Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biophys Chem ; 310: 107255, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38728808

RESUMEN

In solution NMR, chemical shift perturbation (CSP) experiments are widely employed to study intermolecular interactions. However, excluding the nonsignificant peak shift is difficult because little is known about errors in CSP. Here, to address this issue, we introduce a method for estimating errors in CSP based on the noise level. First, we developed a technique that involves line shape fitting to estimate errors in peak position via Monte Carlo simulations. Second, this technique was applied to estimate errors in CSP. In intermolecular interaction analysis of VAP-A with SNX2, error estimation of CSP enabled the evaluation of small but significant changes in peak position and yielded detailed insights that are unattainable with conventional CSP analysis. Third, this technique was successfully applied to estimate errors in residual dipolar couplings. In conclusion, our error estimation method improves CSP analysis by excluding the nonsignificant peak shift.


Asunto(s)
Método de Montecarlo , Nexinas de Clasificación/química , Resonancia Magnética Nuclear Biomolecular , Espectroscopía de Resonancia Magnética/métodos
2.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(6): 1107-1111, 2022 Dec.
Artículo en Chino | MEDLINE | ID: mdl-36373636

RESUMEN

Sorting nexin 16(SNX16),a member of the SNX family,contains a phoxhomology domain that is prone to bind with phosphatidylinositol-3-phosphate domain and a C-terminal coiled-coil domain. SNX16 participates in diverse cellular processes such as endocytosis,protein sorting,and signal transduction. The dysfunctions of SNX16 are demonstrated to be involved in the occurrence of several diseases.Here,we review the structural characteristics and biological functions of SNX16 and discuss the regulatory role of SNX16 in diseases,surveying how SNX16 can be applied to the prevention and treatment of related disorders.


Asunto(s)
Endosomas , Nexinas de Clasificación , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo , Endosomas/metabolismo , Transporte de Proteínas , Transducción de Señal
3.
Structure ; 30(12): 1590-1602.e6, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36302387

RESUMEN

The sorting nexin SNX17 controls endosomal recycling of transmembrane cargo proteins including integrins, the amyloid precursor protein, and lipoprotein receptors. This requires association with the Commander trafficking complex and depends on the C terminus of SNX17 through unknown mechanisms. Using proteomics, we find that the SNX17 C terminus is sufficient for Commander interaction and also associates with members of the PDZ and LIM domain (PDLIM) family. SNX17 contains a type III PDZ binding motif that binds specifically to the PDLIM proteins. The structure of the PDLIM7 PDZ domain bound to the SNX17 C terminus reveals an unconventional perpendicular peptide interaction mediated by electrostatic contacts and a uniquely conserved proline-containing loop sequence in the PDLIM protein family. Our results define the mechanism of SNX17-PDLIM interaction and suggest that the PDLIM proteins may play a role in regulating the activity of SNX17 in conjunction with Commander and actin-rich endosomal trafficking domains.


Asunto(s)
Proteómica , Nexinas de Clasificación , Nexinas de Clasificación/química , Unión Proteica , Secuencia de Aminoácidos , Endosomas/metabolismo
4.
J Mol Biol ; 434(21): 167823, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36103920

RESUMEN

As a subgroup of sorting nexins (SNXs) that contain regulator of G protein signaling homology (RH) domain, SNX-RH proteins, including SNX13, SNX14 and SNX25, were proposed to play bifunctional roles in protein sorting and GPCR signaling regulation. However, mechanistic details of SNX-RH proteins functioning via RH domain remain to be illustrated. Here, we delineate crystal structures of the RH domains of SNX13 and SNX25, revealing a homodimer of SNX13 RH domain mediated by unique extended α4 and α5 helices, and a thiol modulated homodimer of SNX25-RH triggered by a unique cysteine on α6 helix. Further studies showed that RH domains of SNX-RH do not possess binding capacity toward Gα subunits, owing to the lack of critical residues for interaction. Thus, this study identifies a group of novel non-canonical RH domains that can act as a dimerization module in sorting nexins, which provides structural basis for mechanism studies on SNX-RH protein functions.


