Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Nat Commun ; 12(1): 61, 2021 01 04.
Article En | MEDLINE | ID: mdl-33397928

Coat protein complex I (COP-I) mediates the retrograde transport from the Golgi apparatus to the endoplasmic reticulum (ER). Mutation of the COPA gene, encoding one of the COP-I subunits (α-COP), causes an immune dysregulatory disease known as COPA syndrome. The molecular mechanism by which the impaired retrograde transport results in autoinflammation remains poorly understood. Here we report that STING, an innate immunity protein, is a cargo of the retrograde membrane transport. In the presence of the disease-causative α-COP variants, STING cannot be retrieved back to the ER from the Golgi. The forced Golgi residency of STING results in the cGAS-independent and palmitoylation-dependent activation of the STING downstream signaling pathway. Surf4, a protein that circulates between the ER/ ER-Golgi intermediate compartment/ Golgi, binds STING and α-COP, and mediates the retrograde transport of STING to the ER. The STING/Surf4/α-COP complex is disrupted in the presence of the disease-causative α-COP variant. We also find that the STING ligand cGAMP impairs the formation of the STING/Surf4/α-COP complex. Our results suggest a homeostatic regulation of STING at the resting state by retrograde membrane traffic and provide insights into the pathogenesis of COPA syndrome.


Endoplasmic Reticulum/metabolism , Homeostasis , Membrane Proteins/metabolism , Animals , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/ultrastructure , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , HEK293 Cells , Humans , Lipoylation , Luciferases/metabolism , Mice , Nucleotidyltransferases/metabolism , Protein Binding/drug effects , Protein Transport/drug effects
2.
Cell Biol Int ; 45(3): 633-641, 2021 Mar.
Article En | MEDLINE | ID: mdl-33247607

Carbon tetrachloride (CCl4 ) causes hepatotoxicity in mammals, with its hepatocytic metabolism producing radicals that attack the intracellular membrane system and destabilize intracellular vesicle transport. Inhibition of intracellular transport causes lipid droplet retention and abnormal protein distribution. The intracellular transport of synthesized lipids and proteins from the endoplasmic reticulum (ER) to the Golgi apparatus is performed by coat complex II (COPII) vesicle transport, but how CCl4 inhibits COPII vesicle transport has not been elucidated. COPII vesicle formation on the ER membrane is initiated by the recruitment of Sar1 protein from the cytoplasm to the ER membrane, followed by that of the COPII coat constituent proteins (Sec23, Sec24, Sec13, and Sec31). In this study, we evaluated the effect of CCl4 on COPII vesicle formation using the RLC-16 rat hepatocyte cell line. Our results showed that CCl4 suppressed ER-Golgi transport in RLC-16 cells. Using a reconstituted system of rat liver tissue-derived cytoplasm and RLC-16 cell-derived ER membranes, CCl4 treatment inhibited the recruitment of Sar1 and Sec13 from the cytosolic fraction to ER membranes. CCl4 -induced changes in the ER membrane accordingly inhibited the accumulation of COPII vesicle-coated constituent proteins on the ER membrane, as well as the formation of COPII vesicles, which suppressed lipid and protein transport between the ER and Golgi apparatus. Our data suggest that CCl4 inhibits ER-Golgi intracellular transport by inhibiting COPII vesicle formation on the ER membrane in hepatocytes.


COP-Coated Vesicles/metabolism , Carbon Tetrachloride/toxicity , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Hepatocytes/metabolism , Intracellular Membranes/metabolism , Animals , COP-Coated Vesicles/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Hepatocytes/drug effects , Intracellular Membranes/drug effects , Male , Protein Transport/drug effects , Rats, Sprague-Dawley
3.
EMBO J ; 38(8)2019 04 15.
Article En | MEDLINE | ID: mdl-30858281

SREBPs are master regulators of lipid homeostasis and undergo sterol-regulated export from ER to Golgi apparatus for processing and activation via COPII-coated vesicles. While COPII recognizes SREBP through its escort protein SCAP, factor(s) specifically promoting SREBP/SCAP loading to the COPII machinery remains unknown. Here, we show that the ER/lipid droplet-associated protein Cideb selectively promotes the loading of SREBP/SCAP into COPII vesicles. Sterol deprivation releases SCAP from Insig and enhances ER export of SREBP/SCAP by inducing SCAP-Cideb interaction, thereby modulating sterol sensitivity. Moreover, Cideb binds to the guanine nucleotide exchange factor Sec12 to enrich SCAP/SREBP at ER exit sites, where assembling of COPII complex initiates. Loss of Cideb inhibits the cargo loading of SREBP/SCAP, reduces SREBP activation, and alleviates diet-induced hepatic steatosis. Our data point to a linchpin role of Cideb in regulated ER export of SREBP and lipid homeostasis.


Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/physiology , Endoplasmic Reticulum/physiology , Golgi Apparatus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterols/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/physiology , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , HEK293 Cells , Hep G2 Cells , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Transport , Sterol Regulatory Element Binding Protein 1/genetics
4.
Methods Mol Biol ; 1662: 75-86, 2017.
Article En | MEDLINE | ID: mdl-28861818

In plant secretory pathways, the Golgi apparatus serves as the major sorting hub to receive de novo synthesized protein from the endoplasmic reticulum for further sorting to post-Golgi compartments or for residence in the cisternae of Golgi stacks. Meanwhile, Golgi functions as a pivotal biochemical factory to make modifications of N-glycans and to produce mature glycoproteins. Fluorescent tag-based confocal microscopy in combination with the brefeldin A drug or the genetic tools to disturb Golgi function have been shown as powerful approaches to analyze Golgi-mediated protein traffic in transiently expressed plant protoplasts or in stably expressed transgenic plants. Various endoglycosidases like Endo H and PNGase F have been widely used to monitor Golgi-mediated glycosylation of secretory proteins. Here, using fluorescently tagged Golgi-resident proteins and known glycosylated proteins as examples, we describe detailed protocols to analyze Golgi-mediated protein traffic and glycosylation in transiently expressed protoplasts derived from Arabidopsis suspension culture cells and in stably expressed transgenic plants.


Arabidopsis/metabolism , Golgi Apparatus/metabolism , Microscopy, Fluorescence/methods , Plant Cells/metabolism , Protoplasts/metabolism , Secretory Pathway/genetics , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/metabolism , Arabidopsis/drug effects , Arabidopsis/genetics , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/ultrastructure , Cells, Cultured , Dexamethasone/pharmacology , Electroporation/methods , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Glycosylation/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/chemistry , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Plant Cells/drug effects , Plant Cells/ultrastructure , Plants, Genetically Modified , Plasmids/chemistry , Plasmids/metabolism , Protein Transport/drug effects , Protoplasts/drug effects , Protoplasts/ultrastructure , Secretory Pathway/drug effects , Transfection/methods
5.
Elife ; 62017 06 08.
Article En | MEDLINE | ID: mdl-28594326

Native cargo proteins exit the endoplasmic reticulum (ER) in COPII-coated vesicles, whereas resident and misfolded proteins are substantially excluded from vesicles by a retention mechanism that remains unresolved. We probed the ER retention process using the proteostasis regulator 4-phenylbutyrate (4-PBA), which we show targets COPII protein to reduce the stringency of retention. 4-PBA competes with p24 proteins to bind COPII. When p24 protein uptake is blocked, COPII vesicles package resident proteins and an ER-trapped mutant LDL receptor. We further show that 4-PBA triggers the secretion of a KDEL-tagged luminal resident, implying that a compromised retention mechanism causes saturation of the KDEL retrieval system. The results indicate that stringent ER retention requires the COPII coat machinery to actively sort biosynthetic cargo from diffusible misfolded and resident ER proteins.


COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Phenylbutyrates/metabolism , Protein Transport/drug effects , Antineoplastic Agents , Humans , Protein Binding
6.
J Cell Biol ; 215(4): 543-558, 2016 Nov 21.
Article En | MEDLINE | ID: mdl-27872256

Stimulation of cells with epidermal growth factor (EGF) induces internalization and partial degradation of the EGF receptor (EGFR) by the endo-lysosomal pathway. For continuous cell functioning, EGFR plasma membrane levels are maintained by transporting newly synthesized EGFRs to the cell surface. The regulation of this process is largely unknown. In this study, we find that EGF stimulation specifically increases the transport efficiency of newly synthesized EGFRs from the endoplasmic reticulum to the plasma membrane. This coincides with an up-regulation of the inner coat protein complex II (COPII) components SEC23B, SEC24B, and SEC24D, which we show to be specifically required for EGFR transport. Up-regulation of these COPII components requires the transcriptional regulator RNF11, which localizes to early endosomes and appears additionally in the cell nucleus upon continuous EGF stimulation. Collectively, our work identifies a new regulatory mechanism that integrates the degradation and transport of EGFR in order to maintain its physiological levels at the plasma membrane.


Carrier Proteins/metabolism , Endosomes/metabolism , ErbB Receptors/metabolism , Proteolysis , Transcription, Genetic , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins , Endosomes/drug effects , Epidermal Growth Factor/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic/drug effects , Up-Regulation/drug effects
7.
J Biol Chem ; 291(3): 1014-27, 2016 Jan 15.
Article En | MEDLINE | ID: mdl-26546679

The majority of biosynthetic secretory proteins initiate their journey through the endomembrane system from specific subdomains of the endoplasmic reticulum. At these locations, coated transport carriers are generated, with the Sar1 GTPase playing a critical role in membrane bending, recruitment of coat components, and nascent vesicle formation. How these events are appropriately coordinated remains poorly understood. Here, we demonstrate that Sar1 acts as the curvature-sensing component of the COPII coat complex and highlight the ability of Sar1 to bind more avidly to membranes of high curvature. Additionally, using an atomic force microscopy-based approach, we further show that the intrinsic GTPase activity of Sar1 is necessary for remodeling lipid bilayers. Consistent with this idea, Sar1-mediated membrane remodeling is dramatically accelerated in the presence of its guanine nucleotide-activating protein (GAP), Sec23-Sec24, and blocked upon addition of guanosine-5'-[(ß,γ)-imido]triphosphate, a poorly hydrolysable analog of GTP. Our results also indicate that Sar1 GTPase activity is stimulated by membranes that exhibit elevated curvature, potentially enabling Sar1 membrane scission activity to be spatially restricted to highly bent membranes that are characteristic of a bud neck. Taken together, our data support a stepwise model in which the amino-terminal amphipathic helix of GTP-bound Sar1 stably penetrates the endoplasmic reticulum membrane, promoting local membrane deformation. As membrane bending increases, Sar1 membrane binding is elevated, ultimately culminating in GTP hydrolysis, which may destabilize the bilayer sufficiently to facilitate membrane fission.


COP-Coated Vesicles/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Amino Acid Substitution , Animals , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/ultrastructure , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Endoplasmic Reticulum/ultrastructure , Enzyme Inhibitors/pharmacology , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Guanylyl Imidodiphosphate/pharmacology , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Microscopy, Atomic Force , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Mutation , Organelle Shape/drug effects , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
8.
Biochim Biophys Acta ; 1833(3): 487-93, 2013 Mar.
Article En | MEDLINE | ID: mdl-23195223

Protein kinase C epsilon (PKCε) contributes to multiple signaling pathways affecting human disease. The function of PKCε requires it to undergo changes in subcellular distribution in response to signaling events. While the mechanisms underlying this translocation are incompletely understood, it involves the receptor for activated C kinase protein (RACK2/ß'-COP). This receptor also functions as a vesicle coat protein in the secretory pathway where it is regulated by the small GTP-binding protein ADP-ribosylation factor, ARF1. We inhibited ARF1 activation to test the requirement for RACK2/ß'-COP in PKCε localization in NIH3T3 fibroblasts. We found that steady-state localization of PKCε at the Golgi complex requires ARF1-regulated RACK2/ß'-COP function. By contrast, we did not observe any defects in phorbol ester-induced translocation when ARF1 was inhibited. We also found that PKCε bound to isolated membranes through two distinct mechanisms. One mechanism was dependent upon RACK2/ß'-COP while a second was RACK2/ß'-COP-independent and stimulated by phorbol esters. Finally, we show that RACK2/ß'-COP affects the subcellular distribution of a constitutively active form of PKCε, in a manner similar to what we observed for wild-type PKCε. Together, our data support a role for RACK2/ß'-COP in the steady-state localization of PKCε at the Golgi apparatus, which may be independent of its role during PKCε translocation to the cell surface.


ADP-Ribosylation Factor 1/metabolism , COP-Coated Vesicles/metabolism , Coatomer Protein/metabolism , Golgi Apparatus/metabolism , Protein Kinase C-epsilon/metabolism , Receptors, Cell Surface/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , COP-Coated Vesicles/drug effects , Cattle , Fluorescent Antibody Technique , Golgi Apparatus/drug effects , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Mice , NIH 3T3 Cells , Phorbol Esters/pharmacology , Protein Transport/drug effects , Rats , Receptors for Activated C Kinase , Signal Transduction/drug effects
9.
Autophagy ; 7(11): 1392-3, 2011 Nov.
Article En | MEDLINE | ID: mdl-21804352

We now seem to live in a small world, in which everyone is highly interconnected. Cells, too, often also display tremendous interconnectivities in their component systems. As a recent case in point, we have identified a conserved protein complex--the SEA complex--that links the nuclear pore complex (NPC), the COPII vesicle coating complex, vacuoles and autophagy. In this punctum we will discuss the properties of this novel complex.


COP-Coated Vesicles/metabolism , Multiprotein Complexes/metabolism , Nitrogen/deficiency , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , COP-Coated Vesicles/drug effects , Multiprotein Complexes/chemistry , Nitrogen/pharmacology , Nuclear Pore/drug effects , Nuclear Pore/metabolism , Protein Binding/drug effects , Protein Structure, Secondary , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Vacuoles/drug effects
10.
Int J Biochem Cell Biol ; 41(12): 2511-21, 2009 Dec.
Article En | MEDLINE | ID: mdl-19695339

In eukaryotic cells several physiologic and pathologic conditions generate the accumulation of unfolded proteins in the endoplasmic reticulum (ER), leading to ER stress. To restore normal function, some ER transmembrane proteins sense the ER stress and activate coordinated signalling pathways collectively called the Unfolded Protein Response (UPR). Little is known on how the UPR relates to post-ER compartments and to the export from the ER of newly synthesized proteins. Here, we report that the ER stress response induced by either thapsigargin or nitric oxide modifies the dynamics of the intracellular distribution of ERGIC-53 and GM130, two markers of the ER Golgi Intermediate Compartment and of the cis-Golgi, respectively. In addition, induction of ER stress alters the morphology of the ERGIC and the Golgi complex and interferes with the reformation of both compartments. Moreover, ER stress rapidly reduces the transport to the Golgi complex of the temperature sensitive mutant of the Vesicular Stomatitis Virus G Glycoprotein (VSV-G) fused with the Green Fluorescent Protein (ts045G), without apparently decreasing the amount of the protein competent for export. Interestingly, a parallel rapid reduction of the number of Sec31 labelled fluorescent puncta on the ER membranes does occur, thus suggesting that the ER stress alters the ER export and the dynamic of post-ER compartments by rapidly targeting the formation of COPII-coated transport intermediates.


Autoantigens/metabolism , Biomarkers/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Thapsigargin/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Cell Line , Cellular Structures/drug effects , Cellular Structures/metabolism , Cellular Structures/ultrastructure , Endoplasmic Reticulum , Golgi Apparatus , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Membrane Glycoproteins/metabolism , Protein Engineering , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction , Unfolded Protein Response , Viral Envelope Proteins/metabolism
11.
Mol Biol Cell ; 20(20): 4458-70, 2009 Oct.
Article En | MEDLINE | ID: mdl-19710425

Because the functional borders of the intermediate compartment (IC) are not well defined, the spatial map of the transport machineries operating between the endoplasmic reticulum (ER) and the Golgi apparatus remains incomplete. Our previous studies showed that the IC consists of interconnected vacuolar and tubular parts with specific roles in pre-Golgi trafficking. Here, using live cell imaging, we demonstrate that the tubules containing the GTPase Rab1A create a long-lived membrane compartment around the centrosome. Separation of this pericentrosomal domain of the IC from the Golgi ribbon, due to centrosome motility, revealed that it contains a distinct pool of COPI coats and acts as a temperature-sensitive way station in post-ER trafficking. However, unlike the Golgi, the pericentrosomal IC resists the disassembly of COPI coats by brefeldin A, maintaining its juxtaposition with the endocytic recycling compartment, and operation as the focal point of a dynamic tubular network that extends to the cell periphery. These results provide novel insight into the compartmental organization of the secretory pathway and Golgi biogenesis. Moreover, they reveal a direct functional connection between the IC and the endosomal system, which evidently contributes to unconventional transport of the cystic fibrosis transmembrane conductance regulator to the cell surface.


COP-Coated Vesicles/physiology , Centrosome/physiology , Coat Protein Complex I/physiology , Protein Transport/physiology , rab1 GTP-Binding Proteins/metabolism , trans-Golgi Network/physiology , Animals , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/ultrastructure , Cell Line/virology , Centrosome/ultrastructure , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endocytosis , Golgi Apparatus/drug effects , HeLa Cells , Humans , Intracellular Membranes/physiology , Intracellular Membranes/ultrastructure , Kidney/cytology , Mesocricetus , Microscopy, Video , Rats , Recombinant Fusion Proteins/metabolism , Semliki forest virus/physiology , Viral Fusion Proteins/metabolism , rab1 GTP-Binding Proteins/genetics , trans-Golgi Network/ultrastructure
12.
J Neurosci ; 28(47): 12453-64, 2008 Nov 19.
Article En | MEDLINE | ID: mdl-19020038

The GABA transporter-1 (GAT1) is a prototypical protein of the synaptic specialization. Export of GAT1 from the endoplasmic reticulum (ER) is contingent on its interaction with the COPII (coatomer protein-II) coat subunit Sec24D. Here we show that silencing all four Sec24 isoforms strongly inhibits transport of GAT1 to the cell surface. In contrast, transport of GAT1-RL/AS, a mutant that is deficient in Sec24D recruitment, was not inhibited, suggesting a nonconventional, COPII-independent pathway. However, ARFGAP1 bound directly to the C terminus of both GAT1-RL/AS and wild-type GAT1. Surface expression of GAT1-RL/AS involved ARFGAP1. GAT1-RL/AS appeared to bypass the ER-Golgi-intermediate compartment, but its pathway to the plasma membrane still involved passage through the Golgi. Thus, the GAT1-RL/AS mutant allowed to test whether COPII-dependent ER-export is required for correct sorting of GAT1 to the axon terminal in neuronal cells. In contrast to wild-type GAT1, GAT1-RL/AS failed to be specifically enriched at the tip of neurite extensions of CAD.a cells (a neuroblastoma cell line that can be differentiated into a neuron-like phenotype) and in the axon terminals of hippocampal neurons. These findings indicate that correct sorting to the axon is contingent on ER export via the COPII machinery and passage through the ER-Golgi-intermediate compartment.


Axons/physiology , GABA Plasma Membrane Transport Proteins/metabolism , GTPase-Activating Proteins/metabolism , Neurons/cytology , Vesicular Transport Proteins/metabolism , Animals , Animals, Newborn , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/physiology , Cells, Cultured , GABA Plasma Membrane Transport Proteins/genetics , GTPase-Activating Proteins/genetics , Hippocampus/cytology , Humans , Immunoprecipitation/methods , Luminescent Proteins/biosynthesis , Luminescent Proteins/metabolism , Microscopy, Confocal/methods , Protein Transport/drug effects , Protein Transport/physiology , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Serotonin Plasma Membrane Transport Proteins/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Temperature , Transfection/methods , Tritium/metabolism , Vesicular Transport Proteins/genetics , gamma-Aminobutyric Acid/metabolism
13.
Mol Biol Cell ; 19(5): 1976-90, 2008 May.
Article En | MEDLINE | ID: mdl-18287528

Rapidly cycling proteins of the early secretory pathway can operate as cargo receptors. Known cargo receptors are abundant proteins, but it remains mysterious why their inactivation leads to rather limited secretion phenotypes. Studies of Surf4, the human orthologue of the yeast cargo receptor Erv29p, now reveal a novel function of cargo receptors. Surf4 was found to interact with endoplasmic reticulum-Golgi intermediate compartment (ERGIC)-53 and p24 proteins. Silencing Surf4 together with ERGIC-53 or silencing the p24 family member p25 induced an identical phenotype characterized by a reduced number of ERGIC clusters and fragmentation of the Golgi apparatus without effect on anterograde transport. Live imaging showed decreased stability of ERGIC clusters after knockdown of p25. Silencing of Surf4/ERGIC-53 or p25 resulted in partial redistribution of coat protein (COP) I but not Golgi matrix proteins to the cytosol and partial resistance of the cis-Golgi to brefeldin A. These findings imply that cargo receptors are essential for maintaining the architecture of ERGIC and Golgi by controlling COP I recruitment.


Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/ultrastructure , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Cell Survival/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Silencing/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HeLa Cells , Humans , Models, Biological , Protein Binding/drug effects , Protein Transport/drug effects
14.
J Cell Biol ; 179(5): 951-63, 2007 Dec 03.
Article En | MEDLINE | ID: mdl-18056412

Gamma-Secretase is responsible for proteolytic maturation of signaling and cell surface proteins, including amyloid precursor protein (APP). Abnormal processing of APP by gamma-secretase produces a fragment, Abeta(42), that may be responsible for Alzheimer's disease (AD). The biogenesis and trafficking of this important enzyme in relation to aberrant Abeta processing is not well defined. Using a cell-free reaction to monitor the exit of cargo proteins from the endoplasmic reticulum (ER), we have isolated a transient intermediate of gamma-secretase. Here, we provide direct evidence that the gamma-secretase complex is formed in an inactive complex at or before the assembly of an ER transport vesicle dependent on the COPII sorting subunit, Sec24A. Maturation of the holoenzyme is achieved in a subsequent compartment. Two familial AD (FAD)-linked PS1 variants are inefficiently packaged into transport vesicles generated from the ER. Our results suggest that aberrant trafficking of PS1 may contribute to disease pathology.


Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/metabolism , Animals , CHO Cells , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/enzymology , Cell Line , Cricetinae , Cricetulus , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Methylamines/pharmacology , Mutant Proteins/metabolism , Presenilin-1/biosynthesis , Protein Transport/drug effects , Rats , Vesicular Transport Proteins/metabolism
15.
FEBS J ; 274(24): 6365-77, 2007 Dec.
Article En | MEDLINE | ID: mdl-18021253

Angiogenic switch in renal cell carcinoma (RCC) is attributed to the inactivation of the von Hippel-Lindau tumor suppressor, stabilization of hypoxia inducible factor-1 transcription factor and increased vascular endothelial growth factor. To evaluate the role of an angiogenesis inhibitor, thrombopsondin-1 (TSP1), we compared TSP1 production in human RCC and normal tissue and secretion by the normal renal epithelium (human normal kidney, HNK) and RCC cells. Normal and RCC tissues stained positive for TSP1, and the levels of TSP1 mRNA and total protein were similar in RCC and HNK cells. However, HNK cells secreted high TSP1, which rendered them nonangiogenic, whereas RCC cells secreted little TSP1 and were angiogenic. Western blot and immunostaining revealed TSP1 in the cytoplasm of RCC cells on serum withdrawal, whereas, in HNK cells, it was rapidly exported. Seeking mechanisms of defective TSP1 secretion, we discovered impaired calcium uptake by RCC in response to vascular endothelial growth factor. In HNK cells, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester, a calcium chelator, simulated TSP1 retention, mimicking the RCC phenotype. Further analysis revealed a profound decrease in transient receptor potential canonical ion channel 4 (TRPC4) Ca(2+) channel expression in RCC cells. TRPC4 silencing in HNK cells caused TSP1 retention and impaired secretion. Double labeling of the secretory system components revealed TSP1 colocalization with coatomer protein II (COPII) anterograde vesicles in HNK cells. In contrast, in RCC cells, TSP1 colocalized with COPI vesicles, pointing to the retrograde transport to the endoplasmic reticulum caused by misfolding. Our study indicates that TRPC4 loss in RCC leads to impaired Ca(2+) intake, misfolding, retrograde transport and diminished secretion of antiangiogenic TSP1, thus enabling angiogenic switch during RCC progression.


Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Thrombospondin 1/metabolism , Animals , Blotting, Western , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Calcium/metabolism , Calcium/pharmacokinetics , Carcinoma, Renal Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Chelating Agents/pharmacology , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Thrombospondin 1/genetics , Transfection
16.
Adv Drug Deliv Rev ; 59(8): 759-81, 2007 Aug 10.
Article En | MEDLINE | ID: mdl-17681635

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


Drug Delivery Systems , Endoplasmic Reticulum/metabolism , Molecular Chaperones/administration & dosage , Molecular Chaperones/chemistry , Protein Folding , Animals , Apoptosis/drug effects , Biological Transport/drug effects , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Glycosylation/drug effects , Hypoxia/drug therapy , Hypoxia/metabolism , Molecular Chaperones/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Protein Biosynthesis/drug effects , Protein Conformation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Transport Vesicles/drug effects
17.
Curr Biol ; 16(2): 173-9, 2006 Jan 24.
Article En | MEDLINE | ID: mdl-16431369

The COPII coat complex mediates the formation of transport carriers at specialized sites of the endoplasmic reticulum (ERES). It consists of the Sar1p GTPase and the Sec23/24p and the Sec13/31p subcomplexes . Both stimulate the GTPase activity of Sar1p , which itself triggers coat disassembly. This built-in GAP activity makes the COPII complex in principle unstable and raises the question of how sufficient stability required for cargo capture and carrier formation is achieved. To address this, we analyzed COPII turnover at single ERES in living cells. The half times for Sar1p, Sec23p, and Sec24p turnover are 1.1, 3.7, and 3.9 s, respectively. Decreasing the amount of transport-competent cargo in the endoplasmic reticulum accelerates turnover of the Sec23/24p and slows down that of Sar1p. A mathematical model of COPII membrane turnover that reproduces the experimental in vivo FRAP kinetics and is consistent with existing in vitro data predicts that Sec23/24p remains membrane associated even after GTP hydrolysis by Sar1p for a duration that is strongly increased by the presence of cargo. We conclude that secretory cargo retains the COPII complex on membranes, after Sar1p release has occurred, and prevents premature disassembly of COPII during cargo sorting and transport carrier formation.


COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Vesicular Transport Proteins/metabolism , Animals , COP-Coated Vesicles/drug effects , Chlorocebus aethiops , Cycloheximide/pharmacology , Endoplasmic Reticulum/drug effects , Fluorescence Recovery After Photobleaching , Humans , Kinetics , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Protein Subunits/metabolism , Protein Synthesis Inhibitors/pharmacology , Vero Cells
18.
Methods Enzymol ; 404: 115-25, 2005.
Article En | MEDLINE | ID: mdl-16413263

This article describes the use of acyltransferase inhibitors as probes for studying the potential role of lysophospholipid acyltransferases (LPAT) in intracellular membrane trafficking in the secretory and endocytic pathways. The small molecule inhibitors that are described here were originally found as acyl-CoA:cholesterol acyltransferase (ACAT) inhibitors. One of these, CI-976 (2,2-methyl-N-(2,4,6,-trimethoxyphenyl)dodecanamide), was also found to be a potent LPAT inhibitor. CI-976 is a small, hydrophobic, membrane-permeant compound and both in vivo and in vitro studies have shown that it, but not other ACAT inhibitors, has a profound effect on multiple membrane trafficking pathways in eukaryotic cells including: (1) inhibition of COPII vesicle budding from the endoplasmic reticulum (ER), (2) inhibition of transferrin and transferrin receptor export from the endocytic recycling compartment, and (3) stimulation of tubule-mediated retrograde trafficking of Golgi membranes to the ER. Here we describe the use of CI-976 and other ACAT inhibitors for studies with both cultured mammalian cells and in vitro reconstitution assays, with a particular emphasis on COPII vesicle budding from the ER. All of these studies strongly suggest that CI-976-sensitive LPATs play a role in coated vesicle fission, and therefore, CI-976 is a valuable addition to the arsenal of small molecule inhibitors that can be used to study secretory and endocytic membrane trafficking pathways.


Acyltransferases/antagonists & inhibitors , Anilides/pharmacology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/physiology , Protein Transport/drug effects , 1-Acylglycerophosphocholine O-Acyltransferase/physiology , Animals , COP-Coated Vesicles/drug effects , Cells, Cultured , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Imidazoles/pharmacology , Intracellular Membranes/drug effects , Rats , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Urea/pharmacology
19.
Mol Biol Cell ; 14(12): 5011-8, 2003 Dec.
Article En | MEDLINE | ID: mdl-14565973

It is unclear whether the mammalian Golgi apparatus can form de novo from the ER or whether it requires a preassembled Golgi matrix. As a test, we assayed Golgi reassembly after forced redistribution of Golgi matrix proteins into the ER. Two conditions were used. In one, ER redistribution was achieved using a combination of brefeldin A (BFA) to cause Golgi collapse and H89 to block ER export. Unlike brefeldin A alone, which leaves matrix proteins in relatively large remnant structures outside the ER, the addition of H89 to BFA-treated cells caused ER accumulation of all Golgi markers tested. In the other, clofibrate treatment induced ER redistribution of matrix and nonmatrix proteins. Significantly, Golgi reassembly after either treatment was robust, implying that the Golgi has the capacity to form de novo from the ER. Furthermore, matrix proteins reemerged from the ER with faster ER exit rates. This, together with the sensitivity of BFA remnants to ER export blockade, suggests that presence of matrix proteins in BFA remnants is due to cycling via the ER and preferential ER export rather than their stable assembly in a matrix outside the ER. In summary, the Golgi apparatus appears capable of efficient self-assembly.


COP-Coated Vesicles/metabolism , Cell Compartmentation/physiology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Sulfonamides , Animals , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , Cell Compartmentation/drug effects , Cells, Cultured , Clofibrate/pharmacology , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Golgi Apparatus/drug effects , Humans , Hypolipidemic Agents/pharmacology , Isoquinolines/pharmacology , Microscopy, Electron , Microscopy, Fluorescence , Protein Synthesis Inhibitors/pharmacology
20.
Plant Cell ; 15(5): 1242-56, 2003 May.
Article En | MEDLINE | ID: mdl-12724547

We have studied the transport of soluble cargo molecules by inhibiting specific transport steps to and from the Golgi apparatus. Inhibition of export from the Golgi via coexpression of a dominant-negative GTP-restricted ARF1 mutant (Q71L) inhibits the secretion of alpha-amylase and simultaneously induces the secretion of the vacuolar protein phytepsin to the culture medium. By contrast, specific inhibition of endoplasmic reticulum export via overexpression of Sec12p or coexpression of a GTP-restricted form of Sar1p inhibits the anterograde transport of either cargo molecule in a similar manner. Increased secretion of the vacuolar protein was not observed after incubation with the drug brefeldin A or after coexpression of the GDP-restricted mutant of ARF1 (T31N). Therefore, the differential effect of inducing the secretion of one cargo molecule while inhibiting the secretion of another is dependent on the GTP hydrolysis by ARF1p and is not caused by a general inhibition of Golgi-derived COPI vesicle traffic. Moreover, we demonstrate that GTP-restricted ARF1-stimulated secretion is observed only for cargo molecules that are expected to be sorted in a BP80-dependent manner, exhibiting sequence-specific, context-independent, vacuolar sorting signals. Induced secretion of proteins carrying C-terminal vacuolar sorting signals was not observed. This finding suggests that ARF1p influences the BP80-mediated transport route to the vacuole in addition to transport steps of the default secretory pathway to the cell surface.


ADP-Ribosylation Factor 1/metabolism , GTP Phosphohydrolases/metabolism , Vacuoles/metabolism , ADP-Ribosylation Factor 1/genetics , Androstadienes/pharmacology , Brefeldin A/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Coat Protein Complex I/genetics , Coat Protein Complex I/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Plant , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Models, Biological , Mutation , Protein Transport/drug effects , Substrate Specificity , Vacuoles/drug effects , Wortmannin
...