epilepsy at the age of 4 months and no paroxysmal dyskinesia at 15 months. Interestingly, analysis of the fibroblasts with the biallelic variants in PDE2A variants revealed mitochondria network morphology changes. Together with previously reported case, our three patients confirm that biallelic PDE2A variants are a cause of childhood-onset refractory paroxysmal dyskinesia with cognitive impairment, sometimes associated with choreodystonia and interictal baseline EEG abnormalities or epilepsy.


Subject(s)
Chorea/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Adult , Alleles , Cells, Cultured , Child , Chorea/pathology , Codon, Nonsense , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Developmental Disabilities/pathology , Female , Fibroblasts/metabolism , Heterozygote , Homozygote , Humans , Intellectual Disability/pathology , Male , Mitochondria/metabolism , Mitochondria/pathology , Mutation, Missense , Syndrome
7.
Cells ; 9(3)2020 03 10.
Article in English | MEDLINE | ID: mdl-32164332

ABSTRACT

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.


Subject(s)
Actin Cytoskeleton/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Yeasts/metabolism , Humans
8.
Front Genet ; 10: 504, 2019.
Article in English | MEDLINE | ID: mdl-31191616

ABSTRACT

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.

9.
Hum Mutat ; 40(10): 1826-1840, 2019 10.
Article in English | MEDLINE | ID: mdl-31116475

ABSTRACT

Mutations in genes encoding aminoacyl-tRNA synthetases have been reported in several neurological disorders. KARS is a dual localized lysyl-tRNA synthetase and its cytosolic isoform belongs to the multiple aminoacyl-tRNA synthetase complex (MSC). Biallelic mutations in the KARS gene were described in a wide phenotypic spectrum ranging from nonsyndromic deafness to complex impairments. Here, we report on a patient with severe neurological and neurosensory disease investigated by whole-exome sequencing and found to carry biallelic mutations c.683C>T (p.Pro228Leu) and c.871T>G (p.Phe291Val), the second one being novel, in the KARS gene. The patient presented with an atypical clinical presentation with an optic neuropathy not previously reported. At the cellular level, we show that cytoplasmic KARS was expressed at a lower level in patient cells and displayed decreased interaction with MSC. In vitro, these two KARS variants have a decreased aminoacylation activity compared with wild-type KARS, the p.Pro228Leu being the most affected. Our data suggest that dysfunction of cytoplasmic KARS resulted in a decreased level of translation of the nuclear-encoded lysine-rich proteins belonging to the respiratory chain complex, thus impairing mitochondria functions.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Lysine-tRNA Ligase/genetics , Mutation , Nervous System Diseases/complications , Nervous System Diseases/genetics , Optic Nerve Diseases/complications , Sensation Disorders/complications , Sensation Disorders/genetics , Alleles , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/metabolism , Fibroblasts/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/metabolism , Magnetic Resonance Imaging , Models, Molecular , Nervous System Diseases/diagnosis , Optic Nerve Diseases/diagnosis , Pedigree , Protein Binding , Protein Conformation , Sensation Disorders/diagnosis , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Neurol ; 266(2): 353-360, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30515627

ABSTRACT

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.


Subject(s)
Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/physiopathology , Oxidoreductases/genetics , Aged , Female , Finland , Genes, Recessive , Humans , Male , Muscular Dystrophies, Limb-Girdle/pathology , Pedigree , Exome Sequencing
11.
Hum Mutat ; 39(7): 983-992, 2018 07.
Article in English | MEDLINE | ID: mdl-29688594

ABSTRACT

Ciliopathies represent a wide spectrum of rare diseases with overlapping phenotypes and a high genetic heterogeneity. Among those, IFT140 is implicated in a variety of phenotypes ranging from isolated retinis pigmentosa to more syndromic cases. Using whole-genome sequencing in patients with uncharacterized ciliopathies, we identified a novel recurrent tandem duplication of exon 27-30 (6.7 kb) in IFT140, c.3454-488_4182+2588dup p.(Tyr1152_Thr1394dup), missed by whole-exome sequencing. Pathogenicity of the mutation was assessed on the patients' skin fibroblasts. Several hundreds of patients with a ciliopathy phenotype were screened and biallelic mutations were identified in 11 families representing 12 pathogenic variants of which seven are novel. Among those unrelated families especially with a Mainzer-Saldino syndrome, eight carried the same tandem duplication (two at the homozygous state and six at the heterozygous state). In conclusion, we demonstrated the implication of structural variations in IFT140-related diseases expanding its mutation spectrum. We also provide evidences for a unique genomic event mediated by an Alu-Alu recombination occurring on a shared haplotype. We confirm that whole-genome sequencing can be instrumental in the ability to detect structural variants for genomic disorders.


Subject(s)
Carrier Proteins/genetics , Cerebellar Ataxia/genetics , Ciliopathies/genetics , Retinitis Pigmentosa/genetics , Whole Genome Sequencing , Alu Elements/genetics , Cerebellar Ataxia/pathology , Ciliopathies/pathology , Databases, Genetic , Exons/genetics , Female , Heterozygote , Homozygote , Humans , Male , Mutation/genetics , Pedigree , Phenotype , Retinitis Pigmentosa/pathology
12.
J Clin Invest ; 127(12): 4477-4487, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29130937

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dynamin II/metabolism , Muscle Fibers, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Dynamin II/genetics , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/therapy , Nerve Tissue Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 114(44): E9308-E9317, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078390

ABSTRACT

The family of WD40-repeat (WDR) proteins is one of the largest in eukaryotes, but little is known about their function in brain development. Among 26 WDR genes assessed, we found 7 displaying a major impact in neuronal morphology when inactivated in mice. Remarkably, all seven genes showed corpus callosum defects, including thicker (Atg16l1, Coro1c, Dmxl2, and Herc1), thinner (Kif21b and Wdr89), or absent corpus callosum (Wdr47), revealing a common role for WDR genes in brain connectivity. We focused on the poorly studied WDR47 protein sharing structural homology with LIS1, which causes lissencephaly. In a dosage-dependent manner, mice lacking Wdr47 showed lethality, extensive fiber defects, microcephaly, thinner cortices, and sensory motor gating abnormalities. We showed that WDR47 shares functional characteristics with LIS1 and participates in key microtubule-mediated processes, including neural stem cell proliferation, radial migration, and growth cone dynamics. In absence of WDR47, the exhaustion of late cortical progenitors and the consequent decrease of neurogenesis together with the impaired survival of late-born neurons are likely yielding to the worsening of the microcephaly phenotype postnatally. Interestingly, the WDR47-specific C-terminal to LisH (CTLH) domain was associated with functions in autophagy described in mammals. Silencing WDR47 in hypothalamic GT1-7 neuronal cells and yeast models independently recapitulated these findings, showing conserved mechanisms. Finally, our data identified superior cervical ganglion-10 (SCG10) as an interacting partner of WDR47. Taken together, these results provide a starting point for studying the implications of WDR proteins in neuronal regulation of microtubules and autophagy.


Subject(s)
Autophagy/physiology , Brain/growth & development , Brain/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , WD40 Repeats/physiology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Microtubules/physiology , Neurogenesis/physiology , Neurons/metabolism , Neurons/physiology , Phenotype , Stem Cells/metabolism , Stem Cells/physiology
14.
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
15.
Int J Mol Sci ; 18(3)2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28294977

ABSTRACT

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).


Subject(s)
Cell Membrane/metabolism , Lipid Metabolism , Phosphatidylinositols/metabolism , Signal Transduction , Actins/metabolism , Animals , Autophagy , Biological Transport , Endocytosis , Endosomes/metabolism , Humans , Lysosomes/metabolism , Metabolic Networks and Pathways , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Phospholipids/metabolism , Sterols/metabolism
16.
Adv Biol Regul ; 63: 49-58, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27666502

ABSTRACT

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.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Myopathies, Structural, Congenital/genetics , Neoplasms/genetics , Obesity/genetics , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Gene Expression , Humans , Models, Molecular , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Neoplasms/metabolism , Neoplasms/pathology , Obesity/metabolism , Obesity/pathology , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
17.
Methods ; 113: 91-104, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27725303

ABSTRACT

By definition, cytosolic aminoacyl-tRNA synthetases (aaRSs) should be restricted to the cytosol of eukaryotic cells where they supply translating ribosomes with their aminoacyl-tRNA substrates. However, it has been shown that other translationally-active compartments like mitochondria and plastids can simultaneously contain the cytosolic aaRS and its corresponding organellar ortholog suggesting that both forms do not share the same organellar function. In addition, a fair number of cytosolic aaRSs have also been found in the nucleus of cells from several species. Hence, these supposedly cytosolic-restricted enzymes have instead the potential to be multi-localized. As expected, in all examples that were studied so far, when the cytosolic aaRS is imported inside an organelle that already contains its bona fide corresponding organellar-restricted aaRSs, the cytosolic form was proven to exert a nonconventional and essential function. Some of these essential functions include regulating homeostasis and protecting against various stresses. It thus becomes critical to assess meticulously the subcellular localization of each of these cytosolic aaRSs to unravel their additional roles. With this objective in mind, we provide here a review on what is currently known about cytosolic aaRSs multi-compartmentalization and we describe all commonly used protocols and procedures for identifying the compartments in which cytosolic aaRSs relocalize in yeast and human cells.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Cell Nucleus/enzymology , Cytosol/enzymology , Mitochondria/enzymology , Ribosomes/enzymology , Saccharomyces cerevisiae/enzymology , Amino Acyl-tRNA Synthetases/classification , Amino Acyl-tRNA Synthetases/genetics , Antibodies/chemistry , Blotting, Western/methods , Cell Compartmentation , Cell Fractionation/methods , Cell Line , Cell Nucleus/ultrastructure , Cytosol/ultrastructure , Fluorescent Antibody Technique/methods , Gene Expression , Humans , Mitochondria/ultrastructure , Protein Transport , Ribosomes/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure
18.
Nat Commun ; 7: 13586, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882921

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Cilia/metabolism , Ciliopathies/genetics , Golgi Apparatus/metabolism , Vacuolar Sorting Protein VPS15/genetics , Abnormalities, Multiple/genetics , Adolescent , Animals , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Craniofacial Abnormalities/complications , Craniofacial Abnormalities/genetics , Female , Fibroblasts/metabolism , Hand Deformities, Congenital/complications , Hand Deformities, Congenital/genetics , Humans , Learning Disabilities/complications , Learning Disabilities/genetics , Male , Mutation , Mutation, Missense , Renal Insufficiency/complications , Renal Insufficiency/genetics , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/genetics , Saccharomyces cerevisiae , Siblings , Skin/cytology , Young Adult , Zebrafish
19.
Am J Hum Genet ; 99(5): 1086-1105, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27745833

ABSTRACT

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.


Subject(s)
Cell Nucleus/genetics , Distal Myopathies/genetics , Genetic Variation , Myopathies, Structural, Congenital/genetics , Oxidoreductases/genetics , Amino Acid Sequence , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Cohort Studies , Creatine Kinase/genetics , Creatine Kinase/metabolism , Cytoplasm/metabolism , Distal Myopathies/pathology , ELAV-Like Protein 4/genetics , ELAV-Like Protein 4/metabolism , Female , Flavoproteins/metabolism , Gene Deletion , Genome-Wide Association Study , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , HEK293 Cells , Humans , Male , Muscle, Skeletal/pathology , Mutation, Missense , Myopathies, Structural, Congenital/pathology , Oxidoreductases/metabolism , Pedigree , Protein Conformation , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Zebrafish/genetics
20.
Biol Aujourdhui ; 209(1): 97-109, 2015.
Article in French | MEDLINE | ID: mdl-26115715

ABSTRACT

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.


Subject(s)
Intracellular Space/metabolism , Phosphatidylinositols/physiology , Transport Vesicles/physiology , Biological Transport , Cell Membrane/chemistry , Cell Membrane/enzymology , Endocytosis , Endoplasmic Reticulum/chemistry , Endosomes , Golgi Apparatus/chemistry , Humans , Inositol/metabolism , Intracellular Space/chemistry , Microsomes/chemistry , Mitochondria/chemistry , Phosphatidylinositol Phosphates/physiology , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Phosphotransferases/metabolism , Second Messenger Systems , Vacuoles , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...