Asunto(s)
Proteínas de Unión al GTP , Nexinas de Clasificación , Proteínas de Unión al GTP/metabolismo , Transporte de Proteínas , Transducción de Señal , Nexinas de Clasificación/química , Humanos , Multimerización de Proteína , Dominios Proteicos , Compuestos de Sulfhidrilo/química
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35022217

RESUMEN

After binding to its cell surface receptor angiotensin converting enzyme 2 (ACE2), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters the host cell through directly fusing with plasma membrane (cell surface pathway) or undergoing endocytosis traveling to lysosome/late endosome for membrane fusion (endocytic pathway). However, the endocytic entry regulation by host cell remains elusive. Recent studies show ACE2 possesses a type I PDZ binding motif (PBM) through which it could interact with a PDZ domain-containing protein such as sorting nexin 27 (SNX27). In this study, we determined the ACE2-PBM/SNX27-PDZ complex structure, and, through a series of functional analyses, we found SNX27 plays an important role in regulating the homeostasis of ACE2 receptor. More importantly, we demonstrated SNX27, together with retromer complex (the core component of the endosomal protein sorting machinery), prevents ACE2/virus complex from entering lysosome/late endosome, resulting in decreased viral entry in cells where the endocytic pathway dominates. The ACE2/virus retrieval mediated by SNX27-retromer could be considered as a countermeasure against invasion of ACE2 receptor-using SARS coronaviruses.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Endosomas/metabolismo , SARS-CoV-2 , Nexinas de Clasificación/química , COVID-19/virología , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Cristalografía por Rayos X , Citosol/metabolismo , Endocitosis , Perfilación de la Expresión Génica , Células HEK293 , Células HeLa , Homeostasis , Humanos , Lentivirus , Lisosomas/metabolismo , Péptidos/química , Unión Proteica , Conformación Proteica , Dominios Proteicos , Nexinas de Clasificación/metabolismo , Internalización del Virus
6.
J Med Chem ; 65(1): 386-408, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34982557

RESUMEN

The serine protease inhibitor Rv3364c of Mycobacterium tuberculosis (MTB) is highly expressed in cells during MTB exposure. In this study, we showed that the 12WLVSKF17 motif of Rv3364c interacts with the BAR domain of SNX9 and inhibits endosome trafficking to interact with p47phox, thereby suppressing TLR4 inflammatory signaling in macrophages. Derived from the structure of this Rv3364c peptide motif, 2,4-diamino-6-(4-tert-butylphenyl)-1,3,5-trazine, DATPT as a 12WLVSKF17 peptide-mimetic small molecule has been identified. DATPT can block the SNX9-p47phox interaction in the endosome and suppress reactive oxygen species and inflammatory cytokine production; it demonstrated significant therapeutic effects in a mouse model of cecal ligation and puncture-induced sepsis. DATPT has considerably improved potency, with an IC50 500-fold (in vitro) or 2000-fold (in vivo) lower than that of the 12WLVSKF17 peptide. Furthermore, DATPT shows potent antibacterial activities by reduction in ATP production and leakage of intracellular ATP out of bacteria. These results provide evidence for peptide-derived small molecule DATPT with anti-inflammatory and antibacterial functions for the treatment of sepsis.


Asunto(s)
Antibacterianos/farmacología , Mycobacterium tuberculosis/química , Sepsis/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas , Nexinas de Clasificación/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Antibacterianos/química , Citocinas/antagonistas & inhibidores , Endosomas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Noqueados , Fragmentos de Péptidos/efectos de los fármacos , Especies Reactivas de Oxígeno , Sepsis/microbiología , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Transducción de Señal/efectos de los fármacos , Nexinas de Clasificación/química
7.
Mol Biol Rep ; 49(1): 497-510, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34817777

RESUMEN

BACKGROUND: Replication of the influenza A viruses occurs in the cells through the viral RdRP consisting of PB1, PB2, and PA. Several cellular proteins are involved in these processes. This study aims to reveal the interaction between human SNX2 protein and the PA protein and the effects of the SNX2 on the virus replication. RESULTS: To identify potential host interacting proteins to the PA, yeast two-hybrid assay was carried out with HEK293 cell cDNA library and the PA as a bait. We focused on SNX2 protein, which interacts with the PA in the yeast cells. By using the co-immunoprecipitation assays, it has been demonstrated that the amino-terminal part of the PA was important for binding to the SNX2. Immunolocalization of the proteins in HeLa cells supported this interaction. Knockdown of the SNX2 with siRNA in the cells resulted in a significant increase in both viral transcripts and virus growth. However, the increase of SNX2 in transfected cells didn't cause a significant change in the viral RdRP activity in minireplicon assay. This may suggest that the negative effect of SNX2 on the virus replication could be saturated with its authentic intra-cellular amount. CONCLUSIONS: This study revealed that the SNX2 and PA protein interact with each other in both yeast and HEK293 cells, and the SNX2 has a negative regulatory function on the virus replication. However, more knowledge is required to elucidate the action mechanism of the SNX2 on the influenza A virus replication at the molecular level.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/virología , ARN Polimerasa Dependiente del ARN/metabolismo , Nexinas de Clasificación/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Células Cultivadas , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Nexinas de Clasificación/química , Técnicas del Sistema de Dos Híbridos
8.
J Cell Biol ; 221(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-34936700

RESUMEN

We report here two genome-wide CRISPR screens performed to identify genes that, when knocked out, alter levels of lysosomal cholesterol or bis(monoacylglycero)phosphate. In addition, these screens were also performed under conditions of NPC1 inhibition to identify modifiers of NPC1 function in lysosomal cholesterol export. The screens confirm tight coregulation of cholesterol and bis(monoacylglycero)phosphate in cells and reveal an unexpected role for the ER-localized SNX13 protein as a negative regulator of lysosomal cholesterol export and contributor to ER-lysosome membrane contact sites. In the absence of NPC1 function, SNX13 knockdown redistributes lysosomal cholesterol and is accompanied by triacylglycerol-rich lipid droplet accumulation and increased lysosomal bis(monoacylglycero)phosphate. These experiments provide unexpected insight into the regulation of lysosomal lipids and modification of these processes by novel gene products.


Asunto(s)
Sistemas CRISPR-Cas/genética , Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Pruebas Genéticas , Lípidos/química , Lisosomas/metabolismo , Transporte Biológico , Endosomas/metabolismo , Genoma , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células K562 , Dominios Proteicos , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo
9.
Adv Biol Regul ; 83: 100842, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34866035

RESUMEN

Metazoans require the sorting nexin (SNX) protein, SNX27, to recycle hundreds of important transmembrane protein receptors from endosomes to the plasma membrane. Cargo recycling by SNX27 requires its interaction with retromer, a heterotrimer known to assemble on membranes with multiple sorting nexins, including SNX-BAR proteins and SNX3. SNX27 has also been functionally linked to SNX-BARs, but the molecular basis of this interaction has been unknown. We identify a direct biochemical interaction between the conserved and flexible SNX1/SNX2 N-terminus and full-length SNX27 using purified proteins in pulldown experiments. Sequence alignments indicate both SNX1 and SNX2 contain two short and conserved stretches of acidic residues bearing a DxF motif in their flexible N-terminal regions. Biochemical pulldown and mapping experiments reveal forty residues in the N-terminus of either SNX1 or SNX2 can mediate binding to SNX27. SNX27 truncation analysis demonstrates the SNX27 FERM domain binds the SNX1 N-terminus. Calorimetry experiments quantified binding between the SNX1 N-terminus and SNX27 in the low micromolar affinity range (KD ∼10 µM) and suggest the second DxF motif may play a more prominent role in binding. Mutation of either DxF sequence in SNX1 abrogates measurable binding to SNX27 in the calorimeter. Modelling from both predicted and experimentally determined structures suggests the SNX27 FERM domain could accommodate both DxF motifs simultaneously. Together, these data suggest SNX27 is directly linked to specific SNX-BAR proteins through binding acidic motifs in the SNX1 or SNX2 N-terminus.


Asunto(s)
Endosomas , Nexinas de Clasificación , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Endosomas/metabolismo , Humanos , Transporte de Proteínas , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo
10.
PLoS Pathog ; 17(11): e1009409, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34843601

RESUMEN

The HIV-1 accessory protein Vpu modulates membrane protein trafficking and degradation to provide evasion of immune surveillance. Targets of Vpu include CD4, HLAs, and BST-2. Several cellular pathways co-opted by Vpu have been identified, but the picture of Vpu's itinerary and activities within membrane systems remains incomplete. Here, we used fusion proteins of Vpu and the enzyme ascorbate peroxidase (APEX2) to compare the ultrastructural locations and the proximal proteomes of wild type Vpu and Vpu-mutants. The proximity-omes of the proteins correlated with their ultrastructural locations and placed wild type Vpu near both retromer and ESCRT-0 complexes. Hierarchical clustering of protein abundances across the mutants was essential to interpreting the data and identified Vpu degradation-targets including CD4, HLA-C, and SEC12 as well as Vpu-cofactors including HGS, STAM, clathrin, and PTPN23, an ALIX-like protein. The Vpu-directed degradation of BST-2 was supported by STAM and PTPN23 and to a much lesser extent by the retromer subunits Vps35 and SNX3. PTPN23 also supported the Vpu-directed decrease in CD4 at the cell surface. These data suggest that Vpu directs targets from sorting endosomes to degradation at multi-vesicular bodies via ESCRT-0 and PTPN23.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Infecciones por VIH/virología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteoma/metabolismo , Nexinas de Clasificación/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Viroporinas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/fisiología , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Microscopía Electrónica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transporte de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteoma/análisis , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Viroporinas/genética
11.
Viruses ; 13(11)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34835087

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a main receptor for SARS-CoV-2 entry to the host cell. Indeed, the first step in viral entry is the binding of the viral trimeric spike (S) protein to ACE2. Abundantly present in human epithelial cells of many organs, ACE2 is also expressed in the human brain. ACE2 is a type I membrane protein with an extracellular N-terminal peptidase domain and a C-terminal collectrin-like domain that ends with a single transmembrane helix and an intracellular 44-residue segment. This C-terminal segment contains a PDZ-binding motif (PBM) targeting protein-interacting domains called PSD-95/Dlg/ZO-1 (PDZ). Here, we identified the human PDZ specificity profile of the ACE2 PBM using the high-throughput holdup assay and measuring the binding intensities of the PBM of ACE2 against the full human PDZome. We discovered 14 human PDZ binders of ACE2 showing significant binding with dissociation constants' values ranging from 3 to 81 µM. NHERF, SHANK, and SNX27 proteins found in this study are involved in protein trafficking. The PDZ/PBM interactions with ACE2 could play a role in ACE2 internalization and recycling that could be of benefit for the virus entry. Interestingly, most of the ACE2 partners we identified are expressed in neuronal cells, such as SHANK and MAST families, and modifications of the interactions between ACE2 and these neuronal proteins may be involved in the neurological symptoms of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Dominios PDZ , Proteínas/química , Proteínas/metabolismo , Receptores de Coronavirus/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34462354

RESUMEN

The molecular events that determine the recycling versus degradation fates of internalized membrane proteins remain poorly understood. Two of the three members of the SNX-FERM family, SNX17 and SNX31, utilize their FERM domain to mediate endocytic trafficking of cargo proteins harboring the NPxY/NxxY motif. In contrast, SNX27 does not recycle NPxY/NxxY-containing cargo but instead recycles cargo containing PDZ-binding motifs via its PDZ domain. The underlying mechanism governing this divergence in FERM domain binding is poorly understood. Here, we report that the FERM domain of SNX27 is functionally distinct from SNX17 and interacts with a novel DLF motif localized within the N terminus of SNX1/2 instead of the NPxY/NxxY motif in cargo proteins. The SNX27-FERM-SNX1 complex structure reveals that the DLF motif of SNX1 binds to a hydrophobic cave surrounded by positively charged residues on the surface of SNX27. The interaction between SNX27 and SNX1/2 is critical for efficient SNX27 recruitment to endosomes and endocytic recycling of multiple cargoes. Finally, we show that the interaction between SNX27 and SNX1/2 is critical for brain development in zebrafish. Altogether, our study solves a long-standing puzzle in the field and suggests that SNX27 and SNX17 mediate endocytic recycling through fundamentally distinct mechanisms.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Dominios FERM , Nexinas de Clasificación/metabolismo , Animales , Encéfalo/metabolismo , Endocitosis , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Neuronas/citología , Unión Proteica , Transporte de Proteínas , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Nexinas de Clasificación/química , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
13.
Nat Commun ; 12(1): 4552, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315878

RESUMEN

The ability of endolysosomal organelles to move within the cytoplasm is essential for the performance of their functions. Long-range movement involves coupling of the endolysosomes to motor proteins that carry them along microtubule tracks. This movement is influenced by interactions with other organelles, but the mechanisms involved are incompletely understood. Herein we show that the sorting nexin SNX19 tethers endolysosomes to the endoplasmic reticulum (ER), decreasing their motility and contributing to their concentration in the perinuclear area of the cell. Tethering depends on two N-terminal transmembrane domains that anchor SNX19 to the ER, and a PX domain that binds to phosphatidylinositol 3-phosphate on the endolysosomal membrane. Two other domains named PXA and PXC negatively regulate the interaction of SNX19 with endolysosomes. These studies thus identify a mechanism for controlling the motility and positioning of endolysosomes that involves tethering to the ER by a sorting nexin.


Asunto(s)
Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Nexinas de Clasificación/metabolismo , Línea Celular Tumoral , Retículo Endoplásmico/ultraestructura , Endosomas/ultraestructura , Humanos , Lisosomas/ultraestructura , Fosfatos de Fosfatidilinositol/metabolismo , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , Nexinas de Clasificación/química
14.
Autophagy ; 17(11): 3547-3565, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33678121

RESUMEN

Autophagy, in part, is controlled by the repression and activation of autophagy-related (ATG) genes. Here, we describe a new selective autophagy pathway that targets functional transcriptional regulators to control their activity. This pathway is activated in response to nitrogen starvation and recycles transcriptional activators (Msn2 and Rim15) and a repressor (Ssn2/Med13) of ATG expression. Further analysis of Ssn2/Med13 vacuolar proteolysis revealed that this pathway utilizes the core autophagic machinery. However, it is independent of known nucleophagy mechanisms, receptor proteins, and the scaffold protein Atg11. Instead, Ssn2/Med13 exits the nucleus through the nuclear pore complex (NPC) and associates with the cytoplasmic nucleoporin Gle1, a member of the RNA remodeling complex. Dbp5 and Nup159, that act in concert with Gle1, are also required for Ssn2/Med13 clearance. Ssn2/Med13 is retrieved from the nuclear periphery and degraded by Atg17-initiated phagophores anchored to the vacuole. Efficient transfer to phagophores depends on the sorting nexin heterodimer Snx4/Atg24-Atg20, which binds to Atg17, and relocates to the perinucleus following nitrogen starvation. To conclude, this pathway defines a previously undescribed autophagy mechanism that targets select transcriptional regulators for rapid vacuolar proteolysis, utilizing the RNA remodeling complex, the sorting nexin heterodimer Snx4-Atg20, Atg17, and the core autophagic machinery. It is physiologically relevant as this Snx4-assisted vacuolar targeting pathway permits cells to fine-tune the autophagic response by controlling the turnover of both positive and negative regulators of ATG transcription.Abbreviations: AIM: Atg8 interacting motif; ATG: autophagy-related; CKM: CDK8 kinase module; IDR: intrinsically disordered region; IP6: phosphoinositide inositol hexaphosphate; NPC: nuclear pore complex; PAS: phagophore assembly site; UPS: ubiquitin-proteasomal system.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Nexinas de Clasificación/metabolismo , Factores de Transcripción/metabolismo , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Genes Fúngicos , Complejo Mediador/química , Complejo Mediador/genética , Complejo Mediador/metabolismo , Modelos Biológicos , Nitrógeno/metabolismo , Poro Nuclear/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteolisis , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Vacuolas/metabolismo
15.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658379

RESUMEN

The sorting nexin (SNX) family of proteins deform the membrane to generate transport carriers in endosomal pathways. Here, we elucidate how a prototypic member, SNX1, acts in this process. Performing cryoelectron microscopy, we find that SNX1 assembles into a protein lattice that consists of helical rows of SNX1 dimers wrapped around tubular membranes in a crosslinked fashion. We also visualize the details of this structure, which provides a molecular understanding of how various parts of SNX1 contribute to its ability to deform the membrane. Moreover, we have compared the SNX1 structure with a previously elucidated structure of an endosomal coat complex formed by retromer coupled to a SNX, which reveals how the molecular organization of the SNX in this coat complex is affected by retromer. The comparison also suggests insight into intermediary stages of assembly that results in the formation of the retromer-SNX coat complex on the membrane.


Asunto(s)
Membrana Celular/metabolismo , Multimerización de Proteína , Nexinas de Clasificación/metabolismo , Animales , Membrana Celular/química , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Ratones , Estructura Cuaternaria de Proteína , Nexinas de Clasificación/química , Nexinas de Clasificación/ultraestructura
16.
J Cell Physiol ; 236(6): 4207-4215, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33241559

RESUMEN

SNX10 is a member of the phox homology domain-containing family of phosphoinositide-binding proteins. Intracellularly, SNX10 localizes to endosomes where it mediates intracellular trafficking, endosome organization, and protein localization to the centrosome and cilium. It is highly expressed in bone and the gut where it participates in bone mineral and calcium homeostasis through the regulation of osteoclastic bone resorption and gastric acid secretion, respectively. Not surprisingly, patients harboring mutations in SNX10 mutation manifest a phenotype of autosomal recessive osteopetrosis or malignant infantile osteopetrosis, which is clinically characterized by dense bones with increased cortical bone into the medullary space with bone marrow occlusion or depletion, bone marrow failure, and anemia. Accordingly, SNX10 mutant osteoclasts exhibit impaired bone resorptive capacity. Beyond the skeleton, there is emerging evidence implicating SNX10 in cancer development, metabolic disorders, inflammation, and chaperone-mediated autophagy. Understanding the structural basis through which SNX10 exerts its diverse biological functions in both cell and tissue-specific manners may therefore inform new therapeutic opportunities toward the treatment and management of SNX10-related diseases.


Asunto(s)
Endosomas/metabolismo , Neoplasias/metabolismo , Osteopetrosis/metabolismo , Nexinas de Clasificación/metabolismo , Animales , Endosomas/genética , Endosomas/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Mutación , Neoplasias/genética , Neoplasias/patología , Osteopetrosis/genética , Osteopetrosis/patología , Conformación Proteica , Transporte de Proteínas , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Relación Estructura-Actividad
17.
PLoS Pathog ; 16(12): e1009120, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370420

RESUMEN

Positive-strand RNA viruses replicate in host cells by forming large viral replication organelles, which harbor numerous membrane-bound viral replicase complexes (VRCs). In spite of its essential role in viral replication, the biogenesis of the VRCs is not fully understood. The authors identified critical roles of cellular membrane-shaping proteins and PI(3)P (phosphatidylinositol 3-phosphate) phosphoinositide, a minor lipid with key functions in endosomal vesicle trafficking and autophagosome biogenesis, in VRC formation for tomato bushy stunt virus (TBSV). The authors show that TBSV co-opts the endosomal SNX-BAR (sorting nexin with Bin/Amphiphysin/Rvs- BAR domain) proteins, which bind to PI(3)P and have membrane-reshaping function during retromer tubular vesicle formation, directly into the VRCs to boost progeny viral RNA synthesis. We find that the viral replication protein-guided recruitment and pro-viral function of the SNX-BAR proteins depends on enrichment of PI(3)P at the site of viral replication. Depletion of SNX-BAR proteins or PI(3)P renders the viral double-stranded (ds)RNA replication intermediate RNAi-sensitive within the VRCs in the surrogate host yeast and in planta and ribonuclease-sensitive in cell-free replicase reconstitution assays in yeast cell extracts or giant unilamellar vesicles (GUVs). Based on our results, we propose that PI(3)P and the co-opted SNX-BAR proteins are coordinately exploited by tombusviruses to promote VRC formation and to play structural roles and stabilize the VRCs during viral replication. Altogether, the interplay between the co-opted SNX-BAR membrane-shaping proteins, PI(3)P and the viral replication proteins leads to stable VRCs, which provide the essential protection of the viral RNAs against the host antiviral responses.


Asunto(s)
Fosfatos de Fosfatidilinositol/metabolismo , Nexinas de Clasificación/metabolismo , Tombusvirus/fisiología , Proteinas del Complejo de Replicasa Viral/metabolismo , Arabidopsis/metabolismo , Arabidopsis/virología , Células Cultivadas , Interacciones Huésped-Patógeno/genética , Organismos Modificados Genéticamente , Fosfatidilinositoles/metabolismo , Dominios y Motivos de Interacción de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/fisiología , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/virología , Nexinas de Clasificación/química , Nexinas de Clasificación/fisiología , Nicotiana/metabolismo , Nicotiana/virología , Tombusvirus/genética , Tombusvirus/metabolismo , Proteinas del Complejo de Replicasa Viral/fisiología , Replicación Viral/genética
18.
J Biomol NMR ; 74(12): 741-752, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33118136

RESUMEN

Unidirectional coherence transfer is highly efficient in intrinsically disordered proteins (IDPs). Their elevated ps-ns timescale dynamics ensures long transverse (T2) relaxation times allowing sophisticated coherence transfer pathway selection in comparison to folded proteins. 1Hα-detection ensures non-susceptibility to chemical exchange with the solvent and enables chemical shift assignment of consecutive proline residues, typically abundant in IDPs. However, many IDPs undergo a disorder-to-order transition upon interaction with their target protein, which leads to the loss of the favorable relaxation properties. Long coherence transfer routes now result in prohibitively large decrease in sensitivity. We introduce a novel 4D 1Hα-detected experiment HACANCOi, together with its 3D implementation, which warrant high sensitivity for the assignment of proline-rich regions in IDPs in complex with a globular protein. The experiment correlates 1Hαi, 13Cαi, 15Ni and [Formula: see text] spins by transferring the magnetization concomitantly from 13Cαi to 15Ni and [Formula: see text]. The B1 domain of protein G (GB1), and the enteropathogenic E. coli EspF in complex with human SNX9 SH3, serve as model systems to demonstrate the attainable sensitivity and successful sequential assignment.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Resonancia Magnética Nuclear Biomolecular , Humanos , Nexinas de Clasificación/química , Nexinas de Clasificación/metabolismo , Dominios Homologos src
19.
Biochem Soc Trans ; 48(5): 2261-2272, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33125482

RESUMEN

Retromer (VPS26/VPS35/VPS29) is a highly conserved eukaryotic protein complex that localizes to endosomes to sort transmembrane protein cargoes into vesicles and elongated tubules. Retromer mediates retrieval pathways from endosomes to the trans-Golgi network in all eukaryotes and further facilitates recycling pathways to the plasma membrane in metazoans. In cells, retromer engages multiple partners to orchestrate the formation of tubulovesicular structures, including sorting nexin (SNX) proteins, cargo adaptors, GTPases, regulators, and actin remodeling proteins. Retromer-mediated pathways are especially important for sorting cargoes required for neuronal maintenance, which links retromer loss or mutations to multiple human brain diseases and disorders. Structural and biochemical studies have long contributed to the understanding of retromer biology, but recent advances in cryo-electron microscopy and cryo-electron tomography have further uncovered exciting new snapshots of reconstituted retromer structures. These new structures reveal retromer assembles into an arch-shaped scaffold and suggest the scaffold may be flexible and adaptable in cells. Interactions with cargo adaptors, particularly SNXs, likely orient the scaffold with respect to phosphatidylinositol-3-phosphate (PtdIns3P)-enriched membranes. Pharmacological small molecule chaperones have further been shown to stabilize retromer in cultured cell and mouse models, but mechanisms by which these molecules bind remain unknown. This review will emphasize recent structural and biophysical advances in understanding retromer structure as the field moves towards a molecular view of retromer assembly and regulation on membranes.


Asunto(s)
Microscopía por Crioelectrón/métodos , GTP Fosfohidrolasas/química , Aparato de Golgi/metabolismo , Red trans-Golgi/metabolismo , Actinas/metabolismo , Animales , Biofisica , Encéfalo/metabolismo , Tomografía con Microscopio Electrónico , Endosomas/metabolismo , GTP Fosfohidrolasas/metabolismo , Humanos , Fosfatos de Fosfatidilinositol/química , Unión Proteica , Transporte de Proteínas , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Nexinas de Clasificación/química , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo
20.
Protoplasma ; 257(6): 1725-1729, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32780164

RESUMEN

Eukaryotic organisms share many common features in terms of endomembrane trafficking. This fact has helped plant scientists to propose testable hypotheses on how plant intracellular membrane trafficking is achieved and regulated based on knowledge from yeast and mammals. However, when a new compartment has been identified in a plant cell that has a vesicle tethering complex located at a position which is completely different to its counterpart in yeast and mammalian cells, caution is demanded when interpreting possible interactions with other trafficking elements. This is exemplified by the recently discovered EMAC (ER and microtubule-associated compartment). It has been postulated that this compartment is the recipient of vacuolar sorting receptors (VSRs) transported retrogradely via "retromer vesicles" from a post-Golgi location. Unfortunately, this suggestion was based entirely on our knowledge of retromer from yeast and mammalian cells, and did not take into account the available literature on the composition, localization, and function of the plant retromer. It also lacked reference to recent contradictory findings on VSR trafficking. In this short article, we have tried to rectify this situation, pointing out that plant retromer may not function as a pentameric complex of two subunits: the retromer core and the sorting nexins.


Asunto(s)
Transporte Biológico/fisiología , Aparato de Golgi/química , Nexinas de Clasificación/química , Vacuolas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA