Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
EMBO Rep ; 24(12): e57224, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37818801

RESUMO

The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.


Assuntos
Antígeno 2 do Estroma da Médula Óssea , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Liberação de Vírus , Humanos , Antígeno 2 do Estroma da Médula Óssea/antagonistas & inibidores , Antígeno 2 do Estroma da Médula Óssea/metabolismo , COVID-19/virologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/genética
2.
Am J Hum Genet ; 107(6): 1129-1148, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33186545

RESUMO

The endosomal sorting complexes required for transport (ESCRTs) are essential for multiple membrane modeling and membrane-independent cellular processes. Here we describe six unrelated individuals with de novo missense variants affecting the ATPase domain of VPS4A, a critical enzyme regulating ESCRT function. Probands had structural brain abnormalities, severe neurodevelopmental delay, cataracts, growth impairment, and anemia. In cultured cells, overexpression of VPS4A mutants caused enlarged endosomal vacuoles resembling those induced by expression of known dominant-negative ATPase-defective forms of VPS4A. Proband-derived fibroblasts had enlarged endosomal structures with abnormal accumulation of the ESCRT protein IST1 on the limiting membrane. VPS4A function was also required for normal endosomal morphology and IST1 localization in iPSC-derived human neurons. Mutations affected other ESCRT-dependent cellular processes, including regulation of centrosome number, primary cilium morphology, nuclear membrane morphology, chromosome segregation, mitotic spindle formation, and cell cycle progression. We thus characterize a distinct multisystem disorder caused by mutations affecting VPS4A and demonstrate that its normal function is required for multiple human developmental and cellular processes.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento/genética , ATPases Vacuolares Próton-Translocadoras/genética , Alelos , Animais , Encéfalo/anormalidades , Ciclo Celular , Centrossomo/metabolismo , Endossomos/metabolismo , Fibroblastos/metabolismo , Genômica , Células HEK293 , Células HeLa , Humanos , Camundongos , Neurônios/metabolismo , Domínios Proteicos , Transporte Proteico , Fuso Acromático/metabolismo
3.
J Cell Sci ; 134(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34042162

RESUMO

To provide insights into the kiss-and-run and full fusion events resulting in endocytic delivery to lysosomes, we investigated conditions causing increased tethering and pore formation between late endocytic organelles in HeLa cells. Knockout of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) VAMP7 and VAMP8 showed, by electron microscopy, the accumulation of tethered lysosome-associated membrane protein (LAMP)-carrier vesicles around multivesicular bodies, as well as the appearance of 'hourglass' profiles of late endocytic organelles attached by filamentous tethers, but did not prevent endocytic delivery to lysosomal hydrolases. Subsequent depletion of the SNARE YKT6 reduced this delivery, consistent with it compensating for the absence of VAMP7 and VAMP8. We also investigated filamentous tethering between multivesicular bodies and enlarged endolysosomes following depletion of charged multi-vesicular body protein 6 (CHMP6), and provide the first evidence that pore formation commences at the edge of tether arrays, with pore expansion required for full membrane fusion.


Assuntos
Fusão de Membrana , Proteínas SNARE , Endossomos , Células HeLa , Humanos , Lisossomos , Proteínas R-SNARE/genética , Proteínas SNARE/genética
4.
J Cell Sci ; 133(12)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32376785

RESUMO

Optineurin (OPTN) is a multifunctional protein involved in autophagy and secretion, as well as nuclear factor κB (NF-κB) and IRF3 signalling, and OPTN mutations are associated with several human diseases. Here, we show that, in response to viral RNA, OPTN translocates to foci in the perinuclear region, where it negatively regulates NF-κB and IRF3 signalling pathways and downstream pro-inflammatory cytokine secretion. These OPTN foci consist of a tight cluster of small membrane vesicles, which are positive for ATG9A. Disease mutations in OPTN linked to primary open-angle glaucoma (POAG) cause aberrant foci formation in the absence of stimuli, which correlates with the ability of OPTN to inhibit signalling. By using proximity labelling proteomics, we identify the linear ubiquitin assembly complex (LUBAC), CYLD and TBK1 as part of the OPTN interactome and show that these proteins are recruited to this OPTN-positive perinuclear compartment. Our work uncovers a crucial role for OPTN in dampening NF-κB and IRF3 signalling through the sequestration of LUBAC and other positive regulators in this viral RNA-induced compartment, leading to altered pro-inflammatory cytokine secretion.


Assuntos
Glaucoma de Ângulo Aberto , Fator de Transcrição TFIIIA , Proteínas de Ciclo Celular , Citocinas/genética , Humanos , Proteínas de Membrana Transportadoras , NF-kappa B/genética , NF-kappa B/metabolismo , Transporte Proteico , Transdução de Sinais , Fator de Transcrição TFIIIA/genética , Fator de Transcrição TFIIIA/metabolismo
5.
Brain ; 141(12): 3428-3442, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30496365

RESUMO

Mutations in the endosome-associated protein CHMP2B cause frontotemporal dementia and lead to lysosomal storage pathology in neurons. We here report that physiological levels of mutant CHMP2B causes reduced numbers and significantly impaired trafficking of endolysosomes within neuronal dendrites, accompanied by increased dendritic branching. Mechanistically, this is due to the stable incorporation of mutant CHMP2B onto neuronal endolysosomes, which we show renders them unable to traffic within dendrites. This defect is due to the inability of mutant CHMP2B to recruit the ATPase VPS4, which is required for release of CHMP2B from endosomal membranes. Strikingly, both impaired trafficking and the increased dendritic branching were rescued by treatment with antisense oligonucleotides targeting the well validated frontotemporal dementia risk factor TMEM106B, which encodes an endolysosomal protein. This indicates that reducing TMEM106B levels can restore endosomal health in frontotemporal dementia. As TMEM106B is a risk factor for frontotemporal dementia caused by both C9orf72 and progranulin mutations, and antisense oligonucleotides are showing promise as therapeutics for neurodegenerative diseases, our data suggests a potential new strategy for treating the wide range of frontotemporal dementias associated with endolysosomal dysfunction.


Assuntos
Dendritos/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/metabolismo , Demência Frontotemporal/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal
6.
Brain ; 141(5): 1286-1299, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29481671

RESUMO

Many genetic neurological disorders exhibit variable expression within affected families, often exemplified by variations in disease age at onset. Epistatic effects (i.e. effects of modifier genes on the disease gene) may underlie this variation, but the mechanistic basis for such epistatic interactions is rarely understood. Here we report a novel epistatic interaction between SPAST and the contiguous gene DPY30, which modifies age at onset in hereditary spastic paraplegia, a genetic axonopathy. We found that patients with hereditary spastic paraplegia caused by genomic deletions of SPAST that extended into DPY30 had a significantly younger age at onset. We show that, like spastin, the protein encoded by SPAST, the DPY30 protein controls endosomal tubule fission, traffic of mannose 6-phosphate receptors from endosomes to the Golgi, and lysosomal ultrastructural morphology. We propose that additive effects on this pathway explain the reduced age at onset of hereditary spastic paraplegia in patients who are haploinsufficient for both genes.


Assuntos
Epistasia Genética/genética , Mutação/genética , Proteínas Nucleares/genética , Paraplegia Espástica Hereditária/genética , Espastina/genética , Adulto , Idade de Início , Antígenos CD8/genética , Antígenos CD8/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HeLa/metabolismo , Células HeLa/ultraestrutura , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/ultraestrutura , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestrutura , Transporte Proteico/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Traffic ; 17(8): 908-22, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27126738

RESUMO

Krabbe disease is a severe, fatal neurodegenerative disorder caused by defects in the lysosomal enzyme galactocerebrosidase (GALC). The correct targeting of GALC to the lysosome is essential for the degradation of glycosphingolipids including the primary lipid component of myelin. Over 100 different mutations have been identified in GALC that cause Krabbe disease but the mechanisms by which they cause disease remain unclear. We have generated monoclonal antibodies against full-length human GALC and used these to monitor the trafficking and processing of GALC variants in cell-based assays and by immunofluorescence microscopy. Striking differences in the secretion, processing and endosomal targeting of GALC variants allows the classification of these into distinct categories. A subset of GALC variants are not secreted by cells, not proteolytically processed, and remain trapped in the ER; these are likely to cause disease due to protein misfolding and should be targeted for pharmacological chaperone therapies. Other GALC variants can be correctly secreted by cells and cause disease due to catalytic defects in the enzyme active site, inappropriate post-translational modification or a potential inability to bind essential cofactors. The classification of disease pathogenesis presented here provides a molecular framework for appropriate targeting of future Krabbe disease therapies.


Assuntos
Galactosilceramidase/metabolismo , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/metabolismo , Lisossomos/metabolismo , Mutação/genética , Linhagem Celular , Galactosilceramidase/química , Galactosilceramidase/genética , Humanos , Lisossomos/genética , Processamento de Proteína Pós-Traducional
8.
Hum Mol Genet ; 24(17): 4984-96, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26085577

RESUMO

Adaptor proteins (AP 1-5) are heterotetrameric complexes that facilitate specialized cargo sorting in vesicular-mediated trafficking. Mutations in AP5Z1, encoding a subunit of the AP-5 complex, have been reported to cause hereditary spastic paraplegia (HSP), although their impact at the cellular level has not been assessed. Here we characterize three independent fibroblast lines derived from skin biopsies of patients harbouring nonsense mutations in AP5Z1 and presenting with spastic paraplegia accompanied by neuropathy, parkinsonism and/or cognitive impairment. In all three patient-derived lines, we show that there is complete loss of AP-5 ζ protein and a reduction in the associated AP-5 µ5 protein. Using ultrastructural analysis, we show that these patient-derived lines consistently exhibit abundant multilamellar structures that are positive for markers of endolysosomes and are filled with aberrant storage material organized as exaggerated multilamellar whorls, striated belts and 'fingerprint bodies'. This phenotype can be replicated in a HeLa cell culture model by siRNA knockdown of AP-5 ζ. The cellular phenotype bears striking resemblance to features described in a number of lysosomal storage diseases (LSDs). Collectively, these findings reveal an emerging picture of the role of AP-5 in endosomal and lysosomal homeostasis, illuminates a potential pathomechanism that is relevant to the role of AP-5 in neurons and expands the understanding of recessive HSPs. Moreover, the resulting accumulation of storage material in endolysosomes leads us to propose that AP-5 deficiency represents a new type of LSDs.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Endossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/metabolismo , Lisossomos/metabolismo , Mutação , Idoso , Feminino , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Técnicas de Silenciamento de Genes , Estudos de Associação Genética , Células HeLa , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Interferência de RNA
9.
J Cell Sci ; 128(14): 2520-8, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26002056

RESUMO

Intracellular amyloid-ß (Aß) accumulation is a key feature of early Alzheimer's disease and precedes the appearance of Aß in extracellular plaques. Aß is generated through proteolytic processing of amyloid precursor protein (APP), but the intracellular site of Aß production is unclear. APP has been localized to multivesicular bodies (MVBs) where sorting of APP onto intraluminal vesicles (ILVs) could promote amyloidogenic processing, or reduce Aß production or accumulation by sorting APP and processing products to lysosomes for degradation. Here, we show that APP localizes to the ILVs of a subset of MVBs that also traffic EGF receptor (EGFR), and that it is delivered to lysosomes for degradation. Depletion of the endosomal sorting complexes required for transport (ESCRT) components, Hrs (also known as Hgs) or Tsg101, inhibited targeting of APP to ILVs and the subsequent delivery to lysosomes, and led to increased intracellular Aß accumulation. This was accompanied by dramatically decreased Aß secretion. Thus, the early ESCRT machinery has a dual role in limiting intracellular Aß accumulation through targeting of APP and processing products to the lysosome for degradation, and promoting Aß secretion.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular Tumoral , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Endossomos/genética , Endossomos/ultraestrutura , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Lisossomos/genética
10.
BMC Biol ; 14: 46, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27296830

RESUMO

Exosomes are extracellular vesicles first described as such 30 years ago and since implicated in cell-cell communication and the transmission of disease states, and explored as a means of drug discovery. Yet fundamental questions about their biology remain unanswered. Here I explore what exosomes are, highlight the difficulties in studying them and explain the current definition and some of the outstanding issues in exosome biology.


Assuntos
Exossomos/metabolismo , Animais , Biomarcadores/metabolismo , Doença , Exossomos/ultraestrutura , Humanos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA