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










Publication year range
1.
Open Biol ; 13(9): 230122, 2023 09.
Article in English | MEDLINE | ID: mdl-37726093

ABSTRACT

KIF20A is a critical kinesin for cell division and a promising anti-cancer drug target. The mechanisms underlying its cellular roles remain elusive. Interestingly, unusual coupling between the nucleotide- and microtubule-binding sites of this kinesin-6 has been reported, but little is known about how its divergent sequence leads to atypical motility properties. We present here the first high-resolution structure of its motor domain that delineates the highly unusual structural features of this motor, including a long L6 insertion that integrates into the core of the motor domain and that drastically affects allostery and ATPase activity. Together with the high-resolution cryo-electron microscopy microtubule-bound KIF20A structure that reveals the microtubule-binding interface, we dissect the peculiarities of the KIF20A sequence that influence its mechanochemistry, leading to low motility compared to other kinesins. Structural and functional insights from the KIF20A pre-power stroke conformation highlight the role of extended insertions in shaping the motor's mechanochemical cycle. Essential for force production and processivity is the length of the neck linker in kinesins. We highlight here the role of the sequence preceding the neck linker in controlling its backward docking and show that a neck linker four times longer than that in kinesin-1 is required for the activity of this motor.


Subject(s)
Kinesins , Microtubules , Cryoelectron Microscopy , Kinesins/genetics , Binding Sites , Cell Division
2.
EMBO Rep ; 23(10): e54605, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35979738

ABSTRACT

Radial glial (RG) cells are the neural stem cells of the developing neocortex. Apical RG (aRG) cells can delaminate to generate basal RG (bRG) cells, a cell type associated with human brain expansion. Here, we report that aRG delamination is regulated by the post-Golgi secretory pathway. Using in situ subcellular live imaging, we show that post-Golgi transport of RAB6+ vesicles occurs toward the minus ends of microtubules and depends on dynein. We demonstrate that the apical determinant Crumbs3 (CRB3) is also transported by dynein. Double knockout of RAB6A/A' and RAB6B impairs apical localization of CRB3 and induces a retraction of aRG cell apical process, leading to delamination and ectopic division. These defects are phenocopied by knockout of the dynein activator LIS1. Overall, our results identify a RAB6-dynein-LIS1 complex for Golgi to apical surface transport in aRG cells, and highlights the role of this pathway in the maintenance of neuroepithelial integrity.


Subject(s)
Dyneins , rab GTP-Binding Proteins , Dyneins/genetics , Dyneins/metabolism , Golgi Apparatus/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neurons/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
3.
Biol Cell ; 113(7): 311-328, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33666950

ABSTRACT

BACKGROUND INFORMATION: Comprehensive libraries of plasmids for SARS-CoV-2 proteins with various tags (e.g., Strep, HA, Turbo) are now available. They enable the identification of numerous potential protein-protein interactions between the SARS-CoV-2 virus and host proteins. RESULTS: We present here a large library of SARS CoV-2 protein constructs fused with green and red fluorescent proteins and their initial characterisation in various human cell lines including lung epithelial cell models (A549, BEAS-2B), as well as in budding yeast. The localisation of a few SARS-CoV-2 proteins matches their proposed interactions with host proteins. These include the localisation of Nsp13 to the centrosome, Orf3a to late endosomes and Orf9b to mitochondria. CONCLUSIONS AND SIGNIFICANCE: This library should facilitate further cellular investigations, notably by imaging techniques.


Subject(s)
COVID-19/virology , Peptide Library , SARS-CoV-2/metabolism , Viral Proteins/metabolism , A549 Cells , Cell Line , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Host Microbial Interactions/physiology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SARS-CoV-2/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Time-Lapse Imaging , Viral Proteins/genetics , Red Fluorescent Protein
4.
Cells ; 11(1)2021 12 22.
Article in English | MEDLINE | ID: mdl-35011578

ABSTRACT

In the early secretory pathway, the delivery of anterograde cargoes from the endoplasmic reticulum (ER) exit sites (ERES) to the Golgi apparatus is a multi-step transport process occurring via the ER-Golgi intermediate compartment (IC, also called ERGIC). While the role microtubules in ER-to-Golgi transport has been well established, how the actin cytoskeleton contributes to this process remains poorly understood. Here, we report that Arp2/3 inhibition affects the network of acetylated microtubules around the Golgi and induces the accumulation of unusually long RAB1/GM130-positive carriers around the centrosome. These long carriers are less prone to reach the Golgi apparatus, and arrival of anterograde cargoes to the Golgi is decreased upon Arp2/3 inhibition. Our data suggest that Arp2/3-dependent actin polymerization maintains a stable network of acetylated microtubules, which ensures efficient cargo trafficking at the late stage of ER to Golgi transport.


Subject(s)
Actins/metabolism , Endoplasmic Reticulum/metabolism , Secretory Pathway , Acetylation , Actin-Related Protein 2-3 Complex , Autoantigens/metabolism , Centrosome/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Microtubules/metabolism , rab1 GTP-Binding Proteins/metabolism
5.
Development ; 147(19)2020 10 08.
Article in English | MEDLINE | ID: mdl-32895290

ABSTRACT

The Golgi-associated RAB GTPases, RAB6A and RAB6A', regulate anterograde and retrograde transport pathways from and to the Golgi. In vitro, RAB6A/A' control several cellular functions including cell division, migration, adhesion and polarity. However, their role remains poorly described in vivo Here, we generated BlgCre; Rab6aF/F mice presenting a specific deletion of Rab6a in the mammary luminal secretory lineage during gestation and lactation. Rab6a loss severely impaired the differentiation, maturation and maintenance of the secretory tissue, compromising lactation. The mutant epithelium displayed a decreased activation of STAT5, a key regulator of the lactogenic process primarily governed by prolactin. Data obtained with a mammary epithelial cell line suggested that defective STAT5 activation might originate from a perturbed transport of the prolactin receptor, altering its membrane expression and signaling cascade. Despite the major functional defects observed upon Rab6a deletion, the polarized organization of the mammary epithelial bilayer was preserved. Altogether, our data reveal a crucial role for RAB6A/A' in the lactogenic function of the mammary gland and suggest that the trafficking pathways controlled by RAB6A/A' depend on cell-type specialization and tissue context.


Subject(s)
Mammary Glands, Human/metabolism , STAT5 Transcription Factor/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cell Line , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Mammary Glands, Human/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , STAT5 Transcription Factor/genetics , rab GTP-Binding Proteins/genetics
6.
Cell Rep ; 30(13): 4505-4517.e5, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234483

ABSTRACT

TRPM8 is the main ion channel responsible for cold transduction in the somatosensory system. Nerve terminal availability of TRPM8 determines cold sensitivity, but how axonal secretory organelles control channel delivery remains poorly understood. Here we examine the distribution of TRPM8 and trafficking organelles in cold-sensitive peripheral axons and disrupt trafficking by targeting the ARF-GEF GBF1 pharmacologically or the small GTPase RAB6 by optogenetics. In axons of the sciatic nerve, inhibition of GBF1 interrupts TRPM8 trafficking and increases association with the trans-Golgi network, LAMP1, and Golgi satellites, which distribute profusely along the axonal shaft. Accordingly, both TRPM8-dependent ongoing activity and cold-evoked responses reversibly decline upon GBF1 inhibition in nerve endings of corneal cold thermoreceptors. Inhibition of RAB6, which also associates to Golgi satellites, decreases cold-induced responses in vivo. Our results support a non-conventional axonal trafficking mechanism controlling the availability of TRPM8 in axons and cold sensitivity in the peripheral nervous system.


Subject(s)
Axons/metabolism , Cold Temperature , Organelles/metabolism , TRPM Cation Channels/metabolism , Animals , Axons/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Humans , Male , Menthol/pharmacology , Mice , Optogenetics , Organelles/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Thermoreceptors/metabolism , rab GTP-Binding Proteins/metabolism
7.
J Cell Biol ; 218(7): 2215-2231, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31142554

ABSTRACT

To ensure their homeostasis and sustain differentiated functions, cells continuously transport diverse cargos to various cell compartments and in particular to the cell surface. Secreted proteins are transported along intracellular routes from the endoplasmic reticulum through the Golgi complex before reaching the plasma membrane along microtubule tracks. Using a synchronized secretion assay, we report here that exocytosis does not occur randomly at the cell surface but on localized hotspots juxtaposed to focal adhesions. Although microtubules are involved, the RAB6-dependent machinery plays an essential role. We observed that, irrespective of the transported cargos, most post-Golgi carriers are positive for RAB6 and that its inactivation leads to a broad reduction of protein secretion. RAB6 may thus be a general regulator of post-Golgi secretion.


Subject(s)
Focal Adhesions/genetics , Golgi Apparatus/genetics , Microtubules/genetics , rab GTP-Binding Proteins/genetics , Cell Differentiation/genetics , Endoplasmic Reticulum/genetics , Exocytosis/genetics , Golgi Apparatus/metabolism , HeLa Cells , Homeostasis/genetics , Humans
8.
Am J Hum Genet ; 104(2): 348-355, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661770

ABSTRACT

Steroid-resistant nephrotic syndrome (SRNS) is characterized by high-range proteinuria and most often focal and segmental glomerulosclerosis (FSGS). Identification of mutations in genes causing SRNS has improved our understanding of disease mechanisms and highlighted defects in the podocyte, a highly specialized glomerular epithelial cell, as major factors in disease pathogenesis. By exome sequencing, we identified missense mutations in TBC1D8B in two families with an X-linked early-onset SRNS with FSGS. TBC1D8B is an uncharacterized Rab-GTPase-activating protein likely involved in endocytic and recycling pathways. Immunofluorescence studies revealed TBC1D8B presence in human glomeruli, and affected individual podocytes displayed architectural changes associated with migration defects commonly found in FSGS. In zebrafish we demonstrated that both knockdown and knockout of the unique TBC1D8B ortholog-induced proteinuria and that this phenotype was rescued by human TBC1D8B mRNA injection, but not by either of the two mutated mRNAs. We also showed an interaction between TBC1D8B and Rab11b, a key protein in vesicular recycling in cells. Interestingly, both internalization and recycling processes were dramatically decreased in affected individuals' podocytes and fibroblasts, confirming the crucial role of TBC1D8B in the cellular recycling processes, probably as a Rab11b GTPase-activating protein. Altogether, these results confirmed that pathogenic variations in TBC1D8B are involved in X-linked podocytopathy and points to alterations in recycling processes as a mechanism of SRNS.


Subject(s)
Calcium-Binding Proteins/genetics , Genetic Diseases, X-Linked/genetics , Loss of Function Mutation , Nephrotic Syndrome/genetics , Vesicular Transport Proteins/genetics , Zebrafish Proteins/genetics , Animals , Biological Transport/genetics , Calcium-Binding Proteins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Kidney Glomerulus/metabolism , Male , Podocytes/cytology , Podocytes/metabolism , Vesicular Transport Proteins/metabolism , Exome Sequencing , Zebrafish , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/metabolism
9.
Elife ; 72018 09 20.
Article in English | MEDLINE | ID: mdl-30234483

ABSTRACT

One challenge in cell biology is to decipher the biophysical mechanisms governing protein enrichment on curved membranes and the resulting membrane deformation. The ERM protein ezrin is abundant and associated with cellular membranes that are flat, positively or negatively curved. Using in vitro and cell biology approaches, we assess mechanisms of ezrin's enrichment on curved membranes. We evidence that wild-type ezrin (ezrinWT) and its phosphomimetic mutant T567D (ezrinTD) do not deform membranes but self-assemble anti-parallelly, zipping adjacent membranes. EzrinTD's specific conformation reduces intermolecular interactions, allows binding to actin filaments, which reduces membrane tethering, and promotes ezrin binding to positively-curved membranes. While neither ezrinTD nor ezrinWT senses negative curvature alone, we demonstrate that interacting with curvature-sensing I-BAR-domain proteins facilitates ezrin enrichment in negatively-curved membrane protrusions. Overall, our work demonstrates that ezrin can tether membranes, or be targeted to curved membranes, depending on conformations and interactions with actin and curvature-sensing binding partners.


Subject(s)
Cell Membrane/chemistry , Cytoskeletal Proteins/chemistry , Mutant Proteins/chemistry , Protein Conformation , Actins/chemistry , Actins/genetics , Cell Membrane/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation , Protein Binding/genetics , Protein Domains/genetics
10.
Nat Commun ; 8(1): 1254, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29093437

ABSTRACT

The actin and microtubule cytoskeletons play important roles in Golgi structure and function, but how they are connected remain poorly known. In this study, we investigated whether RAB6 GTPase, a Golgi-associated RAB involved in the regulation of several transport steps at the Golgi level, and two of its effectors, Myosin IIA and KIF20A participate in the coupling between actin and microtubule cytoskeleton. We have previously shown that RAB6-Myosin IIA interaction is critical for the fission of RAB6-positive transport carriers from Golgi/TGN membranes. Here we show that KIF20A is also involved in the fission process and serves to anchor RAB6 on Golgi/TGN membranes near microtubule nucleating sites. We provide evidence that the fission events occur at a limited number of hotspots sites. Our results suggest that coupling between actin and microtubule cytoskeletons driven by Myosin II and KIF20A ensures the spatial coordination between RAB6-positive vesicles fission from Golgi/TGN membranes and their exit along microtubules.


Subject(s)
Golgi Apparatus/metabolism , Kinesins/metabolism , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , rab GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Cytoplasmic Vesicles/metabolism , Humans , Microtubules/metabolism , Nonmuscle Myosin Type IIA/metabolism , Rats , trans-Golgi Network/metabolism
11.
Nat Commun ; 8: 15835, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28607494

ABSTRACT

Exocytic carriers convey neo-synthesized components from the Golgi apparatus to the cell surface. While the release and anterograde movement of Golgi-derived vesicles require the small GTPase RAB6, its effector ELKS promotes the targeting and docking of secretory vesicles to particular areas of the plasma membrane. Here, we show that specialized cell types exploit and divert the secretory pathway towards lysosome related organelles. In cultured melanocytes, the secretory route relies on RAB6 and ELKS to directly transport and dock Golgi-derived carriers to melanosomes. By delivering specific cargos, such as MART-1 and TYRP2/ DCT, the RAB6/ELKS-dependent secretory pathway controls the formation and maturation of melanosomes but also pigment synthesis. In addition, pigmentation defects are observed in RAB6 KO mice. Our data together reveal for the first time that the secretory pathway can be directed towards intracellular organelles of endosomal origin to ensure their biogenesis and function.


Subject(s)
Lysosomes/metabolism , Melanocytes/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Endosomes/genetics , Endosomes/metabolism , Female , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Lysosomes/genetics , Male , Melanosomes/genetics , Melanosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Transport , Secretory Pathway , rab GTP-Binding Proteins/genetics
12.
Front Cell Dev Biol ; 4: 2, 2016.
Article in English | MEDLINE | ID: mdl-26925400

ABSTRACT

GTPases of the RAB family are key regulators of multiple steps of membrane trafficking. Several members of the RAB GTPase family have been implicated in mitotic progression. In this review, we will first focus on the function of endosome-associated RAB GTPases reported in early steps of mitosis, spindle pole maturation, and during cytokinesis. Second, we will discuss the role of Golgi-associated RAB GTPases at the metaphase/anaphase transition and during cytokinesis.

13.
Biol Cell ; 107(12): 427-39, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26304202

ABSTRACT

BACKGROUND INFORMATION: Rab6 is one of the most conserved Rab GTPaes throughout evolution and the most abundant Rab protein associated with the Golgi complex. The two ubiquitous Rab isoforms, Rab6A and Rab6A', that are generated by alternative splicing of the RAB6A gene, regulate several transport steps at the Golgi level, including retrograde transport between endosomes and Golgi, anterograde transport between Golgi and the plasma membrane, and intra-Golgi and Golgi to endoplasmic reticulum transport. RESULTS: We have generated mice with a conditional null allele of RAB6A. Mice homozygous for the RAB6A null allele died at an early stage of embryonic development. Mouse embryonic fibroblasts (MEFs) were isolated from RAB6A(loxP/loxP) Rosa26-CreERT2 and incubated with 4-hydroxy tamoxifen, resulting in the efficient depletion of Rab6A and Rab6A'. We show that Rab6 depletion affects cell growth, alters Golgi morphology and decreases the Golgi-associated levels of some known Rab6 effectors such as Bicaudal-D and myosin II. We also show that Rab6 depletion protects MEFs against ricin toxin and delays VSV-G secretion. CONCLUSIONS: Our study shows that RAB6 is an essential gene required for normal embryonic development. We confirm in MEF cells most of the functions previously attributed to the two ubiquitous Rab6 isoforms.


Subject(s)
Alternative Splicing/genetics , Embryonic Development/genetics , Endoplasmic Reticulum/metabolism , rab GTP-Binding Proteins/genetics , Animals , Cell Membrane/drug effects , Cell Membrane/genetics , Endoplasmic Reticulum/drug effects , Fibroblasts/drug effects , Gene Expression Regulation, Developmental/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Humans , Mice , Mice, Knockout , Ricin/toxicity , Tamoxifen/administration & dosage , rab GTP-Binding Proteins/biosynthesis
14.
J Cell Sci ; 128(16): 3068-81, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163492

ABSTRACT

Given their obligate intracellular lifestyle, Chlamydia trachomatis ensure that they have access to multiple host sources of essential lipids by interfering with vesicular transport. These bacteria hijack Rab6-, Rab11- and Rab14-controlled trafficking pathways to acquire sphingomyelin from the Golgi complex. Another important source of sphingolipids, phospholipids and cholesterol are multivesicular bodies (MVBs). Despite their participation in chlamydial inclusion development and bacterial replication, the molecular mechanisms mediating the interaction between MVBs and chlamydial inclusions remain unknown. In the present study, we demonstrate that Rab39a labels a subset of late endocytic vesicles - mainly MVBs - that move along microtubules. Moreover, Rab39a is actively recruited to chlamydial inclusions throughout the pathogen life cycle by a bacterial-driven process that depends on the Rab39a GTP- or GDP-binding state. Interestingly, Rab39a participates in the delivery of MVBs and host sphingolipids to maturing chlamydial inclusions, thereby promoting inclusion growth and bacterial development. Taken together, our findings indicate that Rab39a favours chlamydial replication and infectivity. This is the first report showing that a late endocytic Rab GTPase is involved in chlamydial infection development.


Subject(s)
Chlamydia trachomatis/metabolism , Multivesicular Bodies/metabolism , Sphingolipids/metabolism , rab GTP-Binding Proteins/metabolism , Chlamydia trachomatis/pathogenicity , Cholesterol/metabolism , Golgi Apparatus/chemistry , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Multivesicular Bodies/microbiology , Phospholipids/metabolism , Sphingomyelins/metabolism , Transport Vesicles/metabolism
15.
Methods Mol Biol ; 1298: 73-83, 2015.
Article in English | MEDLINE | ID: mdl-25800833

ABSTRACT

Myosins are actin-based motor proteins that are involved in a wide variety of cellular processes such as membrane transport, muscle contraction, and cell division. Humans have over 40 myosins that can be placed into 18 classes, the malfunctioning of a number of which can lead to disease. There are three members of the human class V myosin family, myosins Va, Vb, and Vc. People lacking functional myosin Va suffer from a rare autosomal recessive disease called Griscelli's Syndrome type I (GS1) that is characterized by severe neurological defects and partial albinism. Mutations in the myosin Vb gene lead to an epithelial disorder called microvillus inclusion disease (MVID) that is often fatal in infants. The class V myosins have been implicated in the transport of diverse cargoes such as melanosomes in pigment cells, synaptic vesicles in neurons, RNA transcripts in a variety of cell types, and organelles such as the endoplasmic reticulum. The Rab GTPases play a critical role in recruiting class V myosins to their cargo. We recently published a study in which we used the yeast two-hybrid system to systematically test myosin Va for its ability to interact with each member of the human Rab GTPase family. We present here a detailed description of this yeast two-hybrid "living chip" assay. Furthermore, we present a protocol for validating positive interactions obtained from this screen by coimmunoprecipitation.


Subject(s)
Myosin Type V/metabolism , Two-Hybrid System Techniques , rab GTP-Binding Proteins/metabolism , Humans , Immunoprecipitation , Myosin Type V/isolation & purification , Protein Binding , Two-Hybrid System Techniques/instrumentation , rab GTP-Binding Proteins/isolation & purification
16.
Cell Rep ; 6(3): 445-54, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24462287

ABSTRACT

Early endosomes consist of vacuolar sorting and tubular recycling domains that segregate components fated for degradation in lysosomes or reuse by recycling to the plasma membrane or Golgi. The tubular transport intermediates that constitute recycling endosomes function in cell polarity, migration, and cytokinesis. Endosomal tubulation and fission require both actin and intact microtubules, but although factors that stabilize recycling endosomal tubules have been identified, those required for tubule generation from vacuolar sorting endosomes (SEs) remain unknown. We show that the microtubule motor KIF13A associates with recycling endosome tubules and controls their morphogenesis. Interfering with KIF13A function impairs the formation of endosomal tubules from SEs with consequent defects in endosome homeostasis and cargo recycling. Moreover, KIF13A interacts and cooperates with RAB11 to generate endosomal tubules. Our data illustrate how a microtubule motor couples early endosome morphogenesis to its motility and function.


Subject(s)
Endocytosis , Endosomes/metabolism , Kinesins/metabolism , Morphogenesis , Endosomes/ultrastructure , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Protein Binding , Protein Transport , rab GTP-Binding Proteins/metabolism
17.
PLoS One ; 8(12): e82223, 2013.
Article in English | MEDLINE | ID: mdl-24312644

ABSTRACT

The Golgi apparatus is an intracellular compartment necessary for post-translational modification, sorting and transport of proteins. It plays a key role in mitotic entry through the Golgi mitotic checkpoint. In order to identify new proteins involved in the Golgi mitotic checkpoint, we combine the results of a knockdown screen for mitotic phenotypes and a localization screen. Using this approach, we identify a new Golgi protein C11ORF24 (NP_071733.1). We show that C11ORF24 has a signal peptide at the N-terminus and a transmembrane domain in the C-terminal region. C11ORF24 is localized on the Golgi apparatus and on the trans-Golgi network. A large part of the protein is present in the lumen of the Golgi apparatus whereas only a short tail extends into the cytosol. This cytosolic tail is well conserved in evolution. By FRAP experiments we show that the dynamics of C11ORF24 in the Golgi membrane are coherent with the presence of a transmembrane domain in the protein. C11ORF24 is not only present on the Golgi apparatus but also cycles to the plasma membrane via endosomes in a pH sensitive manner. Moreover, via video-microscopy studies we show that C11ORF24 is found on transport intermediates and is colocalized with the small GTPase RAB6, a GTPase involved in anterograde transport from the Golgi to the plasma membrane. Knocking down C11ORF24 does not lead to a mitotic phenotype or an intracellular transport defect in our hands. All together, these data suggest that C11ORF24 is present on the Golgi apparatus, transported to the plasma membrane and cycles back through the endosomes by way of RAB6 positive carriers.


Subject(s)
Cell Membrane/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Cell Cycle/physiology , HeLa Cells , Humans , Protein Processing, Post-Translational/physiology
18.
Methods Cell Biol ; 115: 97-108, 2013.
Article in English | MEDLINE | ID: mdl-23973068

ABSTRACT

Mammalian cells show a large diversity in shape and are both shape-changing and mobile when cultured on conventional uniform substrates. The use of micropatterning techniques limits the number of variable parameters, by imposing shape and standardized adhesive areas on the cells, which facilitates analysis. By changing size or shape of the micropattern, for example, forcing a polar axis on the cell, it is possible to study how these parameters impact organelle organization, distribution, and dynamics inside the cell. To study the mitochondrial network, which is composed of dynamic tubular organelles dependent on the microtubule cytoskeleton for its distribution, it is important to be able to distinguish between distinct mitochondria. Here, we present a practical method with which we spread the cells on large patterns created with deep UV technique, which not only makes the cells uniform in size and shape as well as immobile, and therefore easier to compare and analyze, but also expands the mitochondrial network and allows for an easier tracking of appropriately labeled individual mitochondria.


Subject(s)
Cell Shape/physiology , Microtubules/metabolism , Mitochondria/metabolism , Cell Adhesion , Cells, Cultured , Humans , Retinal Pigment Epithelium/cytology , Staining and Labeling/methods
19.
J Neurosci ; 32(41): 14227-41, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23055492

ABSTRACT

Although essential for their neuronal function, the molecular mechanisms underlying the dendritic targeting of serotonin G-protein-coupled receptors are poorly understood. Here, we characterized a Yif1B-dependent vesicular scaffolding complex mediating the intracellular traffic of the rat 5-HT(1A) receptor (5-HT(1A)R) toward dendrites. By combining directed mutagenesis, GST-pull down, and surface plasmon resonance, we identified a tribasic motif in the C-tail of the 5-HT(1A)R on which Yif1B binds directly with high affinity (K(D) ≈ 37 nM). Moreover, we identified Yip1A, Rab6, and Kif5B as new partners of the 5-HT(1A)R/Yif1B complex, and showed that their expression in neurons is also crucial for the dendritic targeting of the 5-HT(1A)R. Live videomicroscopy revealed that 5-HT(1A)R, Yif1B, Yip1A, and Rab6 traffic in vesicles exiting the soma toward the dendritic tree, and also exhibit bidirectional motions, sustaining their role in 5-HT(1A)R dendritic targeting. Hence, we propose a new trafficking pathway model in which Yif1B is the scaffold protein recruiting the 5-HT(1A)R in a complex including Yip1A and Rab6, with Kif5B and dynein as two opposite molecular motors coordinating the traffic of vesicles along dendritic microtubules. This targeting pathway opens new insights for G-protein-coupled receptors trafficking in neurons.


Subject(s)
Dendrites/physiology , Matrix Attachment Regions/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Receptor, Serotonin, 5-HT1A/physiology , Synaptic Vesicles/metabolism , Vesicular Transport Proteins/physiology , Animals , Cells, Cultured , Dendrites/genetics , Gene Targeting/methods , Humans , Matrix Attachment Regions/genetics , Microtubules/metabolism , Microtubules/physiology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Transport/genetics , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Synaptic Vesicles/genetics , Vesicular Transport Proteins/genetics
20.
Nat Genet ; 44(4): 456-60, S1-3, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406640

ABSTRACT

Familial hyperkalemic hypertension (FHHt) is a Mendelian form of arterial hypertension that is partially explained by mutations in WNK1 and WNK4 that lead to increased activity of the Na(+)-Cl(-) cotransporter (NCC) in the distal nephron. Using combined linkage analysis and whole-exome sequencing in two families, we identified KLHL3 as a third gene responsible for FHHt. Direct sequencing of 43 other affected individuals revealed 11 additional missense mutations that were associated with heterogeneous phenotypes and diverse modes of inheritance. Polymorphisms at KLHL3 were not associated with blood pressure. The KLHL3 protein belongs to the BTB-BACK-kelch family of actin-binding proteins that recruit substrates for Cullin3-based ubiquitin ligase complexes. KLHL3 is coexpressed with NCC and downregulates NCC expression at the cell surface. Our study establishes a role for KLHL3 as a new member of the complex signaling pathway regulating ion homeostasis in the distal nephron and indirectly blood pressure.


Subject(s)
Carrier Proteins/genetics , Ion Transport/genetics , Nephrons/metabolism , Pseudohypoaldosteronism/genetics , Sodium Chloride Symporters/metabolism , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Base Sequence , Blood Pressure/genetics , Child , Female , Humans , Kidney/metabolism , Male , Microfilament Proteins , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Pseudohypoaldosteronism/metabolism , Pseudohypoaldosteronism/physiopathology , Sequence Analysis, DNA , Signal Transduction , Sodium Chloride Symporters/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...