Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
1.
J Mol Biol ; 435(12): 168140, 2023 06 15.
Article de Anglais | MEDLINE | ID: mdl-37148997

RÉSUMÉ

Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence is increasing with age. A wealth of genetic evidence indicates that the endo-lysosomal system is a major pathway driving PD pathogenesis with a growing number of genes encoding endo-lysosomal proteins identified as risk factors for PD, making it a promising target for therapeutic intervention. However, detailed knowledge and understanding of the molecular mechanisms linking these genes to the disease are available for only a handful of them (e.g. LRRK2, GBA1, VPS35). Taking on the challenge of studying poorly characterized genes and proteins can be daunting, due to the limited availability of tools and knowledge from previous literature. This review aims at providing a valuable source of molecular and cellular insights into the biology of lesser-studied PD-linked endo-lysosomal genes, to help and encourage researchers in filling the knowledge gap around these less popular genetic players. Specific endo-lysosomal pathways discussed range from endocytosis, sorting, and vesicular trafficking to the regulation of membrane lipids of these membrane-bound organelles and the specific enzymatic activities they contain. We also provide perspectives on future challenges that the community needs to tackle and propose approaches to move forward in our understanding of these poorly studied endo-lysosomal genes. This will help harness their potential in designing innovative and efficient treatments to ultimately re-establish neuronal homeostasis in PD but also other diseases involving endo-lysosomal dysfunction.


Sujet(s)
Maladies neurodégénératives , Maladie de Parkinson , Humains , Maladie de Parkinson/métabolisme , Endocytose , alpha-Synucléine/métabolisme , Maladies neurodégénératives/anatomopathologie , Endosomes/métabolisme , Lysosomes/métabolisme
2.
Brain ; 146(5): 1859-1872, 2023 05 02.
Article de Anglais | MEDLINE | ID: mdl-36370000

RÉSUMÉ

The association between glucocerebrosidase, encoded by GBA, and Parkinson's disease (PD) highlights the role of the lysosome in PD pathogenesis. Genome-wide association studies in PD have revealed multiple associated loci, including the GALC locus on chromosome 14. GALC encodes the lysosomal enzyme galactosylceramidase, which plays a pivotal role in the glycosphingolipid metabolism pathway. It is still unclear whether GALC is the gene driving the association in the chromosome 14 locus and, if so, by which mechanism. We first aimed to examine whether variants in the GALC locus and across the genome are associated with galactosylceramidase activity. We performed a genome-wide association study in two independent cohorts from (i) Columbia University; and (ii) the Parkinson's Progression Markers Initiative study, followed by a meta-analysis with a total of 976 PD patients and 478 controls with available data on galactosylceramidase activity. We further analysed the effects of common GALC variants on expression and galactosylceramidase activity using genomic colocalization methods. Mendelian randomization was used to study whether galactosylceramidase activity may be causal in PD. To study the role of rare GALC variants, we analysed sequencing data from 5028 PD patients and 5422 controls. Additionally, we studied the functional impact of GALC knockout on alpha-synuclein accumulation and on glucocerebrosidase activity in neuronal cell models and performed in silico structural analysis of common GALC variants associated with altered galactosylceramidase activity. The top hit in PD genome-wide association study in the GALC locus, rs979812, is associated with increased galactosylceramidase activity (b = 1.2; SE = 0.06; P = 5.10 × 10-95). No other variants outside the GALC locus were associated with galactosylceramidase activity. Colocalization analysis demonstrated that rs979812 was also associated with increased galactosylceramidase expression. Mendelian randomization suggested that increased galactosylceramidase activity may be causally associated with PD (b = 0.025, SE = 0.007, P = 0.0008). We did not find an association between rare GALC variants and PD. GALC knockout using CRISPR-Cas9 did not lead to alpha-synuclein accumulation, further supporting that increased rather than reduced galactosylceramidase levels may be associated with PD. The structural analysis demonstrated that the common variant p.I562T may lead to improper maturation of galactosylceramidase affecting its activity. Our results nominate GALC as the gene associated with PD in this locus and suggest that the association of variants in the GALC locus may be driven by their effect of increasing galactosylceramidase expression and activity. Whether altering galactosylceramidase activity could be considered as a therapeutic target should be further studied.


Sujet(s)
Maladie de Parkinson , Humains , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme , Galactosylceramidase/génétique , Galactosylceramidase/métabolisme , Glucosylceramidase/génétique , Étude d'association pangénomique , Mutation , Hydrolases/génétique
3.
Sci Rep ; 12(1): 17176, 2022 10 13.
Article de Anglais | MEDLINE | ID: mdl-36229560

RÉSUMÉ

The use of human derived induced pluripotent stem cells (hiPSCs) differentiated to dopaminergic (DA) neurons offers a valuable experimental model to decorticate the cellular and molecular mechanisms of Parkinson's disease (PD) pathogenesis. However, the existing approaches present with several limitations, notably the lengthy time course of the protocols and the high variability in the yield of DA neurons. Here we report on the development of an improved approach that combines neurogenin-2 programming with the use of commercially available midbrain differentiation kits for a rapid, efficient, and reproducible directed differentiation of hiPSCs to mature and functional induced DA (iDA) neurons, with minimum contamination by other brain cell types. Gene expression analysis, associated with functional characterization examining neurotransmitter release and electrical recordings, support the functional identity of the iDA neurons to A9 midbrain neurons. iDA neurons showed selective vulnerability when exposed to 6-hydroxydopamine, thus providing a viable in vitro approach for modeling PD and for the screening of small molecules with neuroprotective proprieties.


Sujet(s)
Cellules souches pluripotentes induites , Maladie de Parkinson , Différenciation cellulaire/génétique , Dopamine/métabolisme , Neurones dopaminergiques/métabolisme , Humains , Mésencéphale/métabolisme , Agents neuromédiateurs/métabolisme , Oxidopamine/métabolisme , Oxidopamine/pharmacologie , Maladie de Parkinson/métabolisme
4.
Cell Rep ; 40(3): 111102, 2022 07 19.
Article de Anglais | MEDLINE | ID: mdl-35858558

RÉSUMÉ

The nervous system spread of alpha-synuclein fibrils is thought to cause Parkinson's disease (PD) and other synucleinopathies; however, the mechanisms underlying internalization and cellular spread are enigmatic. Here, we use confocal and superresolution microscopy, subcellular fractionation, and electron microscopy (EM) of immunogold-labeled α-synuclein preformed fibrils (PFFs) to demonstrate that this form of the protein undergoes rapid internalization and is targeted directly to lysosomes in as little as 2 min. Uptake of PFFs is disrupted by macropinocytic inhibitors and circumvents classical endosomal pathways. Immunogold-labeled PFFs are seen at the highly curved inward edge of membrane ruffles, in newly formed macropinosomes, in multivesicular bodies and in lysosomes. While most fibrils remain in lysosomes, a portion is transferred to neighboring naive cells along with markers of exosomes. These data indicate that PFFs use a unique internalization mechanism as a component of cell-to-cell propagation.


Sujet(s)
Maladie de Parkinson , Synucléinopathies , Endosomes/métabolisme , Humains , Lysosomes/métabolisme , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme
5.
Ann Neurol ; 87(1): 139-153, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31658403

RÉSUMÉ

OBJECTIVE: The TMEM175/GAK/DGKQ locus is the 3rd strongest risk locus in genome-wide association studies of Parkinson disease (PD). We aimed to identify the specific disease-associated variants in this locus, and their potential implications. METHODS: Full sequencing of TMEM175/GAK/DGKQ followed by genotyping of specific associated variants was performed in PD (n = 1,575) and rapid eye movement sleep behavior disorder (RBD) patients (n = 533) and in controls (n = 1,583). Adjusted regression models and a meta-analysis were performed. Association between variants and glucocerebrosidase (GCase) activity was analyzed in 715 individuals with available data. Homology modeling, molecular dynamics simulations, and lysosomal localization experiments were performed on TMEM175 variants to determine their potential effects on structure and function. RESULTS: Two coding variants, TMEM175 p.M393T (odds ratio [OR] = 1.37, p = 0.0003) and p.Q65P (OR = 0.72, p = 0.005), were associated with PD, and p.M393T was also associated with RBD (OR = 1.59, p = 0.001). TMEM175 p.M393T was associated with reduced GCase activity. Homology modeling and normal mode analysis demonstrated that TMEM175 p.M393T creates a polar side-chain in the hydrophobic core of the transmembrane, which could destabilize the domain and thus impair either its assembly, maturation, or trafficking. Molecular dynamics simulations demonstrated that the p.Q65P variant may increase stability and ion conductance of the transmembrane protein, and lysosomal localization was not affected by these variants. INTERPRETATION: Coding variants in TMEM175 are likely to be responsible for the association in the TMEM175/GAK/DGKQ locus, which could be mediated by affecting GCase activity. ANN NEUROL 2020;87:139-153.


Sujet(s)
Canaux potassiques/génétique , Synucléinopathies/génétique , Adulte , Sujet âgé , Études cas-témoins , Femelle , Prédisposition génétique à une maladie/génétique , Génotype , Glucosylceramidase/métabolisme , Humains , Lysosomes/métabolisme , Mâle , Adulte d'âge moyen , Modèles moléculaires , Simulation de dynamique moléculaire , Maladie de Parkinson/génétique , Maladie de Parkinson/physiopathologie , Polymorphisme de nucléotide simple/génétique , Canaux potassiques/physiologie , Trouble du comportement en sommeil paradoxal/génétique , Trouble du comportement en sommeil paradoxal/physiopathologie , Synucléinopathies/physiopathologie
6.
Mov Disord ; 34(4): 526-535, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30788890

RÉSUMÉ

BACKGROUND: SMPD1 (acid-sphingomyelinase) variants have been associated with Parkinson's disease in recent studies. The objective of this study was to further investigate the role of SMPD1 mutations in PD. METHODS: SMPD1 was sequenced in 3 cohorts (Israel Ashkenazi Jewish cohort, Montreal/Montpellier, and New York), including 1592 PD patients and 975 controls. Additional data were available for 10,709 Ashkenazi Jewish controls. Acid-sphingomyelinase activity was measured by a mass spectrometry-based assay in the New York cohort. α-Synuclein levels were measured in vitro following CRISPR/Cas9-mediated knockout and siRNA knockdown of SMPD1 in HeLa and BE(2)-M17 cells. Lysosomal localization of acid-sphingomyelinase with different mutations was studied, and in silico analysis of their effect on acid-sphingomyelinase structure was performed. RESULTS: SMPD1 mutations were associated with PD in the Ashkenazi Jewish cohort, as 1.4% of PD patients carried the p.L302P or p.fsP330 mutation, compared with 0.37% in 10,709 Ashkenazi Jewish controls (OR, 3.7; 95%CI, 1.6-8.2; P = 0.0025). In the Montreal/Montpellier cohort, the p.A487V variant was nominally associated with PD (1.5% versus 0.14%; P = 0.0065, not significant after correction for multiple comparisons). Among PD patients, reduced acid-sphingomyelinase activity was associated with a 3.5- to 5.8-year earlier onset of PD in the lowest quartile versus the highest quartile of acid-sphingomyelinase activity (P = 0.01-0.001). We further demonstrated that SMPD1 knockout and knockdown resulted in increased α-synuclein levels in HeLa and BE(2)-M17 dopaminergic cells and that the p.L302P and p.fsP330 mutations impair the traffic of acid-sphingomyelinase to the lysosome. CONCLUSIONS: Our results support an association between SMPD1 variants, acid-sphingomyelinase activity, and PD. Furthermore, they suggest that reduced acid-sphingomyelinase activity may lead to α-synuclein accumulation. © 2019 International Parkinson and Movement Disorder Society.


Sujet(s)
Encéphale/métabolisme , Prédisposition génétique à une maladie , Maladie de Parkinson/génétique , Sphingomyeline phosphodiesterase/génétique , alpha-Synucléine/métabolisme , Sujet âgé , Encéphale/anatomopathologie , Femelle , Techniques de knock-down de gènes , Cellules HeLa , Humains , Juif/génétique , Mâle , Adulte d'âge moyen , Mutation , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie
7.
Elife ; 62017 10 30.
Article de Anglais | MEDLINE | ID: mdl-29083303

RÉSUMÉ

Recent functional, proteomic and ribosome profiling studies in eukaryotes have concurrently demonstrated the translation of alternative open-reading frames (altORFs) in addition to annotated protein coding sequences (CDSs). We show that a large number of small proteins could in fact be coded by these altORFs. The putative alternative proteins translated from altORFs have orthologs in many species and contain functional domains. Evolutionary analyses indicate that altORFs often show more extreme conservation patterns than their CDSs. Thousands of alternative proteins are detected in proteomic datasets by reanalysis using a database containing predicted alternative proteins. This is illustrated with specific examples, including altMiD51, a 70 amino acid mitochondrial fission-promoting protein encoded in MiD51/Mief1/SMCR7L, a gene encoding an annotated protein promoting mitochondrial fission. Our results suggest that many genes are multicoding genes and code for a large protein and one or several small proteins.


Sujet(s)
Eucaryotes/génétique , Analyse de profil d'expression de gènes , Annotation de séquence moléculaire , Protéines/génétique , Protéines/métabolisme , Cadres ouverts de lecture , Biosynthèse des protéines
8.
Front Mol Neurosci ; 10: 311, 2017.
Article de Anglais | MEDLINE | ID: mdl-29021741

RÉSUMÉ

Synucleinopathies are a family of neurodegenerative disorders that comprises Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Each of these disorders is characterized by devastating motor, cognitive, and autonomic consequences. Current treatments for synucleinopathies are not curative and are limited to improvement of quality of life for affected individuals. Although the underlying causes of these diseases are unknown, a shared pathological hallmark is the presence of proteinaceous inclusions containing the α-synuclein (α-syn) protein in brain tissue. In the past few years, it has been proposed that these inclusions arise from the self-templated, prion-like spreading of misfolded and aggregated forms of α-syn throughout the brain, leading to neuronal dysfunction and death. In this review, we describe how impaired protein homeostasis is a prominent factor in the α-syn aggregation cascade, with alterations in protein quality control (PQC) pathways observed in the brains of patients. We discuss how PQC modulates α-syn accumulation, misfolding and aggregation primarily through chaperoning activity, proteasomal degradation, and lysosome-mediated degradation. Finally, we provide an overview of experimental data indicating that targeting PQC pathways is a promising avenue to explore in the design of novel neuroprotective approaches that could impede the spreading of α-syn pathology and thus provide a curative treatment for synucleinopathies.

9.
J Biol Chem ; 291(32): 16448-61, 2016 08 05.
Article de Anglais | MEDLINE | ID: mdl-27317664

RÉSUMÉ

Selective transport of pyruvate across the inner mitochondrial membrane by the mitochondrial pyruvate carrier (MPC) is a fundamental step that couples cytosolic and mitochondrial metabolism. The recent molecular identification of the MPC complex has revealed two interacting subunits, MPC1 and MPC2. Although in yeast, an additional subunit, MPC3, can functionally replace MPC2, no alternative MPC subunits have been described in higher eukaryotes. Here, we report for the first time the existence of a novel MPC subunit termed MPC1-like (MPC1L), which is present uniquely in placental mammals. MPC1L shares high sequence, structural, and topological homology with MPC1. In addition, we provide several lines of evidence to show that MPC1L is functionally equivalent to MPC1: 1) when co-expressed with MPC2, it rescues pyruvate import in a MPC-deleted yeast strain; 2) in mammalian cells, it can associate with MPC2 to form a functional carrier as assessed by bioluminescence resonance energy transfer; 3) in MPC1 depleted mouse embryonic fibroblasts, MPC1L rescues the loss of pyruvate-driven respiration and stabilizes MPC2 expression; and 4) MPC1- and MPC1L-mediated pyruvate imports show similar efficiency. However, we show that MPC1L has a highly specific expression pattern and is localized almost exclusively in testis and more specifically in postmeiotic spermatids and sperm cells. This is in marked contrast to MPC1/MPC2, which are ubiquitously expressed throughout the organism. To date, the biological importance of this alternative MPC complex during spermatogenesis in placental mammals remains unknown. Nevertheless, these findings open up new avenues for investigating the structure-function relationship within the MPC complex.


Sujet(s)
Transporteurs d'anions/biosynthèse , Régulation de l'expression des gènes/physiologie , Protéines de transport de la membrane mitochondriale/biosynthèse , Spermatides/métabolisme , Spermatogenèse/physiologie , Testicule/métabolisme , Animaux , Transporteurs d'anions/génétique , Femelle , Cellules HEK293 , Humains , Mâle , Souris , Protéines de transport de la membrane mitochondriale/génétique , Protéines mitochondriales/biosynthèse , Protéines mitochondriales/génétique , Transporteurs d'acides monocarboxyliques , Spermatides/cytologie , Testicule/cytologie
10.
PLoS Genet ; 12(5): e1006056, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-27176894

RÉSUMÉ

Mitochondrial import of pyruvate by the mitochondrial pyruvate carrier (MPC) is a central step which links cytosolic and mitochondrial intermediary metabolism. To investigate the role of the MPC in mammalian physiology and development, we generated a mouse strain with complete loss of MPC1 expression. This resulted in embryonic lethality at around E13.5. Mouse embryonic fibroblasts (MEFs) derived from mutant mice displayed defective pyruvate-driven respiration as well as perturbed metabolic profiles, and both defects could be restored by reexpression of MPC1. Labeling experiments using 13C-labeled glucose and glutamine demonstrated that MPC deficiency causes increased glutaminolysis and reduced contribution of glucose-derived pyruvate to the TCA cycle. Morphological defects were observed in mutant embryonic brains, together with major alterations of their metabolome including lactic acidosis, diminished TCA cycle intermediates, energy deficit and a perturbed balance of neurotransmitters. Strikingly, these changes were reversed when the pregnant dams were fed a ketogenic diet, which provides acetyl-CoA directly to the TCA cycle and bypasses the need for a functional MPC. This allowed the normal gestation and development of MPC deficient pups, even though they all died within a few minutes post-delivery. This study establishes the MPC as a key player in regulating the metabolic state necessary for embryonic development, neurotransmitter balance and post-natal survival.


Sujet(s)
Transporteurs d'anions/génétique , Cycle citrique/génétique , Régime cétogène , Mitochondries/génétique , Protéines de transport de la membrane mitochondriale/génétique , Animaux , Transporteurs d'anions/déficit , Femelle , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Gènes létaux , Glucose/métabolisme , Glutamine/métabolisme , Souris , Souris knockout , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Protéines de transport de la membrane mitochondriale/déficit , Transporteurs d'acides monocarboxyliques , Grossesse , Acide pyruvique/métabolisme
11.
Mol Cell ; 59(3): 491-501, 2015 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-26253029

RÉSUMÉ

The transport of pyruvate into mitochondria requires a specific carrier, the mitochondrial pyruvate carrier (MPC). The MPC represents a central node of carbon metabolism, and its activity is likely to play a key role in bioenergetics. Until now, investigation of the MPC activity has been limited. However, the recent molecular identification of the components of the carrier has allowed us to engineer a genetically encoded biosensor and to monitor the activity of the MPC in real time in a cell population or in a single cell. We report that the MPC activity is low in cancer cells, which mainly rely on glycolysis to generate ATP, a characteristic known as the Warburg effect. We show that this low activity can be reversed by increasing the concentration of cytosolic pyruvate, thus increasing oxidative phosphorylation. This biosensor represents a unique tool to investigate carbon metabolism and bioenergetics in various cell types.


Sujet(s)
Techniques de biocapteur/méthodes , Fibroblastes/cytologie , Mesures de luminescence/méthodes , Protéines de transport de la membrane mitochondriale/métabolisme , Acide pyruvique/métabolisme , Animaux , Lignée cellulaire , Embryon de mammifère/cytologie , Transfert d'énergie , Fibroblastes/métabolisme , Cellules HCT116 , Cellules HEK293 , Cellules HeLa , Humains , Cellules MCF-7 , Souris , Analyse sur cellule unique
12.
Curr Opin Cell Biol ; 33: 35-41, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25463844

RÉSUMÉ

Pyruvate metabolism plays a pivotal role in cell homeostasis and energy production. Pyruvate, the end product of glycolysis, is either catabolized in the cytosol, or enters into mitochondria to promote oxidative phosphorylation. The import of pyruvate into mitochondria requires a specific carrier in the inner mitochondrial membrane, the mitochondrial pyruvate carrier (MPC), whose identity was only recently discovered. Here we report our current knowledge of the structure and function of the MPC and we describe how dysfunction of the MPC could participate in various pathologies, including type 2 diabetes and cancer.


Sujet(s)
Homéostasie , Mitochondries/métabolisme , Acide pyruvique/métabolisme , Animaux , Glucose/métabolisme , Humains , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme , Protéines de transport de la membrane mitochondriale , Membranes mitochondriales/métabolisme , Transporteurs d'acides monocarboxyliques , Mutation/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie
13.
PLoS One ; 8(8): e70698, 2013.
Article de Anglais | MEDLINE | ID: mdl-23950983

RÉSUMÉ

A fully mature mRNA is usually associated to a reference open reading frame encoding a single protein. Yet, mature mRNAs contain unconventional alternative open reading frames (AltORFs) located in untranslated regions (UTRs) or overlapping the reference ORFs (RefORFs) in non-canonical +2 and +3 reading frames. Although recent ribosome profiling and footprinting approaches have suggested the significant use of unconventional translation initiation sites in mammals, direct evidence of large-scale alternative protein expression at the proteome level is still lacking. To determine the contribution of alternative proteins to the human proteome, we generated a database of predicted human AltORFs revealing a new proteome mainly composed of small proteins with a median length of 57 amino acids, compared to 344 amino acids for the reference proteome. We experimentally detected a total of 1,259 alternative proteins by mass spectrometry analyses of human cell lines, tissues and fluids. In plasma and serum, alternative proteins represent up to 55% of the proteome and may be a potential unsuspected new source for biomarkers. We observed constitutive co-expression of RefORFs and AltORFs from endogenous genes and from transfected cDNAs, including tumor suppressor p53, and provide evidence that out-of-frame clones representing AltORFs are mistakenly rejected as false positive in cDNAs screening assays. Functional importance of alternative proteins is strongly supported by significant evolutionary conservation in vertebrates, invertebrates, and yeast. Our results imply that coding of multiple proteins in a single gene by the use of AltORFs may be a common feature in eukaryotes, and confirm that translation of unconventional ORFs generates an as yet unexplored proteome.


Sujet(s)
Épissage alternatif , Cadres ouverts de lecture , Biosynthèse des protéines , Protéome , Protéomique , Séquence d'acides aminés , Protéine BRCA1/composition chimique , Protéine BRCA1/génétique , Protéine BRCA1/métabolisme , Lignée cellulaire , Biologie informatique/méthodes , Bases de données génétiques , Expression des gènes , Humains , Données de séquences moléculaires , Initiation de la traduction , Liaison aux protéines , Protéomique/méthodes , Reproductibilité des résultats , Alignement de séquences , Transfection
14.
Database (Oxford) ; 2012: bas025, 2012.
Article de Anglais | MEDLINE | ID: mdl-22613085

RÉSUMÉ

Human alternative open reading frames (HAltORF) is a publicly available and searchable online database referencing putative products of out-of-frame alternative translation initiation (ATI) in human mRNAs. Out-of-frame ATI is a process by which a single mRNA encodes independent proteins, when distinct initiation codons located in different reading frames are recognized by a ribosome to initiate translation. This mechanism is largely used in viruses to increase the coding potential of small viral genomes. There is increasing evidence that out-of-frame ATI is also used in eukaryotes, including human, and may contribute to the diversity of the human proteome. HAltORF is the first web-based searchable database that allows thorough investigation in the human transcriptome of out-of-frame alternative open reading frames with a start codon located in a strong Kozak context, and are thus the more likely to be expressed. It is also the first large scale study on the human transcriptome to successfully predict the expression of out-of-frame ATI protein products that were previously discovered experimentally. HAltORF will be a useful tool for the identification of human genes with multiple coding sequences, and will help to better define and understand the complexity of the human proteome. Database URL: http://haltorf.roucoulab.com/.


Sujet(s)
Fouille de données/méthodes , Bases de données génétiques , Décalage ribosomique , Cadres ouverts de lecture , Séquence d'acides aminés , Séquence nucléotidique , Humains , Données de séquences moléculaires , Initiation de la traduction , Logiciel , Transcriptome
15.
FASEB J ; 25(7): 2373-86, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21478263

RÉSUMÉ

The prion protein gene PRNP directs the synthesis of one of the most intensively studied mammalian proteins, the prion protein (PrP). Yet the physiological function of PrP has remained elusive and has created controversies in the literature. We found a downstream alternative translation initiation AUG codon surrounded by an optimal Kozak sequence in the +3 reading frame of PRNP. The corresponding alternative open reading frame encodes a polypeptide termed alternative prion protein (AltPrP) with a completely different amino acid sequence from PrP. We introduced a hemagglutinin (HA) tag in frame with AltPrP in PrP cDNAs from different species to test the expression of this novel polypeptide using anti-HA antibodies. AltPrP is constitutively coexpressed with human, bovine, sheep, and deer PrP. AltPrP is localized at the mitochondria and is up-regulated by endoplasmic reticulum stress and proteasomal inhibition. Generation of anti-AltPrP antibodies allowed us to test for endogenous expression of AltPrP in wild-type human cells expressing PrP. By transfecting cells with siRNA against PrP mRNA, we repressed expression of both PrP and AltPrP, confirming endogenous expression of AltPrP from PRNP. AltPrP was also detected in human brain homogenate, primary neurons, and peripheral blood mononuclear cells. These results demonstrate an unexpected function for PRNP, which, in addition to plasma membrane-anchored PrP, also encodes a second polypeptide termed AltPrP.


Sujet(s)
Gènes chevauchants/génétique , Cadres ouverts de lecture/génétique , Peptides/génétique , Prions/génétique , Animaux , Séquence nucléotidique , Lignée cellulaire tumorale , Cellules cultivées , ADN complémentaire/génétique , Réticulum endoplasmique/métabolisme , Expression des gènes , Cellules HEK293 , Humains , Immunotransfert , Agranulocytes/cytologie , Agranulocytes/métabolisme , Leupeptines/pharmacologie , Microscopie confocale , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme , Données de séquences moléculaires , Neurones/cytologie , Neurones/métabolisme , Peptides/métabolisme , Protéines prion , Prions/métabolisme , Proteasome endopeptidase complex/métabolisme , Inhibiteurs du protéasome , Interférence par ARN , Similitude de séquences d'acides nucléiques
16.
Biochim Biophys Acta ; 1793(2): 335-45, 2009 Feb.
Article de Anglais | MEDLINE | ID: mdl-19014979

RÉSUMÉ

The observation that PrP is present in the cytosol of some neurons and non-neuronal cells and that the N-terminal signal peptide is slightly inefficient has brought speculations concerning a possible function of the protein in the cytosol. Here, we show that cells expressing a cytosolic form of PrP termed cyPrP display a large juxtanuclear cytoplasmic RNA organelle. Although cyPrP spontaneously forms aggresomes, we used several mutants to demonstrate that the assembly of this RNA organelle is independent from cyPrP aggregation. Components of the organelle fall into three classes: mRNAs; proteins, including the RNAseIII family polymerase Dicer, the decapping enzyme Dcp1a, the DEAD-box RNA helicase DDX6, and the small nuclear ribonucleoprotein-associated proteins SmB/B'/N; and non-coding RNAs, including rRNA 5S, tRNAs, U1 small nuclear RNA, and microRNAs. This composition is similar to RNA granules or chromatoid bodies from germ cells, or planarian stem cells and neurons, which are large ribonucleoprotein complexes predicted to function in RNA processing and posttranscriptional gene regulation. The domain of PrP encompassing residues 30 to 49 is essential for the formation of the RNA particle. Our findings confirm the intriguing relation between PrP and RNA in cells, and underscore an unexpected function for cytosolic PrP: assembling a large RNA processing center which we have termed PrP-RNP for PrP-induced ribonucleoprotein particle.


Sujet(s)
Granulations cytoplasmiques/métabolisme , Régulation de l'expression des gènes , Prions/métabolisme , ARN/métabolisme , Ribonucléoprotéines/métabolisme , Transcription génétique , Animaux , Lignée cellulaire tumorale , Cellules germinales/métabolisme , Souris , Neurones/métabolisme , Pore nucléaire/métabolisme , Planaires/cytologie , Planaires/métabolisme , Liaison aux protéines , ARN ribosomique 18S/métabolisme , ARN ribosomique 28S/métabolisme , Petit ARN nucléaire/métabolisme , ARN de transfert/métabolisme , Ribonucléoprotéines/composition chimique , Cellules souches/métabolisme , Vimentine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...