Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 10: 1153420, 2023.
Article in English | MEDLINE | ID: mdl-37228585

ABSTRACT

Eukaryotic cells intrinsically change their shape, by changing the composition of their membrane and by restructuring their underlying cytoskeleton. We present here further studies and extensions of a minimal physical model, describing a closed vesicle with mobile curved membrane protein complexes. The cytoskeletal forces describe the protrusive force due to actin polymerization which is recruited to the membrane by the curved protein complexes. We characterize the phase diagrams of this model, as function of the magnitude of the active forces, nearest-neighbor protein interactions and the proteins' spontaneous curvature. It was previously shown that this model can explain the formation of lamellipodia-like flat protrusions, and here we explore the regimes where the model can also give rise to filopodia-like tubular protrusions. We extend the simulation with curved components of both convex and concave species, where we find the formation of complex ruffled clusters, as well as internalized invaginations that resemble the process of endocytosis and macropinocytosis. We alter the force model representing the cytoskeleton to simulate the effects of bundled instead of branched structure, resulting in shapes which resemble filopodia.

2.
Adv Sci (Weinh) ; 10(7): e2207368, 2023 03.
Article in English | MEDLINE | ID: mdl-36698307

ABSTRACT

The cell migration cycle, well-established in 2D, proceeds with forming new protrusive structures at the cell membrane and subsequent redistribution of contractile machinery. Three-dimensional (3D) environments are complex and composed of 1D fibers, and 1D fibers are shown to recapitulate essential features of 3D migration. However, the establishment of protrusive activity at the cell membrane and contractility in 1D fibrous environments remains partially understood. Here the role of membrane curvature regulator IRSp53 is examined as a coupler between actin filaments and plasma membrane during cell migration on single, suspended 1D fibers. IRSp53 depletion reduced cell-length spanning actin stress fibers that originate from the cell periphery, protrusive activity, and contractility, leading to uncoupling of the nucleus from cellular movements. A theoretical model capable of predicting the observed transition of IRSp53-depleted cells from rapid stick-slip migration to smooth and slower migration due to reduced actin polymerization at the cell edges is developed, which is verified by direct measurements of retrograde actin flow using speckle microscopy. Overall, it is found that IRSp53 mediates actin recruitment at the cellular tips leading to the establishment of cell-length spanning fibers, thus demonstrating a unique role of IRSp53 in controlling cell migration in 3D.


Subject(s)
Actin Cytoskeleton , Actins , Cell Movement , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Membrane/metabolism , Cell Movement/genetics , Cell Nucleus/metabolism , Pseudopodia/genetics , Pseudopodia/metabolism
3.
Dev Cell ; 56(6): 842-859.e8, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33756122

ABSTRACT

Extracellular vesicles (EVs) are classified as large EVs (l-EVs, or microvesicles) and small EVs (s-EVs, or exosomes). S-EVs are thought to be generated from endosomes through a process that mainly depends on the ESCRT protein complex, including ALG-2 interacting protein X (ALIX). However, the mechanisms of l-EV generation from the plasma membrane have not been identified. Membrane curvatures are generated by the bin-amphiphysin-rvs (BAR) family proteins, among which the inverse BAR (I-BAR) proteins are involved in filopodial protrusions. Here, we show that the I-BAR proteins, including missing in metastasis (MIM), generate l-EVs by scission of filopodia. Interestingly, MIM-containing l-EV production was promoted by in vivo equivalent external forces and by the suppression of ALIX, suggesting an alternative mechanism of vesicle formation to s-EVs. The MIM-dependent l-EVs contained lysophospholipids and proteins, including IRS4 and Rac1, which stimulated the migration of recipient cells through lamellipodia formation. Thus, these filopodia-dependent l-EVs, which we named as filopodia-derived vesicles (FDVs), modify cellular behavior.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Movement , Endocytosis , Endosomal Sorting Complexes Required for Transport/metabolism , Exosomes/metabolism , Extracellular Vesicles/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Pseudopodia/metabolism , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cell Membrane/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HEK293 Cells , Humans , Microfilament Proteins/genetics , Neoplasm Proteins/genetics
4.
Chromosome Res ; 29(1): 37-50, 2021 03.
Article in English | MEDLINE | ID: mdl-33694045

ABSTRACT

There are few technologies that can capture mitotic processes occurring in three-dimensional space with the desired spatiotemporal resolution. Due to such technical limitations, our understanding of mitosis, which has been studied since the early 1880s, is still incomplete with regard to mitotic processes and their regulatory mechanisms at a molecular level. A recently developed high-resolution type of light-sheet microscopy, lattice light-sheet microscopy (LLSM), has achieved unprecedented spatiotemporal resolution scans of intracellular spaces at the whole-cell level. This technology enables experiments that were not possible before (e.g., tracking of growth of every spindle microtubule end and discrimination of individual chromosomes in living cells), thus providing a new avenue for the analysis of mitotic processes. Herein, principles of LLSM technology are introduced, as well as experimental techniques that became possible with LLSM. In addition, issues remaining to be solved for use of this technology in mitosis research, big image data problems, are presented to help guide mitosis research into a new era.


Subject(s)
Microscopy , Mitosis , Microtubules , Spindle Apparatus
5.
J Cell Biol ; 219(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32886101

ABSTRACT

Collective migration of epithelial cells plays crucial roles in various biological processes such as cancer invasion. In migrating epithelial sheets, leader cells form lamellipodia to advance, and follower cells also form similar motile apparatus at cell-cell boundaries, which are called cryptic lamellipodia (c-lamellipodia). Using adenocarcinoma-derived epithelial cells, we investigated how c-lamellipodia form and found that they sporadically grew from around E-cadherin-based adherens junctions (AJs). WAVE and Arp2/3 complexes were localized along the AJs, and silencing them not only interfered with c-lamellipodia formation but also prevented follower cells from trailing the leaders. Disruption of AJs by removing αE-catenin resulted in uncontrolled c-lamellipodia growth, and this was brought about by myosin II activation and the resultant contraction of AJ-associated actomyosin cables. Additional observations indicated that c-lamellipodia tended to grow at mechanically weak sites of the junction. We conclude that AJs not only tie cells together but also support c-lamellipodia formation by recruiting actin regulators, enabling epithelial cells to undergo ordered collective migration.


Subject(s)
Adherens Junctions/genetics , Cell Movement/genetics , Pseudopodia/genetics , Wiskott-Aldrich Syndrome Protein Family/genetics , Actin-Related Protein 2-3 Complex/genetics , Actins/genetics , Cadherins/genetics , Cell Line , Epithelial Cells/metabolism , Humans , Pseudopodia/metabolism
6.
Cells ; 9(5)2020 05 19.
Article in English | MEDLINE | ID: mdl-32438637

ABSTRACT

From cells to organisms, every living system is three-dimensional (3D), but the performance of fluorescence microscopy has been largely limited when attempting to obtain an overview of systems' dynamic processes in three dimensions. Recently, advanced light-sheet illumination technologies, allowing drastic improvement in spatial discrimination, volumetric imaging times, and phototoxicity/photobleaching, have been making live imaging to collect precise and reliable 3D information increasingly feasible. In particular, lattice light-sheet microscopy (LLSM), using an ultrathin light-sheet, enables whole-cell 3D live imaging of cellular processes, including mitosis, at unprecedented spatiotemporal resolution for extended periods of time. This technology produces immense and complex data, including a significant amount of information, raising new challenges for big image data analysis and new possibilities for data utilization. Once the data are digitally archived in a computer, the data can be reused for various purposes by anyone at any time. Such an information science approach has the potential to revolutionize the use of bioimage data, and provides an alternative method for cell biology research in a data-driven manner. In this article, we introduce examples of analyzing digital mitotic spindles and discuss future perspectives in cell biology.


Subject(s)
Computer Simulation , Data Collection , Mitosis , Spindle Apparatus/metabolism , Animals , Embryo, Nonmammalian/anatomy & histology , HeLa Cells , Humans , Imaging, Three-Dimensional , Microtubule-Associated Proteins/metabolism , Zebrafish/embryology
7.
Neuron ; 99(3): 464-479.e7, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30033154

ABSTRACT

Repeated environmental stress has been proposed to induce neural inflammation together with depression and anxiety. Innate immune receptors, such as Toll-like receptors (TLRs), are activated by exogenous or endogenous ligands to evoke inflammation. Here we show that the loss of TLR2 and TLR4 (TLR2/4) abolished repeated social defeat stress (R-SDS)-induced social avoidance and anxiety in mice. TLR2/4 deficiency mitigated R-SDS-induced neuronal response attenuation, dendritic atrophy, and microglial activation in the medial prefrontal cortex (mPFC). Furthermore, mPFC microglia-specific TLR2/4 knockdown blocked social avoidance. Transcriptome analyses revealed that R-SDS induced IL-1α and TNF-α in mPFC microglia in a TLR2/4-dependent manner, and antibody blockade of these cytokines in the mPFC suppressed R-SDS-induced social avoidance. These results identify TLR2/4 as crucial mediators of R-SDS-induced microglial activation in the mPFC, which leads to neuronal and behavioral changes through inflammation-related cytokines, highlighting unexpected pivotal roles of innate immunity in the mPFC in repeated environmental stress-induced behavioral changes. VIDEO ABSTRACT.


Subject(s)
Avoidance Learning/physiology , Microglia/metabolism , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Animals , Cells, Cultured , HEK293 Cells , Humans , Immunity, Innate/physiology , Interpersonal Relations , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/transplantation , Prefrontal Cortex/cytology , Prefrontal Cortex/immunology , Stress, Psychological/immunology , Stress, Psychological/psychology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
8.
Mol Biol Cell ; 28(1): 41-53, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27852897

ABSTRACT

Negatively regulating key signaling pathways is critical to development and altered in cancer. Wnt signaling is kept off by the destruction complex, which is assembled around the tumor suppressors APC and Axin and targets ß-catenin for destruction. Axin and APC are large proteins with many domains and motifs that bind other partners. We hypothesized that if we identified the essential regions required for APC:Axin cooperative function and used these data to design a minimal ß-catenin-destruction machine, we would gain new insights into the core mechanisms of destruction complex function. We identified five key domains/motifs in APC or Axin that are essential for their function in reconstituting Wnt regulation. Strikingly, however, certain APC and Axin mutants that are nonfunctional on their own can complement one another in reducing ß-catenin, revealing that the APC:Axin complex is a highly robust machine. We used these insights to design a minimal ß-catenin-destruction machine, revealing that a minimized chimeric protein covalently linking the five essential regions of APC and Axin reconstitutes destruction complex internal structure, size, and dynamics, restoring efficient ß-catenin destruction in colorectal tumor cells. On the basis of our data, we propose a new model of the mechanistic function of the destruction complex as an integrated machine.


Subject(s)
Axin Protein/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Animals , Axin Protein/physiology , Cell Line, Tumor , Drosophila/metabolism , Humans , Phosphorylation , Protein Domains , Repressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/physiology
9.
J Biomed Opt ; 20(10): 101206, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26527322

ABSTRACT

Mitotic apparatus, which comprises hundreds of microtubules, plays an essential role in cell division, ensuring the correct segregation of chromosomes into each daughter cell. To gain insight into its regulatory mechanisms, it is essential to detect and analyze the behavior of individual microtubule filaments. However, the discrimination of discrete microtubule filaments within the mitotic apparatus is beyond the capabilities of conventional light microscopic technologies. Recently, we detected three-dimensional (3-D) microtubule growth dynamics within the cellular cytoplasmic space using lattice light-sheet microscopy in conjunction with microtubule growth marker protein end-binding 1, a microtubule plus-end-tracking protein, which was fused to green fluorescent protein (EB1-GFP). This technique enables high-resolution 3-D imaging at subsecond intervals. We adapted mathematical computing and geometric representation techniques to analyze spatial variations in microtubule growth dynamics within the mitotic spindle apparatus. Our analytical approach enabled the different dynamic properties of individual microtubules to be determined, including the direction and speed of their growth, and their growth duration within a 3-D spatial map. Our analysis framework provides an important step toward a more comprehensive understanding of the mechanisms driving cellular machinery at the whole-cell level.


Subject(s)
Cell Tracking/methods , Green Fluorescent Proteins/pharmacokinetics , Imaging, Three-Dimensional/methods , Microtubule-Associated Proteins/pharmacokinetics , Microtubules/physiology , Spindle Apparatus/physiology , Cell Enlargement , Fluorescent Dyes/pharmacokinetics , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/genetics , Microtubules/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Spindle Apparatus/ultrastructure
10.
Science ; 346(6208): 1257998, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25342811

ABSTRACT

Although fluorescence microscopy provides a crucial window into the physiology of living specimens, many biological processes are too fragile, are too small, or occur too rapidly to see clearly with existing tools. We crafted ultrathin light sheets from two-dimensional optical lattices that allowed us to image three-dimensional (3D) dynamics for hundreds of volumes, often at subsecond intervals, at the diffraction limit and beyond. We applied this to systems spanning four orders of magnitude in space and time, including the diffusion of single transcription factor molecules in stem cell spheroids, the dynamic instability of mitotic microtubules, the immunological synapse, neutrophil motility in a 3D matrix, and embryogenesis in Caenorhabditis elegans and Drosophila melanogaster. The results provide a visceral reminder of the beauty and the complexity of living systems.


Subject(s)
Caenorhabditis elegans/embryology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/ultrastructure , Imaging, Three-Dimensional/methods , Microscopy/methods , Molecular Imaging/methods , Animals , Cell Communication , Embryonic Stem Cells/ultrastructure , Mice , Spheroids, Cellular/ultrastructure
11.
Dev Cell ; 27(2): 145-160, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24120883

ABSTRACT

Mechanisms controlling microtubule dynamics at the cell cortex play a crucial role in cell morphogenesis and neuronal development. Here, we identified kinesin-4 KIF21A as an inhibitor of microtubule growth at the cell cortex. In vitro, KIF21A suppresses microtubule growth and inhibits catastrophes. In cells, KIF21A restricts microtubule growth and participates in organizing microtubule arrays at the cell edge. KIF21A is recruited to the cortex by KANK1, which coclusters with liprin-α1/ß1 and the components of the LL5ß-containing cortical microtubule attachment complexes. Mutations in KIF21A have been linked to congenital fibrosis of the extraocular muscles type 1 (CFEOM1), a dominant disorder associated with neurodevelopmental defects. CFEOM1-associated mutations relieve autoinhibition of the KIF21A motor, and this results in enhanced KIF21A accumulation in axonal growth cones, aberrant axon morphology, and reduced responsiveness to inhibitory cues. Our study provides mechanistic insight into cortical microtubule regulation and suggests that altered microtubule dynamics contribute to CFEOM1 pathogenesis.


Subject(s)
Eye Diseases, Hereditary/metabolism , Fibrosis/metabolism , Kinesins/metabolism , Microtubules/metabolism , Neurons/metabolism , Ocular Motility Disorders/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , COS Cells , Carrier Proteins/metabolism , Cell Line , Chlorocebus aethiops , Cytoskeletal Proteins , Eye Diseases, Hereditary/genetics , Growth Inhibitors , HEK293 Cells , HeLa Cells , Humans , Kinesins/genetics , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Ophthalmoplegia , RNA Interference , RNA, Small Interfering , Tumor Suppressor Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 110(9): 3399-404, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401517

ABSTRACT

A recent key requirement in life sciences is the observation of biological processes in their natural in vivo context. However, imaging techniques that allow fast imaging with higher resolution in 3D thick specimens are still limited. Spinning disk confocal microscopy using a Yokogawa Confocal Scanner Unit, which offers high-speed multipoint confocal live imaging, has been found to have wide utility among cell biologists. A conventional Confocal Scanner Unit configuration, however, is not optimized for thick specimens, for which the background noise attributed to "pinhole cross-talk," which is unintended pinhole transmission of out-of-focus light, limits overall performance in focal discrimination and reduces confocal capability. Here, we improve spinning disk confocal microscopy by eliminating pinhole cross-talk. First, the amount of pinhole cross-talk is reduced by increasing the interpinhole distance. Second, the generation of out-of-focus light is prevented by two-photon excitation that achieves selective-plane illumination. We evaluate the effect of these modifications and test the applicability to the live imaging of green fluorescent protein-expressing model animals. As demonstrated by visualizing the fine details of the 3D cell shape and submicron-size cytoskeletal structures inside animals, these strategies dramatically improve higher-resolution intravital imaging.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Cell Survival , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Mammalian/cytology , Embryo, Nonmammalian/cytology , Green Fluorescent Proteins , Mice , Photons , Recombinant Fusion Proteins/metabolism
13.
PLoS One ; 7(12): e51442, 2012.
Article in English | MEDLINE | ID: mdl-23251535

ABSTRACT

Recently, the EB1 and XMAP215/TOG families of microtubule binding proteins have been demonstrated to bind autonomously to the growing plus ends of microtubules and regulate their behaviour in in vitro systems. However, their functional redundancy or difference in cells remains obscure. Here, we compared the nanoscale distributions of EB1 and ch-TOG along microtubules using high-resolution microscopy techniques, and also their roles in microtubule organisation in interphase HeLa cells. The ch-TOG accumulation sites protruded ∼100 nm from the EB1 comets. Overexpression experiments showed that ch-TOG and EB1 did not interfere with each other's localisation, confirming that they recognise distinct regions at the ends of microtubules. While both EB1 and ch-TOG showed similar effects on microtubule plus end dynamics and additively increased microtubule dynamicity, only EB1 exhibited microtubule-cell cortex attachment activity. These observations indicate that EB1 and ch-TOG regulate microtubule organisation differently via distinct regions in the plus ends of microtubules.


Subject(s)
Interphase , Microtubule-Associated Proteins/metabolism , Nanoparticles/chemistry , Antibodies/metabolism , Fluorescent Dyes/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Microtubules/metabolism , Protein Transport , Staining and Labeling , Tubulin/metabolism
14.
Development ; 139(19): 3644-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22949618

ABSTRACT

In zebrafish, as in many animals, maternal dorsal determinants are vegetally localized in the egg and are transported after fertilization in a microtubule-dependent manner. However, the organization of early microtubules, their dynamics and their contribution to axis formation are not fully understood. Using live imaging, we identified two populations of microtubules, perpendicular bundles and parallel arrays, which are directionally oriented and detected exclusively at the vegetal cortex before the first cell division. Perpendicular bundles emanate from the vegetal cortex, extend towards the blastoderm, and orient along the animal-vegetal axis. Parallel arrays become asymmetric on the vegetal cortex, and orient towards dorsal. We show that the orientation of microtubules at 20 minutes post-fertilization can predict where the embryonic dorsal structures in zebrafish will form. Furthermore, we find that parallel microtubule arrays colocalize with wnt8a RNA, the candidate maternal dorsal factor. Vegetal cytoplasmic granules are displaced with parallel arrays by ~20°, providing in vivo evidence of a cortical rotation-like process in zebrafish. Cortical displacement requires parallel microtubule arrays, and probably contributes to asymmetric transport of maternal determinants. Formation of parallel arrays depends on Ca(2+) signaling. Thus, microtubule polarity and organization predicts the zebrafish embryonic axis. In addition, our results suggest that cortical rotation-like processes might be more common in early development than previously thought.


Subject(s)
Body Patterning , Cerebral Cortex/embryology , Microtubules/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Blastoderm/embryology , Blastoderm/metabolism , Body Patterning/genetics , Calcium Signaling/physiology , Cerebral Cortex/ultrastructure , Embryo, Nonmammalian , Female , Fertilization/physiology , Forecasting , Green Fluorescent Proteins/genetics , Male , Sperm-Ovum Interactions/physiology , Xenopus , Zebrafish/genetics
15.
Cytoskeleton (Hoboken) ; 68(11): 603-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22021191

ABSTRACT

Microtubules serve as rails for intracellular trafficking and their appropriate organization is critical for the generation of cell polarity, which is a foundation of cell differentiation, tissue morphogenesis, ontogenesis and the maintenance of homeostasis. The microtubule array is not just a static railway network; it undergoes repeated collapse and reassembly in diverse patterns during cell morphogenesis. In the last decade much progress has been made toward understanding the molecular mechanisms governing complex microtubule patterning. This review first revisits the basic principle of microtubule dynamics, and then provides an overview of how microtubules are arranged in highly shaped and functional patterns in cells changing their morphology by factors controlling the fate of microtubule ends.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Cell Polarity/physiology , Humans , Morphogenesis , Protein Transport
16.
EMBO J ; 30(9): 1690-704, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21441895

ABSTRACT

A hallmark of neurogenesis in the vertebrate brain is the apical-basal nuclear oscillation in polarized neural progenitor cells. Known as interkinetic nuclear migration (INM), these movements are synchronized with the cell cycle such that nuclei move basally during G1-phase and apically during G2-phase. However, it is unknown how the direction of movement and the cell cycle are tightly coupled. Here, we show that INM proceeds through the cell cycle-dependent linkage of cell-autonomous and non-autonomous mechanisms. During S to G2 progression, the microtubule-associated protein Tpx2 redistributes from the nucleus to the apical process, and promotes nuclear migration during G2-phase by altering microtubule organization. Thus, Tpx2 links cell-cycle progression and autonomous apical nuclear migration. In contrast, in vivo observations of implanted microbeads, acute S-phase arrest of surrounding cells and computational modelling suggest that the basal migration of G1-phase nuclei depends on a displacement effect by G2-phase nuclei migrating apically. Our model for INM explains how the dynamics of neural progenitors harmonize their extensive proliferation with the epithelial architecture in the developing brain.


Subject(s)
Brain/embryology , Cell Cycle/physiology , Cell Nucleus/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/physiology , Models, Molecular , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Cell Polarity/physiology , Cell Proliferation , DNA Primers/genetics , Gene Knockdown Techniques , Mice , Microscopy, Fluorescence , Microspheres , Protein Transport/physiology , RNA Interference , Time-Lapse Imaging
17.
Biochemistry ; 49(25): 5143-53, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20509626

ABSTRACT

Elucidation of the basis of interactions between biological molecules is essential for the understanding of living systems. Src-homology 3 (SH3) domains play critical roles in interaction networks of proteins by recognizing a proline-rich sequence motif, PxxP. There are, however, several SH3 domains that specifically bind to polypeptide chains without the conventional recognition sequence. The SH3 domain of DDEF1 associates with the SAMP motifs of the adenomatous polyposis coli (APC) tumor suppressor. The SAMP motifs are indispensable for the normal function of APC in tumor suppression. Here we present the structural basis of the interaction between the DDEF1-SH3 domain and the APC-SAMP motifs. We determined the solution structures of the DDEF1-SH3 domain both in a free state and in a complex with APC-SAMP. As the affinity of the interaction was not sufficiently high for the determination of the complex structure in solution by conventional methods, we utilized a fusion protein of the DDEF1-SH3 domain and APC-SAMP. The structures revealed that the SAMP motif adopts a class II polyproline type II helix even though it does not contain the PxxP motif and that a characteristically large hydrophobic pocket of the SH3 domain confers high selectivity to the interaction. Furthermore, investigation into the backbone dynamics of the free and bound systems by NMR spin relaxation experiments demonstrated that the DDEF1-SH3 domain exhibits high flexibility at the peptide recognition site in the absence of the ligand and that most residues of the APC-SAMP motif display extensive local motions even in the stable complex.


Subject(s)
Amino Acid Motifs , Genes, APC , src Homology Domains , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Sequence Homology, Amino Acid
18.
J Cell Biol ; 189(5): 901-17, 2010 May 31.
Article in English | MEDLINE | ID: mdl-20513769

ABSTRACT

LL5beta has been identified as a microtubule-anchoring factor that attaches EB1/CLIP-associating protein (CLASP)-bound microtubule plus ends to the cell cortex. In this study, we show that LL5beta and its homologue LL5alpha (LL5s) colocalize with autocrine laminin-5 and its receptors, integrins alpha3beta1 and alpha6beta4, at the basal side of fully polarized epithelial sheets. Depletion of both laminin receptor integrins abolishes the cortical localization of LL5s, whereas LL5 depletion reduces the amount of integrin alpha3 at the basal cell cortex. Activation of integrin alpha3 is sufficient to initiate LL5 accumulation at the cell cortex. LL5s form a complex with the cytoplasmic tails of these integrins, but their interaction might be indirect. Analysis of the three-dimensional distribution of microtubule growth by visualizing EB1-GFP in epithelial sheets in combination with RNA interference reveals that LL5s are required to maintain the density of growing microtubules selectively at the basal cortex. These findings reveal that signaling from laminin-integrin associations attaches microtubule plus ends to the epithelial basal cell cortex.


Subject(s)
Carrier Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Laminin/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Adhesion , Cell Membrane/metabolism , Cell Polarity/physiology , Female , Humans , Integrin alpha3/genetics , Integrin alpha3/metabolism , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Laminin/genetics , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Nerve Tissue Proteins/genetics , Protein Binding/physiology , RNA, Small Interfering/genetics , Receptors, Laminin/genetics , Receptors, Laminin/metabolism
19.
J Biol Chem ; 285(21): 16145-54, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20308060

ABSTRACT

We have recently shown that beta-catenin-facilitated export of cadherins from the endoplasmic reticulum requires PX-RICS, a beta-catenin-interacting GTPase-activating protein for Cdc42. Here we show that PX-RICS interacts with isoforms of 14-3-3 and couples the N-cadherin-beta-catenin complex to the microtubule-based molecular motor dynein-dynactin. Similar to knockdown of PX-RICS, knockdown of either 14-3-3zeta or - resulted in the disappearance of N-cadherin and beta-catenin from the cell-cell boundaries. Furthermore, we found that PX-RICS and 14-3-3zeta/ are present in a large multiprotein complex that contains dynein-dynactin components as well as N-cadherin and beta-catenin. Both RNAi- and dynamitin-mediated inhibition of dynein-dynactin function also led to the absence of N-cadherin and beta-catenin at the cell-cell contact sites. Our results suggest that the PX-RICS-14-3-3zeta/ complex links the N-cadherin-beta-catenin cargo with the dynein-dynactin motor and thereby mediates its endoplasmic reticulum export.


Subject(s)
14-3-3 Proteins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Dyneins/metabolism , Endoplasmic Reticulum/metabolism , GTPase-Activating Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , beta Catenin/metabolism , 14-3-3 Proteins/genetics , Animals , Antigens, CD/genetics , COS Cells , Cadherins/genetics , Chlorocebus aethiops , Dynactin Complex , Dyneins/genetics , Endoplasmic Reticulum/genetics , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Multiprotein Complexes/genetics , Protein Transport/physiology , beta Catenin/genetics , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
20.
Nat Cell Biol ; 11(9): 1057-68, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668197

ABSTRACT

Orchestrated remodelling of the cytoskeketon is prominent during neurite extension. In contrast with the extensive characterization of actin filament regulation, little is known about the dynamics of microtubules during neurite extension. Here we identify an atypical protein kinase C (aPKC)-Aurora A-NDEL1 pathway that is crucial for the regulation of microtubule organization during neurite extension. aPKC phosphorylates Aurora A at Thr 287 (T287), which augments interaction with TPX2 and facilitates activation of Aurora A at the neurite hillock, followed by phosphorylation of NDEL1 at S251 and recruitment. Suppression of aPKC, Aurora A or TPX2, or disruption of Ndel1, results in severe impairment of neurite extension. Analysis of microtubule dynamics with a microtubule plus-end marker revealed that suppression of the aPKC-Aurora A-NDEL1 pathway resulted in a significant decrease in the frequency of microtubule emanation from the microtubule organizing centre (MTOC), suggesting that Aurora A acts downstream of aPKC. These findings demonstrate a surprising role of aPKC-Aurora A-NDEL1 pathway in microtubule remodelling during neurite extension.


Subject(s)
Carrier Proteins/metabolism , Microtubules/enzymology , Neurites/enzymology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/immunology , Aurora Kinase A , Aurora Kinases , Enzyme Activation , Ganglia, Spinal/enzymology , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/enzymology , Mitosis , Molecular Mimicry , Mutation/genetics , Phosphorylation , Phosphothreonine/metabolism , Protein Binding , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL