Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38991587

RESUMEN

TANGO1, TANGO1-Short, and cTAGE5 form stable complexes at the endoplasmic reticulum exit sites (ERES) to preferably export bulky cargoes. Their C-terminal proline-rich domain (PRD) binds Sec23A and affects COPII assembly. The PRD in TANGO1-Short was replaced with light-responsive domains to control its binding to Sec23A in U2OS cells (human osteosarcoma). TANGO1-ShortΔPRD was dispersed in the ER membrane but relocated rapidly, reversibly, to pre-existing ERES by binding to Sec23A upon light activation. Prolonged binding between the two, concentrated ERES in the juxtanuclear region, blocked cargo export and relocated ERGIC53 into the ER, minimally impacting the Golgi complex organization. Bulky collagen VII and endogenous collagen I were collected at less than 47% of the stalled ERES, whereas small cargo molecules were retained uniformly at almost all the ERES. We suggest that ERES are segregated to handle cargoes based on their size, permitting cells to traffic them simultaneously for optimal secretion.

2.
Cell Rep ; 38(3): 110258, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045300

RESUMEN

The realization that the meticulous organization of cellular organelles is not required for the reconstitution of select intracellular traffic steps has revolutionized cell biology. It transformed the discipline from a morphological one into a molecular one. It helped in defining the activities of COPII and COPI vesicle coats in secretion. The work established the principles of the vesicular traffic model as envisioned by George Palade 50 years ago. However, in recent years, numerous advances in cellular and imaging technologies afforded an unprecedented molecular resolution that sheds new light on COPII activities and biosynthetic traffic between the ER and the Golgi. In the following review, I summarize this new information and attempt to provide a unified physical-molecular-morphological description of this traffic step. This information expands on the simplistic principles of vesicular traffic and provides novel frameworks to examine and explain physiological secretion.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Proteína Coat de Complejo I/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Transporte de Proteínas/fisiología , Animales , Humanos
3.
J Cell Biol ; 220(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33404605
4.
Sci Adv ; 6(14): eaay9572, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32270040

RESUMEN

The endoplasmic reticulum (ER) is a highly dynamic network of membranes. Here, we combine live-cell microscopy with in situ cryo-electron tomography to directly visualize ER dynamics in several secretory cell types including pancreatic ß-cells and neurons under near-native conditions. Using these imaging approaches, we identify a novel, mobile form of ER, ribosome-associated vesicles (RAVs), found primarily in the cell periphery, which is conserved across different cell types and species. We show that RAVs exist as distinct, highly dynamic structures separate from the intact ER reticular architecture that interact with mitochondria via direct intermembrane contacts. These findings describe a new ER subcompartment within cells.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Retículo Endoplásmico/metabolismo , Ribosomas/metabolismo , Animales , Transporte Biológico , Microscopía por Crioelectrón , Vesículas Citoplasmáticas/ultraestructura , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Imagen Molecular , Especificidad de Órganos , Ratas , Ribosomas/ultraestructura , Estrés Fisiológico
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3672-3684, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30251687

RESUMEN

BACKGROUND: The cardiac sodium channel Nav1.5 is essential for the physiological function of the heart and causes cardiac arrhythmias and sudden death when mutated. Many disease-causing mutations in Nav1.5 cause defects in protein trafficking, a cellular process critical to the targeting of Nav1.5 to cell surface. However, the molecular mechanisms underlying the trafficking of Nav1.5, in particular, the exit from the endoplasmic reticulum (ER) for cell surface trafficking, remain poorly understood. METHODS AND RESULTS: Here we investigated the role of the SAR1 GTPases in trafficking of Nav1.5. Overexpression of dominant-negative mutant SAR1A (T39N or H79G) or SAR1B (T39N or H79G) significantly reduces the expression level of Nav1.5 on cell surface, and decreases the peak sodium current density (INa) in HEK/Nav1.5 cells and neonatal rat cardiomyocytes. Simultaneous knockdown of SAR1A and SAR1B expression by siRNAs significantly reduces the INa density, whereas single knockdown of either SAR1A or SAR1B has minimal effect. Computer modeling showed that the three-dimensional structure of SAR1 is similar to RAN. RAN was reported to interact with MOG1, a small protein involved in regulation of the ER exit of Nav1.5. Co-immunoprecipitation showed that SAR1A or SAR1B interacted with MOG1. Interestingly, knockdown of SAR1A and SAR1B expression abolished the MOG1-mediated increases in both cell surface trafficking of Nav1.5 and the density of INa. CONCLUSIONS: These data suggest that SAR1A and SAR1B are the critical regulators of trafficking of Nav1.5. Moreover, SAR1A and SAR1B interact with MOG1, and are required for MOG1-mediated cell surface expression and function of Nav1.5.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Miocitos Cardíacos/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Proteína de Unión al GTP ran/metabolismo , Animales , Animales Recién Nacidos , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/genética , Mutagénesis , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/economía , Técnicas de Placa-Clamp , Cultivo Primario de Células , Transporte de Proteínas/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína de Unión al GTP ran/química
6.
Traffic ; 19(11): 823-839, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29978536

RESUMEN

Our view of the secretory pathway has evolved from a morphological one to one that includes molecular mechanistic understanding of basic traffic components. These components include coat complexes involved in cargo sorting and budding and proteins that mediate targeting, tethering and fusion. The expanding repertoire of regulators that control basic traffic activities begins to paint a unified morphological-molecular view of secretion. The emerging picture provides key insights into the coupling of secretion with physiology. This review examines aspects of morphological-molecular relations that are derived from studies on traffic from the endoplasmic reticulum carried by the coat protein complex II.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Vías Secretoras , Animales , Humanos , Transporte de Proteínas
7.
J Biol Chem ; 291(10): 5206-20, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26740627

RESUMEN

Unesterified cholesterol accumulates in late endosomes in cells expressing the misfolded cystic fibrosis transmembrane conductance regulator (CFTR). CFTR misfolding in the endoplasmic reticulum (ER) or general activation of ER stress led to dynein-mediated clustering of cholesterol-loaded late endosomes at the Golgi region, a process regulated by ER-localized VAMP-associated proteins (VAPs). We hypothesized that VAPs serve as intracellular receptors that couple lipid homeostasis through interactions with two phenylalanines in an acidic track (FFAT) binding signals (found in lipid sorting and sensing proteins, LSS) with proteostasis regulation. VAPB inhibited the degradation of ΔF508-CFTR. The activity was mapped to the ligand-binding major sperm protein (MSP) domain, which was sufficient in regulating CFTR biogenesis. We identified mutations in an unstructured loop within the MSP that uncoupled VAPB-regulated CFTR biogenesis from basic interactions with FFAT. Using this information, we defined functional and physical interactions between VAPB and proteostasis regulators (ligands), including the unfolded protein response sensor ATF6 and the ER degradation cluster that included FAF1, VCP, BAP31, and Derlin-1. VAPB inhibited the degradation of ΔF508-CFTR in the ER through interactions with the RMA1-Derlin-BAP31-VCP pathway. Analysis of pseudoligands containing tandem FFAT signals supports a competitive model for VAP interactions that direct CFTR biogenesis. The results suggest a model in which VAP-ligand binding couples proteostasis and lipid homeostasis leading to observed phenotypes of lipid abnormalities in protein folding diseases.


Asunto(s)
Colesterol/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteolisis , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas , Proteínas Reguladoras de la Apoptosis , Sitios de Unión , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica , Estabilidad Proteica , Ubiquitina-Proteína Ligasas/metabolismo , Proteína que Contiene Valosina
8.
J Cell Sci ; 127(Pt 8): 1765-78, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24522181

RESUMEN

The inner and outer layers of COPII mediate cargo sorting and vesicle biogenesis. Sec16A and p125A (officially known as SEC23IP) proteins interact with both layers to control coat activity, yet the steps directing functional assembly at ER exit sites (ERES) remain undefined. By using temperature blocks, we find that Sec16A is spatially segregated from p125A-COPII-coated ERES prior to ER exit at a step that required p125A. p125A used lipid signals to control ERES assembly. Within p125A, we defined a C-terminal DDHD domain found in phospholipases and PI transfer proteins that recognized PA and phosphatidylinositol phosphates in vitro and was targeted to PI4P-rich membranes in cells. A conserved central SAM domain promoted self-assembly and selective lipid recognition by the DDHD domain. A basic cluster and a hydrophobic interface in the DDHD and SAM domains, respectively, were required for p125A-mediated functional ERES assembly. Lipid recognition by the SAM-DDHD module was used to stabilize membrane association and regulate the spatial segregation of COPII from Sec16A, nucleating the coat at ERES for ER exit.


Asunto(s)
Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Fosfatos de Fosfatidilinositol/fisiología , Proteínas Portadoras/química , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Liposomas/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas de Unión al ARN , Proteínas de Transporte Vesicular/metabolismo
9.
J Cell Biol ; 190(1): 115-28, 2010 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-20624903

RESUMEN

The guanosine triphosphatase Sar1 controls the assembly and fission of COPII vesicles. Sar1 utilizes an amphipathic N-terminal helix as a wedge that inserts into outer membrane leaflets to induce vesicle neck constriction and control fission. We hypothesize that Sar1 organizes on membranes to control constriction as observed with fission proteins like dynamin. Sar1 activation led to membrane-dependent oligomerization that transformed giant unilamellar vesicles into small vesicles connected through highly constricted necks. In contrast, membrane tension provided through membrane attachment led to organization of Sar1 in ordered scaffolds that formed rigid, uniformly nonconstricted lipid tubules to suggest that Sar1 organization regulates membrane constriction. Sar1 organization required conserved residues located on a unique C-terminal loop. Mutations in this loop did not affect Sar1 activation or COPII recruitment and enhanced membrane constriction, yet inhibited Sar1 organization and procollagen transport from the endoplasmic reticulum (ER). Sar1 activity was directed to liquid-disordered lipid phases. Thus, lipid-directed and tether-assisted Sar1 organization controls membrane constriction to regulate ER export.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Transporte Biológico/fisiología , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Células CACO-2 , Membrana Celular/genética , Perros , Retículo Endoplásmico/genética , Activación Enzimática/fisiología , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Multimerización de Proteína/fisiología , Estructura Secundaria de Proteína
10.
Traffic ; 10(11): 1669-84, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19761544

RESUMEN

During neuron development, the biosynthetic needs of the axon initially outweigh those of dendrites. However, although a localized role for the early secretory pathway in dendrite development has been observed, such a role in axon growth remains undefined. We therefore studied the localization of Sar1, a small GTPase that controls ER export, during early stages of neuronal development that are characterized by selective and robust axon growth. At these early stages, Sar1 was selectively targeted to the axon where it gradually concentrated within varicosities in which additional proteins that function in the early secretory pathway were detected. Sar1 targeting to the axon followed axon specification and was dependent on localized actin instability. Changes in Sar1 expression levels at these early development stages modulated axon growth. Specifically, reduced expression of Sar1, which was initially only detectable in the axon, correlated with reduced axon growth, where as overexpression of Sar1 supported the growth of longer axons. In support of the former finding, expression of dominant negative Sar1 inhibited axon growth. Thus, as observed in lower organisms, mammalian cells use temporal and spatial regulation of endoplasmic reticulum exit site (ERES) to address developmental biosynthetic demands. Furthermore, axons, such as dendrites, rely on ERES targeting and assembly for growth.


Asunto(s)
Axones/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Células Cultivadas , Aparato de Golgi/metabolismo , Hipocampo/citología , Ratones , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Neuronas/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley
11.
J Biol Chem ; 284(7): 4168-78, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19098309

RESUMEN

Cysteine string protein (Csp) is a J-domain-containing protein whose overexpression blocks the exit of cystic fibrosis transmembrane conductance regulator (CFTR) from the endoplasmic reticulum (ER). Another method of blocking ER exit, the overexpression of Sar1-GTP, however, yielded twice as much immature CFTR compared with Csp overexpression. This finding suggested that Csp not only inhibits CFTR ER exit but also facilitates the degradation of immature CFTR. This was confirmed by treatment with a proteasome inhibitor, which returned the level of immature CFTR to that found in cells expressing Sar1-GTP only. CspH43Q, which does not interact with Hsc70/Hsp70 efficiently, did not promote CFTR degradation, suggesting that the pro-degradative effect of Csp requires Hsc70/Hsp70 binding/activation. In agreement with this, Csp overexpression increased the amount of Hsc70/Hsp70 co-immunoprecipitated with CFTR, whereas overexpression of CspH43Q did not. The Hsc70/Hsp70 binding partner C terminus of Hsp70-interacting protein (CHIP) can target CFTR for proteasome-mediated degradation. Csp overexpression also increased the amount of CHIP co-immunoprecipitated with CFTR. In addition, CHIP interacted directly with Csp, which was confirmed by in vitro binding experiments. Csp overexpression also increased CFTR ubiquitylation and reduced the half-life of immature CFTR. These findings indicate that Csp not only regulates the exit of CFTR from the ER, but that this action is accompanied by Hsc70/Hsp70 and CHIP-mediated CFTR degradation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de la Membrana/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Retículo Endoplásmico/genética , Proteínas del Choque Térmico HSC70/genética , Proteínas del Choque Térmico HSP40/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Estructura Terciaria de Proteína/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
12.
J Biol Chem ; 282(46): 33475-33483, 2007 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-17855354

RESUMEN

Epithelial sodium channels (ENaCs) are assembled in the endoplasmic reticulum (ER) from alpha, beta, and gamma subunits, each with two transmembrane domains, a large extracellular loop, and cytoplasmic amino and carboxyl termini. ENaC maturation involves transit through the Golgi complex where Asn-linked glycans are processed to complex type and the channel is activated by furin-dependent cleavage of the alpha and gamma subunits. To identify signals in ENaC for ER retention/retrieval or ER exit/release, chimera were prepared with the interleukin alpha subunit (Tac) and each of the three cytoplasmic carboxyl termini of mouse ENaC (Tac-Ct) or with gamma-glutamyltranspeptidase and each of the three cytoplasmic amino termini (Nt-GGT). By monitoring acquisition of endoglycosidase H resistance after metabolic labeling, we found no evidence of ER retention of any chimera when compared with control Tac or GGT, but we did observe enhanced exit of Tac-alphaCt when compared with Tac. ER exit of ENaC was assayed after metabolic labeling by following the appearance of cleaved alpha as cleaved alpha subunit, but not non-cleaved alpha, is endoglycosidase H-resistant. Interestingly ER exit of epitope-tagged and truncated alpha (alphaDelta624-699-V5) with full-length betagamma was similar to wild type alpha (+betagamma), whereas ER exit of ENaC lacking the entire cytoplasmic carboxyl tail of alpha (alphaDelta613-699-V5 +betagamma) was significantly reduced. Subsequent analysis of ER exit for ENaCs with mutations within the intervening sequence (613)HRFRSRYWSPG(623) within the context of the full-length alpha revealed that mutation alphaRSRYW(620) to AAAAA significantly reduced ER exit. These data indicate that ER exit of ENaC is regulated by a signal within the alpha subunit carboxyl cytoplasmic tail.


Asunto(s)
Retículo Endoplásmico/metabolismo , Canales Epiteliales de Sodio/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , Perros , Glicósido Hidrolasas/metabolismo , Aparato de Golgi/metabolismo , Ratones , Modelos Biológicos , Mutación , Estructura Terciaria de Proteína , Factores de Tiempo , gamma-Glutamiltransferasa/metabolismo
13.
Adv Drug Deliv Rev ; 59(8): 759-81, 2007 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-17681635

RESUMEN

The endoplasmic reticulum (ER) is a central processor that controls the expression of functional proteins, required for the communication of the cell with the external environment. Plasma membranes receptors, ion channels, secreted hormones, catabolic and metabolic enzymes are folded and assembled in the ER. Key metabolic functions are also regulated from the ER. Molecular quality control monitors ER processing activities and co-ordinates these activities with cell and organism demands. Recent understandings of the molecular basis for ER processing activities illuminate the key role of the ER in the development of a variety of diseases. ER derived diseases include specific genetic disorders such as cystic fibrosis or highly prevalent diseases including diabetes and a range of neurodegenerative diseases. ER processing also plays a key role in the development of cancer. This review summarizes the molecular basis for ER processing functions and current avenues in ER-targeted drug development.


Asunto(s)
Sistemas de Liberación de Medicamentos , Retículo Endoplásmico/metabolismo , Chaperonas Moleculares/administración & dosificación , Chaperonas Moleculares/química , Pliegue de Proteína , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Vesículas Cubiertas por Proteínas de Revestimiento/efectos de los fármacos , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Glicosilación/efectos de los fármacos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Chaperonas Moleculares/genética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos
14.
J Biol Chem ; 282(24): 17632-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17428803

RESUMEN

Coat protein complex II (COPII)-coated vesicles/carriers, which mediate export of proteins from the endoplasmic reticulum (ER), are formed at special ER subdomains in mammals, termed ER exit sites or transitional ER. The COPII coat consists of a small GTPase, Sar1, and two protein complexes, Sec23-Sec24 and Sec13-Sec31. Sec23-Sec24 and Sec13-Sec31 appear to constitute the inner and the outermost layers of the COPII coat, respectively. We previously isolated two mammalian proteins (p125 and p250) that bind to Sec23. p125 was found to be a mammalian-specific, phospholipase A(1)-like protein that participates in the organization of ER exit sites. Here we show that p250 is encoded by the KIAA0310 clone and has sequence similarity to yeast Sec16 protein. Although KIAA0310p was found to be localized at ER exit sites, subcellular fractionation revealed its predominant presence in the cytosol. Cytosolic KIAA0310p was recruited to ER membranes in a manner dependent on Sar1. Depletion of KIAA0310p mildly caused disorganization of ER exit sites and delayed protein transport from the ER, suggesting its implication in membrane traffic out of the ER. Overexpression of KIAA0310p affected ER exit sites in a manner different from that of p125. Binding experiments suggested that KIAA0310p interacts with both the inner and the outermost layer coat complexes, whereas p125 binds principally to the inner layer complex. Our results suggest that KIAA0310p, a mammalian homologue of yeast Sec16, builds up ER exit sites in cooperation with p125 and plays a role in membrane traffic from the ER.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Transporte Biológico/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células HeLa , Humanos , Ratones , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Transporte Vesicular/genética
15.
Dev Cell ; 11(5): 671-82, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17084359

RESUMEN

The mechanisms that regulate endoplasmic reticulum (ER) exit-site (ERES) assembly and COPII-mediated ER export are currently unknown. We analyzed the role of phosphatidylinositols (PtdIns) in regulating ER export. Utilizing pleckstrin homology domains and a PtdIns phosphatase to specifically sequester or reduce phosphorylated PtdIns levels, we found that PtdIns 4-phosphate (PtsIns4P) is required to promote COPII-mediated ER export. Biochemical and morphological in vitro analysis revealed dynamic and localized PtsIns4P formation at ERES. PtdIns4P was utilized to support Sar1-induced proliferation and constriction of ERES membranes. PtdIns4P also assisted in Sar1-induced COPII nucleation at ERES. Therefore, localized dynamic remodeling of PtdIns marks ERES membranes to regulate COPII-mediated ER export.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/fisiología , Retículo Endoplásmico/metabolismo , Fosfatos de Fosfatidilinositol/biosíntesis , Proteínas de Transporte Vesicular/fisiología , Animales , Transporte Biológico , Línea Celular , Retículo Endoplásmico/ultraestructura , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestructura , Proteínas de la Membrana/fisiología , Proteínas de Unión al GTP Monoméricas/fisiología , Fosforilación , Ratas
16.
J Biol Chem ; 281(16): 11312-21, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16469739

RESUMEN

We examined the role of the cysteine string protein (Csp) in cystic fibrosis transmembrane conductance regulator (CFTR) biogenesis in relation to another J-domain protein, Hdj-2, a recognized CFTR cochaperone. Increased expression of Csp produced a dose-dependent reduction in mature (band C) CFTR and an increase in immature (band B) CFTR. Exogenous expression of Hdj-2 also increased CFTR band B, but unlike Csp, Hdj-2 increased band C as well. The Csp-induced block of CFTR maturation required Hsp70, because a J-domain mutant (H43Q) that interferes with the ability of Csp to stimulate Hsp70 ATPase activity relieved the Csp-induced block of CFTR maturation. Nevertheless, Csp H43Q still increased immature CFTR. Csp-induced band B CFTR was found adjacent to the nucleus, co-localizing with calnexin, and it remained detergent-soluble. These data indicate that Csp did not block CFTR maturation by promoting the aggregation or degradation of immature CFTR. Csp knockdown by RNA interference produced a 5-fold increase in mature CFTR and augmented cAMP-stimulated CFTR currents. Thus, the production of mature CFTR is inversely related to the expression level of Csp. Both Csp and Hdj-2 associated with the CFTR R-domain in vitro, and Hdj-2 binding was displaced by Csp, suggesting common interaction sites. Combined expression of Csp and Hdj-2 mimicked the effect of Csp alone, a block of CFTR maturation. But together, Csp and Hdj-2 produced additive increases in CFTR band B, and this did not depend on their interactions with Hsp70, consistent with direct chaperone actions of these proteins. Like Hdj-2, Csp reduced the aggregation of NBD1 in vitro in the absence of Hsp70. Our data suggest that both Csp and Hdj-2 facilitate the biosynthesis of immature CFTR, acting as direct CFTR chaperones, but in addition, Csp is positioned later in the CFTR biogenesis cascade where it regulates the production of mature CFTR by limiting its exit from the endoplasmic reticulum.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Proteínas del Choque Térmico HSP40/química , Proteínas de la Membrana/química , Actinas/metabolismo , Aniones , Transporte Biológico , Calnexina/metabolismo , Calnexina/farmacología , Línea Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Detergentes/metabolismo , Detergentes/farmacología , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Chaperonas Moleculares/metabolismo , Mutación , Oocitos/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , ARN/metabolismo , Interferencia de ARN , Factores de Tiempo , Transfección
17.
J Cell Biol ; 171(6): 919-24, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16344311

RESUMEN

The mechanisms by which the coat complex II (COPII) coat mediates membrane deformation and vesicle fission are unknown. Sar1 is a structural component of the membrane-binding inner layer of COPII (Bi, X., R.A. Corpina, and J. Goldberg. 2002. Nature. 419:271-277). Using model liposomes we found that Sar1 uses GTP-regulated exposure of its NH2-terminal tail, an amphipathic peptide domain, to bind, deform, constrict, and destabilize membranes. Although Sar1 activation leads to constriction of endoplasmic reticulum (ER) membranes, progression to effective vesicle fission requires a functional Sar1 NH2 terminus and guanosine triphosphate (GTP) hydrolysis. Inhibition of Sar1 GTP hydrolysis, which stabilizes Sar1 membrane binding, resulted in the formation of coated COPII vesicles that fail to detach from the ER. Thus Sar1-mediated GTP binding and hydrolysis regulates the NH2-terminal tail to perturb membrane packing, promote membrane deformation, and control vesicle fission.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Regulación de la Expresión Génica , Guanosina Trifosfato/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Catálisis , Células Cultivadas , Cricetinae , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Hidrólisis , Interacciones Hidrofóbicas e Hidrofílicas , Liposomas/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Unión Proteica , Estructura Terciaria de Proteína/fisiología , Transporte de Proteínas/fisiología , Ratas , Proteínas de Transporte Vesicular/metabolismo
18.
J Biol Chem ; 280(11): 10501-8, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15634669

RESUMEN

Phosphatidylinositol 4-phosphate (PI4P) regulates biosynthetic membrane traffic at multiple steps and differentially affects the surface delivery of apically and basolaterally destined proteins in polarized cells. Two phosphatidylinositol 4-kinases (PI4Ks) have been localized to the Golgi complex in mammalian cells, type III PI4Kbeta (PI4KIIIbeta) and type II PI4Kalpha (PI4KIIalpha). Here we report that PI4KIIIbeta and PI4KIIalpha localize to discrete subcompartments of the Golgi complex in Madin-Darby canine kidney (MDCK) cells. PI4KIIIbeta was enriched in early Golgi compartments, whereas PI4KIIalpha colocalized with markers of the trans-Golgi network (TGN). To understand the temporal and spatial control of PI4P generation across the Golgi complex, we quantitated the steady state distribution of a fluorescent PI4P-binding domain relative to cis/medial Golgi and TGN markers in transiently transfected MDCK cells. The density of the signal from this PI4P reporter was roughly 2-fold greater in the early Golgi compartments compared with that of the TGN. Furthermore, this ratio could be modulated in vivo by overexpression of catalytically inactive PI4KIIIbeta and PI4KIIalpha or in vitro by the PI4KIIIbeta inhibitor wortmannin. Our data suggest that both PI4KIIIbeta and PI4KIIalpha contribute to the compartmental regulation of PI4P synthesis within the Golgi complex. We discuss our results with respect to the kinetic effects of modulating PI4K activity on polarized biosynthetic traffic in MDCK cells.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Aparato de Golgi/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Androstadienos/farmacología , Animales , Proteínas Sanguíneas/química , Catálisis , Línea Celular , Membrana Celular/metabolismo , Perros , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Cinética , Microscopía Confocal , Microscopía Fluorescente , Fosfoproteínas/química , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transfección , Wortmanina , Red trans-Golgi/metabolismo
20.
J Biol Chem ; 280(11): 10141-8, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15623529

RESUMEN

Transport vesicles coated with the COPII complex, which is assembled from Sar1p, Sec23p-Sec24p, and Sec13p-Sec31p, are involved in protein export from the endoplasmic reticulum (ER). We previously identified and characterized a novel Sec23p-interacting protein, p125, that is only expressed in mammals and exhibits sequence homology with phosphatidic acid-preferring phospholipase A(1) (PA-PLA(1)). In this study, we examined the localization and function of p125 in detail. By using immunofluorescence and electron microscopy, we found that p125 is principally localized in ER exit sites where COPII-coated vesicles are produced. Analyses of chimeric proteins comprising p125 and two other members of the mammalian PA-PLA(1) family (PA-PLA(1) and KIAA0725p) showed that, for localization to ER exit sites, the p125-specific N-terminal region is critical, and the putative lipase domain is interchangeable with KIAA0725p but not with PA-PLA(1). RNA interference-mediated depletion of p125 affected the organization of ER exit sites. The structure of the cis-Golgi compartment was also substantially disturbed, whereas the medial-Golgi was not. Protein export from the ER occurred without a significant delay in p125-depleted cells. Our study suggests that p125 is a mammalian-specific component of ER exit sites and participates in the organization of this compartment.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas Portadoras/fisiología , Retículo Endoplásmico/metabolismo , Animales , Sitios de Unión , Transporte Biológico , Proteínas Portadoras/metabolismo , Línea Celular , Membrana Celular/metabolismo , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Immunoblotting , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Complejo Poro Nuclear , Fosfoproteínas/metabolismo , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteínas de Unión al ARN , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Tiempo , Transfección , Proteínas de Transporte Vesicular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...