Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cells ; 9(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31947769

ABSTRACT

In eukaryotic cells, autophagosomes and multivesicular bodies (MVBs) are two closely related partners in the lysosomal/vacuolar protein degradation system. Autophagosomes are double membrane-bound organelles that transport cytoplasmic components, including proteins and organelles for autophagic degradation in the lysosomes/vacuoles. MVBs are single-membrane organelles in the endocytic pathway that contain intraluminal vesicles whose content is either degraded in the lysosomes/vacuoles or recycled to the cell surface. In plants, both autophagosome and MVB pathways play important roles in plant responses to biotic and abiotic stresses. More recent studies have revealed that autophagosomes and MVBs also act together in plant stress responses in a variety of processes, including deployment of defense-related molecules, regulation of cell death, trafficking and degradation of membrane and soluble constituents, and modulation of plant hormone metabolism and signaling. In this review, we discuss these recent findings on the coordination and crosstalk between autophagosome and MVB pathways that contribute to the complex network of plant stress responses.


Subject(s)
Autophagosomes/metabolism , Multivesicular Bodies/metabolism , Plants/metabolism , Stress, Physiological , Autophagosomes/drug effects , Humans , Multivesicular Bodies/drug effects , Plant Growth Regulators/pharmacology , Plants/drug effects , Signal Transduction/drug effects , Stress, Physiological/drug effects
2.
Respir Res ; 20(1): 129, 2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31234850

ABSTRACT

BACKGROUND: Azithromycin (Azm) is a macrolide recognized for its disease-modifying effects and reduction in exacerbation of chronic airway diseases. It is not clear whether the beneficial effects of Azm are due to its anti-microbial activity or other pharmacological actions. We have shown that Azm affects the integrity of the bronchial epithelial barrier measured by increased transepithelial electrical resistance. To better understand these effects of Azm on bronchial epithelia we have investigated global changes in gene expression. METHODS: VA10 bronchial epithelial cells were treated with Azm and cultivated in air-liquid interface conditions for up to 22 days. RNA was isolated at days 4, 10 and 22 and analyzed using high-throughput RNA sequencing. qPCR and immunostaining were used to confirm key findings from bioinformatic analyses. Detailed assessment of cellular changes was done using microscopy, followed by characterization of the lipidomic profiles of the multivesicular bodies present. RESULTS: Bioinformatic analysis revealed that after 10 days of treatment genes encoding effectors of sterol and cholesterol metabolism were prominent. Interestingly, expression of genes associated with epidermal barrier differentiation, KRT1, CRNN, SPINK5 and DSG1, increased significantly at day 22. Together with immunostaining, these results suggest an epidermal differentiation pattern. We also found that Azm induced the formation of multivesicular and lamellar bodies in two different airway epithelial cell lines. Lipidomic analysis revealed that Azm was entrapped in multivesicular bodies linked to different types of lipids, most notably palmitate and stearate. Furthermore, targeted analysis of lipid species showed accumulation of phosphatidylcholines, as well as ceramide derivatives. CONCLUSIONS: Taken together, we demonstrate how Azm might confer its barrier enhancing effects, via activation of epidermal characteristics and changes to intracellular lipid dynamics. These effects of Azm could explain the unexpected clinical benefit observed during Azm-treatment of patients with various lung diseases affecting barrier function.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Cell Differentiation/drug effects , Epidermis/drug effects , Multivesicular Bodies/drug effects , Respiratory Mucosa/drug effects , Cell Differentiation/physiology , Cell Line , Epidermis/metabolism , Humans , Multivesicular Bodies/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
3.
Cell Rep ; 25(12): 3356-3370.e4, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566862

ABSTRACT

The discovery of microRNA (miRNA) sorting into extracellular vesicles (EVs) revealed a novel mode of intercellular communication and uncovered a link between cellular endomembrane compartments and small RNAs in EV-secreting cells. Using a two-step ultracentrifugation procedure to isolate EVs released by T cells, we found that 45% of tRNA fragments (tRFs), but fewer than 1% of miRNAs, were significantly enriched in EVs compared with the corresponding cellular RNA. T cell activation induced the EV-mediated release of a specific set of tRFs derived from the 5' end and 3'-internal region of tRNAs without variable loops. Inhibition of EV biogenesis pathways specifically led to the accumulation of these activation-induced EV-enriched tRFs within multivesicular bodies (MVBs). Introducing antisense oligonucleotides to inhibit these tRFs enhanced T cell activation. Taken together, these results demonstrate that T cells selectively release tRFs into EVs via MVBs and suggest that this process may remove tRFs that repress immune activation.


Subject(s)
Extracellular Vesicles/metabolism , Lymphocyte Activation , RNA Transport , RNA, Transfer/metabolism , T-Lymphocytes/metabolism , Animals , Down-Regulation/drug effects , Extracellular Vesicles/drug effects , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Aggregates/drug effects , RNA Transport/drug effects , RNA, Transfer/chemistry , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/drug effects
4.
J Transl Med ; 16(1): 190, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29986726

ABSTRACT

BACKGROUND: Endoplasmic reticulum (ER) stress and its consequent unfolded protein response (UPR) are believed to be associated with progression, survival and chemoresistance of a variety of tumor cells through multiple cellular processes, including autophagy. Therefore, the ER stress-autophagy pathway presents a potential molecular target for therapeutic intervention. The objective of this study was to evaluate the therapeutic efficacy of ER stress and autophagy modulators in the context of pancreatic ductal adenocarcinoma (PDAC). METHODS: We first targeted IRE1α, an important regulator of the UPR, through STF-083010 treatment in PDAC cell lines in vitro. Chloroquine was then used to target autophagy and an optimal combination treatment was developed using chloroquine, sunitinib and gemcitabine. Apoptosis was analyzed using TUNEL assay, autophagy was estimated using lysotracker staining and electron microscopy, and UPR was analyzed using anti-GRP78 immunostaining and XBP1 splicing. Transplantation of PDAC derived KPCP1 and Panc02 cells in mouse pancreas were performed to study treatment efficacy in vivo. RESULTS: Suppression of the IRE1α by STF-083010 alone resulted in increased lysosomes and reduced viability of PDAC cells. Chloroquine treatment alone inhibited downstream autophagy but was insufficient in reducing PDAC cell growth. However, combining STF-083010 and chloroquine had additive anti-tumor efficacy when used with gemcitabine. Sunitinib alone caused abnormal maturation of the autolysosomes with increased intracellular multivesicular bodies and increased apoptosis evident in PDAC cells. Sunitinib showed a synergistic effect with chloroquine in reducing in vitro PDAC cell viability and significantly increased the efficacy of gemcitabine in human and murine PDAC cell lines. The anti-proliferative effect of gemcitabine was significantly increased when used in combination with sunitinib and/or chloroquine in both in vitro and in vivo PDAC models. The addition of sunitinib and/or chloroquine to gemcitabine, resulted in a significantly increased survival of the animals without noticeably increased toxicity. Sunitinib, gemcitabine and chloroquine treated mice showed a significant reduction of GRP78 expression, reduced cell proliferation and increased apoptosis in pancreas, compatible with a tumor response. CONCLUSIONS: Sunitinib combined with chloroquine reduces tumor growth through suppression of autophagy and increased apoptosis. Co-administration of modulators of ER stress-mediated autophagy with chemotherapy presents a novel therapeutic approach in PDAC.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy , Endoplasmic Reticulum Stress , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Female , Heat-Shock Proteins/metabolism , Humans , Ki-67 Antigen/metabolism , Mice, Inbred C57BL , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Pancreatic Neoplasms/ultrastructure , Sunitinib/pharmacology , Sunitinib/therapeutic use , Survival Analysis , Tumor Burden , Unfolded Protein Response/drug effects , X-Box Binding Protein 1/metabolism , Gemcitabine
5.
J Cell Biol ; 217(3): 1129-1142, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29339438

ABSTRACT

Exosomes are small endosome-derived extracellular vesicles implicated in cell-cell communication and are secreted by living cells when multivesicular bodies (MVBs) fuse with the plasma membrane (PM). Current techniques to study exosome physiology are based on isolation procedures after secretion, precluding direct and dynamic insight into the mechanics of exosome biogenesis and the regulation of their release. In this study, we propose real-time visualization of MVB-PM fusion to overcome these limitations. We designed tetraspanin-based pH-sensitive optical reporters that detect MVB-PM fusion using live total internal reflection fluorescence and dynamic correlative light-electron microscopy. Quantitative analysis demonstrates that MVB-PM fusion frequency is reduced by depleting the target membrane SNAREs SNAP23 and syntaxin-4 but also can be induced in single cells by stimulation of the histamine H1 receptor (H1HR). Interestingly, activation of H1R1 in HeLa cells increases Ser110 phosphorylation of SNAP23, promoting MVB-PM fusion and the release of CD63-enriched exosomes. Using this single-cell resolution approach, we highlight the modulatory dynamics of MVB exocytosis that will help to increase our understanding of exosome physiology and identify druggable targets in exosome-associated pathologies.


Subject(s)
Cell Membrane/physiology , Membrane Fusion/physiology , Multivesicular Bodies/physiology , Receptors, G-Protein-Coupled/metabolism , Cell Communication/drug effects , Cell Membrane/drug effects , Exocytosis/drug effects , HCT116 Cells , HeLa Cells , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Membrane Fusion/drug effects , Multivesicular Bodies/drug effects , Phosphorylation/drug effects , Potassium Chloride/pharmacology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, Histamine H1/drug effects , Single-Cell Analysis , Tetraspanins/genetics , Tetraspanins/metabolism
6.
Cell Death Dis ; 9(2): 96, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367588

ABSTRACT

Type 1 diabetes (T1D) results from ß-cell destruction due to concerted action of both innate and adaptive immune responses. Pro-inflammatory cytokines, such as interleukin-1ß and interferon-γ, secreted by the immune cells invading islets of Langerhans, contribute to pancreatic ß-cell death in T1D. Cytokine-induced endoplasmic reticulum (ER) stress plays a central role in ß-cell demise. ER stress can modulate autophagic response; however, no study addressed the regulation of autophagy during the pathophysiology of T1D. In this study, we document that cytokines activate the AMPK-ULK-1 pathway while inhibiting mTORC1, which stimulates autophagy activity in an ER stress-dependent manner. On the other hand, time-course analysis of LC3-II accumulation in autophagosomes revealed that cytokines block the autophagy flux in an ER stress independent manner, leading to the formation of large dysfunctional autophagosomes and worsening of ER stress. Cytokines rapidly impair lysosome function, leading to lysosome membrane permeabilization, Cathepsin B leakage and lysosomal cell death. Blocking cathepsin activity partially protects against cytokine-induced or torin1-induced apoptosis, whereas blocking autophagy aggravates cytokine-induced CHOP overexpression and ß-cell apoptosis. In conclusion, cytokines stimulate the early steps of autophagy while blocking the autophagic flux, which aggravate ER stress and trigger lysosomal cell death. Restoration of autophagy/lysosomal function may represent a novel strategy to improve ß-cell resistance in the context of T1D.


Subject(s)
Apoptosis , Autophagy , Cytokines/toxicity , Inflammation Mediators/toxicity , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Cathepsin B/metabolism , Cell Line , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Insulin-Secreting Cells/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitophagy/drug effects , Models, Biological , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Transcription Factor CHOP/metabolism
7.
Lab Invest ; 97(11): 1385-1396, 2017 11.
Article in English | MEDLINE | ID: mdl-28892096

ABSTRACT

Primary sclerosing cholangitis (PSC) is an incurable, fibroinflammatory biliary disease for which there is no effective pharmacotherapy. We recently reported cholangiocyte senescence as an important phenotype in PSC while others showed that portal macrophages accumulate in PSC. Unfortunately, our ability to explore cholangiocyte senescence and macrophage accumulation has been hampered by limited in vitro models. Thus, our aim was to develop and characterize a three-dimensional (3D) model of normal and diseased bile ducts (cholangioids) starting with normal human cholangiocytes (NHC), senescent NHC (NHC-sen), and cholangiocytes from PSC patients. In 3D culture, NHCs formed spheroids of ~5000 cells with a central lumen of ~150 µm. By confocal microscopy and western blot, cholangioids retained expression of cholangiocyte proteins (cytokeratin 7/19) and markers of epithelial polarity (secretin receptor and GM130). Cholangioids are functionally active, and upon secretin stimulation, luminal size increased by ~80%. Cholangioids exposed to hydrogen peroxide exhibited cellular senescence and the senescence-associated secretory phenotype (SASP; increased IL-6, p21, SA-ß-Gal, yH2A.x and p16 expression). Furthermore, cholangioids derived from NHC-sen or PSC patients were smaller and had slower growth than the controls. When co-cultured with THP-1 macrophages, the number of macrophages associated with NHC-sen or PSC cholangioids was five- to seven-fold greater compared to co-culture with non-senescent NHC. We observed that NHC-sen and PSC cholangioids release greater number of extracellular vesicles (EVs) compared to controls. Moreover, conditioned media from NHC-sen cholangioids resulted in an ~2-fold increase in macrophage migration. In summary, we developed a method to generate normal and diseased cholangioids, characterized them morphologically and functionally, showed that they can be induced to senescence and SASP, and demonstrated both EV release and macrophage attraction. This novel model mimics several features of PSC, and thus will be useful for studying the pathogenesis of PSC and potentially identifying new therapeutic targets.


Subject(s)
Bile Ducts/pathology , Cholangitis, Sclerosing/pathology , Spheroids, Cellular/pathology , Autoantigens/metabolism , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/ultrastructure , Biomarkers/metabolism , Cell Line , Cells, Cultured , Cellular Senescence/drug effects , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/metabolism , Coculture Techniques , Culture Media, Conditioned , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Extracellular Vesicles/ultrastructure , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/toxicity , Keratin-19/metabolism , Keratin-7/metabolism , Macrophage Activation , Macrophages/cytology , Macrophages/immunology , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Multivesicular Bodies/pathology , Multivesicular Bodies/ultrastructure , Oxidants/toxicity , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/ultrastructure
8.
Blood ; 128(25): 2976-2987, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27742708

ABSTRACT

Chorea-acanthocytosis is one of the hereditary neurodegenerative disorders known as the neuroacanthocytoses. Chorea-acanthocytosis is characterized by circulating acanthocytes deficient in chorein, a protein of unknown function. We report here for the first time that chorea-acanthocytosis red cells are characterized by impaired autophagy, with cytoplasmic accumulation of active Lyn and of autophagy-related proteins Ulk1 and Atg7. In chorea-acanthocytosis erythrocytes, active Lyn is sequestered by HSP90-70 to form high-molecular-weight complexes that stabilize and protect Lyn from its proteasomal degradation, contributing to toxic Lyn accumulation. An interplay between accumulation of active Lyn and autophagy was found in chorea-acanthocytosis based on Lyn coimmunoprecipitation with Ulk1 and Atg7 and on the presence of Ulk1 in Lyn-containing high-molecular-weight complexes. In addition, chorein associated with Atg7 in healthy but not in chorea-acanthocytosis erythrocytes. Electron microscopy detected multivesicular bodies and membrane remnants only in circulating chorea-acanthocytosis red cells. In addition, reticulocyte-enriched chorea-acanthocytosis red cell fractions exhibited delayed clearance of mitochondria and lysosomes, further supporting the impairment of authophagic flux. Because autophagy is also important in erythropoiesis, we studied in vitro CD34+-derived erythroid precursors. In chorea-acanthocytosis, we found (1) dyserythropoiesis; (2) increased active Lyn; (3) accumulation of a marker of autophagic flux and autolysososme degradation; (4) accumlation of Lamp1, a lysosmal membrane protein, and LAMP1-positive aggregates; and (5) reduced clearance of lysosomes and mitochondria. Our results uncover in chorea-acanthocytosis erythroid cells an association between accumulation of active Lyn and impaired autophagy, suggesting a link between chorein and autophagic vesicle trafficking in erythroid maturation.


Subject(s)
Autophagy , Erythroid Cells/pathology , Neuroacanthocytosis/pathology , Adult , Anion Exchange Protein 1, Erythrocyte/metabolism , Autophagy/drug effects , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Benzoquinones/pharmacology , Bortezomib/pharmacology , Cell Differentiation/drug effects , Cytosol/drug effects , Cytosol/metabolism , Demography , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/pathology , Erythrocytes/ultrastructure , Erythroid Cells/drug effects , Erythroid Cells/ultrastructure , Erythropoiesis/drug effects , Female , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Male , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Weight , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , src-Family Kinases/metabolism
9.
J Virol ; 90(24): 11181-11196, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27707921

ABSTRACT

Hepatitis C virus (HCV) particles are described as lipoviroparticles which are released similarly to very-low-density lipoproteins (VLDLs). However, the release mechanism is still poorly understood; the canonical endoplasmic reticulum-Golgi intermediate compartment (ERGIC) pathway as well as endosome-dependent release has been proposed. Recently, the role of exosomes in the transmission of HCV has been reported. Only a minor fraction of the de novo-synthesized lipoviroparticles is released by the infected cell. To investigate the relevance of multivesicular bodies (MVBs) for viral morphogenesis and release, the MVB inhibitor U18666A was used. Intracellular trafficking was analyzed by confocal microscopy and electron microscopy. Moreover, an mCherry-tagged HCV variant was used. Conditions were established that enable U18666A-dependent inhibition of MVBs without affecting viral replication. Under these conditions, significant inhibition of the HCV release was observed. The assembly of viral particles is not affected. In U18666A-treated cells, intact infectious viral particles accumulate in CD63-positive exosomal structures and large dysfunctional lysosomal structures (multilamellar bodies). These retained particles possess a lower density, reflecting a misloading with lipids. Our data indicate that at least a fraction of HCV particles leaves the cell via the endosomal pathway. Endosomes facilitate the sorting of HCV particles for release or degradation. IMPORTANCE: There are still a variety of open questions regarding morphogenesis and release of hepatitis C virus. The HCV-infected cell produces significant more viral particles that are released, raising the question about the fate of the nonreleased particles. Moreover, the relevance of the endosomal pathway for the release of HCV is under debate. Use of the MVB (multivesicular body) inhibitor U18666A enabled a detailed analysis of the impact of MVBs for viral morphogenesis and release. It was revealed that infectious, fully assembled HCV particles are either MVB-dependently released or intracellularly degraded by the lysosome. Our data indicate that at least a fraction of HCV particles leaves the cell via the endosomal pathway independent from the constitutive secretory pathway. Our study describes a so-far-unprecedented cross talk between two pathways regulating on the one hand the release of infectious viral particles and on the other hand the intracellular degradation of nonreleased particles.


Subject(s)
Androstenes/pharmacology , Anticholesteremic Agents/pharmacology , Exosomes/drug effects , Hepacivirus/drug effects , Hepatocytes/drug effects , Virus Release/drug effects , Animals , Biological Transport/drug effects , Cell Line, Tumor , Cholesterol/metabolism , Exosomes/ultrastructure , Exosomes/virology , Gene Expression , Genes, Reporter , Hepacivirus/physiology , Hepacivirus/ultrastructure , Hepatocytes/ultrastructure , Hepatocytes/virology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Multivesicular Bodies/drug effects , Multivesicular Bodies/ultrastructure , Multivesicular Bodies/virology , Virion/drug effects , Virion/physiology , Virion/ultrastructure , Virus Assembly/physiology , Red Fluorescent Protein
10.
Cell Mol Life Sci ; 73(24): 4717-4737, 2016 12.
Article in English | MEDLINE | ID: mdl-27438886

ABSTRACT

Exosomes are vesicles released from cells by fusion of multivesicular bodies (MVBs) with the plasma membrane. This study aimed to investigate whether the phosphoinositide kinase PIKfyve affects this process. Our results show that in PC-3 cells inhibition of PIKfyve by apilimod or depletion by siRNA increased the secretion of the exosomal fraction. Moreover, quantitative electron microscopy analysis showed that cells treated with apilimod contained more MVBs per cell and more intraluminal vesicles per MVB. Interestingly, mass spectrometry analysis revealed a considerable enrichment of autophagy-related proteins (NBR1, p62, LC3, WIPI2) in exosomal fractions released by apilimod-treated cells, a result that was confirmed by immunoblotting. When the exosome preparations were investigated by electron microscopy a small population of p62-labelled electron dense structures was observed together with CD63-containing exosomes. The p62-positive structures were found in less dense fractions than exosomes in density gradients. Inside the cells, p62 and CD63 were found in the same MVB-like organelles. Finally, both the degradation of EGF and long-lived proteins were shown to be reduced by apilimod. In conclusion, inhibition of PIKfyve increases secretion of exosomes and induces secretory autophagy, showing that these pathways are closely linked. We suggest this is due to impaired fusion of lysosomes with both MVBs and autophagosomes, and possibly increased fusion of MVBs with autophagosomes, and that the cells respond by secreting the content of these organelles to maintain cellular homeostasis.


Subject(s)
Autophagy , Exosomes/metabolism , Phosphoinositide-3 Kinase Inhibitors , Secretory Pathway , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Exosomes/drug effects , Exosomes/ultrastructure , Gene Knockdown Techniques , Humans , Hydrazones , Lysosomes/drug effects , Lysosomes/metabolism , Morpholines/pharmacology , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Phosphatidylinositol 3-Kinases/metabolism , Proteolysis/drug effects , Proteomics , Pyrimidines , RNA-Binding Proteins/metabolism , Secretory Pathway/drug effects , Tetraspanin 30/metabolism , Triazines/pharmacology , Ubiquitin/metabolism , Ubiquitination/drug effects , Up-Regulation/drug effects
11.
Dev Cell ; 37(5): 473-83, 2016 Jun 06.
Article in English | MEDLINE | ID: mdl-27270042

ABSTRACT

Membrane contact sites between the ER and multivesicular endosomes/bodies (MVBs) play important roles in endosome positioning and fission and in neurite outgrowth. ER-MVB contacts additionally function in epidermal growth factor receptor (EGFR) tyrosine kinase downregulation by providing sites where the ER-localized phosphatase, PTP1B, interacts with endocytosed EGFR before the receptor is sorted onto intraluminal vesicles (ILVs). Here we show that these contacts are tethered by annexin A1 and its Ca(2+)-dependent ligand, S100A11, and form a subpopulation of differentially regulated contact sites between the ER and endocytic organelles. Annexin A1-regulated contacts function in the transfer of ER-derived cholesterol to the MVB when low-density lipoprotein-cholesterol in endosomes is low. This sterol traffic depends on interaction between ER-localized VAP and endosomal oxysterol-binding protein ORP1L, and is required for the formation of ILVs within the MVB and thus for the spatial regulation of EGFR signaling.


Subject(s)
Annexin A1/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Biological Transport/drug effects , Endocytosis/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Endosomes/drug effects , Endosomes/ultrastructure , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , HeLa Cells , Humans , Lipoproteins, LDL/pharmacology , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptors, Steroid/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Vesicular Transport Proteins/metabolism
12.
Cell Rep ; 15(10): 2239-2250, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27239031

ABSTRACT

Complexin (Cplx) proteins modulate the core SNARE complex to regulate exocytosis. To understand the contributions of Cplx to signaling in a well-characterized neural circuit, we investigated how Cplx3, a retina-specific paralog, shapes transmission at rod bipolar (RB)→AII amacrine cell synapses in the mouse retina. Knockout of Cplx3 strongly attenuated fast, phasic Ca(2+)-dependent transmission, dependent on local [Ca(2+)] nanodomains, but enhanced slower Ca(2+)-dependent transmission, dependent on global intraterminal [Ca(2+)] ([Ca(2+)]I). Surprisingly, coordinated multivesicular release persisted at Cplx3(-/-) synapses, although its onset was slowed. Light-dependent signaling at Cplx3(-/-) RB→AII synapses was sluggish, owing largely to increased asynchronous release at light offset. Consequently, propagation of RB output to retinal ganglion cells was suppressed dramatically. Our study links Cplx3 expression with synapse and circuit function in a specific retinal pathway and reveals a role for asynchronous release in circuit gain control.


Subject(s)
Exocytosis , Eye Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retina/cytology , Retina/metabolism , Signal Transduction , Synapses/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium/pharmacology , Exocytosis/drug effects , Mice, Inbred C57BL , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Nerve Tissue Proteins/deficiency , Retina/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects
13.
Plant Physiol ; 169(1): 497-511, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26229051

ABSTRACT

Multivesicular bodies (MVBs) are unique endosomes containing vesicles in the lumen and play critical roles in many cellular processes. We have recently shown that Arabidopsis (Arabidopsis thaliana) Lyst-Interacting Protein5 (LIP5), a positive regulator of the Suppressor of K(+) Transport Growth Defect1 (SKD1) AAA ATPase in MVB biogenesis, is a critical target of the mitogen-activated protein kinases MPK3 and MPK6 and plays an important role in the plant immune system. In this study, we report that the LIP5-regulated MVB pathway also plays a critical role in plant responses to abiotic stresses. Disruption of LIP5 causes compromised tolerance to both heat and salt stresses. The critical role of LIP5 in plant tolerance to abiotic stresses is dependent on its ability to interact with Suppressor of K(+) Transport Growth Defect1. When compared with wild-type plants, lip5 mutants accumulate increased levels of ubiquitinated protein aggregates and NaCl under heat and salt stresses, respectively. Further analysis using fluorescent dye and MVB markers reveals that abiotic stress increases the formation of endocytic vesicles and MVBs in a largely LIP5-dependent manner. LIP5 is also required for the salt-induced increase of intracellular reactive oxygen species, which have been implicated in signaling of salt stress responses. Basal levels of LIP5 phosphorylation by MPKs and the stability of LIP5 are elevated by salt stress, and mutation of MPK phosphorylation sites in LIP5 reduces the stability and compromises the ability to complement the lip5 salt-sensitive mutant phenotype. These results collectively indicate that the MVB pathway is positively regulated by pathogen/stress-responsive MPK3/6 through LIP5 phosphorylation and plays a critical role in broad plant responses to biotic and abiotic stresses.


Subject(s)
Arabidopsis Proteins/metabolism , Hot Temperature , Multivesicular Bodies/metabolism , Mutation/genetics , Organelle Biogenesis , Sodium Chloride/pharmacology , Stress, Physiological/drug effects , Adenosine Triphosphatases/metabolism , Endocytosis/drug effects , Green Fluorescent Proteins/metabolism , Ions , Multivesicular Bodies/drug effects , Phenotype , Phosphorylation/drug effects , Plant Roots/drug effects , Plant Roots/physiology , Protein Binding/drug effects , Protein Stability/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Salt Tolerance/drug effects , Sodium/metabolism , Subcellular Fractions/metabolism , Ubiquitinated Proteins/metabolism
14.
J Virol ; 90(7): 3330-41, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26719264

ABSTRACT

UNLABELLED: In addition to infectious viral particles, hepatitis B virus-replicating cells secrete large amounts of subviral particles assembled by the surface proteins, but lacking any capsid and genome. Subviral particles form spheres (22-nm particles) and filaments. Filaments contain a much larger amount of the large surface protein (LHBs) compared to spheres. Spheres are released via the constitutive secretory pathway, while viral particles are ESCRT-dependently released via multivesicular bodies (MVBs). The interaction of virions with the ESCRT machinery is mediated by α-taxilin that connects the viral surface protein LHBs with the ESCRT component tsg101. Since filaments in contrast to spheres contain a significant amount of LHBs, it is unclear whether filaments are released like spheres or like virions. To study the release of subviral particles in the absence of virion formation, a core-deficient HBV mutant was generated. Confocal microscopy, immune electron microscopy of ultrathin sections and isolation of MVBs revealed that filaments enter MVBs. Inhibition of MVB biogenesis by the small-molecule inhibitor U18666A or inhibition of ESCRT functionality by coexpression of transdominant negative mutants (Vps4A, Vps4B, and CHMP3) abolishes the release of filaments while the secretion of spheres is not affected. These data indicate that in contrast to spheres which are secreted via the secretory pathway, filaments are released via ESCRT/MVB pathway like infectious viral particles. IMPORTANCE: This study revises the current model describing the release of subviral particles by showing that in contrast to spheres, which are secreted via the secretory pathway, filaments are released via the ESCRT/MVB pathway like infectious viral particles. These data significantly contribute to a better understanding of the viral morphogenesis and might be helpful for the design of novel antiviral strategies.


Subject(s)
DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Hepatitis B virus/metabolism , Multivesicular Bodies/metabolism , Transcription Factors/metabolism , Virus Release/physiology , Androstenes/pharmacology , Cell Line, Tumor , Cytoskeleton/metabolism , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , Hep G2 Cells , Hepatitis B virus/genetics , Hepatocytes/virology , Humans , Microscopy, Confocal , Microscopy, Electron , Multivesicular Bodies/drug effects , Vesicular Transport Proteins/metabolism , Viral Core Proteins/deficiency , Viral Core Proteins/genetics
15.
Traffic ; 15(2): 197-211, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24279430

ABSTRACT

Multivesicular endosomes/bodies (MVBs) contain intraluminal vesicles (ILVs) that bud away from the cytoplasm. Multiple mechanisms of ILV formation have been identified, but the relationship between different populations of ILVs and MVBs remains unclear. Here, we show in HeLa cells that different ILV subpopulations can be distinguished by size. EGF stimulation promotes the formation of large ESCRT-dependent ILVs, whereas depletion of the ESCRT-0 component, Hrs, promotes the formation of a uniformly sized population of small ILVs, the formation of which requires CD63. CD63 has previously been implicated in ESCRT-independent sorting of PMEL in MVBs and transfected PMEL is present on the small ILVs that form on Hrs depletion. Upregulation of CD63-dependent ILV formation by Hrs depletion indicates that Hrs and CD63 regulate competing machineries required for the generation of distinct ILV subpopulations. Taken together our results indicate that ILV size is influenced by their cargo and mechanism of formation and suggest a competitive relationship between ESCRT-dependent and -independent mechanisms of ILV formation within single MVBs.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Multivesicular Bodies/metabolism , Phosphoproteins/metabolism , Tetraspanin 30/metabolism , Epidermal Growth Factor/pharmacology , HeLa Cells , Humans , Multivesicular Bodies/drug effects , Multivesicular Bodies/ultrastructure , Protein Transport , gp100 Melanoma Antigen/metabolism
16.
Mol Biol Cell ; 24(2): 129-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23154999

ABSTRACT

The ErbB2 receptor is a clinically validated cancer target whose internalization and trafficking mechanisms remain poorly understood. HSP90 inhibitors, such as geldanamycin (GA), have been developed to target the receptor to degradation or to modulate downstream signaling. Despite intense investigations, the entry route and postendocytic sorting of ErbB2 upon GA stimulation have remained controversial. We report that ErbB2 levels inversely impact cell clathrin-mediated endocytosis (CME) capacity. Indeed, the high levels of the receptor are responsible for its own low internalization rate. GA treatment does not directly modulate ErbB2 CME rate but it affects ErbB2 recycling fate, routing the receptor to modified multivesicular endosomes (MVBs) and lysosomal compartments, by perturbing early/recycling endosome structure and sorting capacity. This activity occurs irrespective of the cargo interaction with HSP90, as both ErbB2 and the constitutively recycled, HSP90-independent, transferrin receptor are found within modified endosomes, and within aberrant, elongated recycling tubules, leading to modified MVBs/lysosomes. We propose that GA, as part of its anticancer activity, perturbs early/recycling endosome sorting, routing recycling cargoes toward mixed endosomal compartments.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Lysosomes/metabolism , Multivesicular Bodies/metabolism , Receptor, ErbB-2/metabolism , Transferrin/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Clathrin/physiology , Clathrin-Coated Vesicles/metabolism , Dynamins/metabolism , Electron Microscope Tomography , Endocytosis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Mice , Microscopy, Fluorescence , Multivesicular Bodies/drug effects , Multivesicular Bodies/ultrastructure , Protein Transport/drug effects , Single-Cell Analysis
17.
PLoS One ; 7(7): e40673, 2012.
Article in English | MEDLINE | ID: mdl-22815786

ABSTRACT

BACKGROUND: In addition to lowering cholesterol, statins are thought to beneficially modulate inflammation. Several chemokines including CXCL1/growth-related oncogene (GRO)-α, CXCL8/interleukin (IL)-8 and CCL2/monocyte chemoattractant protein (MCP)-1 are important in the pathogenesis of atherosclerosis and can be influenced by statin-treatment. Recently, we observed that atorvastatin-treatment alters the intracellular content and subcellular distribution of GRO-α in cultured human umbilical vein endothelial cells (HUVECs). The objective of this study was to investigate the mechanisms involved in this phenomenon. METHODOLOGY/ PRINCIPAL FINDINGS: The effect of atorvastatin on secretion levels and subcellular distribution of GRO-α, IL-8 and MCP-1 in HUVECs activated by interleukin (IL)-1ß were evaluated by ELISA, confocal microscopy and immunoelectron microscopy. Atorvastatin increased the intracellular contents of GRO-α, IL-8, and MCP-1 and induced colocalization with E-selectin in multivesicular bodies. This effect was prevented by adding the isoprenylation substrate GGPP, but not the cholesterol precursor squalene, indicating that atorvastatin exerts these effects by inhibiting isoprenylation rather than depleting the cells of cholesterol. CONCLUSIONS/ SIGNIFICANCE: Atorvastatin targets inflammatory chemokines to the endocytic pathway and multivesicular bodies and may contribute to explain the anti-inflammatory effect of statins at the level of endothelial cell function.


Subject(s)
Chemokines/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Multivesicular Bodies/metabolism , Atorvastatin , Cell Compartmentation/drug effects , Chemokine CXCL1/metabolism , E-Selectin/metabolism , Endocytosis/drug effects , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Heptanoic Acids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Indoles/pharmacology , Interleukin-1beta/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Multivesicular Bodies/drug effects , Pravastatin/pharmacology , Prenylation/drug effects , Pyrroles/pharmacology , Simvastatin/pharmacology , Solubility , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tetraspanin 30/metabolism
18.
Proc Natl Acad Sci U S A ; 109(22): 8382-7, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586080

ABSTRACT

A primary method for studying cellular function is to examine cell morphology after a given manipulation. Fluorescent markers attached to proteins/intracellular structures of interest in conjunction with 3D fluorescent microscopy are frequently exploited for functional analysis. Despite the central role of morphology comparisons in cell biological approaches, few statistical tools are available that allow biological scientists without a high level of statistical training to quantify the similarity or difference of fluorescent images containing multifactorial information. We transform intracellular structures into kernels and develop a multivariate two-sample test that is nonparametric and asymptotically normal to directly and quantitatively compare cellular morphologies. The asymptotic normality bypasses the computationally intensive calculations used by the usual resampling techniques to compute the P-value. Because all parameters required for the statistical test are estimated directly from the data, it does not require any subjective decisions. Thus, we provide a black-box method for unbiased, automated comparison of cell morphology. We validate the performance of our test statistic for finite synthetic samples and experimental data. Employing our test for the comparison of the morphology of intracellular multivesicular bodies, we detect changes in their distribution after disruption of the cellular microtubule cytoskeleton with high statistical significance in fixed samples and live cell analysis. These results demonstrate that density-based comparison of multivariate image information is a powerful tool for automated detection of cell morphology changes. Moreover, the underlying mathematics of our test statistic is a general technique, which can be applied in situations where two data samples are compared.


Subject(s)
Algorithms , Intracellular Space/metabolism , Microscopy, Fluorescence/methods , Models, Biological , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Intracellular Space/drug effects , Microtubules/drug effects , Microtubules/metabolism , Molecular Dynamics Simulation , Multivariate Analysis , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Nocodazole/pharmacology , Reproducibility of Results , Tetraspanin 30/genetics , Tetraspanin 30/metabolism , Transfection , Tubulin Modulators/pharmacology
19.
Plant Signal Behav ; 7(4): 450-3, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22499175

ABSTRACT

Protein storage vacuoles (PSVs) are the primarily storage organelles in cotyledon cells for protein preservation in seeds. Storage proteins are transported from the endoplasmic reticulum (ER) to the Golgi apparatus for subsequent delivery to PSVs via presumably Golgi-derived dense vesicles (DVs). However, recent studies demonstrated that storage proteins in early stage of developing cotyledon of mung beans reached the multivesicular bodies (MVBs) prior to the detection of DVs, indicating the possible involvement of MVBs in mediating transport of storage proteins during the early stage of seed development. Here, we further show that the MVBs in developing tobacco seeds are functionally and biochemically equivalent to those in developing mung beans. Thus, MVBs in developing tobacco seeds are structurally distinct from DVs, contain both vacuolar sorting receptors (VSRs) and storage proteins, and they are insensitive to treatments of wortmannin and brefeldin A (BFA).


Subject(s)
Fabaceae/metabolism , Multivesicular Bodies/metabolism , Nicotiana/embryology , Nicotiana/metabolism , Seeds/metabolism , Androstadienes/pharmacology , Blotting, Western , Brefeldin A/pharmacology , Cotyledon/drug effects , Cotyledon/metabolism , Fabaceae/drug effects , Multivesicular Bodies/drug effects , Multivesicular Bodies/ultrastructure , Seeds/drug effects , Seeds/ultrastructure , Nicotiana/drug effects , Nicotiana/ultrastructure , Wortmannin
20.
J Proteome Res ; 11(6): 3127-42, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22524784

ABSTRACT

Wortmannin is a widely used pharmaceutical compound which is employed to define vesicular trafficking routes of particular proteins or cellular compounds. It targets phosphatidylinositol 3-kinase and phosphatidylinositol 4-kinases in a dose-dependent manner leading to the inhibition of protein vacuolar sorting and endocytosis. Combined proteomics and cell biological approaches have been used in this study to explore the effects of wortmannin on Arabidopsis root cells, especially on proteome and endomembrane trafficking. On the subcellular level, wortmannin caused clustering, fusion, and swelling of trans-Golgi network (TGN) vesicles and multivesicular bodies (MVBs) leading to the formation of wortmannin-induced multivesicular compartments. Appearance of wortmannin-induced compartments was associated with depletion of TGN as revealed by electron microscopy. On the proteome level, wortmannin induced massive changes in protein abundance profiles. Wortmannin-sensitive proteins belonged to various functional classes. An inhibition of vacuolar trafficking by wortmannin was related to the downregulation of proteins targeted to the vacuole, as showed for vacuolar proteases. A small GTPase, RabA1d, which regulates vesicular trafficking at TGN, was identified as a new protein negatively affected by wortmannin. In addition, Sec14 was upregulated and PLD1 alpha was downregulated by wortmannin.


Subject(s)
Androstadienes/pharmacology , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Plant Roots/metabolism , Proteome/metabolism , trans-Golgi Network/metabolism , Arabidopsis/cytology , Arabidopsis/drug effects , Multivesicular Bodies/drug effects , Multivesicular Bodies/metabolism , Phospholipase D/metabolism , Phospholipid Transfer Proteins/metabolism , Plant Roots/cytology , Plant Roots/drug effects , Protein Transport/drug effects , Wortmannin , rab GTP-Binding Proteins/metabolism , trans-Golgi Network/drug effects , trans-Golgi Network/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...