Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters











Publication year range
1.
EMBO J ; 43(15): 3141-3174, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38877304

ABSTRACT

Migrating cells preferentially breach and integrate epithelial and endothelial monolayers at multicellular vertices. These sites are amenable to forces produced by the migrating cell and subsequent opening of the junctions. However, the cues that guide migrating cells to these entry portals, and eventually drive the transmigration process, are poorly understood. Here, we show that lymphatic endothelium multicellular junctions are the preferred sites of dendritic cell transmigration in both primary cell co-cultures and in mouse dermal explants. Dendritic cell guidance to multicellular junctions was dependent on the dendritic cell receptor CCR7, whose ligand, lymphatic endothelial chemokine CCL21, was exocytosed at multicellular junctions. Characterization of lymphatic endothelial secretory routes indicated Golgi-derived RAB6+ vesicles and RAB3+/27+ dense core secretory granules as intracellular CCL21 storage vesicles. Of these, RAB6+ vesicles trafficked CCL21 to the multicellular junctions, which were enriched with RAB6 docking factor ELKS (ERC1). Importantly, inhibition of RAB6 vesicle exocytosis attenuated dendritic cell transmigration. These data exemplify how spatially-restricted exocytosis of guidance cues helps to determine where dendritic cells transmigrate.


Subject(s)
Chemokine CCL21 , Dendritic Cells , Exocytosis , Receptors, CCR7 , rab GTP-Binding Proteins , Animals , Mice , Chemokine CCL21/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Dendritic Cells/metabolism , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Intercellular Junctions/metabolism , Transendothelial and Transepithelial Migration , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/cytology , Endothelial Cells/metabolism , Mice, Inbred C57BL , Humans , Coculture Techniques , Cells, Cultured , Cell Movement
2.
Nat Commun ; 15(1): 2547, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38514695

ABSTRACT

Focal adhesions (FAs) connect inner workings of cell to the extracellular matrix to control cell adhesion, migration and mechanosensing. Previous studies demonstrated that FAs contain three vertical layers, which connect extracellular matrix to the cytoskeleton. By using super-resolution iPALM microscopy, we identify two additional nanoscale layers within FAs, specified by actin filaments bound to tropomyosin isoforms Tpm1.6 and Tpm3.2. The Tpm1.6-actin filaments, beneath the previously identified α-actinin cross-linked actin filaments, appear critical for adhesion maturation and controlled cell motility, whereas the adjacent Tpm3.2-actin filament layer beneath seems to facilitate adhesion disassembly. Mechanistically, Tpm3.2 stabilizes ACF-7/MACF1 and KANK-family proteins at adhesions, and hence targets microtubule plus-ends to FAs to catalyse their disassembly. Tpm3.2 depletion leads to disorganized microtubule network, abnormally stable FAs, and defects in tail retraction during migration. Thus, FAs are composed of distinct actin filament layers, and each may have specific roles in coupling adhesions to the cytoskeleton, or in controlling adhesion dynamics.


Subject(s)
Actins , Focal Adhesions , Actins/metabolism , Focal Adhesions/metabolism , Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Protein Isoforms/metabolism
3.
Cancer Res ; 82(21): 3932-3949, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36054547

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest malignancies and potentially curable only with radical surgical resection at early stages. The tumor microenvironment has been shown to be central to the development and progression of PDAC. A better understanding of how early human PDAC metabolically communicates with its environment and differs from healthy pancreas could help improve PDAC diagnosis and treatment. Here we performed deep proteomic analyses from diagnostic specimens of operable, treatment-naïve PDAC patients (n = 14), isolating four tissue compartments by laser-capture microdissection: PDAC lesions, tumor-adjacent but morphologically benign exocrine glands, and connective tissues neighboring each of these compartments. Protein and pathway levels were compared between compartments and with control pancreatic proteomes. Selected targets were studied immunohistochemically in the 14 patients and in additional tumor microarrays, and lipid deposition was assessed by nonlinear label-free imaging (n = 16). Widespread downregulation of pancreatic secretory functions was observed, which was paralleled by high cholesterol biosynthetic activity without prominent lipid storage in the neoplastic cells. Stromal compartments harbored ample blood apolipoproteins, indicating abundant microvasculature at the time of tumor removal. The features best differentiating the tumor-adjacent exocrine tissue from healthy control pancreas were defined by upregulation of proteins related to lipid transport. Importantly, histologically benign exocrine regions harbored the most significant prognostic pathways, with proteins involved in lipid transport and metabolism, such as neutral cholesteryl ester hydrolase 1, associating with shorter survival. In conclusion, this study reveals prognostic molecular changes in the exocrine tissue neighboring pancreatic cancer and identifies enhanced lipid transport and metabolism as its defining features. SIGNIFICANCE: In clinically operable pancreatic cancer, regions distant from malignant cells already display proteomic changes related to lipid transport and metabolism that affect prognosis and may be pharmacologically targeted.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Proteomics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Lipids , Biomarkers, Tumor/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
4.
Traffic ; 21(5): 386-397, 2020 05.
Article in English | MEDLINE | ID: mdl-32144825

ABSTRACT

The human Niemann-Pick C1 (NPC1) gene encoding a 1278 amino acid protein is very heterogeneous. While some variants represent benign polymorphisms, NPC disease carriers and patients may possess rare variants, whose functional importance remains unknown. An NPC1 cDNA construct known as NPC1 wild-type variant (WT-V), distributed between laboratories and used as a WT control in several studies, also contains changes regarding specific amino acids compared to the NPC1 Genbank reference sequence. To improve the dissection of subtle functional differences, we generated human cells stably expressing NPC1 variants from the AAVS1 safe-harbor locus on an NPC1-null background engineered by CRISPR/Cas9 editing. We then employed high-content imaging with automated image analysis to quantitatively assess LDL-induced, time-dependent changes in lysosomal cholesterol content and lipid droplet formation. Our results indicate that the L472P change present in NPC1 WT-V compromises NPC1 functionality in lysosomal cholesterol export. All-atom molecular dynamics simulations suggest that the L472P change alters the relative position of the NPC1 middle and the C-terminal luminal domains, disrupting the recently characterized cholesterol efflux tunnel. These results reveal functional defects in NPC1 WT-V and highlight the strength of simulations and quantitative imaging upon stable protein expression in elucidating subtle differences in protein function.


Subject(s)
Cholesterol , Intracellular Signaling Peptides and Proteins , Proteins , Biological Transport , Cholesterol/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/metabolism , Molecular Dynamics Simulation , Niemann-Pick C1 Protein , Proteins/metabolism
5.
Dev Cell ; 50(4): 478-493.e9, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31178403

ABSTRACT

Seipin is an oligomeric integral endoplasmic reticulum (ER) protein involved in lipid droplet (LD) biogenesis. To study the role of seipin in LD formation, we relocalized it to the nuclear envelope and found that LDs formed at these new seipin-defined sites. The sites were characterized by uniform seipin-mediated ER-LD necks. At low seipin content, LDs only grew at seipin sites, and tiny, growth-incompetent LDs appeared in a Rab18-dependent manner. When seipin was removed from ER-LD contacts within 1 h, no lipid metabolic defects were observed, but LDs became heterogeneous in size. Studies in seipin-ablated cells and model membranes revealed that this heterogeneity arises via a biophysical ripening process, with triglycerides partitioning from smaller to larger LDs through droplet-bilayer contacts. These results suggest that seipin supports the formation of structurally uniform ER-LD contacts and facilitates the delivery of triglycerides from ER to LDs. This counteracts ripening-induced shrinkage of small LDs.


Subject(s)
Endoplasmic Reticulum/genetics , GTP-Binding Protein gamma Subunits/genetics , Lipid Droplets/metabolism , Fibroblasts/metabolism , Humans , Lipid Metabolism/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Primary Cell Culture , Triglycerides/genetics , Triglycerides/metabolism
6.
Biochimie ; 158: 90-101, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30590084

ABSTRACT

ORP2 is a sterol-binding protein with documented functions in lipid and glucose metabolism, Akt signaling, steroidogenesis, cell adhesion, migration and proliferation. Here we investigate the interactions of ORP2 with phosphoinositides (PIPs) by surface plasmon resonance (SPR), its affinity for cholesterol with a pull-down assay, and its capacity to transfer sterol in vitro. Moreover, we determine the effects of wild-type (wt) ORP2 and a mutant with attenuated PIP binding, ORP2(mHHK), on the subcellular distribution of cholesterol, and analyze the interaction of ORP2 with the related cholesterol transporter ORP1L. ORP2 showed specific affinity for PI(4,5)P2, PI(3,4,5)P3 and PI(4)P, with suggestive Kd values in the µM range. Also binding of cholesterol by ORP2 was detectable, but a Kd could not be determined. Wt ORP2 was in HeLa cells mainly detected in the cytosol, ER, late endosomes, and occasionally on lipid droplets (LDs), while ORP2(mHHK) displayed an enhanced LD localization. Overexpression of wt ORP2 shifted the D4H cholesterol probe away from endosomes, while ORP2(mHHK) caused endosomal accumulation of the probe. Although ORP2 failed to transfer dehydroergosterol in an in vitro assay where OSBP is active, its knock-down resulted in the accumulation of cholesterol in late endocytic compartments, as detected by both D4H and filipin probes. Interestingly, ORP2 was shown to interact and partially co-localize on late endosomes with ORP1L, a cholesterol transporter/sensor at ER-late endosome junctions. Our data demonstrates that ORP2 binds several phosphoinositides, both PI(4)P and multiply phosphorylated species. ORP2 regulates the subcellular distribution of cholesterol dependent on its PIP-binding capacity. The interaction of ORP2 with ORP1L suggests a concerted action of the two ORPs.


Subject(s)
Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lipid Droplets/metabolism , Phosphatidylinositols/metabolism , Receptors, Steroid/metabolism , Cholesterol/genetics , Endoplasmic Reticulum/genetics , Endosomes/genetics , HeLa Cells , Humans , Phosphatidylinositols/genetics , Receptors, Steroid/genetics
7.
J Cell Sci ; 130(6): 1147-1157, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28137756

ABSTRACT

Adaptation of cell shape and polarization through the formation and retraction of cellular protrusions requires balancing of endocytosis and exocytosis combined with fine-tuning of the local activity of small GTPases like Rab8. Here, we show that endocytic turnover of the plasma membrane at protrusions is directly coupled to surface removal and inactivation of Rab8. Removal is induced by reduced membrane tension and mediated by the GTPase regulator associated with focal adhesion kinase-1 (GRAF1, also known as ARHGAP26), a regulator of clathrin-independent endocytosis. GRAF1-depleted cells were deficient in multi-directional spreading and displayed elevated levels of GTP-loaded Rab8, which was accumulated at the tips of static protrusions. Furthermore, GRAF1 depletion impaired lumen formation and spindle orientation in a 3D cell culture system, indicating that GRAF1 activity regulates polarity establishment. Our data suggest that GRAF1-mediated removal of Rab8 from the cell surface restricts its activity during protrusion formation, thereby facilitating dynamic adjustment of the polarity axis.


Subject(s)
Cell Polarity , Endocytosis , rab GTP-Binding Proteins/metabolism , Animals , Cell Surface Extensions/metabolism , Dogs , GTPase-Activating Proteins/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Matrix Metalloproteinase 14/metabolism , Protein Binding , Protein Transport , Spindle Apparatus/metabolism
8.
J Cell Sci ; 130(5): 892-902, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28096473

ABSTRACT

The actin and intermediate filament cytoskeletons contribute to numerous cellular processes, including morphogenesis, cytokinesis and migration. These two cytoskeletal systems associate with each other, but the underlying mechanisms of this interaction are incompletely understood. Here, we show that inactivation of vimentin leads to increased actin stress fiber assembly and contractility, and consequent elevation of myosin light chain phosphorylation and stabilization of tropomyosin-4.2 (see Geeves et al., 2015). The vimentin-knockout phenotypes can be rescued by re-expression of wild-type vimentin, but not by the non-filamentous 'unit length form' vimentin, demonstrating that intact vimentin intermediate filaments are required to facilitate the effects on the actin cytoskeleton. Finally, we provide evidence that the effects of vimentin on stress fibers are mediated by activation of RhoA through its guanine nucleotide exchange factor GEF-H1 (also known as ARHGEF2). Vimentin depletion induces phosphorylation of the microtubule-associated GEF-H1 on Ser886, and thereby promotes RhoA activity and actin stress fiber assembly. Taken together, these data reveal a new mechanism by which intermediate filaments regulate contractile actomyosin bundles, and may explain why elevated vimentin expression levels correlate with increased migration and invasion of cancer cells.


Subject(s)
Actins/metabolism , Intermediate Filaments/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Stress Fibers/metabolism , Vimentin/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Phosphorylation
9.
Neurobiol Dis ; 96: 335-345, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27425888

ABSTRACT

In Parkinson's disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinson's disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue.


Subject(s)
Drug Design , Genetic Variation/genetics , Neurturin/chemistry , Neurturin/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Amphetamine/pharmacology , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Humans , Macaca fascicularis , Male , Models, Molecular , Neurturin/genetics , Oxidopamine/toxicity , Parkinson Disease/complications , Parkinson Disease/etiology , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Rats , Rats, Wistar , Stereotyped Behavior/drug effects , Sympatholytics/toxicity , Tyrosine 3-Monooxygenase/metabolism
10.
Curr Opin Lipidol ; 27(3): 282-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27054443

ABSTRACT

PURPOSE OF REVIEW: In this article, we summarize the present information related to the export of LDL-derived cholesterol from late endosomes, with a focus on Nieman-Pick disease, type C1 (NPC1) cholesterol delivery toward the endoplasmic reticulum (ER). We review data suggesting that several pathways may operate in parallel, including membrane transport routes and membrane contact sites (MCSs). RECENT FINDINGS: There is increasing appreciation that MCSs provide an important mechanism for intermembrane lipid transfer. In late endosome-ER contacts, three protein bridges involving oxysterol binding protein related protein (ORP)1L-vesicle associated membrane protein-associated protein (VAP), steroidogenic acute regulatory protein (StAR)D3-VAP and ORP5-NPC1 proteins have been reported. How much they contribute to the flux of LDL-cholesterol to the ER is currently open. Studies for lipid transfer via MCSs have been most advanced in Saccharomyces cerevisiae. Recently, a new sterol-binding protein family conserved between yeast and man was identified. Its members localize at MCSs and were named lipid transfer protein anchored at membrane contact sites (Lam) proteins. In yeast, sterol transfer between the ER and the yeast lysosome may be facilitated by a Lam protein. SUMMARY: Increasing insights into the role of MCSs in directional sterol delivery between membranes propose that they might provide routes for LDL-cholesterol transfer to the ER. Future work should reveal which specific contacts may operate for this, and how they are controlled by cholesterol homeostatic machineries.


Subject(s)
Cholesterol, LDL/metabolism , Endoplasmic Reticulum/metabolism , Animals , Biological Transport , Endosomes/metabolism , Humans , Intracellular Membranes/metabolism
11.
Biochim Biophys Acta ; 1863(5): 1006-13, 2016 May.
Article in English | MEDLINE | ID: mdl-26775587

ABSTRACT

In this review article, we summarize current knowledge on peroxisome biogenesis/functions and the role that small GTPases may play in these processes. Precise intracellular distribution of cell organelles requires their regulated association to microtubules and the actin cytoskeleton. In this respect, RhoGDP/RhoGTP favor binding of peroxisomes to microtubules and actin filaments. In its GTP-bound form, RhoA activates a regulatory cascade involving Rho kinaseII and non-muscle myosinIIA. Such interactions frequently depend on phosphoinositides (PIs) of which PI4P, PI(4,5)P2, and PI(3,5)P2 were found to be present in the peroxisomal membrane. PIs are pivotal determinants of intracellular signaling and known to regulate a wide range of cellular functions. In many of these functions, small GTPases are implicated. The small GTPase ADP-ribosylation factor 1 (Arf1), for example, is known to stimulate synthesis of PI4P and PI(4,5)P2 on the Golgi to regulate protein and lipid sorting. In vitro binding assays localized Arf1 and the COPI complex to peroxisomes. In light of the recent discussion of pre-peroxisomal vesicle generation at the ER, peroxisomal Arf1-COPI vesicles may serve retrograde transport of ER-resident components. A mass spectrometric screen localized various Rab proteins to peroxisomes. Overexpression of these proteins in combination with laser-scanning fluorescence microscopy co-localized Rab6, Rab8, Rab10, Rab14, and Rab18 with peroxisomal structures. By analogy to the role these proteins play in other organelle dynamics, we may envisage what the function of these proteins may be in relation to the peroxisomal compartment.


Subject(s)
Peroxisomes/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Gene Expression Regulation , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Humans , Microtubules/chemistry , Microtubules/metabolism , Nonmuscle Myosin Type IIA/genetics , Nonmuscle Myosin Type IIA/metabolism , Peroxisomes/chemistry , Phosphatidylinositols/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , rab GTP-Binding Proteins/genetics
12.
Methods Mol Biol ; 1298: 99-106, 2015.
Article in English | MEDLINE | ID: mdl-25800835

ABSTRACT

The Rab family of small GTPases acts as molecular switches that control various stages of vesicular transport. Rab8 functions in exocytic trafficking from the trans-Golgi network (TGN) and recycling endosomes to the plasma membrane. Rabin8 is a major guanine nucleotide exchange factor (GEF) for Rab8. It activates Rab8 by catalyzing its GDP release for subsequent GTP loading. However, how Rabin8 itself is activated in cells is unclear. Recently, it was found that Rabin8 is a downstream effector of Rab11, which controls vesicle exit from the recycling endosomes. Rab11, in its GTP-bound form, stimulates the GEF activity of Rabin8. The Rab11-Rabin8-Rab8 interactions thus couple vesicle generation from the donor compartment to its delivery to plasma membrane. Here we describe the methods we used to express and purify several Rab proteins, and to assay for the effect of Rab11 in the kinetic activation of Rabin8 GEF activity.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Protein Interaction Mapping/methods , rab GTP-Binding Proteins/metabolism , Humans , Kinetics , Mutation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/isolation & purification
13.
Hum Mol Genet ; 24(8): 2185-200, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25552655

ABSTRACT

Proper functioning of cilia, hair-like structures responsible for sensation and locomotion, requires nephrocystin-5 (NPHP5) and a multi-subunit complex called the Bardet-Biedl syndrome (BBS)ome, but their precise relationship is not understood. The BBSome is involved in the trafficking of membrane cargos to cilia. While it is known that a loss of any single subunit prevents ciliary trafficking of the BBSome and its cargos, the mechanisms underlying ciliary entry of this complex are not well characterized. Here, we report that a transition zone protein NPHP5 contains two separate BBS-binding sites and interacts with the BBSome to mediate its integrity. Depletion of NPHP5, or expression of NPHP5 mutant missing one binding site, specifically leads to dissociation of BBS2 and BBS5 from the BBSome and loss of ciliary BBS2 and BBS5 without compromising the ability of the other subunits to traffic into cilia. Depletion of Cep290, another transition zone protein that directly binds to NPHP5, causes additional dissociation of BBS8 and loss of ciliary BBS8. Furthermore, delivery of BBSome cargos, smoothened, VPAC2 and Rab8a, to the ciliary compartment is completely disabled in the absence of single BBS subunits, but is selectively impaired in the absence of NPHP5 or Cep290. These findings define a new role of NPHP5 and Cep290 in controlling integrity and ciliary trafficking of the BBSome, which in turn impinge on the delivery of ciliary cargo.


Subject(s)
Antigens, Neoplasm/metabolism , Bardet-Biedl Syndrome/metabolism , Calmodulin-Binding Proteins/metabolism , Cilia/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Antigens, Neoplasm/genetics , Bardet-Biedl Syndrome/genetics , Calmodulin-Binding Proteins/genetics , Cell Cycle Proteins , Cilia/genetics , Cytoskeletal Proteins , Humans , Multiprotein Complexes/genetics , Neoplasm Proteins/genetics , Protein Transport
14.
Proc Natl Acad Sci U S A ; 112(1): 148-53, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535387

ABSTRACT

Exocytosis is tightly regulated in many cellular processes, from neurite expansion to tumor proliferation. Rab8, a member of the Rab family of small GTPases, plays an important role in membrane trafficking from the trans-Golgi network and recycling endosomes to the plasma membrane. Rabin8 is a guanine nucleotide exchange factor (GEF) and major activator of Rab8. Investigating how Rabin8 is activated in cells is thus pivotal to the understanding of the regulation of exocytosis. Here we show that phosphorylation serves as an important mechanism for Rabin8 activation. We identified Rabin8 as a direct phospho-substrate of ERK1/2 in response to EGF signaling. At the molecular level, ERK phosphorylation relieves the autoinhibition of Rabin8, thus promoting its GEF activity. We further demonstrate that blocking ERK1/2-mediated phosphorylation of Rabin8 inhibits transferrin recycling to the plasma membrane. Together, our results suggest that ERK1/2 activate Rabin8 to regulate vesicular trafficking to the plasma membrane in response to extracellular signaling.


Subject(s)
Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Epidermal Growth Factor/pharmacology , Germinal Center Kinases , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Signal Transduction/drug effects , Transferrin/metabolism
15.
Exp Cell Res ; 331(2): 278-91, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25447204

ABSTRACT

ORP3 is an R-Ras interacting oxysterol-binding protein homolog that regulates cell adhesion and is overexpressed in several cancers. We investigated here a novel function of ORP3 dependent on its targeting to both the endoplasmic reticulum (ER) and the plasma membrane (PM). Using biochemical and cell imaging techniques we demonstrate the mechanistic requirements for the subcellular targeting and function of ORP3 in control of R-Ras activity. We show that hyperphosphorylated ORP3 (ORP3-P) selectively interacts with the ER membrane protein VAPA, and ORP3-VAPA complexes are targeted to PM sites via the ORP3 pleckstrin homology (PH) domain. A novel FFAT (two phenylalanines in an acidic tract)-like motif was identified in ORP3; only disruption of both the FFAT-like and canonical FFAT motif abolished the phorbol-12-myristate-13-acetate (PMA) stimulated interaction of ORP3-P with VAPA. Co-expression of ORP3 and VAPA induced R-Ras activation, dependent on the interactions of ORP3 with VAPA and the PM. Consistently, downstream AktS473 phosphorylation and ß1-integrin activity were enhanced by ORP3-VAPA. To conclude, phosphorylation of ORP3 controls its association with VAPA. Furthermore, we present evidence that ORP3-VAPA complexes stimulate R-Ras signaling.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Vesicular Transport Proteins/metabolism , ras Proteins/metabolism , Amino Acid Motifs , Carrier Proteins/biosynthesis , Cell Line, Tumor , Enzyme Activation , Fatty Acid-Binding Proteins , HEK293 Cells , Humans , Integrin beta1/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Vesicular Transport Proteins/biosynthesis
16.
Cell Mol Life Sci ; 72(10): 1967-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25420878

ABSTRACT

Oxysterol-binding protein/OSBP-related proteins (ORPs) constitute a conserved family of sterol/phospholipid-binding proteins with lipid transporter or sensor functions. We investigated the spatial occurrence and regulation of the interactions of human OSBP/ORPs or the S. cerevisiae orthologs, the Osh (OSBP homolog) proteins, with their endoplasmic reticulum (ER) anchors, the VAMP-associated proteins (VAPs), by employing bimolecular fluorescence complementation and pull-down set-ups. The ORP-VAP interactions localize frequently at distinct subcellular sites, shown in several cases to represent membrane contact sites (MCSs). Using established ORP ligand-binding domain mutants and pull-down assays with recombinant proteins, we show that ORP liganding regulates the ORP-VAP association, alters the subcellular targeting of ORP-VAP complexes, or modifies organelle morphology. There is distinct protein specificity in the effects of the mutants on subcellular targeting of ORP-VAP complexes. We provide evidence that complexes of human ORP2 and VAPs at ER-lipid droplet interfaces regulate the hydrolysis of triglycerides and lipid droplet turnover. The data suggest evolutionarily conserved, complex ligand-dependent functions of ORP-VAP complexes at MCSs, with implications for cellular lipid homeostasis and signaling.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Multiprotein Complexes/metabolism , Receptors, Steroid/metabolism , Recombinant Proteins/metabolism , Triglycerides/metabolism , Cell Line, Tumor , Fluorescent Antibody Technique , Genetic Complementation Test , Humans , Hydrolysis , Microscopy, Electron, Transmission , Microscopy, Fluorescence , RNA Interference , Yeasts
17.
Springerplus ; 3: 502, 2014.
Article in English | MEDLINE | ID: mdl-25279294

ABSTRACT

Brain-derived neurotrophic factor (BDNF) regulates multiple biological processes ranging from central nervous system development and function to neuroinflammation and myogenic differentiation and repair. While coordination of BDNF levels is central in determining the biological outcome, mechanisms involved in controlling BDNF levels are not fully understood. Here we find that both short (BDNF-S) and long (BDNF-L) BDNF 3'UTR isoforms contain conserved adenylate- and uridylate rich elements (AREs) that may serve as binding sites for RNA-binding proteins (ARE-BPs). We demonstrate that ARE-BPs tristetraprolin (TTP) and its family members butyrate response factor 1 (BRF1) and 2 (BRF2) negatively regulate expression from both BDNF-S and BDNF-L containing transcripts in several cell-lines and that interaction between TTP and AU-rich region in proximal 5' end of BDNF 3'UTR is direct. In line with the above, endogenous BDNF mRNA co-immunoprecipitates with endogenous TTP in differentiated mouse myoblast C2C12 cells and TTP overexpression destabilizes BDNF-S containing transcript. Finally, RNAi-mediated knock-down of TTP increases the levels of endogenous BDNF protein in C2C12 cells. Our findings uncover TTP as a novel regulator of BDNF assisting future studies in different physiological and pathological contexts.

18.
PLoS Genet ; 10(3): e1004193, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603431

ABSTRACT

Although the growth factor (GF) signaling guiding renal branching is well characterized, the intracellular cascades mediating GF functions are poorly understood. We studied mitogen-activated protein kinase (MAPK) pathway specifically in the branching epithelia of developing kidney by genetically abrogating the pathway activity in mice lacking simultaneously dual-specificity protein kinases Mek1 and Mek2. Our data show that MAPK pathway is heterogeneously activated in the subset of G1- and S-phase epithelial cells, and its tissue-specific deletion results in severe renal hypodysplasia. Consequently to the deletion of Mek1/2, the activation of ERK1/2 in the epithelium is lost and normal branching pattern in mutant kidneys is substituted with elongation-only phenotype, in which the epithelium is largely unable to form novel branches and complex three-dimensional patterns, but able to grow without primary defects in mitosis. Cellular characterization of double mutant epithelium showed increased E-cadherin at the cell surfaces with its particular accumulation at baso-lateral locations. This indicates changes in cellular adhesion, which were revealed by electron microscopic analysis demonstrating intercellular gaps and increased extracellular space in double mutant epithelium. When challenged to form monolayer cultures, the mutant epithelial cells were impaired in spreading and displayed strong focal adhesions in addition to spiky E-cadherin. Inhibition of MAPK activity reduced paxillin phosphorylation on serine 83 while remnants of phospho-paxillin, together with another focal adhesion (FA) protein vinculin, were augmented at cell surface contacts. We show that MAPK activity is required for branching morphogenesis, and propose that it promotes cell cycle progression and higher cellular motility through remodeling of cellular adhesions.


Subject(s)
Focal Adhesions/genetics , Kidney/growth & development , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Animals , Epithelial Cells/metabolism , Kidney/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinases/genetics , Morphogenesis/genetics , Phosphorylation , Signal Transduction/genetics , Vinculin/metabolism
19.
Dev Cell ; 27(3): 249-62, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24209575

ABSTRACT

Mammalian cells acquire cholesterol, a major membrane constituent, via low-density lipoprotein (LDL) uptake. However, the mechanisms by which LDL cholesterol reaches the plasma membrane (PM) have remained obscure. Here, we applied LDL labeled with BODIPY cholesteryl linoleate to identify this pathway in living cells. The egress of BODIPY cholesterol (BC) from late endosomal (LE) organelles was dependent on acid lipase and Niemann-Pick C1 (NPC1) protein, as for natural cholesterol. We show that NPC1 was needed to recruit Rab8a to BC-containing LEs, and Rab8a enhanced the motility and segregation of BC- and CD63-positive organelles from lysosomes. The BC carriers docked to the cortical actin by a Rab8a- and Myosin5b (Myo5b)-dependent mechanism, typically in the proximity of focal adhesions (FAs). LDL increased the number and dynamics of FAs and stimulated cell migration in an acid lipase, NPC1, and Rab8a-dependent fashion, providing evidence that this cholesterol delivery route to the PM is important for cell movement.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cholesterol, LDL/metabolism , Myosins/metabolism , rab GTP-Binding Proteins/metabolism , Biological Transport , Carrier Proteins/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Fluorescent Antibody Technique , Focal Adhesions/physiology , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Microscopy, Immunoelectron , Niemann-Pick C1 Protein , Porphobilinogen/analogs & derivatives , Porphobilinogen/pharmacology , Tetraspanin 30/metabolism , Wound Healing/drug effects
20.
Brain Behav ; 3(2): 75-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23532969

ABSTRACT

Cerebral dopamine neurotrophic factor (CDNF) protein has been shown to protect the nigrostriatal dopaminergic pathway when given as intrastriatal infusions in rat and mouse models of Parkinson's disease (PD). In this study, we assessed the neuroprotective effect of CDNF delivered with a recombinant adeno-associated viral (AAV) serotype 2 vector in a rat 6-hydroxydopamine (6-OHDA) model of PD. AAV2 vectors encoding CDNF, glial cell line-derived neurotrophic factor (GDNF), or green fluorescent protein were injected into the rat striatum. Protein expression analysis showed that our AAV2 vector efficiently delivered the neurotrophic factor genes into the brain and gave rise to a long-lasting expression of the proteins. Two weeks after AAV2 vector injection, 6-OHDA was injected into the rat striatum, creating a progressive degeneration of the nigrostriatal dopaminergic system. Treatment with AAV2-CDNF resulted in a marked decrease in amphetamine-induced ipsilateral rotations while it provided only partial protection of tyrosine hydroxylase (TH)-immunoreactive cells in the rat substantia nigra pars compacta and TH-reactive fibers in the striatum. Results from this study provide additional evidence that CDNF can be considered a potential treatment of Parkinson's disease.

SELECTION OF CITATIONS
SEARCH DETAIL