Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(16)2023 Aug 19.
Article in English | MEDLINE | ID: mdl-37629161

ABSTRACT

Autophagy is a tightly regulated catabolic process involved in the degradation and recycling of proteins and organelles. Ubiquitination plays an important role in the regulation of autophagy. Vacuole Membrane Protein 1 (VMP1) is an essential autophagy protein. The expression of VMP1 in pancreatic cancer stem cells carrying the activated Kirsten rat sarcoma viral oncogene homolog (KRAS) triggers autophagy and enables therapy resistance. Using biochemical and cellular approaches, we identified ubiquitination as a post-translational modification of VMP1 from the initial steps in autophagosome biogenesis. VMP1 remains ubiquitinated as part of the autophagosome membrane throughout autophagic flux until autolysosome formation. However, VMP1 is not degraded by autophagy, nor by the ubiquitin-proteasomal system. Mass spectrometry and immunoprecipitation showed that the cell division cycle protein cdt2 (Cdt2), the substrate recognition subunit of the E3 ligase complex associated with cancer, cullin-RING ubiquitin ligase complex 4 (CRL4), is a novel interactor of VMP1 and is involved in VMP1 ubiquitination. VMP1 ubiquitination decreases under the CRL inhibitor MLN4924 and increases with Cdt2 overexpression. Moreover, VMP1 recruitment and autophagosome formation is significantly affected by CRL inhibition. Our results indicate that ubiquitination is a novel post-translational modification of VMP1 during autophagy in human tumor cells. VMP1 ubiquitination may be of clinical relevance in tumor-cell-therapy resistance.


Subject(s)
Membrane Proteins , Neoplasms , Protein Processing, Post-Translational , Humans , Autophagy/genetics , Macroautophagy , Membrane Proteins/metabolism , Ubiquitin , Ubiquitination
2.
J Vis Exp ; (194)2023 04 28.
Article in English | MEDLINE | ID: mdl-37184277

ABSTRACT

Autophagy is a specialized catabolic process that selectively degrades cytoplasmic components, including proteins and damaged organelles. Autophagy allows cells to physiologically respond to stress stimuli and, thus, maintain cellular homeostasis. Cancer cells might modulate their autophagy levels to adapt to adverse conditions such as hypoxia, nutrient deficiency, or damage caused by chemotherapy. Ductal pancreatic adenocarcinoma is one of the deadliest types of cancer. Pancreatic cancer cells have high autophagy activity due to the transcriptional upregulation and post-translational activation of autophagy proteins. Here, the PANC-1 cell line was used as a model of pancreatic human cancer cells, and the AR42J pancreatic acinar cell line was used as a physiological model of highly differentiated mammalian cells. This study used the immunofluorescence of microtubule-associated protein light chain 3 (LC3) as an indicator of the status of autophagy activation. LC3 is an autophagy protein that, in basal conditions, shows a diffuse pattern of distribution in the cytoplasm (known as LC3-I in this condition). Autophagy induction triggers the conjugation of LC3 to phosphatidylethanolamine on the surface of newly formed autophagosomes to form LC3-II, a membrane-bound protein that aids in the formation and expansion of autophagosomes. To quantify the number of labeled autophagic structures, the open-source software FIJI was utilized with the aid of the "3D Objects Counter" tool. The measure of the autophagic levels both in physiological conditions and in cancer cells allows us to study the modulation of autophagy under diverse conditions such as hypoxia, chemotherapy treatment, or the knockdown of certain proteins.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Animals , Humans , HeLa Cells , Autophagy/physiology , Microtubule-Associated Proteins/metabolism , Fluorescent Antibody Technique , Hypoxia , Mammals/metabolism , Pancreatic Neoplasms
3.
Cell Rep ; 40(3): 111092, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858571

ABSTRACT

The integrated stress response (ISR) plays a pivotal role in adaptation of translation machinery to cellular stress. Here, we demonstrate an ISR-independent osmoadaptation mechanism involving reprogramming of translation via coordinated but independent actions of mTOR and plasma membrane amino acid transporter SNAT2. This biphasic response entails reduced global protein synthesis and mTOR signaling followed by translation of SNAT2. Induction of SNAT2 leads to accumulation of amino acids and reactivation of mTOR and global protein synthesis, paralleled by partial reversal of the early-phase, stress-induced translatome. We propose SNAT2 functions as a molecular switch between inhibition of protein synthesis and establishment of an osmoadaptive translation program involving the formation of cytoplasmic condensates of SNAT2-regulated RNA-binding proteins DDX3X and FUS. In summary, we define key roles of SNAT2 in osmotolerance.


Subject(s)
Amino Acid Transport System A , Amino Acids , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism
4.
Autophagy ; 18(4): 841-859, 2022 04.
Article in English | MEDLINE | ID: mdl-34424124

ABSTRACT

The Mycobacterium ulcerans exotoxin, mycolactone, is responsible for the immunosuppression and tissue necrosis that characterizes Buruli ulcer. Mycolactone inhibits SEC61-dependent co-translational translocation of proteins into the endoplasmic reticulum and the resultant cytosolic translation triggers degradation of mislocalized proteins by the ubiquitin-proteasome system. Inhibition of SEC61 by mycolactone also activates multiple EIF2S1/eIF2α kinases in the integrated stress response (ISR). Here we show mycolactone increased canonical markers of selective macroautophagy/autophagy LC3B-II, ubiquitin and SQSTM1/p62 in diverse disease-relevant primary cells and cell lines. Increased formation of puncta positive for the early autophagy markers WIPI2, RB1CC1/FIP200 and ATG16L1 indicates increased initiation of autophagy. The mycolactone response was SEC61A1-dependent and involved a pathway that required RB1CC1 but not ULK. Deletion of Sqstm1 reduced cell survival in the presence of mycolactone, suggesting this response protects against the increased cytosolic protein burden caused by the toxin. However, reconstitution of baseline SQSTM1 expression in cells lacking all autophagy receptor proteins could not rescue viability. Translational regulation by EIF2S1 in the ISR plays a key role in the autophagic response to mycolactone. Mycolactone-dependent induction of SQSTM1 was reduced in eif2ak3-/-/perk-/- cells while the p-EIF2S1 antagonist ISRIB reversed the upregulation of SQSTM1 and reduced RB1CC1, WIPI2 and LC3B puncta formation. Increased SQSTM1 staining could be seen in Buruli ulcer patient skin biopsy samples, reinforcing genetic data that suggests autophagy is relevant to disease pathology. Since selective autophagy and the ISR are both implicated in neurodegeneration, cancer and inflammation, the pathway uncovered here may have a broad relevance to human disease.Abbreviations: ATF4: activating transcription factor 4; ATG: autophagy related; BAF: bafilomycin A1; ATG16L1: autophagy related 16 like 1; BU: Buruli ulcer; CQ: chloroquine; EIF2AK3: eukaryotic translation initiation factor 2 alpha kinase 3; CALCOCO2: calcium binding and coiled-coil domain 2; DMSO: dimethyl sulfoxide; EIF2S1: eukaryotic translation initiation factor 2 subunit alpha; ER: endoplasmic reticulum; GFP: green fluorescent protein; HDMEC: human dermal microvascular endothelial cells; HFFF: human fetal foreskin fibroblasts; ISR: integrated stress response; ISRIB: integrated stress response inhibitor; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MEF: mouse embryonic fibroblast; Myco: mycolactone; NBR1: NBR1 autophagy cargo receptor; NFE2L2: nuclear factor, erythroid 2 like 2; OPTN: optineurin; PFA: paraformaldehyde; PtdIns3P: phosphatidylinositol-3-phosphate; RB1CC1: RB1-inducible coiled coil 1; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; ULK: unc-51 like autophagy activating kinase; UPS: ubiquitin-proteasome system; WIPI: WD repeat domain, phosphoinositide interacting; WT: wild type.


Subject(s)
Autophagy , Buruli Ulcer , Eukaryotic Initiation Factor-2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Macrolides , Mice , Prokaryotic Initiation Factor-2/metabolism , Proteasome Endopeptidase Complex/metabolism , SEC Translocation Channels/metabolism , Sequestosome-1 Protein/metabolism , Ubiquitin/metabolism
5.
EMBO J ; 40(10): e103563, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33932238

ABSTRACT

The early secretory pathway and autophagy are two essential and evolutionarily conserved endomembrane processes that are finely interlinked. Although growing evidence suggests that intracellular trafficking is important for autophagosome biogenesis, the molecular regulatory network involved is still not fully defined. In this study, we demonstrate a crucial effect of the COPII vesicle-related protein TFG (Trk-fused gene) on ULK1 puncta number and localization during autophagy induction. This, in turn, affects formation of the isolation membrane, as well as the correct dynamics of association between LC3B and early ATG proteins, leading to the proper formation of both omegasomes and autophagosomes. Consistently, fibroblasts derived from a hereditary spastic paraparesis (HSP) patient carrying mutated TFG (R106C) show defects in both autophagy and ULK1 puncta accumulation. In addition, we demonstrate that TFG activity in autophagy depends on its interaction with the ATG8 protein LC3C through a canonical LIR motif, thereby favouring LC3C-ULK1 binding. Altogether, our results uncover a link between TFG and autophagy and identify TFG as a molecular scaffold linking the early secretion pathway to autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Blotting, Western , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Proteins/genetics , RNA Interference
6.
Autophagy ; 17(5): 1131-1141, 2021 05.
Article in English | MEDLINE | ID: mdl-32320309

ABSTRACT

During macroautophagy/autophagy, the ULK complex nucleates autophagic precursors, which give rise to autophagosomes. We analyzed, by live imaging and mathematical modeling, the translocation of ATG13 (part of the ULK complex) to the autophagic puncta in starvation-induced autophagy and ivermectin-induced mitophagy. In nonselective autophagy, the intensity and duration of ATG13 translocation approximated a normal distribution, whereas wortmannin reduced this effect and shifted to a log-normal distribution. During mitophagy, multiple translocations of ATG13 with increasing time between peaks were observed. We hypothesized that these multiple translocations arise because the engulfment of mitochondrial fragments required successive nucleation of phagophores on the same target, and a mathematical model based on this idea reproduced the oscillatory behavior. Significantly, model and experimental data were also in agreement that the number of ATG13 translocations is directly proportional to the diameter of the targeted mitochondrial fragments. Thus, our data provide novel insights into the early dynamics of selective and nonselective autophagy.Abbreviations: ATG: autophagy related 13; CFP: cyan fluorescent protein; dsRED: Discosoma red fluorescent protein; GABARAP: GABA type A receptor-associated protein; GFP: green fluorescent protein; IVM: ivermectin; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3P: PtdIns-3-phosphate; ULK: unc-51 like autophagy activating kinase.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/physiology , Mitophagy/physiology , Models, Theoretical , Autophagosomes/metabolism , HEK293 Cells , Humans , Microtubule-Associated Proteins/metabolism
8.
Cell Metab ; 32(4): 591-604.e7, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32738205

ABSTRACT

Regulatory T cells (Tregs) are vital for the maintenance of immune homeostasis, while their dysfunction constitutes a cardinal feature of autoimmunity. Under steady-state conditions, mitochondrial metabolism is critical for Treg function; however, the metabolic adaptations of Tregs during autoimmunity are ill-defined. Herein, we report that elevated mitochondrial oxidative stress and a robust DNA damage response (DDR) associated with cell death occur in Tregs in individuals with autoimmunity. In an experimental autoimmune encephalitis (EAE) mouse model of autoimmunity, we found a Treg dysfunction recapitulating the features of autoimmune Tregs with a prominent mtROS signature. Scavenging of mtROS in Tregs of EAE mice reversed the DDR and prevented Treg death, while attenuating the Th1 and Th17 autoimmune responses. These findings highlight an unrecognized role of mitochondrial oxidative stress in defining Treg fate during autoimmunity, which may facilitate the design of novel immunotherapies for diseases with disturbed immune tolerance.


Subject(s)
Autoimmunity/immunology , Mitochondria/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
9.
Dev Cell ; 50(5): 627-643.e5, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31353311

ABSTRACT

The dynamics and coordination between autophagy machinery and selective receptors during mitophagy are unknown. Also unknown is whether mitophagy depends on pre-existing membranes or is triggered on the surface of damaged mitochondria. Using a ubiquitin-dependent mitophagy inducer, the lactone ivermectin, we have combined genetic and imaging experiments to address these questions. Ubiquitination of mitochondrial fragments is required the earliest, followed by auto-phosphorylation of TBK1. Next, early essential autophagy proteins FIP200 and ATG13 act at different steps, whereas ULK1 and ULK2 are dispensable. Receptors act temporally and mechanistically upstream of ATG13 but downstream of FIP200. The VPS34 complex functions at the omegasome step. ATG13 and optineurin target mitochondria in a discontinuous oscillatory way, suggesting multiple initiation events. Targeted ubiquitinated mitochondria are cradled by endoplasmic reticulum (ER) strands even without functional autophagy machinery and mitophagy adaptors. We propose that damaged mitochondria are ubiquitinated and dynamically encased in ER strands, providing platforms for formation of the mitophagosomes.


Subject(s)
Endoplasmic Reticulum/metabolism , Mitophagy , Ubiquitination , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Cells, Cultured , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , TNF Receptor-Associated Factor 2/metabolism
10.
Elife ; 52016 10 11.
Article in English | MEDLINE | ID: mdl-27725083

ABSTRACT

Amino acids are essential activators of mTORC1 via a complex containing RAG GTPases, RAGULATOR and the vacuolar ATPase. Sensing of amino acids causes translocation of mTORC1 to lysosomes, an obligate step for activation. To examine the spatial and temporal dynamics of this translocation, we used live imaging of the mTORC1 component RAPTOR and a cell permeant fluorescent analogue of di-leucine methyl ester. Translocation to lysosomes is a transient event, occurring within 2 min of aa addition and peaking within 5 min. It is temporally coupled with fluorescent leucine appearance in lysosomes and is sustained in comparison to aa stimulation. Sestrin2 and the vacuolar ATPase are negative and positive regulators of mTORC1 activity in our experimental system. Of note, phosphorylation of canonical mTORC1 targets is delayed compared to lysosomal translocation suggesting a dynamic and transient passage of mTORC1 from the lysosomal surface before targetting its substrates elsewhere.


Subject(s)
Amino Acids/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , HEK293 Cells , Humans , Intravital Microscopy , Nuclear Proteins/metabolism , Protein Transport , Regulatory-Associated Protein of mTOR/analysis , Spatio-Temporal Analysis , Vacuolar Proton-Translocating ATPases/metabolism
11.
Nat Commun ; 7: 12420, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510922

ABSTRACT

Autophagosome formation requires sequential translocation of autophagy-specific proteins to membranes enriched in PI3P and connected to the ER. Preceding this, the earliest autophagy-specific structure forming de novo is a small punctum of the ULK1 complex. The provenance of this structure and its mode of formation are unknown. We show that the ULK1 structure emerges from regions, where ATG9 vesicles align with the ER and its formation requires ER exit and coatomer function. Super-resolution microscopy reveals that the ULK1 compartment consists of regularly assembled punctate elements that cluster in progressively larger spherical structures and associates uniquely with the early autophagy machinery. Correlative electron microscopy after live imaging shows tubulovesicular membranes present at the locus of this structure. We propose that the nucleation of autophagosomes occurs in regions, where the ULK1 complex coalesces with ER and the ATG9 compartment.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Autophagosomes/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Intracellular Membranes/metabolism , Lysosomes/metabolism , Microscopy, Confocal , Microscopy, Electron , Mitochondria/metabolism , Plasmids/metabolism , Protein Transport
12.
Biochem Soc Trans ; 42(5): 1389-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25233420

ABSTRACT

Autophagosomes form in eukaryotic cells in response to starvation or to other stress conditions brought about by the unwanted presence in the cytosol of pathogens, damaged organelles or aggregated protein assemblies. The uniqueness of autophagosomes is that they form de novo and that they are the only double-membraned vesicles known in cells, having arisen from flat membrane sheets which have expanded and self-closed. The various steps describing their formation as well as most of the protein and lipid components involved have been identified. Furthermore, the hierarchical relationships among the components are well documented, and the mechanistic rationale for some of these hierarchies has been revealed. In the present review, we try to provide a current view of the process of autophagosome formation in mammalian cells, emphasizing along the way gaps in our knowledge that need additional work.


Subject(s)
Autophagy , Eukaryotic Cells/physiology , Models, Biological , Phagosomes/metabolism , Animals , Humans , Kinetics , Signal Transduction
13.
Curr Protoc Cytom ; 69: 12.34.1-12.34.16, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24984962

ABSTRACT

Autophagy is a membrane-trafficking pathway activated to deliver cytosolic material for degradation to lysosomes through a novel membrane compartment, the autophagosome. Fluorescence microscopy is the most common method used to visualize proteins inside cells, and it is widely used in the autophagy field. To distinguish it from the cellular background, the protein of interest (POI) is either fused with a genetically encoded fluorescent protein or stained with an antibody that is conjugated to an inorganic fluorescent compound. Genetic tagging of the POI allows its visualization in live cells, while immunostaining of the POI requires the fixation of cells and the permeabilization of cell membranes. Here we describe detailed protocols on how to visualize autophagy dynamics using fluorescence microscopy in live and fixed cells. We discuss the critical parameters of each technique, their advantages, and why the robustness is increased when they are used in tandem.


Subject(s)
Autophagy/physiology , Cell Membrane/metabolism , Biological Transport, Active/physiology , Cell Line , Cell Membrane/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Microscopy, Fluorescence/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
14.
J Cell Sci ; 126(Pt 22): 5224-38, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24013547

ABSTRACT

Induction of autophagy requires the ULK1 protein kinase complex and the Vps34 lipid kinase complex. PtdIns3P synthesised by Vps34 accumulates in omegasomes, membrane extensions of the ER within which some autophagosomes form. The ULK1 complex is thought to target autophagosomes independently of PtdIns3P, and its functional relationship to omegasomes is unclear. Here we show that the ULK1 complex colocalises with omegasomes in a PtdIns3P-dependent way. Live-cell imaging of Atg13 (a ULK1 complex component), omegasomes and LC3 establishes and annotates for the first time a complete sequence of steps leading to autophagosome formation, as follows. Upon starvation, the ULK1 complex forms puncta associated with the ER and sporadically with mitochondria. If PtdIns3P is available, these puncta become omegasomes. Subsequently, the ULK1 complex exits omegasomes and autophagosomes bud off. If PtdIns3P is unavailable, ULK1 puncta are greatly reduced in number and duration. Atg13 contains a region with affinity for acidic phospholipids, required for translocation to punctate structures and autophagy progression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Class III Phosphatidylinositol 3-Kinases/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phagosomes/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Vacuoles/metabolism
15.
J Vis Exp ; (77)2013 Jul 27.
Article in English | MEDLINE | ID: mdl-23929131

ABSTRACT

Autophagy is a cellular response triggered by the lack of nutrients, especially the absence of amino acids. Autophagy is defined by the formation of double membrane structures, called autophagosomes, that sequester cytoplasm, long-lived proteins and protein aggregates, defective organelles, and even viruses or bacteria. Autophagosomes eventually fuse with lysosomes leading to bulk degradation of their content, with the produced nutrients being recycled back to the cytoplasm. Therefore, autophagy is crucial for cell homeostasis, and dysregulation of autophagy can lead to disease, most notably neurodegeneration, ageing and cancer. Autophagosome formation is a very elaborate process, for which cells have allocated a specific group of proteins, called the core autophagy machinery. The core autophagy machinery is functionally complemented by additional proteins involved in diverse cellular processes, e.g. in membrane trafficking, in mitochondrial and lysosomal biology. Coordination of these proteins for the formation and degradation of autophagosomes constitutes the highly dynamic and sophisticated response of autophagy. Live cell imaging allows one to follow the molecular contribution of each autophagy-related protein down to the level of a single autophagosome formation event and in real time, therefore this technique offers a high temporal and spatial resolution. Here we use a cell line stably expressing GFP-DFCP1, to establish a spatial and temporal context for our analysis. DFCP1 marks omegasomes, which are precursor structures leading to autophagosomes formation. A protein of interest (POI) can be marked with either a red or cyan fluorescent tag. Different organelles, like the ER, mitochondria and lysosomes, are all involved in different steps of autophagosome formation, and can be marked using a specific tracker dye. Time-lapse microscopy of autophagy in this experimental set up, allows information to be extracted about the fourth dimension, i.e. time. Hence we can follow the contribution of the POI to autophagy in space and time.


Subject(s)
Autophagy/physiology , Phagosomes/chemistry , Single-Cell Analysis/methods , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Carrier Proteins/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Microscopy, Fluorescence/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Transfection
16.
Autophagy ; 9(9): 1407-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23800949

ABSTRACT

Basal autophagy-here defined as macroautophagic activity during cellular growth in normal medium containing amino acids and serum-appears to be highly active in many cell types and in animal tissues. Here we characterized this pathway in mammalian HEK 293 cells. First, we examined, side by side, three compounds that are widely used to reveal basal autophagy by blocking maturation of autophagosomes: bafilomycin A 1 (BafA1), chloroquine and vinblastine. Only BafA1 appeared to be without complicating side effects. Chloroquine partially inhibited mechanistic target of rapamycin (MTOR) activity, which would induce autophagy induction as well as block autophagosome maturation. Vinblastine caused the distribution of early omegasome components into punctate phagophore assembly sites, and therefore it would also induce autophagy, complicating interpretation. Basal autophagy was significantly sensitive to inhibition by wortmannin, and therefore required formation of phosphatidylinositol 3-phosphate (PtdIns3P), but it was twice as resistant to wortmannin as starvation-induced autophagy. We also determined that basal autophagy was significantly suppressed by MTOR activation brought about by overexpression of RHEB or activated RAGs. Finally we investigated the spatial relationship of nascent autophagosomes to the endoplasmic reticulum (ER) or to mitochondria by live imaging experiments under conditions that reveal basal autophagy (with BafA1 treatment), or upon MTOR inactivation (which would result in autophagy induction). Side-by-side comparison showed that under both basal and induced autophagy, 100% of autophagosomes first appeared in close proximity to ER strands. In parallel measurements, 40% were in close proximity to mitochondria under both conditions. We concluded that in HEK 293 cells, basal autophagy is mechanistically similar to that induced by MTOR inactivation in all aspects examined.


Subject(s)
Autophagy , Androstadienes/pharmacology , Biomarkers/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Activation/drug effects , HEK293 Cells , Humans , Macrolides/pharmacology , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , TOR Serine-Threonine Kinases/metabolism , Wortmannin
17.
Biochem Soc Trans ; 40(1): 37-43, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260663

ABSTRACT

The simple phosphoinositide PtdIns3P has been shown to control cell growth downstream of amino acid signalling and autophagy downstream of amino acid withdrawal. These opposing effects depend in part on the existence of distinct complexes of Vps34 (vacuolar protein sorting 34), the kinase responsible for the majority of PtdIns3P synthesis in cells: one complex is activated after amino acid withdrawal to induce autophagy and another regulates mTORC1 (mammalian target of rapamycin complex 1) activation when amino acids are present. However, lipid-dependent signalling almost always exhibits a spatial dimension, related to the site of formation of the lipid signal. In the case of PtdIns3P-regulated autophagy induction, recent data suggest that PtdIns3P accumulates in a membrane compartment dynamically connected to the endoplasmic reticulum that constitutes a platform for the formation of some autophagosomes. For PtdIns3P-regulated mTORC1 activity, a spatial context is not yet known: several possibilities can be envisaged based on the known effects of PtdIns3P on the endocytic system and on recent data suggesting that activation of mTORC1 depends on its localization on lysosomes.


Subject(s)
Amino Acids/metabolism , Autophagy , Cell Proliferation , Class III Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/physiology , Amino Acids/deficiency , Animals , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphatidylinositols/metabolism , Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
18.
Autophagy ; 7(11): 1335-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21799305

ABSTRACT

Autophagy is a cellular response to starvation which generates autophagosomes to carry cellular organelles and long-lived proteins to lysosomes for degradation. Degradation through autophagy can provide an innate defence against virus infection, or conversely autophagosomes can promote infection by facilitating assembly of replicase proteins. We demonstrate that the avian coronavirus, Infectious Bronchitis Virus (IBV) activates autophagy. A screen of individual IBV non-structural proteins (nsps) showed that autophagy was activated by IBV nsp6. This property was shared with nsp6 of mammalian coronaviruses Mouse Hepatitis Virus, and Severe Acute Respiratory Syndrome Virus, and the equivalent nsp5-7 of the arterivirus Porcine Reproductive and Respiratory Syndrome Virus. These multiple-spanning transmembrane proteins located to the endoplasmic reticulum (ER) where they generated Atg5 and LC3II-positive vesicles, and vesicle formation was dependent on Atg5 and class III PI3 kinase. The vesicles recruited double FYVE-domain containing protein (DFCP) indicating localised concentration of phosphatidylinositol 3 phosphate, and therefore shared many features with omegasomes formed from the ER in response to starvation. Omegasomes induced by viral nsp6 matured into autophagosomes that delivered LC3 to lysosomes and therefore recruited and recycled the proteins needed for autophagosome nucleation, expansion, cellular trafficking and delivery of cargo to lysosomes. The coronavirus nsp6 proteins activated omegasome and autophagosome formation independently of starvation, but activation did not involve direct inhibition of mTOR signalling, activation of sirtuin1 or induction of ER stress.


Subject(s)
Autophagy , Endoplasmic Reticulum/metabolism , Infectious bronchitis virus/metabolism , Phagosomes/metabolism , Viral Nonstructural Proteins/metabolism , Androstadienes/pharmacology , Animals , Arterivirus/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5 , Cell Line , Coronavirus Infections/pathology , Coronavirus Infections/virology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/virology , Endoplasmic Reticulum Stress/drug effects , Genome, Viral/genetics , Humans , Infectious bronchitis virus/genetics , Membrane Fusion/drug effects , Mice , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol Phosphates/pharmacology , Protein Structure, Tertiary , Sequence Deletion , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor CHOP/metabolism , Viral Nonstructural Proteins/chemistry , Wortmannin
19.
Biochim Biophys Acta ; 1791(9): 889-97, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19345277

ABSTRACT

Phospholipase D (PLD) catalyses the hydrolysis of phosphatidylcholine to generate phosphatidic acid and choline. Historically, much PLD work has been conducted in mammalian settings although genes encoding enzymes of this family have been identified in all eukaryotic organisms. Recently, important insights on PLD function are emerging from work in yeast, but much less is known about PLD in other organisms. In this review we will summarize what is known about phospholipase D in several model organisms, including C. elegans, D. discoideum, D. rerio and D. melanogaster. In the cases where knockouts are available (C. elegans, Dictyostelium and Drosophila) the PLD gene(s) appear not to be essential for viability, but several studies are beginning to identify pathways where this activity has a role. Given that the proteins in model organisms are very similar to their mammalian counterparts, we expect that future studies in model organisms will complement and extend ongoing work in mammalian settings. At the end of this review we will also provide a short update on phosphatidic acid targets, a topic last reviewed in 2006.


Subject(s)
Models, Animal , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phospholipase D/chemistry , Phospholipase D/genetics , Phylogeny
20.
J Cell Biol ; 185(1): 129-45, 2009 Apr 06.
Article in English | MEDLINE | ID: mdl-19349583

ABSTRACT

Phosphatidic acid (PA) is postulated to have both structural and signaling functions during membrane dynamics in animal cells. In this study, we show that before a critical time period during rhabdomere biogenesis in Drosophila melanogaster photoreceptors, elevated levels of PA disrupt membrane transport to the apical domain. Lipidomic analysis shows that this effect is associated with an increase in the abundance of a single, relatively minor molecular species of PA. These transport defects are dependent on the activation state of Arf1. Transport defects via PA generated by phospholipase D require the activity of type I phosphatidylinositol (PI) 4 phosphate 5 kinase, are phenocopied by knockdown of PI 4 kinase, and are associated with normal endoplasmic reticulum to Golgi transport. We propose that PA levels are critical for apical membrane transport events required for rhabdomere biogenesis.


Subject(s)
Drosophila melanogaster/ultrastructure , Phosphatidic Acids/metabolism , Photoreceptor Cells/ultrastructure , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/physiology , ADP-Ribosylation Factor 1/physiology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/physiology , Adaptor Protein Complex alpha Subunits/antagonists & inhibitors , Adaptor Protein Complex alpha Subunits/physiology , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Diacylglycerol Cholinephosphotransferase/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Dynamins/genetics , Dynamins/metabolism , Dynamins/physiology , Membrane Lipids/metabolism , Microscopy, Electron, Transmission , Phenotype , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/metabolism , Phospholipase D/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/physiology , Photoreceptor Cells/metabolism , Photoreceptor Cells/physiology , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...