Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(44): E9308-E9317, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29078390

RESUMO

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.


Assuntos
Autofagia/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Repetições WD40/fisiologia , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Microtúbulos/fisiologia , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Fenótipo , Células-Tronco/metabolismo , Células-Tronco/fisiologia
2.
Hum Mutat ; 40(10): 1826-1840, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31116475

RESUMO

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.


Assuntos
Aminoacil-tRNA Sintetases/genética , Lisina-tRNA Ligase/genética , Mutação , Doenças do Sistema Nervoso/complicações , Doenças do Sistema Nervoso/genética , Doenças do Nervo Óptico/complicações , Transtornos de Sensação/complicações , Transtornos de Sensação/genética , Alelos , Sequência de Aminoácidos , Aminoacil-tRNA Sintetases/química , Aminoacil-tRNA Sintetases/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Fibroblastos/metabolismo , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Lisina-tRNA Ligase/química , Lisina-tRNA Ligase/metabolismo , Imageamento por Ressonância Magnética , Modelos Moleculares , Doenças do Sistema Nervoso/diagnóstico , Doenças do Nervo Óptico/diagnóstico , Linhagem , Ligação Proteica , Conformação Proteica , Transtornos de Sensação/diagnóstico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Hum Mutat ; 39(7): 983-992, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29688594

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Ataxia Cerebelar/genética , Ciliopatias/genética , Retinose Pigmentar/genética , Sequenciamento Completo do Genoma , Elementos Alu/genética , Ataxia Cerebelar/patologia , Ciliopatias/patologia , Bases de Dados Genéticas , Éxons/genética , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Mutação/genética , Linhagem , Fenótipo , Retinose Pigmentar/patologia
4.
Methods ; 113: 91-104, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27725303

RESUMO

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.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Núcleo Celular/enzimologia , Citosol/enzimologia , Mitocôndrias/enzimologia , Ribossomos/enzimologia , Saccharomyces cerevisiae/enzimologia , Aminoacil-tRNA Sintetases/classificação , Aminoacil-tRNA Sintetases/genética , Anticorpos/química , Western Blotting/métodos , Compartimento Celular , Fracionamento Celular/métodos , Linhagem Celular , Núcleo Celular/ultraestrutura , Citosol/ultraestrutura , Imunofluorescência/métodos , Expressão Gênica , Humanos , Mitocôndrias/ultraestrutura , Transporte Proteico , Ribossomos/ultraestrutura , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/ultraestrutura
5.
PLoS Pathog ; 11(3): e1004703, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25742010

RESUMO

The intrinsic oncotropism and oncosuppressive activities of rodent protoparvoviruses (PVs) are opening new prospects for cancer virotherapy. Virus propagation, cytolytic activity, and spread are tightly connected to activation of the PDK1 signaling cascade, which delays stress-induced cell death and sustains functioning of the parvoviral protein NS1 through PKC(η)-driven modifications. Here we reveal a new PV-induced intracellular loop-back mechanism whereby PKCη/Rdx phosphorylates mouse PDK1:S138 and activates it independently of PI3-kinase signaling. The corresponding human PDK1phosphoS135 appears as a hallmark of highly aggressive brain tumors and may contribute to the very effective targeting of human gliomas by H-1PV. Strikingly, although H-1PV does not trigger PDK1 activation in normal human cells, such cells show enhanced viral DNA amplification and NS1-induced death upon expression of a constitutively active PDK1 mimicking PDK1phosphoS135. This modification thus appears as a marker of human glioma malignant progression and sensitivity to H-1PV-induced tumor cell killing.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/virologia , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Western Blotting , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virologia , Glioma/terapia , Glioma/virologia , Parvovirus H-1 , Humanos , Imunoprecipitação , Camundongos , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Parvovirus , Fosforilação , Piruvato Desidrogenase Quinase de Transferência de Acetil , Transfecção
6.
Int J Mol Sci ; 18(3)2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28294977

RESUMO

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).


Assuntos
Membrana Celular/metabolismo , Metabolismo dos Lipídeos , Fosfatidilinositóis/metabolismo , Transdução de Sinais , Actinas/metabolismo , Animais , Autofagia , Transporte Biológico , Endocitose , Endossomos/metabolismo , Humanos , Lisossomos/metabolismo , Redes e Vias Metabólicas , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipídeos/metabolismo , Esteróis/metabolismo
7.
PLoS Pathog ; 9(9): e1003605, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068925

RESUMO

Progeny particles of non-enveloped lytic parvoviruses were previously shown to be actively transported to the cell periphery through vesicles in a gelsolin-dependent manner. This process involves rearrangement and destruction of actin filaments, while microtubules become protected throughout the infection. Here the focus is on the intracellular egress pathway, as well as its impact on the properties and release of progeny virions. By colocalization with cellular marker proteins and specific modulation of the pathways through over-expression of variant effector genes transduced by recombinant adeno-associated virus vectors, we show that progeny PV particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane. Besides known factors like sar1, sec24, rab1, the ERM family proteins, radixin and moesin play (an) essential role(s) in the formation/loading and targeting of virus-containing COPII-vesicles. These proteins also contribute to the transport through ER and Golgi of the well described analogue of cellular proteins, the secreted Gaussia luciferase in absence of virus infection. It is therefore likely that radixin and moesin also serve for a more general function in cellular exocytosis. Finally, parvovirus egress via ER and Golgi appears to be necessary for virions to gain full infectivity through post-assembly modifications (e.g. phosphorylation). While not being absolutely required for cytolysis and progeny virus release, vesicular transport of parvoviruses through ER and Golgi significantly accelerates these processes pointing to a regulatory role of this transport pathway.


Assuntos
Linfócitos B/virologia , Retículo Endoplasmático/virologia , Complexo de Golgi/virologia , Interações Hospedeiro-Patógeno , Parvovirus/fisiologia , Vírion/fisiologia , Montagem de Vírus , Substituição de Aminoácidos , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/ultraestrutura , Transporte Biológico , Proteínas do Capsídeo/metabolismo , Efeito Citopatogênico Viral , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Hibridomas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mutação , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Parvovirus/imunologia , Parvovirus/ultraestrutura , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/metabolismo , Vírion/imunologia , Vírion/ultraestrutura , Liberação de Vírus
8.
Int J Mol Sci ; 16(1): 1509-25, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25584613

RESUMO

The yeast Saccharomyces cerevisiae is one of the best characterized eukaryotic models. The secretory pathway was the first trafficking pathway clearly understood mainly thanks to the work done in the laboratory of Randy Schekman in the 1980s. They have isolated yeast sec mutants unable to secrete an extracellular enzyme and these SEC genes were identified as encoding key effectors of the secretory machinery. For this work, the 2013 Nobel Prize in Physiology and Medicine has been awarded to Randy Schekman; the prize is shared with James Rothman and Thomas Südhof. Here, we present the different trafficking pathways of yeast S. cerevisiae. At the Golgi apparatus newly synthesized proteins are sorted between those transported to the plasma membrane (PM), or the external medium, via the exocytosis or secretory pathway (SEC), and those targeted to the vacuole either through endosomes (vacuolar protein sorting or VPS pathway) or directly (alkaline phosphatase or ALP pathway). Plasma membrane proteins can be internalized by endocytosis (END) and transported to endosomes where they are sorted between those targeted for vacuolar degradation and those redirected to the Golgi (recycling or RCY pathway). Studies in yeast S. cerevisiae allowed the identification of most of the known effectors, protein complexes, and trafficking pathways in eukaryotic cells, and most of them are conserved among eukaryotes.


Assuntos
Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Membrana Celular/metabolismo , Endocitose , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Vesículas Secretórias/metabolismo
9.
Swiss Med Wkly ; 152: w30115, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35262317

RESUMO

AIM OF THE STUDY: Previous literature suggests that ambient temperature may play a role in increasing the risk of suicide. Although in Switzerland suicides are an important cause of death, limited research exists on risk factors for suicidal behaviour, including ambient temperature. We aimed to assess the short-term association between ambient temperature and suicide risk in Switzerland between 1995 and 2016, and the differences by region, individual characteristics and method of suicide. METHODS: We collected daily data on suicides and mean temperatures in each canton of Switzerland. We used a two-stage approach, consisting of a case time series analysis using conditional quasi-Poisson and distributed lag non-linear models followed by a multivariate meta-regression analysis. We conducted subgroup analyses by sex, age (<35, 35-65 and >65 years) and method of suicide (violent or nonviolent). RESULTS: Between 1995 and 2016, there were a total of 24,067 suicides in Switzerland. Overall, we found a positive and non-linear temperature-suicide association in all regions. On average, the risk of suicide increased by 34% (1.34 relative risk [95% confidence interval: 1.19-1.52]) from the 10th to the 99th temperature percentile in Switzerland (lag period of 0-2 days). Indications of larger risks were mostly found in females, younger individuals (<35 years) and with nonviolent methods. Regional risks ranged from 24% (East region) to 55% (North-West region). CONCLUSIONS: Our findings suggest that increasing temperatures could be considered a risk factor for suicidal behaviour in Switzerland. Knowledge of the profile of people committing suicide could help us to understand the mechanisms behind this association and thus support policymakers in suicide prevention.


Assuntos
Suicídio , Idoso , Feminino , Humanos , Fatores de Risco , Suíça/epidemiologia , Temperatura , Violência
10.
Biochem Biophys Res Commun ; 394(4): 904-8, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20230797

RESUMO

The conserved disulfide-bonded region (DSR) of the human immunodeficiency virus type 1 (HIV-1) fusion glycoprotein, gp41, mediates association with the receptor-binding glycoprotein, gp120. Interactions between gp120, CD4 and chemokine receptors activate the fusion activity of gp41. The introduction of W596L and W610F mutations to the DSR of HIV-1(QH1549.13) blocked viral entry and hemifusion without affecting gp120-gp41 association. The fusion defect correlated with inhibition of CD4-triggered gp41 pre-hairpin formation, consistent with the DSR mutations having decoupled receptor-induced conformational changes in gp120 from gp41 activation. Our data implicate the DSR in sensing conformational changes in the gp120-gp41 complex that lead to fusion activation.


Assuntos
Antígenos CD4/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , HIV-1/fisiologia , Fusão de Membrana , Internalização do Vírus , Substituição de Aminoácidos , Cristalografia por Raios X , Cisteína/genética , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/genética , HIV-1/genética , Humanos , Mutação , Conformação Proteica
11.
J Virol ; 83(11): 5854-63, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19321616

RESUMO

The propagation of autonomous parvoviruses is strongly dependent on the phosphorylation of the major nonstructural protein NS1 by members of the protein kinase C (PKC) family. Minute virus of mice (MVM) replication is accompanied by changes in the overall phosphorylation pattern of NS1, which is newly modified at consensus PKC sites. These changes result, at least in part, from the ability of MVM to modulate the PDK-1/PKC pathway, leading to activation and redistribution of both PDK-1 and PKCeta. We show that proteins of the ezrin-radixin-moesin (ERM) family are essential for virus propagation and spreading through their functions as adaptors for PKCeta. MVM infection led to redistribution of radixin and moesin in the cell, resulting in increased colocalization of these proteins with PKCeta. Radixin was found to control the PKCeta-driven phosphorylation of NS1 and newly synthesized capsids in vivo. Conversely, radixin phosphorylation and activation were driven by the NS1/CKIIalpha complex. Altogether, these data argue for ERM proteins being both targets and modulators of parvovirus infection.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Parvovirus/metabolismo , Replicação Viral , Animais , Caseína Quinase II/metabolismo , Linhagem Celular , Proteínas do Citoesqueleto/genética , Humanos , Proteínas de Membrana/genética , Camundongos , Proteínas dos Microfilamentos/genética , Parvovirus/genética , Fosforilação , Ligação Proteica , Proteína Quinase C/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
12.
PLoS Pathog ; 4(8): e1000126, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18704167

RESUMO

The autonomous parvovirus Minute Virus of Mice (MVM) induces specific changes in the cytoskeleton filaments of infected permissive cells, causing in particular the degradation of actin fibers and the generation of "actin patches." This is attributed to a virus-induced imbalance between the polymerization factor N-WASP (Wiscott-Aldrich syndrome protein) and gelsolin, a multifunctional protein cleaving actin filaments. Here, the focus is on the involvement of gelsolin in parvovirus propagation and virus-induced actin processing. Gelsolin activity was knocked-down, and consequences thereof were determined for virus replication and egress and for actin network integrity. Though not required for virus replication or progeny particle assembly, gelsolin was found to control MVM (and related H1-PV) transport from the nucleus to the cell periphery and release into the culture medium. Gelsolin-dependent actin degradation and progeny virus release were both controlled by (NS1)/CKIIalpha, a recently identified complex between a cellular protein kinase and a MVM non-structural protein. Furthermore, the export of newly synthesized virions through the cytoplasm appeared to be mediated by (virus-modified) lysomal/late endosomal vesicles. By showing that MVM release, like entry, is guided by the cytoskeleton and mediated by vesicles, these results challenge the current view that egress of non-enveloped lytic viruses is a passive process.


Assuntos
Núcleo Celular/metabolismo , Gelsolina/metabolismo , Vírus Miúdo do Camundongo/fisiologia , Replicação Viral/fisiologia , Citoesqueleto de Actina/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/virologia , Endossomos/metabolismo , Endossomos/virologia , Humanos , Lisossomos/metabolismo , Lisossomos/virologia , Infecções por Parvoviridae/metabolismo , Proteínas não Estruturais Virais , Vírion/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
13.
Eur J Hum Genet ; 28(10): 1403-1413, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32467598

RESUMO

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.


Assuntos
Coreia/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Adulto , Alelos , Células Cultivadas , Criança , Coreia/patologia , Códon sem Sentido , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Deficiências do Desenvolvimento/patologia , Feminino , Fibroblastos/metabolismo , Heterozigoto , Homozigoto , Humanos , Deficiência Intelectual/patologia , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação de Sentido Incorreto , Síndrome
14.
EMBO Mol Med ; 12(7): e11861, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32500975

RESUMO

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.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/genética , Catarata/genética , Surdez/genética , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Proteólise , Estresse Fisiológico , Proteínas de Peixe-Zebra/genética , Adolescente , Animais , Catarata/patologia , Criança , Pré-Escolar , Consanguinidade , Surdez/fisiopatologia , Feminino , Humanos , Lactente , Masculino , Fator 1 Nuclear Respiratório/genética , Linhagem , Fenótipo , Inibidores de Proteassoma/farmacologia , Proteólise/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Síndrome , Ubiquitina/metabolismo , Peixe-Zebra/genética
15.
Cell Microbiol ; 10(3): 755-69, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18042254

RESUMO

Autonomous parvoviruses are strongly dependent on the phosphorylation of the major non-structural protein NS1 by members of the protein kinase C (PKC) family. Besides being accompanied with changes in the overall phosphorylation pattern of NS1 and acquiring new modifications at consensus PKC sites, ongoing minute virus of mice (MVM) infections lead to the appearance of new phosphorylated cellular protein species. This prompted us to investigate whether MVM actively interferes with phosphoinositol-dependent kinase (PDK)/PKC signalling. The activity, subcellular localization and phosphorylation status of the protein kinases PDK1, PKCeta and PKClambda were measured in A9 cells in the presence or absence of MVM infection. Parvovirus infection was found to result in activation of both PDK1 and PKCeta, as evidenced by changes in their subcellular distribution and overall (auto)phosphorylation. We show evidence that activation of PKCeta by PDK1 is driven by atypical PKClambda. By modifying the hydrophobic motif of PKCeta, PKClambda appeared to control docking and consecutive phosphorylation of PKCeta's activation-loop by PDK1, a process that was inhibited in vivo in the presence of a dominant-negative PKClambda mutant.


Assuntos
Fibroblastos/virologia , Vírus Miúdo do Camundongo/fisiologia , Proteína Quinase C/metabolismo , Animais , Linhagem Celular , Núcleo Celular/química , Citoplasma/química , Isoenzimas/metabolismo , Camundongos , Microscopia de Fluorescência , Fosforilação , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil
16.
Front Genet ; 10: 504, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191616

RESUMO

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.

17.
Nat Commun ; 7: 13586, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27882921

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Cílios/metabolismo , Ciliopatias/genética , Complexo de Golgi/metabolismo , Proteína VPS15 de Distribuição Vacuolar/genética , Anormalidades Múltiplas/genética , Adolescente , Animais , Estudos de Casos e Controles , Células Cultivadas , Criança , Pré-Escolar , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/genética , Feminino , Fibroblastos/metabolismo , Deformidades Congênitas da Mão/complicações , Deformidades Congênitas da Mão/genética , Humanos , Deficiências da Aprendizagem/complicações , Deficiências da Aprendizagem/genética , Masculino , Mutação , Mutação de Sentido Incorreto , Insuficiência Renal/complicações , Insuficiência Renal/genética , Retinose Pigmentar/complicações , Retinose Pigmentar/genética , Saccharomyces cerevisiae , Irmãos , Pele/citologia , Adulto Jovem , Peixe-Zebra
18.
Biol Aujourdhui ; 209(1): 97-109, 2015.
Artigo em Francês | MEDLINE | ID: mdl-26115715

RESUMO

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.


Assuntos
Espaço Intracelular/metabolismo , Fosfatidilinositóis/fisiologia , Vesículas Transportadoras/fisiologia , Transporte Biológico , Membrana Celular/química , Membrana Celular/enzimologia , Endocitose , Retículo Endoplasmático/química , Endossomos , Complexo de Golgi/química , Humanos , Inositol/metabolismo , Espaço Intracelular/química , Microssomos/química , Mitocôndrias/química , Fosfatos de Fosfatidilinositol/fisiologia , Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Fosfotransferases/metabolismo , Sistemas do Segundo Mensageiro , Vacúolos , Proteínas de Transporte Vesicular
19.
Biochem Biophys Res Commun ; 359(4): 1037-43, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17577584

RESUMO

Retroviral transmembrane proteins (TMs) contain a glycine-rich segment linking the N-terminal fusion peptide and coiled coil core. Previously, we reported that the glycine-rich segment (Met-326-Ser-337) of the human T-cell leukemia virus type 1 (HTLV-1) TM, gp21, is a determinant of membrane fusion function [K.A. Wilson, S. Bär, A.L. Maerz, M. Alizon, P. Poumbourios, The conserved glycine-rich segment linking the N-terminal fusion peptide to the coiled coil of human T-cell leukemia virus type 1 transmembrane glycoprotein gp21 is a determinant of membrane fusion function, J. Virol. 79 (2005) 4533-4539]. Here we show that the reduced fusion activity of an I334A mutant correlated with a decrease in stability of the gp21 trimer of hairpins conformation, in the context of a maltose-binding protein-gp21 chimera. The stabilizing influence of Ile-334 required the C-terminal membrane-proximal sequence Trp-431-Ser-436. Proline substitution of four of five Gly residues altered gp21 trimer of hairpins stability. Our data indicate that flexibility within and hydrophobic interactions mediated by this region are determinants of gp21 stability and membrane fusion function.


Assuntos
Produtos do Gene env/química , Glicina/química , Proteínas Oncogênicas de Retroviridae/química , Retroviridae/química , Sequência de Aminoácidos , Estabilidade de Medicamentos , Dados de Sequência Molecular , Conformação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Produtos do Gene env do Vírus da Imunodeficiência Humana
20.
J Virol ; 80(6): 2815-22, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501090

RESUMO

Ebola viruses (EboV) are enveloped RNA viruses infecting cells by a pH-dependent process mediated by viral glycoproteins (GP) involving endocytosis of virions and their routing into acidic endosomes. As with well-characterized pH-dependent viral entry proteins, in particular influenza virus hemagglutinin, it is thought that EboV GP require activation by low pH in order to mediate fusion of the viral envelope with the membrane of endosomes. However, it has not yet been possible to confirm the direct role of EboV GP in membrane fusion and the requirement for low-pH activation. It was in particular not possible to induce formation of syncytia by exposing cells expressing EboV GP to acidic medium. Here, we have used an assay based on the induction of a beta-galactosidase (lacZ) reporter gene in target cells to detect cytoplasmic exchanges, indicating membrane fusion, with cells expressing EboV GP (Zaire species). Acidic activation of GP-expressing cells was required for efficient fusion with target cells. The direct role of EboV GP in this process is indicated by its inhibition by anti-GP antibodies and by the lack of activity of mutant GP normally expressed at the cell surface but defective for virus entry. Fusion was not observed when target cells underwent acidic treatment, for example, when they were placed in coculture with GP-expressing cells before the activation step. This unexpected feature, possibly related to the nature of the EboV receptor, could explain the impossibility of inducing formation of syncytia among GP-expressing cells.


Assuntos
Ebolavirus/fisiologia , Glicoproteínas/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Animais , Células COS , Fusão Celular , Chlorocebus aethiops , Glicoproteínas/genética , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA