Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 589(7842): 456-461, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33328639

RESUMEN

Autophagy, a process of degradation that occurs via the lysosomal pathway, has an essential role in multiple aspects of immunity, including immune system development, regulation of innate and adaptive immune and inflammatory responses, selective degradation of intracellular microorganisms, and host protection against infectious diseases1,2. Autophagy is known to be induced by stimuli such as nutrient deprivation and suppression of mTOR, but little is known about how autophagosomal biogenesis is initiated in mammalian cells in response to viral infection. Here, using genome-wide short interfering RNA screens, we find that the endosomal protein sorting nexin 5 (SNX5)3,4 is essential for virus-induced, but not for basal, stress- or endosome-induced, autophagy. We show that SNX5 deletion increases cellular susceptibility to viral infection in vitro, and that Snx5 knockout in mice enhances lethality after infection with several human viruses. Mechanistically, SNX5 interacts with beclin 1 and ATG14-containing class III phosphatidylinositol-3-kinase (PI3KC3) complex 1 (PI3KC3-C1), increases the lipid kinase activity of purified PI3KC3-C1, and is required for endosomal generation of phosphatidylinositol-3-phosphate (PtdIns(3)P) and recruitment of the PtdIns(3)P-binding protein WIPI2 to virion-containing endosomes. These findings identify a context- and organelle-specific mechanism-SNX5-dependent PI3KC3-C1 activation at endosomes-for initiation of autophagy during viral infection.


Asunto(s)
Autofagia/inmunología , Nexinas de Clasificación/metabolismo , Virus/inmunología , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/metabolismo , Línea Celular , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endosomas/metabolismo , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Nexinas de Clasificación/deficiencia , Nexinas de Clasificación/genética , Proteínas de Transporte Vesicular/metabolismo
2.
Traffic ; 24(6): 234-250, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37089068

RESUMEN

Several functions of the human cell, such as sensing nutrients, cell movement and interaction with the surrounding environment, depend on a myriad of transmembrane proteins and their associated proteins and lipids (collectively termed "cargoes"). To successfully perform their tasks, cargo must be sorted and delivered to the right place, at the right time, and in the right amount. To achieve this, eukaryotic cells have evolved a highly organized sorting platform, the endosomal network. Here, a variety of specialized multiprotein complexes sort cargo into itineraries leading to either their degradation or their recycling to various organelles for further rounds of reuse. A key sorting complex is the Endosomal SNX-BAR Sorting Complex for Promoting Exit (ESCPE-1) that promotes the recycling of an array of cargos to the plasma membrane and/or the trans-Golgi network. ESCPE-1 recognizes a hydrophobic-based sorting motif in numerous cargoes and orchestrates their packaging into tubular carriers that pinch off from the endosome and travel to the target organelle. A wide range of pathogens mimic this sorting motif to hijack ESCPE-1 transport to promote their invasion and survival within infected cells. In other instances, ESCPE-1 exerts restrictive functions against pathogens by limiting their replication and infection. In this review, we discuss ESCPE-1 assembly and functions, with a particular focus on recent advances in the understanding of its role in membrane trafficking, cellular homeostasis and host-pathogen interaction.


Asunto(s)
Endosomas , Interacciones Huésped-Patógeno , Complejos Multiproteicos , Receptores de Superficie Celular , Nexinas de Clasificación , Humanos , Nexinas de Clasificación/metabolismo , Endosomas/metabolismo , Complejos Multiproteicos/metabolismo , Red trans-Golgi/metabolismo , Salmonella typhimurium/metabolismo , Chlamydia trachomatis/metabolismo , Virus/metabolismo , Receptores de Superficie Celular/metabolismo , Transporte de Proteínas
3.
PLoS Biol ; 20(4): e3001601, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35417450

RESUMEN

Coat complexes coordinate cargo recognition through cargo adaptors with biogenesis of transport carriers during integral membrane protein trafficking. Here, we combine biochemical, structural, and cellular analyses to establish the mechanistic basis through which SNX27-Retromer, a major endosomal cargo adaptor, couples to the membrane remodeling endosomal SNX-BAR sorting complex for promoting exit 1 (ESCPE-1). In showing that the SNX27 FERM (4.1/ezrin/radixin/moesin) domain directly binds acidic-Asp-Leu-Phe (aDLF) motifs in the SNX1/SNX2 subunits of ESCPE-1, we propose a handover model where SNX27-Retromer captured cargo proteins are transferred into ESCPE-1 transport carriers to promote endosome-to-plasma membrane recycling. By revealing that assembly of the SNX27:Retromer:ESCPE-1 coat evolved in a stepwise manner during early metazoan evolution, likely reflecting the increasing complexity of endosome-to-plasma membrane recycling from the ancestral opisthokont to modern animals, we provide further evidence of the functional diversification of yeast pentameric Retromer in the recycling of hundreds of integral membrane proteins in metazoans.


Asunto(s)
Endosomas , Nexinas de Clasificación , Animales , Membrana Celular/metabolismo , Endosomas/metabolismo , Transporte de Proteínas , Nexinas de Clasificación/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(25): e2201980119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696571

RESUMEN

Endosomal sorting maintains cellular homeostasis by recycling transmembrane proteins and associated proteins and lipids (termed "cargoes") from the endosomal network to multiple subcellular destinations, including retrograde traffic to the trans-Golgi network (TGN). Viral and bacterial pathogens subvert retrograde trafficking machinery to facilitate infectivity. Here, we develop a proteomic screen to identify retrograde cargo proteins of the endosomal SNX-BAR sorting complex promoting exit 1 (ESCPE-1). Using this methodology, we identify Neuropilin-1 (NRP1), a recently characterized host factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as a cargo directly bound and trafficked by ESCPE-1. ESCPE-1 mediates retrograde trafficking of engineered nanoparticles functionalized with the NRP1-interacting peptide of the SARS-CoV-2 spike (S) protein. CRISPR-Cas9 deletion of ESCPE-1 subunits reduces SARS-CoV-2 infection levels in cell culture. ESCPE-1 sorting of NRP1 may therefore play a role in the intracellular membrane trafficking of NRP1-interacting viruses such as SARS-CoV-2.


Asunto(s)
COVID-19 , Endosomas , Interacciones Huésped-Patógeno , Neuropilina-1 , SARS-CoV-2 , COVID-19/metabolismo , COVID-19/virología , Sistemas CRISPR-Cas , Endosomas/virología , Eliminación de Gen , Humanos , Nanopartículas , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteómica , SARS-CoV-2/metabolismo , Nexinas de Clasificación/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
5.
J Cell Sci ; 133(15)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32747499

RESUMEN

Human retromer, a heterotrimer of VPS26 (VPS26A or VPS26B), VPS35 and VPS29, orchestrates the endosomal retrieval of internalised cargo and promotes their cell surface recycling, a prototypical cargo being the glucose transporter GLUT1 (also known as SLC2A1). The role of retromer in the retrograde sorting of the cation-independent mannose 6-phosphate receptor (CI-MPR, also known as IGF2R) from endosomes back to the trans-Golgi network remains controversial. Here, by applying knocksideways technology, we develop a method for acute retromer inactivation. While retromer knocksideways in HeLa and H4 human neuroglioma cells resulted in time-resolved defects in cell surface sorting of GLUT1, we failed to observe a quantifiable defect in CI-MPR sorting. In contrast, knocksideways of the ESCPE-1 complex - a key regulator of retrograde CI-MPR sorting - revealed time-resolved defects in CI-MPR sorting. Together, these data are consistent with a comparatively limited role for retromer in ESCPE-1-mediated CI-MPR retrograde sorting, and establish a methodology for acute retromer and ESCPE-1 inactivation that will aid the time-resolved dissection of their functional roles in endosomal cargo sorting.


Asunto(s)
Nexinas de Clasificación , Proteínas de Transporte Vesicular , Endosomas/metabolismo , Células HeLa , Humanos , Transporte de Proteínas , Nexinas de Clasificación/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Red trans-Golgi/metabolismo
6.
J Cell Sci ; 133(14)2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32513819

RESUMEN

The sorting nexins (SNXs) are a family of peripheral membrane proteins that direct protein trafficking decisions within the endocytic network. Emerging evidence in yeast and mammalian cells implicates a subgroup of SNXs in selective and non-selective forms of autophagy. Using siRNA and CRISPR-Cas9, we demonstrate that the SNX-BAR protein SNX4 is needed for efficient LC3 (also known as MAP1LC3) lipidation and autophagosome assembly in mammalian cells. SNX-BARs exist as homo- and hetero-dimers, and we show that SNX4 forms functional heterodimers with either SNX7 or SNX30 that associate with tubulovesicular endocytic membranes. Detailed image-based analysis during the early stages of autophagosome assembly reveals that SNX4-SNX7 is an autophagy-specific SNX-BAR heterodimer, required for efficient recruitment and/or retention of core autophagy regulators at the nascent isolation membrane. SNX4 partially colocalises with juxtanuclear ATG9A-positive membranes, with our data linking the autophagy defect upon SNX4 disruption to the mis-trafficking and/or retention of ATG9A in the Golgi region. Taken together, our findings show that the SNX4-SNX7 heterodimer coordinates ATG9A trafficking within the endocytic network to establish productive autophagosome assembly sites, thus extending knowledge of SNXs as positive regulators of autophagy.


Asunto(s)
Autofagosomas , Endosomas , Nexinas de Clasificación , Animales , Autofagosomas/metabolismo , Autofagia , Endosomas/metabolismo , Transporte de Proteínas , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo
7.
bioRxiv ; 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37503045

RESUMEN

T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3 and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and a biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.

8.
Cells ; 12(21)2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37947636

RESUMEN

T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to a persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3, and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct, with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.


Asunto(s)
Neoplasias , Linfocitos T , Ratones , Animales , Antígeno CTLA-4/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias/metabolismo , Inmunoterapia
9.
Science ; 370(6518): 861-865, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33082294

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), uses the viral spike (S) protein for host cell attachment and entry. The host protease furin cleaves the full-length precursor S glycoprotein into two associated polypeptides: S1 and S2. Cleavage of S generates a polybasic Arg-Arg-Ala-Arg carboxyl-terminal sequence on S1, which conforms to a C-end rule (CendR) motif that binds to cell surface neuropilin-1 (NRP1) and NRP2 receptors. We used x-ray crystallography and biochemical approaches to show that the S1 CendR motif directly bound NRP1. Blocking this interaction by RNA interference or selective inhibitors reduced SARS-CoV-2 entry and infectivity in cell culture. NRP1 thus serves as a host factor for SARS-CoV-2 infection and may potentially provide a therapeutic target for COVID-19.


Asunto(s)
Betacoronavirus/fisiología , Neuropilina-1/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , COVID-19 , Células CACO-2 , Infecciones por Coronavirus/virología , Cristalografía por Rayos X , Furina/metabolismo , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/química , Neuropilina-1/genética , Pandemias , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
10.
Curr Opin Cell Biol ; 56: 22-33, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30227382

RESUMEN

Endosomes constitute major sorting compartments within the cell. There, a myriad of transmembrane proteins (cargoes) are delivered to the lysosome for degradation or retrieved from this fate and recycled through tubulo-vesicular transport carriers to different cellular destinations. Retrieval and recycling are orchestrated by multi-protein assemblies that include retromer and retriever, sorting nexins, and the Arp2/3 activating WASH complex. Fine-tuned control of actin polymerization on endosomes is fundamental for the retrieval and recycling of cargoes. Recent advances in the field have highlighted several roles that actin plays in this process including the binding to cargoes, stabilization of endosomal subdomains, generation of the remodeling forces required for the biogenesis of cargo-enriched transport carriers and short-range motility of the transport carriers.


Asunto(s)
Actinas/metabolismo , Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Proteínas Portadoras/metabolismo , Cortactina/metabolismo , Humanos , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Transporte de Proteínas , Proteínas de Transporte Vesicular/metabolismo , Proteína del Síndrome de Wiskott-Aldrich
11.
Nat Cell Biol ; 21(10): 1219-1233, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31576058

RESUMEN

Protein trafficking requires coat complexes that couple recognition of sorting motifs in transmembrane cargoes with biogenesis of transport carriers. The mechanisms of cargo transport through the endosomal network are poorly understood. Here, we identify a sorting motif for endosomal recycling of cargoes, including the cation-independent mannose-6-phosphate receptor and semaphorin 4C, by the membrane tubulating BAR domain-containing sorting nexins SNX5 and SNX6. Crystal structures establish that this motif folds into a ß-hairpin, which binds a site in the SNX5/SNX6 phox homology domains. Over sixty cargoes share this motif and require SNX5/SNX6 for their recycling. These include cargoes involved in neuronal migration and a Drosophila snx6 mutant displays defects in axonal guidance. These studies identify a sorting motif and provide molecular insight into an evolutionary conserved coat complex, the 'Endosomal SNX-BAR sorting complex for promoting exit 1' (ESCPE-1), which couples sorting motif recognition to the BAR-domain-mediated biogenesis of cargo-enriched tubulo-vesicular transport carriers.


Asunto(s)
Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo , Secuencias de Aminoácidos/genética , Animales , Drosophila melanogaster , Células HEK293 , Células HeLa , Humanos , Dominios Proteicos/genética , Transporte de Proteínas/fisiología , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Nexinas de Clasificación/genética
12.
Curr Biol ; 28(23): R1350-R1352, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30513333

RESUMEN

Retromer is a master regulator of endosomal cargo sorting. Using cryo-EM, a new study now reveals how, in yeast, this multiprotein complex is assembled on tubular membranes to form a coat complex that orchestrates the process of tubular-based cargo sorting.


Asunto(s)
Tomografía con Microscopio Electrónico , Nexinas de Clasificación , Endosomas , Transporte de Proteínas , Proteínas de Transporte Vesicular
13.
J Cell Biol ; 216(11): 3695-3712, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28935633

RESUMEN

Endosomal recycling of transmembrane proteins requires sequence-dependent recognition of motifs present within their intracellular cytosolic domains. In this study, we have reexamined the role of retromer in the sequence-dependent endosome-to-trans-Golgi network (TGN) transport of the cation-independent mannose 6-phosphate receptor (CI-MPR). Although the knockdown or knockout of retromer does not perturb CI-MPR transport, the targeting of the retromer-linked sorting nexin (SNX)-Bin, Amphiphysin, and Rvs (BAR) proteins leads to a pronounced defect in CI-MPR endosome-to-TGN transport. The retromer-linked SNX-BAR proteins comprise heterodimeric combinations of SNX1 or SNX2 with SNX5 or SNX6 and serve to regulate the biogenesis of tubular endosomal sorting profiles. We establish that SNX5 and SNX6 associate with the CI-MPR through recognition of a specific WLM endosome-to-TGN sorting motif. From validating the CI-MPR dependency of SNX1/2-SNX5/6 tubular profile formation, we provide a mechanism for coupling sequence-dependent cargo recognition with the biogenesis of tubular profiles required for endosome-to-TGN transport. Therefore, the data presented in this study reappraise retromer's role in CI-MPR transport.


Asunto(s)
Receptor IGF Tipo 2/metabolismo , Nexinas de Clasificación/metabolismo , Sistemas CRISPR-Cas , Endosomas/metabolismo , Células HeLa , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/genética , Epitelio Pigmentado de la Retina/metabolismo , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Factores de Tiempo , Transfección , Red trans-Golgi/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA