Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
NPJ Vaccines ; 9(1): 76, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38594284

RESUMEN

Dendritic cell (DC)-targeted vaccination is a new mode of antigen delivery that relies on the use of monoclonal antibodies (mAb) to target antigen to specific DC subsets. The neonatal Fc receptor (FcRn) is a non-classical Fc receptor that binds to immunoglobulin G (IgG) in acidified endosomes and controls its intracellular transport and recycling. FcRn is known to participate in the antigen presentation of immune complexes, however its contribution to DC-targeted vaccination has not previously been examined. Here we have investigated the role of FcRn in antigen presentation using antigen conjugated to IgG mAb which target specific DC receptors, including DEC205 and Clec9A expressed by the conventional DC 1 (cDC1) subset. We show that FcRn is expressed at high levels by cDC1, both at steady-state and following activation and plays a significant role in MHC I cross-presentation and MHC II presentation of antigens that are targeted to cDC1 via mAb specific for DEC205. This effect of FcRn is intrinsic to cDC1 and FcRn impacts the efficacy of anti-DEC205-mediated vaccination against B cell lymphoma. In contrast, FcRn does not impact presentation of antigens targeted to Clec9A and does not regulate presentation of cell-associated antigen. These data highlight a new and unique role of FcRn in controlling the immunogenicity of anti-DEC205-based vaccination, with consequences for exploiting this pathway to improve DC-targeted vaccine outcomes.

2.
Commun Biol ; 7(1): 209, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378743

RESUMEN

Autophagy-related genes have been closely associated with intestinal homeostasis. BECLIN1 is a component of Class III phosphatidylinositol 3-kinase complexes that orchestrate autophagy initiation and endocytic trafficking. Here we show intestinal epithelium-specific BECLIN1 deletion in adult mice leads to rapid fatal enteritis with compromised gut barrier integrity, highlighting its intrinsic critical role in gut maintenance. BECLIN1-deficient intestinal epithelial cells exhibit extensive apoptosis, impaired autophagy, and stressed endoplasmic reticulum and mitochondria. Remaining absorptive enterocytes and secretory cells display morphological abnormalities. Deletion of the autophagy regulator, ATG7, fails to elicit similar effects, suggesting additional novel autophagy-independent functions of BECLIN1 distinct from ATG7. Indeed, organoids derived from BECLIN1 KO mice show E-CADHERIN mislocalisation associated with abnormalities in the endocytic trafficking pathway. This provides a mechanism linking endocytic trafficking mediated by BECLIN1 and loss of intestinal barrier integrity. Our findings establish an indispensable role of BECLIN1 in maintaining mammalian intestinal homeostasis and uncover its involvement in endocytic trafficking in this process. Hence, this study has important implications for our understanding of intestinal pathophysiology.


Asunto(s)
Apoptosis , Células Epiteliales , Ratones , Animales , Beclina-1/genética , Beclina-1/metabolismo , Apoptosis/genética , Células Epiteliales/metabolismo , Autofagia/genética , Homeostasis , Mamíferos
3.
Mol Biol Cell ; 33(1): ar6, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34731029

RESUMEN

The neonatal Fc receptor (FcRn) is responsible for the recycling of endocytosed albumin and IgG, and contributes to their long plasma half-life. We recently identified an FcRn-dependent recycling pathway from macropinosomes in macrophages; however, little is known about the dynamics of intracellular FcRn-ligand interactions to promote recycling. Here we demonstrate a multiplexed biophysical fluorescent microscopy approach to resolve the spatiotemporal dynamics of albumin-FcRn interactions in living bone marrow-derived macrophages (BMDMs). We used the phasor approach to fluorescence lifetime imaging microscopy (FLIM) of Förster resonance energy transfer (FRET) to detect the interaction of a FcRn-mCherry fusion protein with endocytosed Alexa Fluor 488-labeled human serum albumin (HSA-AF488) in BMDMs, and raster image correlation spectroscopy (RICS) analysis of single fluorescent-labeled albumin molecules to monitor the diffusion kinetics of internalized albumin. Our data identified a major fraction of immobile HSA-AF488 molecules in endosomal structures of human FcRn-positive mouse macrophages and an increase in FLIM-FRET following endocytosis, including detection of FRET in tubular-like structures. A nonbinding mutant of albumin showed minimum FLIM-FRET and high mobility. These data reveal the kinetics of FcRn-ligand binding within endosomal structures for recruitment into transport carriers for recycling. These approaches have wide applicability for analyses of intracellular ligand-receptor interactions.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Macrófagos/metabolismo , Receptores Fc/metabolismo , Albúminas/metabolismo , Animales , Endocitosis/fisiología , Endosomas/metabolismo , Femenino , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia/métodos , Semivida , Células HeLa , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Cinética , Ligandos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente/métodos , Unión Proteica , Receptores Fc/fisiología
4.
Fac Rev ; 10: 50, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34195689

RESUMEN

Mechanistic (or mammalian) target of rapamycin complex 1 (mTORC1) is a major signalling kinase in cells that regulates proliferation and metabolism and is controlled by extrinsic and intrinsic signals. The lysosome has received considerable attention as a major hub of mTORC1 activation. However, mTOR has also been located to a variety of other intracellular sites, indicating the possibility of spatial regulation of mTORC1 signalling within cells. In particular, there have been numerous recent reports of mTORC1 activation associated with the Golgi apparatus. Here, we review the evidence for the regulation of mTORC1 signalling at the Golgi in mammalian cells. mTORC1 signalling is closely linked to the morphology of the Golgi architecture; a number of Golgi membrane tethers/scaffolds that influence Golgi architecture in mammalian cells that directly or indirectly regulate mTORC1 activation have been identified. Perturbation of the Golgi mTORC1 pathway arising from fragmentation of the Golgi has been shown to promote oncogenesis. Here, we highlight the potential mechanisms for the activation mTORC1 at the Golgi, which is emerging as a major site for mTORC1 signalling.

5.
Membranes (Basel) ; 10(8)2020 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756454

RESUMEN

Macropinocytosis is a unique pathway of endocytosis characterised by the nonspecific internalisation of large amounts of extracellular fluid, solutes and membrane in large endocytic vesicles known as macropinosomes. Macropinocytosis is important in a range of physiological processes, including antigen presentation, nutrient sensing, recycling of plasma proteins, migration and signalling. It has become apparent in recent years from the study of specialised cells that there are multiple pathways of macropinocytosis utilised by different cell types, and some of these pathways are triggered by different stimuli. Understanding the physiological function of macropinocytosis requires knowledge of the regulation and fate of the macropinocytosis pathways in a range of cell types. Here, we compare the mechanisms of macropinocytosis in different primary and immortalised cells, identify the gaps in knowledge in the field and discuss the potential approaches to analyse the function of macropinocytosis in vivo.

6.
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
7.
FEBS Lett ; 593(17): 2289-2305, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31378930

RESUMEN

In addition to the classical functions of the Golgi in membrane transport and glycosylation, the Golgi apparatus of mammalian cells is now recognised to contribute to the regulation of a range of cellular processes, including mitosis, DNA repair, stress responses, autophagy, apoptosis and inflammation. These processes are often mediated, either directly or indirectly, by membrane scaffold molecules, such as golgins and GRASPs which are located on Golgi membranes. In many cases, these scaffold molecules also link the actin and microtubule cytoskeleton and influence Golgi morphology. An emerging theme is a strong relationship between the morphology of the Golgi and regulation of a variety of signalling pathways. Here, we review the molecular regulation of the morphology of the Golgi, especially the role of the golgins and other scaffolds in the interaction with the microtubule and actin networks. In addition, we discuss the impact of the modulation of the Golgi ribbon in various diseases, such as neurodegeneration and cancer, to the pathology of disease.


Asunto(s)
Citoesqueleto/metabolismo , Aparato de Golgi/metabolismo , Transducción de Señal , Animales , Humanos
8.
Front Cell Dev Biol ; 7: 112, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31334231

RESUMEN

In vertebrate cells the Golgi consists of individual stacks fused together into a compact ribbon structure. The function of the ribbon structure of the Golgi has only begun to be appreciated (De Matteis et al., 2008; Gosavi and Gleeson, 2017; Wei and Seemann, 2017). Recent advances have identified a role for the Golgi in the regulation of a broad range of cellular processes and of particular interest is that the modulation of the Golgi ribbon is associated with regulation of a number of signaling pathways (Makhoul et al., 2018). Various cell responses, such as inflammation, and various disorders and diseases, including neurodegeneration and cancer, are associated with the loss of the Golgi ribbon and the appearance of a dispersed or semi-dispersed Golgi. Often the dispersed Golgi is referred to as a "fragmented" morphology. However, the description of a dispersed Golgi ribbon as "fragmented" is inadequate as it does not accurately define the morphological state of the Golgi. This issue is particularly relevant as there are an increasing number of reports describing Golgi fragmentation under physiological and pathological conditions. Knowledge of the precise Golgi architecture is relevant to an appreciation of the functional status of the Golgi apparatus and the underlying molecular mechanism for the contribution of the Golgi to different cellular processes. Here we propose a classification to define the various morphological states of the non-ribbon architecture of the Golgi in mammalian cells as a guide to more precisely define the relationship between the morphological and functional status of this organelle.

9.
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
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.
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
12.
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
13.
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
14.
Eur J Immunol ; 44(7): 2048-58, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24740292

RESUMEN

The expression of the Ikaros transcription factor family member, Helios, has been shown to be associated with T-cell tolerance in both the thymus and the periphery. To better understand the importance of Helios in tolerance pathways, we have examined the expression of Helios in TCR-transgenic T cells specific for the gastric H(+) /K(+) ATPase, the autoantigen target in autoimmune gastritis. Analysis of H(+) /K(+) ATPase-specific T cells in mice with different patterns of H(+) /K(+) ATPase expression revealed that, in addition to the expression of Helios in CD4(+) Foxp3(+) regulatory T (Treg) cells, Helios is expressed by a large proportion of CD4(+) Foxp3(-) T cells in both the thymus and the paragastric lymph node (PgLN), which drains the stomach. In the thymus, Helios was expressed by H(+) /K(+) ATPase-specific thymocytes that were undergoing negative selection. In the periphery, Helios was expressed in H(+) /K(+) ATPase-specific CD4(+) T cells following H(+) /K(+) ATPase presentation and was more highly expressed when T-cell activation occurred in the absence of inflammation. Analysis of purified H(+) /K(+) ATPase-specific CD4(+) Foxp3(-) Helios(+) T cells demonstrated that they were functionally anergic. These results demonstrate that Helios is expressed by thymic and peripheral T cells that are being driven to tolerance in response to a genuine autoantigen.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Tolerancia Inmunológica , Linfocitos T Reguladores/inmunología , Timo/inmunología , Factores de Transcripción/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/análisis , Gastritis/inmunología , ATPasa Intercambiadora de Hidrógeno-Potásio/fisiología , Ratones , Ratones Endogámicos BALB C
15.
Traffic ; 15(5): 572-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24612275

RESUMEN

Current methods for the quantitation of membrane protein trafficking rely heavily on microscopy, which has limited quantitative capacity for analyses of cell populations and is cumbersome to perform. Here we describe a simple flow cytometry-based method that circumvents these limitations. The method utilizes fluorescent pulse-width measurements as a highly sensitive indicator to monitor the changes in intracellular distributions of a fluorescently labelled molecule in a cell. Pulse-width analysis enabled us to discriminate cells with target proteins in different intracellular locations including Golgi, lyso-endosomal network and the plasma membrane, as well as detecting morphological changes in organelles such as Golgi perturbation. The movement of endogenous and exogenous retrograde cargo was tracked from the plasma membrane-to-endosomes-to-Golgi, by decreasing pulse-width values. A block in transport upon RNAi-mediated ablation of transport machinery was readily quantified, demonstrating the versatility of this technique to identify pathway inhibitors. We also showed that pulse-width can be exploited to sort and recover cells based on different intracellular staining patterns, e.g. early endosomes and Golgi, opening up novel downstream applications. Overall, the method provides new capabilities for viewing membrane transport in thousands of cells per minute, unbiased analysis of the trafficking of cargo, and the potential for rapid screening of inhibitors of trafficking pathways.


Asunto(s)
Endosomas/metabolismo , Citometría de Flujo/métodos , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/fisiología , Endosomas/fisiología , Aparato de Golgi/fisiología , Células HeLa , Humanos , Lisosomas/fisiología
16.
Traffic ; 14(9): 997-1013, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23773724

RESUMEN

ß-Secretase (BACE1) cleavage of the amyloid precursor protein (APP) represents the initial step in the formation of the Alzheimer's disease associated amyloidogenic Aß peptide. Substantive evidence indicates that APP processing by BACE1 is dependent on intracellular sorting of this enzyme. Nonetheless, knowledge of the intracellular trafficking pathway of internalised BACE1 remains in doubt. Here we show that cell surface BACE1 is rapidly internalised by the AP2/clathrin dependent pathway in transfected cells and traffics to early endosomes and Rab11-positive, juxtanuclear recycling endosomes, with very little transported to the TGN as has been previously suggested. Moreover, BACE1 is predominantly localised to the early and recycling endosome compartments in different cell types, including neuronal cells. In contrast, the majority of internalised wild-type APP traffics to late endosomes/lysosomes. To explore the relevance of the itinerary of BACE1 on APP processing, we generated a BACE1 chimera containing the cytoplasmic tail of TGN38 (BACE/TGN38), which cycles between the cell surface and TGN in an AP2-dependent manner. Wild-type BACE1 is less efficient in Aß production than the BACE/TGN38 chimera, highlighting the relevance of the itinerary of BACE1 on APP processing. Overall the data suggests that internalised BACE1 and APP diverge at early endosomes and that Aß biogenesis is regulated in part by the recycling itinerary of BACE1.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/biosíntesis , ARN Largo no Codificante/metabolismo , Animales , Células CHO , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Clatrina/metabolismo , Cricetulus , Citoplasma/metabolismo , Endosomas/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Transporte de Proteínas
17.
PLoS One ; 8(2): e57034, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437303

RESUMEN

Lipopolysaccharide (LPS)-activated macrophages secrete pro-inflammatory cytokines, including tumor necrosis factor (TNF) to elicit innate immune responses. Secretion of these cytokines is also a major contributing factor in chronic inflammatory disease. In previous studies we have begun to elucidate the pathways and molecules that mediate the intracellular trafficking and secretion of TNF. Rab6a and Rab6a' (collectively Rab6) are trans-Golgi-localized GTPases known for roles in maintaining Golgi structure and Golgi-associated trafficking. We found that induction of TNF secretion by LPS promoted the selective increase of Rab6 expression. Depletion of Rab6 (via siRNA and shRNA) resulted in reorganization of the Golgi ribbon into more compact structures that at the resolution of electron microcopy consisted of elongated Golgi stacks that likely arose from fusion of smaller Golgi elements. Concomitantly, the delivery of TNF to the cell surface and subsequent release into the media was reduced. Dominant negative mutants of Rab6 had similar effects in disrupting TNF secretion. In live cells, Rab6-GFP were localized on trans-Golgi network (TGN)-derived tubular carriers demarked by the golgin p230. Rab6 depletion and inactive mutants altered carrier egress and partially reduced p230 membrane association. Our results show that Rab6 acts on TNF trafficking at the level of TGN exit in tubular carriers and our findings suggest Rab6 may stabilize p230 on the tubules to facilitate TNF transport. Both Rab6 isoforms are needed in macrophages for Golgi stack organization and for the efficient post-Golgi transport of TNF. This work provides new insights into Rab6 function and into the role of the Golgi complex in cytokine secretion in inflammatory macrophages.


Asunto(s)
Aparato de Golgi/metabolismo , Macrófagos/metabolismo , Factores de Necrosis Tumoral/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Autoantígenos/metabolismo , Brefeldino A/farmacología , Línea Celular , Membrana Celular/metabolismo , Proteínas de la Matriz de Golgi , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Estabilidad Proteica , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al GTP rab/genética
18.
Eur J Immunol ; 43(5): 1286-96, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23420509

RESUMEN

Autoimmune disease can be prevented with immunosuppressive agents; however, the effectiveness of these treatments in advanced stage of disease and the fate of pathogenic T cells following such treatments are not clear. In this study we demonstrate that a single dose of in vitro-induced Treg cells (iTreg cells) resulted in the functional repair and restitution of stomach tissue that had been severely damaged in advanced autoimmune gastritis. iTreg cells caused depletion or inactivation of autoreactive naïve T cells that were antigen inexperienced, however, autoreactive effector/memory T cells persisted in treated mice, resulting in residual cellular infiltrates within the repaired stomach tissue. The persisting autoreactive T cells were able to rapidly cause autoimmune disease if iTreg cells were removed. Similar data were obtained from mice treated continuously with corticosteroid, in that there was substantial restitution of the gastric mucosa; however, effector T cells persisted and rapidly caused pathology following drug removal. Therefore, iTreg cells or corticosteroid can suppress pathogenic autoreactive cells in advanced autoimmune disease, reversing tissue damage and improving tissue function. However, the persistence of pathogenic T cells represents a disease risk.


Asunto(s)
Enfermedades Autoinmunes/patología , Gastritis/patología , Inmunoterapia Adoptiva , Linfocitos T Citotóxicos/patología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Corticoesteroides/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Comunicación Celular , Modelos Animales de Enfermedad , Gastritis/inmunología , Gastritis/terapia , Terapia de Inmunosupresión , Activación de Linfocitos , Ratones , Prednisolona/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/trasplante
19.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23154976

RESUMEN

Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.


Asunto(s)
Ácido Gástrico/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Canal de Potasio KCNQ1/metabolismo , Células Parietales Gástricas/enzimología , Animales , Membrana Celular/enzimología , Cromanos/farmacología , Citoplasma/enzimología , Endosomas/enzimología , Transferencia Resonante de Energía de Fluorescencia , Histamina/metabolismo , Canal de Potasio KCNQ1/efectos de los fármacos , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Transporte de Proteínas , Sulfonamidas/farmacología , Factores de Tiempo , Proteínas de Unión al GTP rab/metabolismo
20.
Nat Methods ; 9(5): 467-70, 2012 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-22426490

RESUMEN

We applied pulse-shape analysis (PulSA) to monitor protein localization changes in mammalian cells by flow cytometry. PulSA enabled high-throughput tracking of protein aggregation, translocation from the cytoplasm to the nucleus and trafficking from the plasma membrane to the Golgi as well as stress-granule formation. Combining PulSA with tetracysteine-based oligomer sensors in a cell model of Huntington's disease enabled further separation of cells enriched with monomers, oligomers and inclusion bodies.


Asunto(s)
Citometría de Flujo/métodos , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Aparato de Golgi/metabolismo , Humanos , Proteína Huntingtina , Cuerpos de Inclusión/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA