Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS Biol ; 20(11): e3001855, 2022 11.
Article in English | MEDLINE | ID: mdl-36395330

ABSTRACT

The neuronal microtubule cytoskeleton is key to establish axon-dendrite polarity. Dendrites are characterized by the presence of minus-end out microtubules. However, the mechanisms that organize these microtubules with the correct orientation are still poorly understood. Using Caenorhabditis elegans as a model system for microtubule organization, we characterized the role of 2 microtubule minus-end related proteins in this process, the microtubule minus-end stabilizing protein calmodulin-regulated spectrin-associated protein (CAMSAP/PTRN-1), and the NINEIN homologue, NOCA-2 (noncentrosomal microtubule array). We found that CAMSAP and NINEIN function in parallel to mediate microtubule organization in dendrites. During dendrite outgrowth, RAB-11-positive vesicles localized to the dendrite tip to nucleate microtubules and function as a microtubule organizing center (MTOC). In the absence of either CAMSAP or NINEIN, we observed a low penetrance MTOC vesicles mislocalization to the cell body, and a nearly fully penetrant phenotype in double mutant animals. This suggests that both proteins are important for localizing the MTOC vesicles to the growing dendrite tip to organize microtubules minus-end out. Whereas NINEIN localizes to the MTOC vesicles where it is important for the recruitment of the microtubule nucleator γ-tubulin, CAMSAP localizes around the MTOC vesicles and is cotranslocated forward with the MTOC vesicles upon dendritic growth. Together, these results indicate that microtubule nucleation from the MTOC vesicles and microtubule stabilization are both important to localize the MTOC vesicles distally to organize dendritic microtubules minus-end out.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Microtubules , Microtubule-Organizing Center , Tubulin , Dendrites , Microtubule-Associated Proteins , Caenorhabditis elegans Proteins/genetics
2.
J Neurosci ; 39(25): 4864-4873, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30967428

ABSTRACT

Selective cargo transport into axons and dendrites over the microtubule network is essential for neuron polarization. The axon initial segment (AIS) separates the axon from the somatodendritic compartment and controls the microtubule-dependent transport into the axon. Interestingly, the AIS has a characteristic microtubule organization; it contains bundles of closely spaced microtubules with electron dense cross-bridges, referred to as microtubule fascicles. The microtubule binding protein TRIM46 localizes to the AIS and when overexpressed in non-neuronal cells forms microtubule arrays that closely resemble AIS fascicles in neurons. However, the precise role of TRIM46 in microtubule fasciculation in neurons has not been studied. Here we developed a novel correlative light and electron microscopy approach to study AIS microtubule organization. We show that in cultured rat hippocampal neurons of both sexes, TRIM46 levels steadily increase at the AIS during early neuronal differentiation and at the same time closely spaced microtubules form, whereas the fasciculated microtubules appear at later developmental stages. Moreover, we localized TRIM46 to the electron dense cross-bridges and show that depletion of TRIM46 causes loss of cross-bridges and increased microtubule spacing. These data indicate that TRIM46 has an essential role in organizing microtubule fascicles in the AIS.SIGNIFICANCE STATEMENT The axon initial segment (AIS) is a specialized region at the proximal axon where the action potential is initiated. In addition the AIS separates the axon from the somatodendritic compartment, where it controls protein transport to establish and maintain neuron polarity. Cargo vesicles destined for the axon recognize specialized microtubule tracks that enter the AIS. Interestingly the microtubules entering the AIS form crosslinked bundles, called microtubule fascicules. Recently we found that the microtubule-binding protein TRIM46 localizes to the AIS, where it may organize the AIS microtubules. In the present study we developed a novel correlative light and electron microscopy approach to study the AIS microtubules during neuron development and identified an essential role for TRIM46 in microtubule fasciculation.


Subject(s)
Axon Fasciculation/physiology , Axon Initial Segment/metabolism , Microtubules/metabolism , Neurons/metabolism , Tripartite Motif Proteins/metabolism , Animals , Cell Polarity/physiology , Cells, Cultured , Cytoskeleton/metabolism , Female , Hippocampus/cytology , Hippocampus/metabolism , Male , Neurons/cytology , Rats , Tripartite Motif Proteins/genetics
3.
J Cell Sci ; 131(20)2018 10 22.
Article in English | MEDLINE | ID: mdl-30254025

ABSTRACT

The specific organization of the neuronal microtubule cytoskeleton in axons and dendrites is an evolutionarily conserved determinant of neuronal polarity that allows for selective cargo sorting. However, how dendritic microtubules are organized and whether local differences influence cargo transport remains largely unknown. Here, we use live-cell imaging to systematically probe the microtubule organization in Caenorhabditiselegans neurons, and demonstrate the contribution of distinct mechanisms in the organization of dendritic microtubules. We found that most non-ciliated neurons depend on unc-116 (kinesin-1), unc-33 (CRMP) and unc-44 (ankyrin) for correct microtubule organization and polarized cargo transport, as previously reported. Ciliated neurons and the URX neuron, however, use an additional pathway to nucleate microtubules at the tip of the dendrite, from the base of the cilium in ciliated neurons. Since inhibition of distal microtubule nucleation affects distal dendritic transport, we propose a model in which the presence of a microtubule-organizing center at the dendrite tip ensures correct dendritic cargo transport.


Subject(s)
Caenorhabditis elegans/metabolism , Dendrites/metabolism , Microtubules/metabolism , Protein Transport/physiology , Animals , Cells, Cultured
4.
Nat Methods ; 14(5): 479-482, 2017 May.
Article in English | MEDLINE | ID: mdl-28394337

ABSTRACT

The actin cytoskeleton is essential for many fundamental biological processes, but tools for directly manipulating actin dynamics are limited to cell-permeable drugs that preclude single-cell perturbations. Here we describe DeActs, genetically encoded actin-modifying polypeptides, which effectively induce actin disassembly in eukaryotic cells. We demonstrate that DeActs are universal tools for studying the actin cytoskeleton in single cells in culture, tissues, and multicellular organisms including various neurodevelopmental model systems.


Subject(s)
ADP Ribose Transferases/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Gelsolin/genetics , Peptides/genetics , Recombinant Fusion Proteins/genetics , Virulence Factors/genetics , Actin Cytoskeleton/genetics , Actins/genetics , Animals , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Rats , Transfection
5.
BMC Biol ; 14: 66, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27506200

ABSTRACT

BACKGROUND: Affinity purification followed by mass spectrometry (AP/MS) is a widely used approach to identify protein interactions and complexes. In multicellular organisms, the accurate identification of protein complexes by AP/MS is complicated by the potential heterogeneity of complexes in different tissues. Here, we present an in vivo biotinylation-based approach for the tissue-specific purification of protein complexes from Caenorhabditis elegans. Tissue-specific biotinylation is achieved by the expression in select tissues of the bacterial biotin ligase BirA, which biotinylates proteins tagged with the Avi peptide. RESULTS: We generated N- and C-terminal tags combining GFP with the Avi peptide sequence, as well as four BirA driver lines expressing BirA ubiquitously and specifically in the seam and hyp7 epidermal cells, intestine, or neurons. We validated the ability of our approach to identify bona fide protein interactions by identifying the known LGL-1 interaction partners PAR-6 and PKC-3. Purification of the Discs large protein DLG-1 identified several candidate interaction partners, including the AAA-type ATPase ATAD-3 and the uncharacterized protein MAPH-1.1. We have identified the domains that mediate the DLG-1/ATAD-3 interaction, and show that this interaction contributes to C. elegans development. MAPH-1.1 co-purified specifically with DLG-1 purified from neurons, and shared limited homology with the microtubule-associated protein MAP1A, a known neuronal interaction partner of mammalian DLG4/PSD95. A CRISPR/Cas9-engineered GFP::MAPH-1.1 fusion was broadly expressed and co-localized with microtubules. CONCLUSIONS: The method we present here is able to purify protein complexes from specific tissues. We uncovered a series of DLG-1 interactors, and conclude that ATAD-3 is a biologically relevant interaction partner of DLG-1. Finally, we conclude that MAPH-1.1 is a microtubule-associated protein of the MAP1 family and a candidate neuron-specific interaction partner of DLG-1.


Subject(s)
Caenorhabditis elegans Proteins/isolation & purification , Caenorhabditis elegans/metabolism , Guanylate Kinases/metabolism , Organ Specificity , Protein Interaction Mapping/methods , Amino Acid Sequence , Animals , Biotinylation , Caenorhabditis elegans Proteins/metabolism , Fluorescent Antibody Technique , Multiprotein Complexes/isolation & purification , Neurons/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Reproducibility of Results
6.
EMBO J ; 29(24): 4094-105, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21076391

ABSTRACT

Wnt proteins are lipid-modified glycoproteins that have important roles in development, adult tissue homeostasis and disease. Secretion of Wnt proteins from producing cells is mediated by the Wnt-binding protein MIG-14/Wls, which binds Wnt in the Golgi network and transports it to the cell surface for release. It has recently been shown that recycling of MIG-14/Wls from the plasma membrane to the trans-Golgi network is required for efficient Wnt secretion, but the mechanism of this retrograde transport pathway is still poorly understood. In this study, we report the identification of MTM-6 and MTM-9 as novel regulators of MIG-14/Wls trafficking in Caenorhabditis elegans. MTM-6 and MTM-9 are myotubularin lipid phosphatases that function as a complex to dephosphorylate phosphatidylinositol-3-phosphate, a central regulator of endosomal trafficking. We show that mutation of mtm-6 or mtm-9 leads to defects in several Wnt-dependent processes and demonstrate that MTM-6 is required in Wnt-producing cells as part of the MIG-14/Wls-recycling pathway. This function is evolutionarily conserved, as the MTM-6 orthologue DMtm6 is required for Wls stability and Wg secretion in Drosophila. We conclude that regulation of endosomal trafficking by the MTM-6/MTM-9 myotubularin complex is required for the retromer-dependent recycling of MIG-14/Wls and Wnt secretion.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Carrier Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Drosophila/enzymology , Gene Knockdown Techniques , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Wnt Proteins/metabolism
7.
Development ; 138(14): 2915-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21653614

ABSTRACT

The migration of neuroblasts along the anteroposterior body axis of C. elegans is controlled by multiple Wnts that act partially redundantly to guide cells to their precisely defined final destinations. How positional information is specified by this system is, however, still largely unknown. Here, we used a novel fluorescent in situ hybridization methods to generate a quantitative spatiotemporal expression map of the C. elegans Wnt genes. We found that the five Wnt genes are expressed in a series of partially overlapping domains along the anteroposterior axis, with a predominant expression in the posterior half of the body. Furthermore, we show that a secreted Frizzled-related protein is expressed at the anterior end of the body axis, where it inhibits Wnt signaling to control neuroblast migration. Our findings reveal that a system of regionalized Wnt gene expression and anterior Wnt inhibition guides the highly stereotypic migration of neuroblasts in C. elegans. Opposing expression of Wnts and Wnt inhibitors has been observed in basal metazoans and in the vertebrate neurectoderm. Our results in C. elegans support the notion that a system of posterior Wnt signaling and anterior Wnt inhibition is an evolutionarily conserved principle of primary body axis specification.


Subject(s)
Body Patterning/physiology , Caenorhabditis elegans/embryology , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Glycoproteins/metabolism , Neurons/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Cloning, Molecular , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Neurons/cytology , Plasmids/genetics
8.
PLoS Genet ; 5(5): e1000494, 2009 May.
Article in English | MEDLINE | ID: mdl-19461891

ABSTRACT

Post-transcriptional regulatory mechanisms are widely used to influence cell fate decisions in germ cells, early embryos, and neurons. Many conserved cytoplasmic RNA regulatory proteins associate with each other and assemble on target mRNAs, forming ribonucleoprotein (RNP) complexes, to control the mRNAs translational output. How these RNA regulatory networks are orchestrated during development to regulate cell fate decisions remains elusive. We addressed this problem by focusing on Caenorhabditis elegans germline development, an exemplar of post-transcriptional control mechanisms. Here, we report the discovery of GLS-1, a new factor required for many aspects of germline development, including the oocyte cell fate in hermaphrodites and germline survival. We find that GLS-1 is a cytoplasmic protein that localizes in germ cells dynamically to germplasm (P) granules. Furthermore, its functions depend on its ability to form a protein complex with the RNA-binding Bicaudal-C ortholog GLD-3, a translational activator and P granule component important for similar germ cell fate decisions. Based on genetic epistasis experiments and in vitro competition experiments, we suggest that GLS-1 releases FBF/Pumilio from GLD-3 repression. This facilitates the sperm-to-oocyte switch, as liberated FBF represses the translation of mRNAs encoding spermatogenesis-promoting factors. Our proposed molecular mechanism is based on the GLS-1 protein acting as a molecular mimic of FBF/Pumilio. Furthermore, we suggest that a maternal GLS-1/GLD-3 complex in early embryos promotes the expression of mRNAs encoding germline survival factors. Our work identifies GLS-1 as a fundamental regulator of germline development. GLS-1 directs germ cell fate decisions by modulating the availability and activity of a single translational network component, GLD-3. Hence, the elucidation of the mechanisms underlying GLS-1 functions provides a new example of how conserved machinery can be developmentally manipulated to influence cell fate decisions and tissue development.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Germ Cells/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Cell Survival , Chromosome Mapping , Cytoplasmic Granules/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Helminth , Germ Cells/cytology , Male , Models, Biological , Molecular Sequence Data , Mutation , Oocytes/cytology , Oocytes/metabolism , Oogenesis/genetics , Protein Binding , RNA Processing, Post-Transcriptional , RNA, Helminth/genetics , RNA, Helminth/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sex Determination Processes , Spermatozoa/cytology , Spermatozoa/metabolism
9.
Curr Biol ; 30(5): 899-908.e6, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32084403

ABSTRACT

Neuronal dendrites are characterized by an anti-parallel microtubule organization. The mixed oriented microtubules promote dendrite development and facilitate polarized cargo trafficking; however, the mechanism that regulates dendritic microtubule organization is still unclear. Here, we found that the kinesin-14 motor KIFC3 is important for organizing dendritic microtubules and to control dendrite development. The kinesin-14 motor proteins (Drosophila melanogaster Ncd, Saccharomyces cerevisiae Kar3, Saccharomyces pombe Pkl1, and Xenopus laevis XCTK2) are characterized by a C-terminal motor domain and are well described to organize the spindle microtubule during mitosis using an additional microtubule binding site in the N terminus [1-4]. In mammals, there are three kinesin-14 members, KIFC1, KIFC2, and KIFC3. It was recently shown that KIFC1 is important for organizing axonal microtubules in neurons, a process that depends on the two microtubule-interacting domains [5]. Unlike KIFC1, KIFC2 and KIFC3 lack the N-terminal microtubule binding domain and only have one microtubule-interacting domain, the motor domain [6, 7]. Thus, in order to regulate microtubule-microtubule crosslinking or sliding, KIFC2 and KIFC3 need to interact with additional microtubule binding proteins to connect two microtubules. We found that KIFC3 has a dendrite-specific distribution and interacts with microtubule minus-end binding protein CAMSAP2. Depletion of KIFC3 or CAMSAP2 results in increased microtubule dynamics during dendritic development. We propose a model in which CAMSAP2 anchors KIFC3 at microtubule minus ends and immobilizes microtubule arrays in dendrites.


Subject(s)
Kinesins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Binding , Protein Transport
10.
Elife ; 92020 04 15.
Article in English | MEDLINE | ID: mdl-32293562

ABSTRACT

The development of a polarized neuron relies on the selective transport of proteins to axons and dendrites. Although it is well known that the microtubule cytoskeleton has a central role in establishing neuronal polarity, how its specific organization is established and maintained is poorly understood. Using the in vivo model system Caenorhabditis elegans, we found that the highly conserved UNC-119 protein provides a link between the membrane-associated Ankyrin (UNC-44) and the microtubule-associated CRMP (UNC-33). Together they form a periodic membrane-associated complex that anchors axonal and dendritic microtubule bundles to the cortex. This anchoring is critical to maintain microtubule organization by opposing kinesin-1 powered microtubule sliding. Disturbing this molecular complex alters neuronal polarity and causes strong developmental defects of the nervous system leading to severely paralyzed animals.


Subject(s)
Cell Polarity/physiology , Cytoskeleton/physiology , Microtubules/physiology , Neurons/physiology , Animals , Ankyrins/physiology , Caenorhabditis elegans , Caenorhabditis elegans Proteins/physiology , Cells, Cultured , Cerebral Cortex/physiology , Locomotion , Nerve Growth Factors/physiology , Nerve Tissue Proteins
11.
Cell Rep ; 26(8): 1988-1999.e6, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784582

ABSTRACT

The motor protein kinesin-1 plays an important role in polarized sorting of transport vesicles to the axon. However, the mechanism by which the axonal entry of kinesin-1-dependent cargo transport is regulated remains unclear. Microtubule-associated protein MAP7 (ensconsin in Drosophila) is an essential kinesin-1 cofactor and promotes kinesin-1 recruitment to microtubules. Here, we found that MAP7 family member MAP7D2 concentrates at the proximal axon, where it overlaps with the axon initial segment and interacts with kinesin-1. Depletion of MAP7D2 results in reduced axonal cargo entry and defects in axon development and neuronal migration. We propose a model in which MAP7D2 in the proximal axon locally promotes kinesin-1-mediated cargo entry into the axon.


Subject(s)
Axonal Transport , Axons/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Binding Sites , COS Cells , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Kinesins/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Protein Binding , Rats , Rats, Wistar
12.
Neuron ; 104(2): 305-321.e8, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31474508

ABSTRACT

The axon initial segment (AIS) is a unique neuronal compartment that plays a crucial role in the generation of action potential and neuronal polarity. The assembly of the AIS requires membrane, scaffolding, and cytoskeletal proteins, including Ankyrin-G and TRIM46. How these components cooperate in AIS formation is currently poorly understood. Here, we show that Ankyrin-G acts as a scaffold interacting with End-Binding (EB) proteins and membrane proteins such as Neurofascin-186 to recruit TRIM46-positive microtubules to the plasma membrane. Using in vitro reconstitution and cellular assays, we demonstrate that TRIM46 forms parallel microtubule bundles and stabilizes them by acting as a rescue factor. TRIM46-labeled microtubules drive retrograde transport of Neurofascin-186 to the proximal axon, where Ankyrin-G prevents its endocytosis, resulting in stable accumulation of Neurofascin-186 at the AIS. Neurofascin-186 enrichment in turn reinforces membrane anchoring of Ankyrin-G and subsequent recruitment of TRIM46-decorated microtubules. Our study reveals feedback-based mechanisms driving AIS assembly.


Subject(s)
Ankyrins/metabolism , Axon Initial Segment/metabolism , Cell Adhesion Molecules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Animals , Axon Initial Segment/ultrastructure , Axonal Transport , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cytoskeleton , Endocytosis , Feedback, Physiological , HEK293 Cells , Hippocampus/cytology , Humans , Microtubules/ultrastructure , Neurons/ultrastructure , Rats , Tripartite Motif Proteins/metabolism
13.
Curr Biol ; 26(4): R153-4, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26906482

ABSTRACT

To establish and maintain their complex morphology and function, neurons and other polarized cells exploit cytoskeletal motor proteins to distribute cargoes to specific compartments. Recent studies in cultured cells have used inducible motor protein recruitment to explore how different motors contribute to polarized transport and to control the subcellular positioning of organelles. Such approaches also seem promising avenues for studying motor activity and organelle positioning within more complex cellular assemblies, but their applicability to multicellular in vivo systems has so far remained unexplored. Here, we report the development of an optogenetic organelle transport strategy in the in vivo model system Caenorhabditis elegans. We demonstrate that movement and pausing of various organelles can be achieved by recruiting the proper cytoskeletal motor protein with light. In neurons, we find that kinesin and dynein exclusively target the axon and dendrite, respectively, revealing the basic principles for polarized transport. In vivo control of motor attachment and organelle distributions will be widely useful in exploring the mechanisms that govern the dynamic morphogenesis of cells and tissues, within the context of a developing animal.


Subject(s)
Caenorhabditis elegans/metabolism , Dyneins/metabolism , Kinesins/metabolism , Light , Organelles/metabolism , Protein Multimerization , Animals , Biological Transport , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/cytology , Dyneins/chemistry , Kinesins/chemistry , Optogenetics , Organelles/chemistry
14.
Curr Biol ; 26(7): 849-61, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-26948876

ABSTRACT

Kinesin motor proteins play a fundamental role for normal neuronal development by controlling intracellular cargo transport and microtubule (MT) cytoskeleton organization. Regulating kinesin activity is important to ensure their proper functioning, and their misregulation often leads to severe human neurological disorders. Homozygous nonsense mutations in kinesin-binding protein (KBP)/KIAA1279 cause the neurological disorder Goldberg-Shprintzen syndrome (GOSHS), which is characterized by intellectual disability, microcephaly, and axonal neuropathy. Here, we show that KBP regulates kinesin activity by interacting with the motor domains of a specific subset of kinesins to prevent their association with the MT cytoskeleton. The KBP-interacting kinesins include cargo-transporting motors such as kinesin-3/KIF1A and MT-depolymerizing motor kinesin-8/KIF18A. We found that KBP blocks KIF1A/UNC-104-mediated synaptic vesicle transport in cultured hippocampal neurons and in C. elegans PVD sensory neurons. In contrast, depletion of KBP results in the accumulation of KIF1A motors and synaptic vesicles in the axonal growth cone. We also show that KBP regulates neuronal MT dynamics by controlling KIF18A activity. Our data suggest that KBP functions as a kinesin inhibitor that modulates MT-based cargo motility and depolymerizing activity of a subset of kinesin motors. We propose that misregulation of KBP-controlled kinesin motors may represent the underlying molecular mechanism that contributes to the neuropathological defects observed in GOSHS patients.


Subject(s)
Craniofacial Abnormalities/metabolism , Hirschsprung Disease/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Animals , Caenorhabditis elegans/metabolism , Carrier Proteins/metabolism , Kinesins/chemistry , Kinesins/metabolism , Mice , Neurons/metabolism , Synaptic Vesicles/metabolism
15.
Neuron ; 88(6): 1208-1226, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671463

ABSTRACT

Axon formation, the initial step in establishing neuronal polarity, critically depends on local microtubule reorganization and is characterized by the formation of parallel microtubule bundles. How uniform microtubule polarity is achieved during axonal development remains an outstanding question. Here, we show that the tripartite motif containing (TRIM) protein TRIM46 plays an instructive role in the initial polarization of neuronal cells. TRIM46 is specifically localized to the newly specified axon and, at later stages, partly overlaps with the axon initial segment (AIS). TRIM46 specifically forms closely spaced parallel microtubule bundles oriented with their plus-end out. Without TRIM46, all neurites have a dendrite-like mixed microtubule organization resulting in Tau missorting and altered cargo trafficking. By forming uniform microtubule bundles in the axon, TRIM46 is required for neuronal polarity and axon specification in vitro and in vivo. Thus, TRIM46 defines a unique axonal cytoskeletal compartment for regulating microtubule organization during neuronal development.


Subject(s)
Axons/physiology , Axons/ultrastructure , Cell Polarity/physiology , Microtubules/physiology , Microtubules/ultrastructure , Nerve Tissue Proteins/physiology , Nerve Tissue Proteins/ultrastructure , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Cerebral Cortex/ultrastructure , Chlorocebus aethiops , Female , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/physiology , Neurons/ultrastructure , Pregnancy , Rats , Repressor Proteins/physiology , Repressor Proteins/ultrastructure
16.
Cell Signal ; 26(1): 19-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24056045

ABSTRACT

Secretion of Wnt proteins is mediated by the Wnt sorting receptor Wls, which transports Wnt from the Golgi to the cell surface for release. To maintain efficient Wnt secretion, Wls is recycled back to the trans-Golgi network (TGN) through a retromer dependent endosome to TGN retrieval pathway. It has recently been shown that this is mediated by an alternative retromer pathway in which the sorting nexin SNX3 interacts with the cargo-selective subcomplex of the retromer to sort Wls into a retrieval pathway that is morphologically distinct from the classical SNX-BAR dependent retromer pathway. Here, we investigated how sorting of Wls between the two different retromer pathways is specified. We found that when the function of the cargo-selective subcomplex of the retromer is partially disrupted, Wnt secretion can be restored by interfering with the maturation of late endosomes to lysosomes. This leads to an accumulation of Wls in late endosomes and facilitates the retrieval of Wls through a SNX-BAR dependent retromer pathway. Our results are consistent with a model in which spatial separation of the SNX3 and SNX-BAR retromer complexes along the endosomal maturation pathway as well as cargo-specific mechanisms contribute to the selective retrieval of Wls through the SNX3 retromer pathway.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , Endosomes/metabolism , Mutation/genetics , Wnt Proteins/metabolism , Animals , Caenorhabditis elegans/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Knockdown Techniques , Genes, Dominant , Models, Biological , Protein Subunits/genetics , Signal Transduction , Transgenes
17.
Neuron ; 82(5): 1058-73, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24908486

ABSTRACT

In neurons, most microtubules are not associated with a central microtubule-organizing center (MTOC), and therefore, both the minus and plus-ends of these non-centrosomal microtubules are found throughout the cell. Microtubule plus-ends are well established as dynamic regulatory sites in numerous processes, but the role of microtubule minus-ends has remained poorly understood. Using live-cell imaging, high-resolution microscopy, and laser-based microsurgery techniques, we show that the CAMSAP/Nezha/Patronin family protein CAMSAP2 specifically localizes to non-centrosomal microtubule minus-ends and is required for proper microtubule organization in neurons. CAMSAP2 stabilizes non-centrosomal microtubules and is required for neuronal polarity, axon specification, and dendritic branch formation in vitro and in vivo. Furthermore, we found that non-centrosomal microtubules in dendrites are largely generated by γ-Tubulin-dependent nucleation. We propose a two-step model in which γ-Tubulin initiates the formation of non-centrosomal microtubules and CAMSAP2 stabilizes the free microtubule minus-ends in order to control neuronal polarity and development.


Subject(s)
Axons/metabolism , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Microtubules/metabolism , Pyramidal Cells/metabolism , Animals , Axons/ultrastructure , Dendrites/ultrastructure , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Microtubule-Associated Proteins , Microtubules/ultrastructure , Pyramidal Cells/ultrastructure , Rats
18.
Dev Cell ; 26(4): 326-8, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23987508

ABSTRACT

Molecular motors employ specific adaptor proteins to dock on transport cargos. Reporting in The Journal of Cell Biology, Fu and Holzbaur (2013) show that the adaptor JNK interacting protein 1 (JIP1) binds kinesin-1 and dynactin and controls bidirectional axonal amyloid precursor protein trafficking, suggesting a regulatory role for adaptors during cargo transport.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/metabolism , Axonal Transport , Dyneins/metabolism , Kinesins/metabolism , Animals
19.
Acta Neuropathol Commun ; 1: 24, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-24252306

ABSTRACT

BACKGROUND: Protein aggregation and the formation of intracellular inclusions are a central feature of many neurodegenerative disorders, but precise knowledge about their pathogenic role is lacking in most instances. Here we have characterized inclusions formed in transgenic mice carrying the P56S mutant form of VAPB that causes various motor neuron syndromes including ALS8. RESULTS: Inclusions in motor neurons of VAPB-P56S transgenic mice are characterized by the presence of smooth ER-like tubular profiles, and are immunoreactive for factors that operate in the ER associated degradation (ERAD) pathway, including p97/VCP, Derlin-1, and the ER membrane chaperone BAP31. The presence of these inclusions does not correlate with signs of axonal and neuronal degeneration, and axotomy leads to their gradual disappearance, indicating that they represent reversible structures. Inhibition of the proteasome and knockdown of the ER membrane chaperone BAP31 increased the size of mutant VAPB inclusions in primary neuron cultures, while knockdown of TEB4, an ERAD ubiquitin-protein ligase, reduced their size. Mutant VAPB did not codistribute with mutant forms of seipin that are associated with an autosomal dominant motor neuron disease, and accumulate in a protective ER derived compartment termed ERPO (ER protective organelle) in neurons. CONCLUSIONS: The data indicate that the VAPB-P56S inclusions represent a novel reversible ER quality control compartment that is formed when the amount of mutant VAPB exceeds the capacity of the ERAD pathway and that isolates misfolded and aggregated VAPB from the rest of the ER. The presence of this quality control compartment reveals an additional level of flexibility of neurons to cope with misfolded protein stress in the ER.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Endoplasmic Reticulum/physiology , Inclusion Bodies/physiology , Motor Neurons/physiology , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Axons/ultrastructure , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum-Associated Degradation/physiology , Gene Knockdown Techniques , Hippocampus/physiopathology , Hippocampus/ultrastructure , Inclusion Bodies/ultrastructure , Mice, Transgenic , Motor Neurons/ultrastructure , Mutation , Rats , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sciatic Nerve/ultrastructure , Vesicular Transport Proteins/genetics
20.
Nat Cell Biol ; 13(8): 914-923, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725319

ABSTRACT

Wnt proteins are lipid-modified glycoproteins that play a central role in development, adult tissue homeostasis and disease. Secretion of Wnt proteins is mediated by the Wnt-binding protein Wntless (Wls), which transports Wnt from the Golgi network to the cell surface for release. It has recently been shown that recycling of Wls through a retromer-dependent endosome-to-Golgi trafficking pathway is required for efficient Wnt secretion, but the mechanism of this retrograde transport pathway is poorly understood. Here, we report that Wls recycling is mediated through a retromer pathway that is independent of the retromer sorting nexins SNX1-SNX2 and SNX5-SNX6. We have found that the unrelated sorting nexin, SNX3, has an evolutionarily conserved function in Wls recycling and Wnt secretion and show that SNX3 interacts directly with the cargo-selective subcomplex of the retromer to sort Wls into a morphologically distinct retrieval pathway. These results demonstrate that SNX3 is part of an alternative retromer pathway that functionally separates the retrograde transport of Wls from other retromer cargo.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Sorting Nexins/metabolism , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified , Biological Transport, Active , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Endosomes/metabolism , HeLa Cells , Humans , Models, Biological , RNA Interference , Signal Transduction , Sorting Nexins/antagonists & inhibitors , Sorting Nexins/genetics , trans-Golgi Network/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL