Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
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 ; 25(3): e12932, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38528836

RESUMEN

Alzheimer's disease is associated with increased levels of amyloid beta (Aß) generated by sequential intracellular cleavage of amyloid precursor protein (APP) by membrane-bound secretases. However, the spatial and temporal APP cleavage events along the trafficking pathways are poorly defined. Here, we use the Retention Using Selective Hooks (RUSH) to compare in real time the anterograde trafficking and temporal cleavage events of wild-type APP (APPwt) with the pathogenic Swedish APP (APPswe) and the disease-protective Icelandic APP (APPice). The analyses revealed differences in the trafficking profiles and processing between APPwt and the APP familial mutations. While APPwt was predominantly processed by the ß-secretase, BACE1, following Golgi transport to the early endosomes, the transit of APPswe through the Golgi was prolonged and associated with enhanced amyloidogenic APP processing and Aß secretion. A 20°C block in cargo exit from the Golgi confirmed ß- and γ-secretase processing of APPswe in the Golgi. Inhibition of the ß-secretase, BACE1, restored APPswe anterograde trafficking profile to that of APPwt. APPice was transported rapidly through the Golgi to the early endosomes with low levels of Aß production. This study has revealed different intracellular locations for the preferential cleavage of APPwt and APPswe and Aß production, and the Golgi as the major processing site for APPswe, findings relevant to understand the molecular basis of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Suecia , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Mutación
3.
J Cell Sci ; 136(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37565427

RESUMEN

Human serum albumin (HSA) has a long circulatory half-life owing, in part, to interaction with the neonatal Fc receptor (FcRn or FCGRT) in acidic endosomes and recycling of internalised albumin. Vascular endothelial and innate immune cells are considered the most relevant cells for FcRn-mediated albumin homeostasis in vivo. However, little is known about endocytic trafficking of FcRn-albumin complexes in primary human endothelial cells. To investigate FcRn-albumin trafficking in physiologically relevant endothelial cells, we generated primary human vascular endothelial cell lines from blood endothelial precursors, known as blood outgrowth endothelial cells (BOECs). We mapped the endosomal system in BOECs and showed that BOECs efficiently internalise fluorescently labelled HSA predominantly by fluid-phase macropinocytosis. Pulse-chase studies revealed that intracellular HSA molecules co-localised with FcRn in acidic endosomal structures and that the wildtype HSA, but not the non-FcRn-binding HSAH464Q mutant, was excluded from late endosomes and/or lysosomes. Live imaging revealed that HSA is partitioned into FcRn-positive tubules derived from maturing macropinosomes, which are then transported towards the plasma membrane. These findings identify the FcRn-albumin trafficking pathway in primary vascular endothelial cells, relevant to albumin homeostasis.


Asunto(s)
Albúminas , Células Endoteliales , Humanos , Albúminas/metabolismo , Línea Celular , Endosomas/metabolismo , Células Endoteliales/metabolismo , Semivida , Antígenos de Histocompatibilidad Clase I/metabolismo
4.
Traffic ; 23(3): 158-173, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35076977

RESUMEN

The intracellular trafficking of ß-site amyloid precursor protein (APP) cleaving enzyme (BACE1) and APP regulates amyloid-ß production. Our previous work demonstrated that newly synthesized BACE1 and APP are segregated into distinct trafficking pathways from the trans-Golgi network (TGN), and that alterations in their trafficking lead to an increase in Aß production in non-neuronal and neuronal cells. However, it is not known whether BACE1 and APP are transported through the Golgi stacks together and sorted at the TGN or segregated prior to arrival at the TGN. To address this question, we have used high-resolution Airyscan technology followed by Huygens deconvolution to quantify the overlap of BACE1 and APP in Golgi subcompartments in HeLa cells and primary neurons. Here, we show that APP and BACE1 are segregated, on exit from the endoplasmic reticulum and in the cis-Golgi and throughout the Golgi stack. In contrast, the transferrin receptor, which exits the TGN in AP-1 mediated transport carriers as for BACE1, colocalizes with BACE1, but not APP, throughout the Golgi stack. The segregation of APP and BACE1 is independent of the Golgi ribbon structure and the cytoplasmic domain of the cargo. Overall, our findings reveal the segregation of different membrane cargoes early in the secretory pathway, a finding relevant to the regulation of APP processing events.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Transporte de Proteínas/fisiología
5.
Cell ; 138(4): 787-94, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19703403

RESUMEN

Mammalian golgins of the trans-Golgi network (TGN) are small G protein effectors that are required for membrane transport and contain a Golgi targeting C-terminal GRIP domain. The localization of two TGN golgins, p230/golgin-245 and golgin-97, is mediated by the small GTPase Arl1, whereas recruitment of the TGN golgin GCC185 is controversial. Recently, GCC185 was proposed to localize to the Golgi by the co-operation of two small GTPases, Rab6A/A' and Arl1 (Burguete et al., 2008), a model based predominantly on in vitro interactions. Here we demonstrate that Golgi recruitment of endogenous GCC185 does not involve Rab6A/A' and Arl1. We find minimal colocalization between Rab6A/A' and endogenous GCC185 on Golgi membranes and failed to detect an interaction between Rab6A/A' and C-terminal domains of GCC185 by yeast two-hybrid analyses. Moreover, depletion of both Rab6A/A' and Arl1 also had no effect on the localization of endogenous GCC185 or the isolated GRIP domain of GCC185.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Aparato de Golgi/química , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Factores de Ribosilacion-ADP/análisis , Citosol , Proteínas de la Matriz de Golgi , Células HeLa , Humanos , Proteínas de la Membrana/análisis , Transporte de Proteínas , Proteínas de Unión al GTP rab/análisis
6.
Traffic ; 22(9): 319-327, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34189821

RESUMEN

ß-amyloid peptides (Aß) are generated in intracellular compartments of neurons and secreted to form cytotoxic fibrils and plaques. Dysfunctional membrane trafficking contributes to aberrant Aß production and Alzheimer's disease. Endosomes represent one of the major sites for Aß production and recently the Golgi has re-emerged also as a major location for amyloid precursor protein (APP) processing and Aß production. Based on recent findings, here we propose that APP processing in the Golgi is finely tuned by segregating newly-synthesised APP and the ß-secretase BACE1 within the Golgi and into distinct trans-Golgi network transport pathways. We hypothesise that there are multiple mechanisms responsible for segregating APP and BACE1 during transit through the Golgi, and that perturbation in Golgi morphology associated with Alzheimer's disease, and or changes in cholesterol metabolism associated with Alzheimer's disease risk factors, may lead to a loss of partitioning and enhanced Aß production.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Humanos , Neuronas/metabolismo , Transporte de Proteínas , Vías Secretoras
7.
Exp Cell Res ; 399(2): 112442, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359467

RESUMEN

Rab30 is a poorly characterized small GTPase. Here we show that Rab30 is localised primarily to the TGN and recycling endosomes in a range of cell types, including primary neurons; minor levels of Rab30 were also detected throughout the Golgi stack and early endosomes. Silencing of Rab30 resulted in the dispersal of both early and recycling endosomes and TGN compartments in HeLa cells. By analyzing cargo trafficking in Rab30-silenced and Rab30-overexpressing HeLa cells, we demonstrate that Rab30 plays a role in retrograde trafficking of TGN38 from endosomes to the Golgi, but has no apparent role in the endocytic recycling of the transferrin receptor to the plasma membrane. Five interactive partners with Rab30 were identified by pull-down and MS analysis using GFP-tagged Rab30 mutant, Rab30(Q68L). Two of the interactive partners identified were Arf1 and Arf4, known regulators of endosome to TGN retrograde transport. Knockdown of Arf1 and Arf4 results in GFP-Rab30 decorated tubules arising from the recycling endosomes, suggesting association of Rab30 with tubular carriers. Overall our data demonstrates a role for Rab30 in regulating retrograde transport to the TGN and maintenance of endosomal-TGN organization.


Asunto(s)
Endosomas/metabolismo , Proteínas de Unión al GTP rab/fisiología , Red trans-Golgi/genética , Antígenos CD/metabolismo , Endosomas/genética , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Transporte de Proteínas/genética , Receptores de Transferrina/metabolismo , Proteínas de Unión al GTP rab/genética , Red trans-Golgi/metabolismo
8.
J Cell Sci ; 133(5)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31444284

RESUMEN

The neonatal Fc receptor (FcRn) rescues albumin and IgG from degradation following endocytosis and thereby extends the half-life of these plasma proteins. However, the pathways for the uptake of these soluble FcRn ligands, and the recycling itinerary of the FcRn-ligand complexes, have not been identified in primary cells. Here, we have defined the recycling of human albumin and IgG in primary mouse macrophages selectively expressing the human FcRn. Albumin is internalised by macropinocytosis; in the absence of FcRn, internalised albumin is rapidly degraded, while in the presence of FcRn albumin colocalises to SNX5-positive membrane domains and is partitioned into tubules emanating from early macropinosomes for delivery in transport carriers to the plasma membrane. Soluble monomeric IgG was also internalised by macropinocytosis and rapidly recycled by the same pathway. In contrast, the fate of IgG bound to surface Fcγ receptors differed from monomeric IgG endocytosed by macropinocytosis. Overall, our findings identify a rapid recycling pathway for FcRn ligands from early macropinosomes to the cell surface of primary cells.


Asunto(s)
Albúminas/metabolismo , Antígenos de Histocompatibilidad Clase I/fisiología , Inmunoglobulina G/metabolismo , Macrófagos/metabolismo , Pinocitosis , Receptores Fc/fisiología , Animales , Línea Celular , Endocitosis , Endosomas/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Ratones , Ratones Noqueados , Transporte de Proteínas , Receptores Fc/genética
9.
J Biol Chem ; 294(5): 1618-1631, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30545942

RESUMEN

Amyloid precursor protein (APP) is processed along the amyloidogenic pathway by the ß-secretase, BACE1, generating ß-amyloid (Aß), or along the nonamyloidogenic pathway by α-secretase, precluding Aß production. The plasma membrane is considered the major site for α-secretase-mediated APP cleavage, but other cellular locations have not been rigorously investigated. Here, we report that APP is processed by endogenous α-secretase at the trans-Golgi network (TGN) of both transfected HeLa cells and mouse primary neurons. We have previously shown the adaptor protein complex, AP-4, and small G protein ADP-ribosylation factor-like GTPase 5b (Arl5b) are required for efficient post-Golgi transport of APP to endosomes. We found here that AP-4 or Arl5b depletion results in Golgi accumulation of APP and increased secretion of the soluble α-secretase cleavage product sAPPα. Moreover, inhibition of γ-secretase following APP accumulation in the TGN increases the levels of the membrane-bound C-terminal fragments of APP from both α-secretase cleavage (α-CTF, named C83 according to its band size) and BACE1 cleavage (ß-CTF/C99). The level of C83 was ∼4 times higher than that of C99, indicating that α-secretase processing is the major pathway and that BACE1 processing is the minor pathway in the TGN. AP-4 silencing in mouse primary neurons also resulted in the accumulation of endogenous APP in the TGN and enhanced α-secretase processing. These findings identify the TGN as a major site for α-secretase processing in HeLa cells and primary neurons and indicate that both APP processing pathways can occur within the TGN compartment along the secretory pathway.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Neuronas/metabolismo , Red trans-Golgi/metabolismo , Animales , Células Cultivadas , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/citología
10.
J Cell Sci ; 131(3)2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29361552

RESUMEN

In vertebrates, individual Golgi stacks are joined into a compact ribbon structure; however, the relevance of a ribbon structure has been elusive. Here, we exploit the finding that the membrane tether of the trans-Golgi network, GCC88 (encoded by GCC1), regulates the balance between Golgi mini-stacks and the Golgi ribbon. Loss of Golgi ribbons in stable cells overexpressing GCC88 resulted in compromised mechanistic target of rapamycin (mTOR) signaling and a dramatic increase in LC3-II-positive autophagosomes, whereas RNAi-mediated depletion of GCC88 restored the Golgi ribbon and reduced autophagy. mTOR was absent from dispersed Golgi mini-stacks whereas recruitment of mTOR to lysosomes was unaffected. We show that the Golgi ribbon is a site for localization and activation of mTOR, a process dependent on the ribbon structure. We demonstrate a strict temporal sequence of fragmentation of Golgi ribbon, loss of Golgi mTOR and subsequent increased autophagy. Golgi ribbon fragmentation has been reported in various neurodegenerative diseases and we demonstrate the potential relevance of our findings in neuronal cells using a model of neurodegeneration. Overall, this study highlights a role for the Golgi ribbon in pathways central to cellular homeostasis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Autofagia , Aparato de Golgi/metabolismo , Mamíferos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Aparato de Golgi/ultraestructura , Proteínas de la Matriz de Golgi/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neuroblastoma/metabolismo , Transducción de Señal , Factores de Tiempo , Proteínas tau/metabolismo , Red trans-Golgi/metabolismo , Red trans-Golgi/ultraestructura
11.
Immunol Cell Biol ; 98(1): 28-41, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31654430

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid beta (Aß) accumulation, tau pathology and neuroinflammation. Recently, there has been considerable interest in the role of neuroinflammation in directly contributing to the progression of AD. Studies in mice and humans have identified a role for microglial cells, the resident innate immune cells of the central nervous system, in AD. Activated microglia are a key hallmark of the disease and the secretion of proinflammatory cytokines by microglia may result in a positive feedback loop between neurons and microglia, resulting in ongoing low-grade inflammation. Traditionally, the pathways of Aß production and neuroinflammation have been considered independently; however, recent studies suggest that these processes may converge to promote the pathology associated with AD. Here we review the importance of inflammation and microglia in AD development and effects of inflammatory responses on cellular pathways of neurons, including Aß generation.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Inmunidad Innata , Microglía/inmunología , Neuronas/inmunología , Enfermedad de Alzheimer/patología , Animales , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones , Microglía/patología , Neuronas/patología
12.
Exp Cell Res ; 380(1): 55-68, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30981667

RESUMEN

The small GTPases Rab11a and 11b are key regulators of membrane transport, localised to the recycling endosomes and also early endosomes. The function of Rab11 within the recycling pathway has been well defined, however, the role of Rab11 at the early endosomes remains poorly characterised. Here, we have generated HeLa cell lines devoid of either Rab11a or Rab11b using CRISPR/Cas9 to functionally dissect the roles of these two Rab11 family members in recycling and in the endosomal-lysosomal system. Both Rab11a and Rab11b contribute to the dynamics of tubulation arising from recycling endosomes whereas Rab11a has the major role in recycling of transferrin receptor. Deletion of either Rab11a or Rab11b resulted in the formation of enlarged early endosomes and perturbation of the endosomal-lysosomal pathway. Strikingly, Rab11a knock-out cells showed an increased density of functional late endosomes/lysosomes as well as lysotracker-positive organelles which were primarily concentrated in a perinuclear location, indicating that the homeostasis of the endosome/lysosome pathway had been perturbed. Moreover, in Rab11a knockout cells there was a functional defect in the intracellular recycling of the cation-independent mannose 6-phosphate receptor (CI-M6PR) between the late endosomes and the TGN, a defect associated with enhanced degradation of CI-M6PR. Expression of wild-type Rab11a in Rab11a knockout cells rescued the late endosome/lysosome phenotype. Overall, these results indicate that Rab11a and Rab11b have overlapping and distinct functions and that Rab11a, unexpectedly, plays a central role in the homeostasis of endosomal-lysosomal biogenesis.


Asunto(s)
Endosomas/genética , Lisosomas/genética , Proteínas de Unión al GTP rab/genética , Sistemas CRISPR-Cas/genética , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Receptor IGF Tipo 2/genética , Transducción de Señal/genética
13.
Traffic ; 18(3): 159-175, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28000370

RESUMEN

The intracellular trafficking and proteolytic processing of the membrane-bound amyloid precursor protein (APP) are coordinated events leading to the generation of pathogenic amyloid-beta (Aß) peptides. The membrane transport of newly synthesized APP from the Golgi to the endolysosomal system is not well defined, yet it is likely to be critical for regulating its processing by ß-secretase (BACE1) and γ-secretase. Here, we show that the majority of newly synthesized APP is transported from the trans-Golgi network (TGN) directly to early endosomes and then subsequently to the late endosomes/lysosomes with very little transported to the cell surface. We show that Arl5b, a small G protein localized to the TGN, and AP4 are essential for the post-Golgi transport of APP to early endosomes. Arl5b is physically associated with AP4 and is required for the recruitment of AP4, but not AP1, to the TGN. Depletion of either Arl5b or AP4 results in the accumulation of APP, but not BACE1, in the Golgi, and an increase in APP processing and Aß secretion. These findings demonstrate that APP is diverted from BACE1 at the TGN for direct transport to early endosomes and that the TGN represents a site for APP processing with the subsequent secretion of Aß.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Endosomas/metabolismo , Aparato de Golgi/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proteínas de Unión al ADN , Células HeLa , Humanos , Lisosomas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Transporte de Proteínas/fisiología , Proteínas de Unión al ARN , Vesículas Transportadoras/metabolismo , Red trans-Golgi/metabolismo
14.
J Biol Chem ; 293(17): 6363-6373, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29523681

RESUMEN

The neonatal Fc receptor (FcRn) has a pivotal role in albumin and IgG homeostasis. Internalized IgG captured by FcRn under acidic endosomal conditions is recycled to the cell surface where exocytosis and a shift to neutral pH promote extracellular IgG release. Although a similar mechanism is proposed for FcRn-mediated albumin intracellular trafficking and recycling, this pathway is less well defined but is relevant to the development of therapeutics exploiting FcRn to extend the half-life of short-lived plasma proteins. Recently, a long-acting recombinant coagulation factor IX-albumin fusion protein (rIX-FP) has been approved for the management of hemophilia B. Fusion to albumin potentially enables internalized proteins to engage FcRn and escape lysosomal degradation. In this study, we present for the first time a detailed investigation of the FcRn-mediated recycling of albumin and the albumin fusion protein rIX-FP. We demonstrate that following internalization via FcRn at low pH, rIX-FP, like albumin, is detectable within the early endosome and rapidly (within 10-15 min) traffics into the Rab11+ recycling endosomes, from where it is exported from the cell. Similarly, rIX-FP and albumin taken up by fluid-phase endocytosis at physiological pH traffics into the Rab11+ recycling compartment in FcRn-positive cells but into the lysosomal compartment in FcRn-negative cells. As expected, recombinant factor IX (without albumin fusion) and an FcRn interaction-defective albumin variant localized to the lysosomal compartments of both FcRn-expressing and nonexpressing cells. These results indicate that FcRn-mediated recycling via the albumin moiety is a mechanism for the half-life extension of rIX-FP observed in clinical studies.


Asunto(s)
Factor IX , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión , Albúmina Sérica Humana , Línea Celular , Factor IX/genética , Factor IX/farmacocinética , Factor IX/farmacología , Semivida , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemofilia B/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica Humana/genética , Albúmina Sérica Humana/farmacocinética , Albúmina Sérica Humana/farmacología
15.
Bioessays ; 39(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28984991

RESUMEN

The Golgi apparatus in vertebrate cells consists of individual Golgi stacks fused together in a continuous ribbon structure. The ribbon structure per se is not required to mediate the classical functions of this organelle and the relevance of the "ribbon" structure has been a mystery since first identified ultrastructurally in the 1950s. Recent advances recognize a role for the Golgi apparatus in a range of cellular processes, some mediated by signaling networks which are regulated at the Golgi. Here we review the cellular processes and signaling events regulated by the Golgi apparatus and, in particular, explore an emerging theme that the ribbon structure of the Golgi contributes directly to the regulation of these higher order functions.


Asunto(s)
Aparato de Golgi/fisiología , Animales , Aparato de Golgi/metabolismo , Humanos , Transducción de Señal , Vertebrados/metabolismo , Vertebrados/fisiología
16.
Eur J Immunol ; 47(1): 155-167, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27759162

RESUMEN

How the immune system maintains peripheral tolerance under inflammatory conditions is poorly understood. Here we assessed the fate of gastritogenic T cells following inflammatory activation in vivo. Self-reactive T cells (A23 T cells) specific for the gastric H+ /K+ ATPase α subunit (HKα) were transferred into immunosufficient recipient mice and immunised at a site distant to the stomach with adjuvant containing the cognate HKα peptide antigen. Activation of A23 T cells by immunisation did not impact on either immune tolerance or protection from gastric autoimmunity in wild-type BALB/c mice. However, increased presentation of endogenously derived HKα epitopes by dendritic cells (DCs) in the gastric lymph node of IE-H+ /K+ ß transgenic mice (IEß) reduces A23 T-cell tolerance to gastric antigens after inflammatory activation, with subsequent development of gastritis. While HKα-specific A23 T cells from immunised wild-type mice were poorly responsive to in vitro antigen specific activation, A23 T cells from immunised IEß transgenic mice were readily re-activated, indicating loss of T-cell anergy. These findings show that DCs of gastric lymph nodes can maintain tolerance of pathogenic T cells following inflammatory stimulation and that the density of endogenous antigen presented to self-reactive T cells is critical in the balance between tolerance and autoimmunity.


Asunto(s)
Presentación de Antígeno , Autoantígenos/inmunología , Autoinmunidad , Susceptibilidad a Enfermedades , Gastritis/inmunología , Animales , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Anergia Clonal/genética , Anergia Clonal/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Gastritis/metabolismo , Gastritis/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Inmunofenotipificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
17.
Biochem Soc Trans ; 46(5): 1063-1072, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30242119

RESUMEN

An array of signalling molecules are located at the Golgi apparatus, including phosphoinositides, small GTPases, kinases, and phosphatases, which are linked to multiple signalling pathways. Initially considered to be associated predominantly with membrane trafficking, signalling pathways at the Golgi are now recognised to regulate a diverse range of higher-order functions. Many of these signalling pathways are influenced by the architecture of the Golgi. In vertebrate cells, the Golgi consists of individual stacks fused together into a compact ribbon structure and the function of this ribbon structure has been enigmatic. Notably, recent advances have identified a role for the Golgi ribbon in regulation of cellular processes. Fragmentation of the Golgi ribbon results in modulation of many signalling pathways. Various diseases and disorders, including cancer and neurodegeneration, are associated with the loss of the Golgi ribbon and the appearance of a dispersed fragmented Golgi. Here, we review the emerging theme of the Golgi as a cell sensor and highlight the relationship between the morphological status of the Golgi in vertebrate cells and the modulation of signalling networks.


Asunto(s)
Aparato de Golgi/metabolismo , Transporte de Proteínas , Transducción de Señal , Animales , Membrana Celular/metabolismo , Proliferación Celular , Citoesqueleto/metabolismo , Daño del ADN , Glicosilación , Homeostasis , Humanos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mitosis , Fosfatidilinositoles/química , Serina-Treonina Quinasas TOR/metabolismo
18.
J Cell Sci ; 128(23): 4407-19, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26459636

RESUMEN

The regulation of macropinocytosis, a specialised endocytosis pathway, is important for immune cell function. However, it is not known whether the biogenesis of macropinosomes involves one or more distinct pathways. We previously identified sorting nexin 5 (SNX5) as a regulator of macropinocytosis in macrophages. Here, we show that bone-marrow-derived macrophages from SNX5-knockout mice had a 60-70% reduction in macropinocytic uptake of dextran or ovalbumin, whereas phagocytosis and retrograde transport from the plasma membrane to the Golgi was unaffected. In contrast, deficiency of SNX5 had no effect on macropinocytosis or antigen presentation by dendritic cells. Activation of macrophages with CSF-1 resulted in a localisation of SNX5 to actin-rich ruffles in a manner dependent on receptor tyrosine kinases. SNX5-deficient macrophages showed a dramatic reduction in ruffling on the dorsal surface following CSF-1 receptor activation, whereas peripheral ruffling and cell migration were unaffected. We demonstrate that SNX5 is acting upstream of actin polymerisation following CSF-1 receptor activation. Overall, our findings reveal the important contribution of dorsal ruffing to receptor-activated macropinocytosis in primary macrophages and show that SNX5 selectively regulates macropinosomes derived from the dorsal ruffles.


Asunto(s)
Presentación de Antígeno/fisiología , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Pinocitosis/fisiología , Nexinas de Clasificación/metabolismo , Animales , Células Dendríticas/citología , Macrófagos/citología , Ratones , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Nexinas de Clasificación/genética
20.
Int Immunol ; 28(10): 513-518, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371614

RESUMEN

Thymus-derived regulatory T cells (Tregs) are essential for the maintenance of immunological tolerance. Diverse signalling pathways contribute to thymic Treg cells (tTregs) development; however, the role of mammalian target of rapamycin (mTOR) remains unclear. Rapamycin is a well-characterized inhibitor of mTOR complex 1 signalling and a potent inducer of Treg cells in the periphery. However, the effect of rapamycin on the development of tTregs is poorly defined. Here we have used thymic organ culture to investigate the effect of rapamycin on tTreg development. We show that, contrary to its effect in the periphery, rapamycin inhibits the development of tTregs in wild-type thymi. The inhibition of tTregs by rapamycin could be rescued by a deficiency of Bim. However, rapamycin did not inhibit the development of antigen-specific TCR transgenic tTregs in response to exogenous peptide antigen, indicating that the development of thymic Foxp3+CD4+ cells was not intrinsically inhibited by rapamycin. Collectively our data demonstrate that rapamycin results in a reduction of tTregs because of Bim-mediated apoptosis of immature tTregs via a cell extrinsic mechanism. These findings are important not only for understanding the mechanism of tTreg induction but also for an appreciation of the impact of the clinical application of rapamycin.


Asunto(s)
Proteína 11 Similar a Bcl2/metabolismo , Sirolimus/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Timo/efectos de los fármacos , Timo/inmunología , Animales , Proteína 11 Similar a Bcl2/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Técnicas de Cultivo de Órganos , Timo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA