Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(22): eade7753, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809969

ABSTRACT

Lipid droplets (LDs) comprise a triglyceride core surrounded by a lipid monolayer enriched with proteins, many of which function in LD homeostasis. How proteins are targeted to the growing LD is still unclear. Rab1b, a GTPase regulating secretory transport, was recently associated with targeting proteins to LDs in a Drosophila RNAi screen. LD formation was prevented in human hepatoma cells overexpressing dominant-negative Rab1b. We thus hypothesized that Rab1b recruits lipid-synthesizing enzymes, facilitating LD growth. Here, FRET between diacylglycerol acyltransferase 2 (DGAT2) and Rab1b and activity mutants of the latter demonstrated that Rab1b promotes DGAT2 ER to the LD surface redistribution. Last, alterations in LD metabolism and DGAT2 redistribution, consistent with Rab1b activity, were caused by mutations in the Rab1b-GTPase activating protein TBC1D20 in Warburg Micro syndrome (WARBM) model mice fibroblasts. These data contribute to our understanding of the mechanism of Rab1b in LD homeostasis and WARBM, a devastating autosomal-recessive disorder caused by mutations in TBC1D20.


Subject(s)
Diacylglycerol O-Acyltransferase , Endoplasmic Reticulum , Lipid Droplets , rab1 GTP-Binding Proteins , Lipid Droplets/metabolism , Animals , Humans , rab1 GTP-Binding Proteins/metabolism , rab1 GTP-Binding Proteins/genetics , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/genetics , Mice , Endoplasmic Reticulum/metabolism , Mutation , Lipid Metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics
2.
J Mol Biol ; 435(15): 168171, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37285900

ABSTRACT

Carboxypeptidase E (CPE), an essential enzyme in the biosynthetic production line of most peptide hormones and neuropeptides, is predominantly expressed in endocrine tissues and in the nervous system. CPE is active in acidic environments where it cleaves the C'-terminal basic residues of peptide precursors to generate their bioactive form. Consequently, this highly conserved enzyme regulates numerous fundamental biological processes. Here, we combined live-cell microscopy and molecular analysis to examine the intracellular distribution and secretion dynamics of fluorescently tagged CPE. We show that, in non-endocrine cells, tagged-CPE is a soluble luminal protein that is efficiently exported from the ER via the Golgi apparatus to lysosomes. The C'-terminal conserved amphipathic helix serves as a lysosomal and secretory granule targeting and a secretion motif. Following secretion, CPE may be reinternalized into the lysosomes of neighboring cells.


Subject(s)
Carboxypeptidase H , Lysosomes , Carboxypeptidase H/genetics , Carboxypeptidase H/metabolism , Golgi Apparatus/enzymology , Lysosomes/enzymology , Neuropeptides/metabolism
3.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835119

ABSTRACT

The opportunistic fungus Aspergillus fumigatus is the primary invasive mold pathogen in humans, and is responsible for an estimated 200,000 yearly deaths worldwide. Most fatalities occur in immunocompromised patients who lack the cellular and humoral defenses necessary to halt the pathogen's advance, primarily in the lungs. One of the cellular responses used by macrophages to counteract fungal infection is the accumulation of high phagolysosomal Cu levels to destroy ingested pathogens. A. fumigatus responds by activating high expression levels of crpA, which encodes a Cu+ P-type ATPase that actively transports excess Cu from the cytoplasm to the extracellular environment. In this study, we used a bioinformatics approach to identify two fungal-unique regions in CrpA that we studied by deletion/replacement, subcellular localization, Cu sensitivity in vitro, killing by mouse alveolar macrophages, and virulence in a mouse model of invasive pulmonary aspergillosis. Deletion of CrpA fungal-unique amino acids 1-211 containing two N-terminal Cu-binding sites, moderately increased Cu-sensitivity but did not affect expression or localization to the endoplasmic reticulum (ER) and cell surface. Replacement of CrpA fungal-unique amino acids 542-556 consisting of an intracellular loop between the second and third transmembrane helices resulted in ER retention of the protein and strongly increased Cu-sensitivity. Deleting CrpA N-terminal amino acids 1-211 or replacing amino acids 542-556 also increased sensitivity to killing by mouse alveolar macrophages. Surprisingly, the two mutations did not affect virulence in a mouse model of infection, suggesting that even weak Cu-efflux activity by mutated CrpA preserves fungal virulence.


Subject(s)
Aspergillus fumigatus , Fungal Proteins , Humans , Animals , Mice , Aspergillus fumigatus/genetics , Virulence , Fungal Proteins/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism
4.
Bioessays ; 44(10): e2200064, 2022 10.
Article in English | MEDLINE | ID: mdl-35986435

ABSTRACT

An entirely different mechanism and localization were recently proposed for the COPII coat complex, challenging its well-accepted function to select and concentrate cargo into small COPII-coated spherical transport vesicles. Instead, the COPII complex is suggested to form a dynamic yet stationary collar that forms a boundary between the ER and the ER export membrane domain. This membrane domain, the ER exit site (ERES), is the site of COPII-mediated sorting and concentration of transport competent proteins. Subsequently, the ERES is implicated to mature and bud to form a sizeable pleiomorphic transport carrier that translocate on microtubules to fuse with the Golgi apparatus. Despite this drastic mechanistic dogma shift, most of the underlying protein-protein and protein-membrane interactions remain unchanged. Here, we attempt to provide a detailed description of the newly proposed model of how ER to Golgi transport works by describing the role of several essential proteins of the transport machinery.


Subject(s)
COP-Coated Vesicles , Golgi Apparatus , COP-Coated Vesicles/metabolism , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Protein Transport
5.
Front Immunol ; 13: 865797, 2022.
Article in English | MEDLINE | ID: mdl-35720342

ABSTRACT

Sandfly fever viruses are emerging Phleboviruses typically causing mild febrile illness. Some strains, however, can cause severe and occasionally fatal neuro-invasive disease. Like most viruses, Phleboviruses have devised various strategies to inhibit the type I interferon (IFN) response to support a productive infection. Still, most of the strategies identified so far focus on inhibiting the sensing arm of the IFN response. In contrast, the effect of sandfly virus infection on signaling from the IFN receptor is less characterized. Therefore, we tested the effect of sandfly fever virus Naples (SFNV) and Sicily (SFSV) infection on IFN signaling. We found that infection with either of these viruses inhibits signaling from the IFN receptor by inhibiting STAT1 phosphorylation and nuclear localization. We show that the viral nonstructural protein NSs mediates these effects, but only NSs from SFNV was found to interact with STAT1 directly. Thus, we tested the upstream IFN signaling components and found that Janus kinase 1 (Jak1) phosphorylation is also impaired by infection. Furthermore, the NSs proteins from both viruses directly interacted with Jak1. Last, we show that IFN inhibition by SFNV and SFSV is most likely downstream of the IFN receptor at the Jak1 level. Overall, our results reveal the multiple strategies used by these related viruses to overcome host defenses.


Subject(s)
Interferon Type I , Phlebotomus Fever , Phlebovirus , Humans , Interferon Type I/metabolism , Janus Kinase 1 , Phosphorylation , STAT1 Transcription Factor , Viral Nonstructural Proteins/metabolism
6.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33852719

ABSTRACT

COPII and COPI mediate the formation of membrane vesicles translocating in opposite directions within the secretory pathway. Live-cell and electron microscopy revealed a novel mode of function for COPII during cargo export from the ER. COPII is recruited to membranes defining the boundary between the ER and ER exit sites, facilitating selective cargo concentration. Using direct observation of living cells, we monitored cargo selection processes, accumulation, and fission of COPII-free ERES membranes. CRISPR/Cas12a tagging, the RUSH system, and pharmaceutical and genetic perturbations of ER-Golgi transport demonstrated that the COPII coat remains bound to the ER-ERES boundary during protein export. Manipulation of the cargo-binding domain in COPII Sec24B prohibits cargo accumulation in ERES. These findings suggest a role for COPII in selecting and concentrating exported cargo rather than coating Golgi-bound carriers. These findings transform our understanding of coat proteins' role in ER-to-Golgi transport.


Subject(s)
COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , HeLa Cells , Humans , Protein Transport
7.
Traffic ; 22(3): 64-77, 2021 03.
Article in English | MEDLINE | ID: mdl-33314495

ABSTRACT

The endoplasmic reticulum (ER) is involved in biogenesis, modification and transport of secreted and membrane proteins. The ER membranes are spread throughout the cell cytoplasm as well as the export domains known as ER exit sites (ERES). A subpopulation of ERES is centrally localized proximal to the Golgi apparatus. The significance of this subpopulation on ER-to-Golgi transport remains unclear. Transport carriers (TCs) form at the ERES via a COPII-dependent mechanism and move to Golgi on microtubule (MT) tracks. It was shown previously that ERES are distributed along MTs and undergo chaotic short-range movements and sporadic rapid long-range movements. The long-range movements of ERES are impaired by either depolymerization of MTs or inhibition of dynein, suggesting that ERES central concentration is mediated by dynein activity. We demonstrate that the processive movements of ERES are frequently coupled with the TC departure. Using the Sar1a[H79G]-induced ERES clustering at the perinuclear region, we identified BicaudalD2 (BicD2) and Rab6 as components of the dynein adaptor complex which drives perinuclear ERES concentration at the cell center. BicD2 partially colocalized with ERES and with TC. Peri-Golgi ERES localization was significantly affected by inhibition of BicD2 function with its N-terminal fragment or inhibition of Rab6 function with its dominant-negative mutant. Golgi accumulation of secretory protein was delayed by inhibition of Rab6 and BicD2. Thus, we conclude that a BicD2/Rab6 dynein adaptor is required for maintenance of Golgi-associated ERES. We propose that Golgi-associated ERES may enhance the efficiency of the ER-to-Golgi transport.


Subject(s)
Endoplasmic Reticulum , Golgi Apparatus , Biological Transport , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes , Microtubules , Protein Transport
8.
J Vis Exp ; (136)2018 06 20.
Article in English | MEDLINE | ID: mdl-29985342

ABSTRACT

Regulated exocytosis is a process by which cargo, which is stored in secretory granules (SGs), is released in response to a secretory trigger. Regulated exocytosis is fundamental for intercellular communication and is a key mechanism for the secretion of neurotransmitters, hormones, inflammatory mediators, and other compounds, by a variety of cells. At least three distinct mechanisms are known for regulated exocytosis: full exocytosis, where a single SG fully fuses with the plasma membrane, kiss-and-run exocytosis, where a single SG transiently fuses with the plasma membrane, and compound exocytosis, where several SGs fuse with each other, prior to or after SG fusion with the plasma membrane. The type of regulated exocytosis undertaken by a cell is often dictated by the type of secretory trigger. However, in many cells, a single secretory trigger can activate multiple modes of regulated exocytosis simultaneously. Despite their abundance and importance across cell types and species, the mechanisms that determine the different modes of secretion are largely unresolved. One of the main challenges in investigating the different modes of regulated exocytosis, is the difficulty in distinguishing between them as well as exploring them separately. Here we describe the use of fluorescein isothiocyanate (FITC)-dextran as an exocytosis reporter, and live cell imaging, to differentiate between the different pathways of regulated exocytosis, focusing on compound exocytosis, based on the robustness and duration of the exocytic events.


Subject(s)
Biological Transport/physiology , Dextrans/chemistry , Exocytosis/physiology , Fluorescein-5-isothiocyanate/analogs & derivatives , Animals , Fluorescein-5-isothiocyanate/chemistry
9.
Sci Rep ; 7(1): 15315, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127297

ABSTRACT

Compound exocytosis is considered the most massive mode of exocytosis, during which the membranes of secretory granules (SGs) fuse with each other to form a channel through which the entire contents of their granules is released. The underlying mechanisms of compound exocytosis remain largely unresolved. Here we show that the small GTPase Rab5, a known regulator of endocytosis, is pivotal for compound exocytosis in mast cells. Silencing of Rab5 shifts receptor-triggered secretion from a compound to a full exocytosis mode, in which SGs individually fuse with the plasma membrane. Moreover, we show that Rab5 is essential for FcεRI-triggered association of the SNARE protein SNAP23 with the SGs. Direct evidence is provided for SNAP23 involvement in homotypic SG fusion that occurs in the activated cells. Finally, we show that this fusion event is prevented by inhibition of the IKKß2 kinase, however, neither a phosphorylation-deficient nor a phosphomimetic mutant of SNAP23 can mediate homotypic SG fusion in triggered cells. Taken together our findings identify Rab5 as a heretofore-unrecognized regulator of compound exocytosis that is essential for SNAP23-mediated granule-granule fusion. Our results also implicate phosphorylation cycles in controlling SNAP23 SNARE function in homotypic SG fusion.


Subject(s)
Cell Membrane/metabolism , Exocytosis/physiology , Membrane Fusion/physiology , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Secretory Vesicles/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Line , Cell Membrane/genetics , Humans , Phosphorylation/physiology , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Secretory Vesicles/genetics , rab5 GTP-Binding Proteins/genetics
10.
Methods ; 127: 30-36, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28526563

ABSTRACT

Lipid droplets (LDs) are regulated neutral lipid storage organelles having a central role in numerous cellular processes as well as in various pathologies such as metabolic disorders, immune responses and during pathogen infection. Due to the growing significance of LDs, extensive efforts are made to study the mechanism and the dynamics of their formation and life history and how are these diverted or modified by pathogens. Real-time visualization of lipid droplet biogenesis can assist in clarifying these and other important issues and may have implications towards understanding the pathogenesis of the associated diseases. Typically, LDs are post-experimentally stained using lipophilic dyes and are visualized under a microscope. Alternatively, overexpression of LD-associated proteins or immunofluorescence analyses are used to identify and follow LDs. These experimental approaches only examine a single end point of the experiment and cannot answer questions regarding LD dynamics. Here, we describe a simple and novel experimental setting that allows real-time fluorescence staining and detection of LDs in cultured living as well as infected cells. This method is quick and simple and is not restricted to a specific dye or cell line. Using this system, the biogenesis of LDs and their growth is demonstrated in cells infected with hepatitis C virus (HCV), confirming the strength of this method and the wide range of its applications.


Subject(s)
Hepatitis C/metabolism , Lipid Droplets/metabolism , Virology/methods , Animals , Cell Line , Chlorocebus aethiops/metabolism , Chlorocebus aethiops/virology , Fluorescent Antibody Technique/methods , Host-Pathogen Interactions , Humans , Microbiological Techniques/methods
11.
J Cell Sci ; 129(20): 3868-3877, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27587840

ABSTRACT

Export out of the endoplasmic reticulum (ER) involves the Sar1 and COPII machinery acting at ER exit sites (ERES). Whether and how cargo proteins are recruited upstream of Sar1 and COPII is unclear. Two models are conceivable, a recruitment model where cargo is actively transported through a transport factor and handed over to the Sar1 and COPII machinery in ERES, and a capture model, where cargo freely diffuses into ERES where it is captured by the Sar1 and COPII machinery. Using the novel secretion inhibitor FLI-06, we show that recruitment of the cargo VSVG to ERES is an active process upstream of Sar1 and COPII. Applying FLI-06 before concentration of VSVG in ERES completely abolishes its recruitment. In contrast, applying FLI-06 after VSVG concentration in ERES does not lead to dispersal of the concentrated VSVG, arguing that it inhibits recruitment to ERES as opposed to capture in ERES. FLI-06 also inhibits export out of the trans-Golgi network (TGN), suggesting that similar mechanisms might orchestrate cargo selection and concentration at the ER and TGN. FLI-06 does not inhibit autophagosome biogenesis and the ER-peroxisomal transport route, suggesting that these rely on different mechanisms.


Subject(s)
Endoplasmic Reticulum/metabolism , Quinolines/pharmacology , trans-Golgi Network/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Endocytosis/drug effects , Exocytosis/drug effects , HeLa Cells , Humans , Peroxisomes/drug effects , Peroxisomes/metabolism , Protein Folding/drug effects , Protein Transport/drug effects , trans-Golgi Network/drug effects
12.
J Cell Sci ; 128(13): 2293-302, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002055

ABSTRACT

Myelin comprises a compactly stacked massive surface area of protein-poor thick membrane that insulates axons to allow fast signal propagation. Increasing levels of the myelin protein plasmolipin (PLLP) were correlated with post-natal myelination; however, its function is unknown. Here, the intracellular localization and dynamics of PLLP were characterized in primary glial and cultured cells using fluorescently labeled PLLP and antibodies against PLLP. PLLP localized to and recycled between the plasma membrane and the Golgi complex. In the Golgi complex, PLLP forms oligomers based on fluorescence resonance energy transfer (FRET) analyses. PLLP oligomers blocked Golgi to plasma membrane transport of the secretory protein vesicular stomatitis virus G protein (VSVG), but not of a VSVG mutant with an elongated transmembrane domain. Laurdan staining analysis showed that this block is associated with PLLP-induced proliferation of liquid-ordered membranes. These findings show the capacity of PLLP to assemble potential myelin membrane precursor domains at the Golgi complex through its oligomerization and ability to attract liquid-ordered lipids. These data support a model in which PLLP functions in myelin biogenesis through organization of myelin liquid-ordered membranes in the Golgi complex.


Subject(s)
Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Myelin Sheath/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Protein Multimerization , Proteolipids/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Dogs , Endocytosis , Intracellular Space/metabolism , Madin Darby Canine Kidney Cells , Molecular Sequence Data , Myelin and Lymphocyte-Associated Proteolipid Proteins/chemistry , Protein Structure, Tertiary , Protein Transport , Proteolipids/chemistry
13.
Virology ; 475: 139-49, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25462354

ABSTRACT

Hepatitis C virus (HCV) replicates in membrane associated, highly ordered replication complexes (RCs). These complexes include viral and host proteins necessary for viral RNA genome replication. The interaction network among viral and host proteins underlying the formation of these RCs is yet to be thoroughly characterized. Here, we investigated the association between NS4B and NS5A, two critical RC components. We characterized the interaction between these proteins using fluorescence resonance energy transfer and a mammalian two-hybrid system. Specific tryptophan residues within the C-terminal domain (CTD) of NS4B were shown to mediate this interaction. Domain I of NS5A, was sufficient to mediate its interaction with NS4B. Mutations in the NS4B CTD tryptophan residues abolished viral replication. Moreover, one of these mutations also affected NS5A hyperphosphorylation. These findings provide new insights into the importance of the NS4B-NS5A interaction and serve as a starting point for studying the complex interactions between the replicase subunits.


Subject(s)
Gene Expression Regulation, Viral/physiology , Hepacivirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Endoplasmic Reticulum/physiology , Fluorescence Resonance Energy Transfer , Hepacivirus/genetics , Hepacivirus/metabolism , Humans , RNA, Viral , Viral Nonstructural Proteins/genetics
14.
J Cell Sci ; 125(Pt 15): 3545-56, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22492786

ABSTRACT

Occludin (Ocln), a MARVEL-motif-containing protein, is found in all tight junctions. MARVEL motifs are comprised of four transmembrane helices associated with the localization to or formation of diverse membrane subdomains by interacting with the proximal lipid environment. The functions of the Ocln MARVEL motif are unknown. Bioinformatics sequence- and structure-based analyses demonstrated that the MARVEL domain of Ocln family proteins has distinct evolutionarily conserved sequence features that are consistent with its basolateral membrane localization. Live-cell microscopy, fluorescence resonance energy transfer (FRET) and bimolecular fluorescence complementation (BiFC) were used to analyze the intracellular distribution and self-association of fluorescent-protein-tagged full-length human Ocln or the Ocln MARVEL motif excluding the cytosolic C- and N-termini (amino acids 60-269, FP-MARVEL-Ocln). FP-MARVEL-Ocln efficiently arrived at the plasma membrane (PM) and was sorted to the basolateral PM in filter-grown polarized MDCK cells. A series of conserved aromatic amino acids within the MARVEL domain were found to be associated with Ocln dimerization using BiFC. FP-MARVEL-Ocln inhibited membrane pore growth during Triton-X-100-induced solubilization and was shown to increase the membrane-ordered state using Laurdan, a lipid dye. These data demonstrate that the Ocln MARVEL domain mediates self-association and correct sorting to the basolateral membrane.


Subject(s)
Occludin/chemistry , Occludin/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Cell Culture Techniques , Cell Membrane/metabolism , Chlorocebus aethiops , Computational Biology , Dogs , Epithelium/metabolism , Humans , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Occludin/genetics , Transfection
15.
J Virol ; 86(12): 6491-502, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22491470

ABSTRACT

Replication and assembly of hepatitis C virus (HCV) depend on the host's secretory and lipid-biosynthetic machinery. Viral replication occurs on endoplasmic reticulum (ER)-derived modified membranes, while viral assembly is thought to occur on lipid droplets (LDs). A physical association and coordination between the viral replication and assembly complexes are prerequisites for efficient viral production. Nonstructural protein 5A (NS5A), which localizes both to the ER and LDs, is an ideal candidate for this function. Here, the interaction of NS5A with host cell membranes and binding partners was characterized in living cells. The binding of NS5A to LDs is apparently irreversible, both in HCV-infected cells and when ectopically expressed. In HCV-infected cells, NS5A fluorescence was observed around the LDs and in perinuclear structures that were incorporated into a highly immobile platform superimposed over the ER membrane. Moreover, TBC1D20 and its cognate GTPase Rab1 are recruited by NS5A to LDs. The NS5A-TBC1D20 interaction was shown to be essential for the viral life cycle. In cells, expression of the Rab1 dominant negative (Rab1DN) GTPase mutant abolished steady-state LDs. In infected cells, Rab1DN induced the elimination of NS5A from viral replication sites. Our results demonstrate the significance of the localization of NS5A to LDs and support a model whereby its interaction with TBC1D20 and Rab1 affects lipid droplet metabolism to promote the viral life cycle.


Subject(s)
Hepacivirus/physiology , Hepatitis C/enzymology , Lipid Bilayers/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , rab1 GTP-Binding Proteins/metabolism , Cell Line , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Hepacivirus/genetics , Hepatitis C/genetics , Hepatitis C/metabolism , Humans , Protein Binding , Protein Transport , Viral Nonstructural Proteins/genetics , rab1 GTP-Binding Proteins/genetics
16.
Biochem J ; 439(3): 433-42, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21756249

ABSTRACT

EHDs [EH (Eps15 homology)-domain-containing proteins] participate in different stages of endocytosis. EHD2 is a plasma-membrane-associated EHD which regulates trafficking from the plasma membrane and recycling. EHD2 has a role in nucleotide-dependent membrane remodelling and its ATP-binding domain is involved in dimerization, which creates a membrane-binding region. Nucleotide binding is important for association of EHD2 with the plasma membrane, since a nucleotide-free mutant (EHD2 T72A) failed to associate. To elucidate the possible function of EHD2 during endocytic trafficking, we attempted to unravel proteins that interact with EHD2, using the yeast two-hybrid system. A novel interaction was found between EHD2 and Nek3 [NIMA (never in mitosis in Aspergillus nidulans)-related kinase 3], a serine/threonine kinase. EHD2 was also found in association with Vav1, a Nek3-regulated GEF (guanine-nucleotide-exchange factor) for Rho GTPases. Since Vav1 regulates Rac1 activity and promotes actin polymerization, the impact of overexpression of EHD2 on Rac1 activity was tested. The results indicated that wt (wild-type) EHD2, but not its P-loop mutants, reduced Rac1 activity. The inhibitory effect of EHD2 overexpression was partially rescued by co-expression of Rac1 as measured using a cholera toxin trafficking assay. The results of the present study strongly indicate that EHD2 regulates trafficking from the plasma membrane by controlling Rac1 activity.


Subject(s)
Carrier Proteins/physiology , Cell Membrane/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-vav/metabolism , rac1 GTP-Binding Protein/metabolism , Amino Acid Sequence , Enzyme Activation/physiology , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , NIMA-Related Kinases , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein Transport/physiology , Proto-Oncogene Proteins c-vav/genetics , rac1 GTP-Binding Protein/biosynthesis , rac1 GTP-Binding Protein/genetics
17.
J Biol Chem ; 286(15): 13470-80, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21266584

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is expressed as two isozymes α and ß. They share high similarity in their catalytic domains but differ in their N- and C-terminal regions, with GSK-3α having an extended glycine-rich N terminus. Here, we undertook live cell imaging combined with molecular and bioinformatic studies to understand the distinct functions of the GSK-3 isozymes focusing on GSK-3α N-terminal region. We found that unlike GSK-3ß, which shuttles between the nucleus and cytoplasm, GSK-3α was excluded from the nucleus. Deletion of the N-terminal region of GSK-3α resulted in nuclear localization, and treatment with leptomycin B resulted in GSK-3α accumulation in the nucleus. GSK-3α rapidly accumulated in the nucleus in response to calcium or serum deprivation, and accumulation was strongly inhibited by the calpain inhibitor calpeptin. This nuclear accumulation was not mediated by cleavage of the N-terminal region or phosphorylation of GSK-3α. Rather, we show that calcium-induced GSK-3α nuclear accumulation was governed by GSK-3α binding with as yet unknown calpain-sensitive protein or proteins; this binding was mediated by the N-terminal region. Bioinformatic and experimental analyses indicated that nuclear exclusion of GSK-3α was likely an exclusive characteristic of mammalian GSK-3α. Finally, we show that nuclear localization of GSK-3α reduced the nuclear pool of ß-catenin and its target cyclin D1. Taken together, these data suggest that the N-terminal region of GSK-3α is responsible for its nuclear exclusion and that binding with a calcium/calpain-sensitive product enables GSK-3α nuclear retention. We further uncovered a novel link between calcium and nuclear GSK-3α-mediated inhibition of the canonical Wnt/ß-catenin pathway.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Calpain/metabolism , Cell Nucleus/enzymology , Glycogen Synthase Kinase 3/metabolism , Active Transport, Cell Nucleus/physiology , Animals , COS Cells , Calpain/genetics , Chlorocebus aethiops , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Structure, Tertiary , Rats , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
18.
J Cell Biochem ; 110(3): 598-608, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20512921

ABSTRACT

The aim of this study was to investigate the impact of increased mRNA levels encoding GIRK1 in breast tumours on GIRK protein expression. mRNA levels encoding hGIRK1 and hGIRK4 in the MCF7, MCF10A and MDA-MB-453 breast cancer cell lines were assessed and the corresponding proteins detected using Western blots. cDNAs encoding for four hGIRK1 splice variants (hGIRK1a, 1c, 1d and 1e) were cloned from the MCF7 cell line. Subcellular localisation of fluorescence labelled hGIRK1a-e and hGIRK4 and of endogenous GIRK1 and GIRK4 subunits was monitored in the MCF7 cell line. All hGIRK1 splice variants and hGIRK4 were predominantly located within the endoplasmic reticulum. Heterologous expression in Xenopus laevis oocytes and two electrode voltage clamp experiments together with confocal microscopy were performed. Only the hGIRK1a subunit was able to form functional GIRK channels in connection with hGIRK4. The other splice variants are expressed, but exert a dominant negative effect on heterooligomeric channel function. Hence, alternative splicing of the KCNJ3 gene transcript in the MCF7 cell line leads to a family of mRNA's, encoding truncated versions of the hGIRK1 protein. The very high abundance of mRNA's encoding GIRK1 together with the presence of GIRK1 protein suggests a pathophysiological role in breast cancer.


Subject(s)
Breast Neoplasms/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , RNA Editing/genetics , Animals , Base Sequence , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Clone Cells , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Immunohistochemistry , Microscopy, Confocal , Molecular Sequence Data , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
19.
Mol Biol Cell ; 20(16): 3751-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19553470

ABSTRACT

MAL, a compact hydrophobic, four-transmembrane-domain apical protein that copurifies with detergent-resistant membranes is obligatory for the machinery that sorts glycophosphatidylinositol (GPI)-anchored proteins and others to the apical membrane in epithelia. The mechanism of MAL function in lipid-raft-mediated apical sorting is unknown. We report that MAL clusters formed by two independent procedures-spontaneous clustering of MAL tagged with the tandem dimer DiHcRED (DiHcRED-MAL) in the plasma membrane of COS7 cells and antibody-mediated cross-linking of FLAG-tagged MAL-laterally concentrate markers of sphingolipid rafts and exclude a fluorescent analogue of phosphatidylethanolamine. Site-directed mutagenesis and bimolecular fluorescence complementation analysis demonstrate that MAL forms oligomers via xx intramembrane protein-protein binding motifs. Furthermore, results from membrane modulation by using exogenously added cholesterol or ceramides support the hypothesis that MAL-mediated association with raft lipids is driven at least in part by positive hydrophobic mismatch between the lengths of the transmembrane helices of MAL and membrane lipids. These data place MAL as a key component in the organization of membrane domains that could potentially serve as membrane sorting platforms.


Subject(s)
Membrane Lipids/metabolism , Membrane Microdomains/chemistry , Membrane Transport Proteins/metabolism , Myelin Proteins/metabolism , Proteolipids/metabolism , Animals , COS Cells , Chlorocebus aethiops , Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Humans , Membrane Lipids/chemistry , Membrane Microdomains/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Myelin Proteins/chemistry , Myelin Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins , Protein Conformation , Proteolipids/chemistry , Proteolipids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
J Cell Sci ; 122(Pt 11): 1759-67, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435807

ABSTRACT

The cellular destination of secretory proteins is determined by interactions of their targeting motifs with coat-protein complexes. The transmembrane domain (TMD) of secretory proteins also plays a central role in their transport and targeting. However, a comprehensive model that considers both TMD- and targeting-sequence-mediated transport has never been advanced. We focused on the secretory transport of two fluorescently tagged membrane proteins: vesicular stomatitis virus G tsO45 (VSVG), which is a cargo protein that is a thermoreversible mutant, and the Golgi-resident protein GalT-CFP. A quantitative approach was applied to analyze, in living cells, secretory transport dynamics, as well as cargo concentration of YFP-tagged VSVG mutants with one, three, five, seven, eight or nine amino acids deleted from their TMD, as well as two or four amino acids added to their TMD. Changes in TMD length affected secretory transport dynamics and the extent of cargo concentration in the ER exit sites, demonstrating that the capacity of the transport machinery to concentrate cargo depends on the length of the TMD of the cargo protein.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Protein Transport/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...