Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Commun Chem ; 6(1): 80, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37100870

ABSTRACT

By facilitating a water/water phase separation (w/wPS), crowded biopolymers in cells form droplets that contribute to the spatial localization of biological components and their biochemical reactions. However, their influence on mechanical processes driven by protein motors has not been well studied. Here, we show that the w/wPS droplet spontaneously entraps kinesins as well as microtubules (MTs) and generates a micrometre-scale vortex flow inside the droplet. Active droplets with a size of 10-100 µm are generated through w/wPS of dextran and polyethylene glycol mixed with MTs, molecular-engineered chimeric four-headed kinesins and ATP after mechanical mixing. MTs and kinesin rapidly created contractile network accumulated at the interface of the droplet and gradually generated vortical flow, which can drive translational motion of a droplet. Our work reveals that the interface of w/wPS contributes not only to chemical processes but also produces mechanical motion by assembling species of protein motors in a functioning manner.

2.
Small ; 18(32): e2107854, 2022 08.
Article in English | MEDLINE | ID: mdl-35815940

ABSTRACT

Cilia and flagella are beating rod-like organelles that enable the directional movement of microorganisms in fluids and fluid transport along the surface of biological organisms or inside organs. The molecular motor axonemal dynein drives their beating by interacting with microtubules. Constructing synthetic beating systems with axonemal dynein capable of mimicking ciliary beating still represents a major challenge. Here, the bottom-up engineering of a sustained beating synthoneme consisting of a pair of microtubules connected by a series of periodic arrays of approximately eight axonemal dyneins is reported. A model leads to the understanding of the motion through the cooperative, cyclic association-dissociation of the molecular motor from the microtubules. The synthoneme represents a bottom-up self-organized bio-molecular machine at the nanoscale with cilia-like properties.


Subject(s)
Axonemal Dyneins , Axoneme , Axonemal Dyneins/metabolism , Axoneme/metabolism , Cilia/metabolism , Dyneins/metabolism , Flagella/metabolism , Microtubules/metabolism
3.
Science ; 375(6585): 1159-1164, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35271337

ABSTRACT

Intracellular transport is the basis of microscale logistics within cells and is powered by biomolecular motors. Mimicking transport for in vitro applications has been widely studied; however, the inflexibility in track design and control has hindered practical applications. Here, we developed protein-based motors that move on DNA nanotubes by combining a biomolecular motor dynein and DNA binding proteins. The new motors and DNA-based nanoarchitectures enabled us to arrange the binding sites on the track, locally control the direction of movement, and achieve multiplexed cargo transport by different motors. The integration of these technologies realized microscale cargo sorters and integrators that automatically transport molecules as programmed in DNA sequences on a branched DNA nanotube. Our system should provide a versatile, controllable platform for future applications.


Subject(s)
Biological Transport , DNA-Binding Proteins/chemistry , DNA/chemistry , Dyneins/metabolism , Nanotubes , Protein Engineering , Dyneins/chemistry , Nucleic Acid Conformation , Protein Binding , Protein Domains
4.
Chemistry ; 28(30): e202200807, 2022 May 25.
Article in English | MEDLINE | ID: mdl-35332959

ABSTRACT

Adenosine triphosphate (ATP) is the energy source for various biochemical processes and biomolecular motors in living things. Development of ATP antagonists and their stimuli-controlled actions offer a novel approach to regulate biological processes. Herein, we developed azobenzene-based photoswitchable ATP antagonists for controlling the activity of motor proteins; cytoplasmic and axonemal dyneins. The new ATP antagonists showed reversible photoswitching of cytoplasmic dynein activity in an in vitro dynein-microtubule system due to the trans and cis photoisomerization of their azobenzene segment. Importantly, our ATP antagonists reversibly regulated the axonemal dynein motor activity for the force generation in a demembranated model of Chlamydomonas reinhardtii. We found that the trans and cis isomers of ATP antagonists significantly differ in their affinity to the ATP binding site.


Subject(s)
Adenosine Triphosphate , Dyneins , Adenosine Triphosphate/metabolism , Binding Sites , Cytoplasm/metabolism , Dyneins/chemistry , Dyneins/metabolism , Microtubules
5.
Sci Rep ; 12(1): 2965, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194068

ABSTRACT

Chemical sensing is vital to the survival of all organisms. Bacterial chemotaxis is conducted by multiple receptors that sense chemicals to regulate a single signalling system controlling the transition between the direction (clockwise vs. counterclockwise) of flagellar rotation. Such an integrated system seems better suited to judge chemicals as either favourable or unfavourable, but not for identification purposes though differences in their affinities to the receptors may cause difference in response strength. Here, an experimental setup was developed to monitor behaviours of multiple cells stimulated simultaneously as well as a statistical framework based on Bayesian inferences. Although responses of individual cells varied substantially, ensemble averaging of the time courses seemed characteristic to attractant species, indicating we can extract information of input chemical species from responses of the bacterium. Furthermore, two similar, but distinct, beverages elicited attractant responses of cells with profiles distinguishable with the Bayesian procedure. These results provide a basis for novel bio-inspired sensors that could be used with other cell types to sense wider ranges of chemicals.

6.
J Cell Biol ; 220(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33929515

ABSTRACT

Multiciliated cells (MCCs) in tracheas generate mucociliary clearance through coordinated ciliary beating. Apical microtubules (MTs) play a crucial role in this process by organizing the planar cell polarity (PCP)-dependent orientation of ciliary basal bodies (BBs), for which the underlying molecular basis remains elusive. Herein, we found that the deficiency of Daple, a dishevelled-associating protein, in tracheal MCCs impaired the planar polarized apical MTs without affecting the core PCP proteins, causing significant defects in the BB orientation at the cell level but not the tissue level. Using live-cell imaging and ultra-high voltage electron microscope tomography, we found that the apical MTs accumulated and were stabilized by side-by-side association with one side of the apical junctional complex, to which Daple was localized. In vitro binding and single-molecule imaging revealed that Daple directly bound to, bundled, and stabilized MTs through its dimerization. These features convey a PCP-related molecular basis for the polarization of apical MTs, which coordinate ciliary beating in tracheal MCCs.


Subject(s)
Carrier Proteins/genetics , Cilia/genetics , Mucociliary Clearance/genetics , Trachea/growth & development , Animals , Basal Bodies/metabolism , Cell Polarity/genetics , Epithelial Cells/metabolism , Mice , Mice, Knockout , Microtubules/genetics , Trachea/metabolism
7.
Int J Mol Sci ; 21(8)2020 Apr 18.
Article in English | MEDLINE | ID: mdl-32325779

ABSTRACT

In eukaryotic cilia and flagella, various types of axonemal dyneins orchestrate their distinct functions to generate oscillatory bending of axonemes. The force-generating mechanism of dyneins has recently been well elucidated, mainly in cytoplasmic dyneins, thanks to progress in single-molecule measurements, X-ray crystallography, and advanced electron microscopy. These techniques have shed light on several important questions concerning what conformational changes accompany ATP hydrolysis and whether multiple motor domains are coordinated in the movements of dynein. However, due to the lack of a proper expression system for axonemal dyneins, no atomic coordinates of the entire motor domain of axonemal dynein have been reported. Therefore, a substantial amount of knowledge on the molecular architecture of axonemal dynein has been derived from electron microscopic observations on dynein arms in axonemes or on isolated axonemal dynein molecules. This review describes our current knowledge and perspectives of the force-generating mechanism of axonemal dyneins in solo and in ensemble.


Subject(s)
Adenosine Triphosphate/metabolism , Axonemal Dyneins/chemistry , Flagella/metabolism , Microtubules/metabolism , Animals , Axonemal Dyneins/metabolism , Axonemal Dyneins/ultrastructure , Axoneme/chemistry , Axoneme/metabolism , Cilia/metabolism , Crystallography, X-Ray , Cytoplasmic Dyneins/metabolism , Flagella/ultrastructure
8.
Phys Rev E ; 101(3-1): 032607, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32289972

ABSTRACT

Emergence and collapse of coherent motions of self-propelled particles are affected more by particle motions and interactions than by their material or biological details. In the reconstructed systems of biofilaments and molecular motors, several types of collective motion including a global-order pattern emerge due to the alignment interaction. Meanwhile, earlier studies show that the alignment interaction of a binary collision of biofilaments is too weak to form the global order. The multiple collision is revealed to be important to achieve global order, but it is still unclear what kind of multifilament collision is actually involved. In this study, we demonstrate that not only alignment but also crossing of two filaments is essential to produce an effective multiple-particle interaction and the global order. We design the reconstructed system of biofilaments and molecular motors to vary a probability of the crossing of biofilaments on a collision and thus control the effect of volume exclusion. In this system, biofilaments glide along their polar strands on the turf of molecular motors and can align themselves nematically when they collide with each other. Our experiments show the counterintuitive result, in which the global order is achieved only when the crossing is allowed. When the crossing is prohibited, the cluster pattern emerges instead. We also investigate the numerical model in which we can change the strength of the volume exclusion effect and find that the global orientational order and clusters emerge with weak and strong volume exclusion effects, respectively. With those results and simple theory, we conclude that not only alignment but also finite crossing probability are necessary for the effective multiple-particles interaction forming the global order. Additionally, we describe the chiral symmetry breaking of a microtubule motion which causes a rotation of global alignment.

9.
Sci Adv ; 6(4): eaax7413, 2020 01.
Article in English | MEDLINE | ID: mdl-32010782

ABSTRACT

Kinesin is a motor protein that plays important roles in a variety of cellular functions. In vivo, multiple kinesin molecules are bound to cargo and work as a team to produce larger forces or higher speeds than a single kinesin. However, the coordination of kinesins remains poorly understood because of the experimental difficulty in controlling the number and arrangement of kinesins, which are considered to affect their coordination. Here, we report that both the number and spacing significantly influence the velocity of microtubules driven by nonprocessive kinesin-14 (Ncd), whereas neither the number nor the spacing changes the velocity in the case of highly processive kinesin-1. This result was realized by the optimum nanopatterning method of kinesins that enables immobilization of a single kinesin on a nanopillar. Our proposed method enables us to study the individual effects of the number and spacing of motors on the collective dynamics of multiple motors.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Molecular Motor Proteins/metabolism , Gold/chemistry , Humans , Kinetics , Molecular Imaging , Nanofibers/chemistry , Spectrum Analysis
10.
Biochem Biophys Res Commun ; 523(4): 1014-1019, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31973818

ABSTRACT

Dynein motor proteins usually work as a group in vesicle transport, mitosis, and ciliary/flagellar beating inside cells. Despite the obvious importance of the functions of dynein, the effect of inter-dynein interactions on collective motility remains poorly understood due to the difficulty in building large dynein ensembles with defined geometry. Here, we describe a method to build dynein ensembles to investigate the collective motility of dynein on microtubules. Using electron microscopy, we show that tens to hundreds of cytoplasmic dynein monomers were anchored along a 4- or 10-helix DNA nanotube with an average periodicity of 19 or 44 nm (a programmed periodicity of 14 or 28 nm, respectively). They drove the sliding movement of DNA nanotubes along microtubules at a velocity of 170-620 nm/s. Reducing the stiffness of DNA nanotubes made the nanotube movement discontinuous and considerably slower. Decreasing the spacing between motors simply slowed down the nanotube movement. This slowdown was independent of the number of motors involved but heavily dependent on motor-motor distance. This suggests that steric hindrance or mechanical coupling between dynein molecules was responsible for the slowdown. Furthermore, we observed cyclical buckling of DNA nanotubes on microtubules, reminiscent of ciliary/flagellar beating. These results highlight the importance of the geometric arrangement of dynein motors on their collective motility.


Subject(s)
DNA/metabolism , Dyneins/metabolism , Nanotubes/chemistry , DNA/ultrastructure , Dyneins/ultrastructure , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Nanotubes/ultrastructure , Protein Transport , Recombinant Proteins/metabolism
11.
ACS Appl Bio Mater ; 3(4): 1875-1883, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-35025310

ABSTRACT

Mechanical stress on cells has profound influences on biological processes, such as cell shape regulation, the formation of tissue patterns, and development. Recently, mechanosensing properties of the microtubule, an important cytoskeletal component, have drawn attention. In this work, we studied cargo transport by dynein, a microtubule-associated motor protein, along microtubules deformed under mechanical stress. We reveal that the microtubule deformation took place as a response to the applied stress and that the deformation of microtubules facilitated the transport of dynein-driven quantum dots. This finding will provide opportunities to explore the role of microtubules as molecular mechanotransducers in cellular processes.

12.
Nanoscale ; 11(20): 9879-9887, 2019 May 28.
Article in English | MEDLINE | ID: mdl-30888373

ABSTRACT

Motor proteins function in in vivo ensembles to achieve cargo transport, flagellum motion, and mitotic cell division. Although the cooperativity of multiple motors is indispensable for physiological function, reconstituting the arrangement of motors in vitro is challenging, so detailed analysis of the functions of motor ensembles has not yet been achieved. Here, we developed an assay platform to study the motility of microtubules driven by a defined number of kinesin motors spaced in a definite manner. Gold (Au) nano-pillar arrays were fabricated on a silicon/silicon dioxide (Si/SiO2) substrate with spacings of 100 nm to 500 nm. The thiol-polyethylene glycol (PEG)-biotin self-assembled monolayer (SAM) and silane-PEG-CH3 SAM were then selectively formed on the pillars and SiO2 surface, respectively. This allowed for both immobilization of kinesin molecules on Au nano-pillars in a precise manner and repulsion of kinesins from the SiO2 surface. Using arrayed kinesin motors, we report that motor number and spacing do not influence the motility of microtubules driven by kinesin-1 motors. This assay platform is applicable to all kinds of biotinylated motors, allows the study of the effects of motor number and spacing, and is expected to reveal novel behaviors of motor proteins.


Subject(s)
Gold/chemistry , Kinesins/chemistry , Biotin/chemistry , Immobilized Proteins/chemistry , Polyethylene Glycols/chemistry , Silicon Dioxide/chemistry , Sulfhydryl Compounds/chemistry , Surface Properties
13.
Adv Biosyst ; 3(9): e1900130, 2019 09.
Article in English | MEDLINE | ID: mdl-32648655

ABSTRACT

Multielectrode arrays (MEAs) are versatile tools that are used for chronic recording and stimulation of neural cells and tissues. Driven by the recent progress in understanding of how neuronal growth and function respond to scaffold stiffness, development of MEAs with a soft cell-to-device interface has gained importance not only for in vivo but also for in vitro applications. However, the passivation layer, which constitutes the majority of the cell-device interface, is typically prepared with stiff materials. Herein, a fabrication of an MEA device with an ultrasoft passivation layer is described, which takes advantage of inkjet printing and a polydimethylsiloxane (PDMS) gel with a stiffness comparable to that of the brain. The major challenge in using the PDMS gel is that it cannot be patterned to expose the sensing area of the electrode. This issue is resolved by printing 3D micropillars at the electrode tip. Primary cortical neurons are grown on the fabricated device, and effective stimulation of the culture confirms functional cell-device coupling. The 3D MEA device with an ultrasoft interface provides a novel platform for investigating evoked activity and drug responses of living neuronal networks cultured in a biomimetic environment for both fundamental research and pharmaceutical applications.


Subject(s)
Biosensing Techniques/instrumentation , Calcium/metabolism , Neurons/metabolism , Silicone Gels/chemistry , Animals , Biomimetic Materials/chemistry , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dimethylpolysiloxanes/chemistry , Electric Stimulation , Electrochemical Techniques , Embryo, Mammalian , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Gold/chemistry , Microelectrodes , Neurons/ultrastructure , Optical Imaging , Primary Cell Culture , Printing, Three-Dimensional/instrumentation , Rats , Rats, Sprague-Dawley
14.
Sci Rep ; 8(1): 17991, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30559483

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

15.
Sci Rep ; 8(1): 15550, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341325

ABSTRACT

Cytoskeletal organization is essential for the precise morphogenesis of cells, tissues, and organs. Cytoskeletons, bound to scaffolding proteins, regulate the apical junction complex (AJC), which is composed of tight and adherens junctions, and located at the apical side of epithelial cell sheets. Cingulin is a tight junction-associated protein that binds to both actin filaments and microtubules. However, how cingulin binds to microtubules and whether cingulin can bind to actin and microtubules simultaneously are unclear. Here we examined the mechanisms behind cingulin's cytoskeleton-binding properties. First, using total internal reflection fluorescence microscopy, we detected cingulin at microtubule cross points. We then found the interdomain interactions in cingulin molecules. Notably, we found that this interaction was regulated by AMPK-dependent phosphorylation and changed cingulin's conformation and binding properties to actin filaments and microtubules. Finally, we found that the AMPK-regulated cingulin properties regulated the barrier functions of epithelial cell sheets. We propose that the cellular metabolic state, which involves AMPK, can contribute to the organization and maintenance of epithelial tissues through cingulin's tight junction/cytoskeleton regulation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Actin Cytoskeleton/metabolism , Membrane Proteins/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Animals , Membrane Proteins/chemistry , Mice , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Protein Conformation
16.
Cell Struct Funct ; 43(1): 1-14, 2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29311430

ABSTRACT

Genomics and proteomics studies in Chlamydomonas have revealed that an axoneme is composed of 200-600 types of proteins, including uncharacterized proteins collectively named flagellar-associated proteins (FAPs). Nine FAPs contain the EF-hand motif; however, they have not yet been well characterized. To find components responsible for Chlamydomonas-specific waveform changes coupled with intracellular Ca2+ concentrations, we focused on FAP85, an EF-hand motif-containing FAP specific to Chlamydomonas and its relatives. We cloned the cDNA encoding FAP85, expressed it in Escherichia coli cells, and generated a polyclonal antibody against the expressed protein. Immunoblotting showed that FAP85 was present in every axoneme of several flagellar mutants lacking major axonemal components. Immuno-electron microscopy revealed that anti-FAP85 antibodies were found only on the inner wall of A-tubules of the doublets exposed by N-lauroylsarcosine (Sarkosyl) treatment. The zero-length cross-linker 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) applied to 0.6 M KCl-extracted axonemes generated a 75-kDa complex containing ß-tubulin and FAP85. Further characterization of FAP85 and its effects on microtubule dynamics showed that FAP85 binds to tubulin and stabilized microtubules. According to these results, we conclude that FAP85 is a novel member of microtubule-binding proteins, localizing on the inner wall of the A-tubule and stabilizing microtubules.Key words: Chlamydomonas, flagella, doublet microtubule, microtubule inner proteins.


Subject(s)
Chlamydomonas reinhardtii/metabolism , Flagella/metabolism , Microtubule Proteins/metabolism , Microtubules/metabolism , Plant Proteins/metabolism , Axoneme/chemistry , Axoneme/metabolism , Chlamydomonas reinhardtii/genetics , Cloning, Molecular , Microscopy, Immunoelectron , Microtubule Proteins/chemistry , Microtubule Proteins/genetics , Microtubules/drug effects , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Binding , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Tubulin/chemistry , Tubulin/metabolism
17.
J Biol Chem ; 292(26): 10998-11008, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28476885

ABSTRACT

Myosin Vc (myoVc) is unique among vertebrate class V myosin isoforms in that it requires teams of motors to move continuously on single actin filaments. Single molecules of myoVc cannot take multiple hand-over-hand steps from one actin-binding site to the next without dissociating, in stark contrast to the well studied myosin Va (myoVa) isoform. At low salt, single myoVc motors can, however, move processively on actin bundles, and at physiologic ionic strength, even teams of myoVc motors require actin bundles to sustain continuous motion. Here, we linked defined numbers of myoVc or myoVa molecules to DNA nanostructures as synthetic cargos. Using total internal reflectance fluorescence microscopy, we compared the stepping behavior of myoVc versus myoVa ensembles and myoVc stepping patterns on single actin filaments versus actin bundles. Run lengths of both myoVc and myoVa teams increased with motor number, but only multiple myoVc motors showed a run-length enhancement on actin bundles compared with actin filaments. By resolving the stepping behavior of individual myoVc motors with a quantum dot bound to the motor domain, we found that coupling of two myoVc motors significantly decreased the futile back and side steps that were frequently observed for single myoVc motors. Changes in the inter-motor distance between two coupled myoVc motors affected stepping dynamics, suggesting that mechanical tension coordinates the stepping behavior of two myoVc motors for efficient directional motion. Our study provides a molecular basis to explain how teams of myoVc motors are suited to transport cargos such as zymogen granules on actin bundles.


Subject(s)
Actin Cytoskeleton/chemistry , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Quantum Dots/chemistry , Secretory Vesicles/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Biological Transport, Active , Mice , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Secretory Vesicles/genetics , Secretory Vesicles/metabolism
18.
Jpn J Ophthalmol ; 61(3): 260-266, 2017 May.
Article in English | MEDLINE | ID: mdl-28154953

ABSTRACT

PURPOSE: To evaluate the effect of half-dose verteporfin photodynamic therapy (hPDT) on the physiology of the macula determined by focal macular electroretinograms (FMERGs) in eyes with chronic central serous chorioretinopathy (CSC). METHODS: Fourteen eyes of 13 patients with chronic CSC were treated with hPDT. The best-corrected visual acuity (BCVA) was measured, and optical coherence tomography (OCT) and FMERGs were performed at the baseline, and at 4 days, 1, 3, 6, and 12 months after the hPDT. RESULTS: The subreitnal fluid was resolved in 12 of the 14 eyes after the hPDT. The amplitude of the a-wave at 12 months was significantly increased by 1.28 times over that at the baseline. The amplitude of the b-wave was also increased but not significantly (P = 0.055). The implicit time of the a-wave was significantly reduced at 6 months, and that of the b-wave at 3 months. The amplitudes of the oscillatory potentials did not change significantly during the 12-month follow-up period. CONCLUSIONS: hPDT led to an improvement in the FMERGs for at least 12 months without a transient depression of the FMERGs in eyes with chronic CSC. hPDT can be used safely to treat eyes with CSC.


Subject(s)
Central Serous Chorioretinopathy/drug therapy , Electroretinography/methods , Macula Lutea/physiopathology , Photochemotherapy/methods , Porphyrins/administration & dosage , Visual Acuity , Adult , Aged , Central Serous Chorioretinopathy/diagnosis , Central Serous Chorioretinopathy/physiopathology , Dose-Response Relationship, Drug , Female , Fluorescein Angiography/methods , Follow-Up Studies , Fundus Oculi , Humans , Injections, Intravenous , Macula Lutea/pathology , Male , Middle Aged , Photosensitizing Agents/administration & dosage , Retrospective Studies , Time Factors , Tomography, Optical Coherence/methods , Treatment Outcome , Verteporfin
19.
Sci Rep ; 7: 39902, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28079116

ABSTRACT

Human mutations in KATNB1 (p80) cause severe congenital cortical malformations, which encompass the clinical features of both microcephaly and lissencephaly. Although p80 plays critical roles during brain development, the underlying mechanisms remain predominately unknown. Here, we demonstrate that p80 regulates microtubule (MT) remodeling in combination with NuMA (nuclear mitotic apparatus protein) and cytoplasmic dynein. We show that p80 shuttles between the nucleus and spindle pole in synchrony with the cell cycle. Interestingly, this striking feature is shared with NuMA. Importantly, p80 is essential for aster formation and maintenance in vitro. siRNA-mediated depletion of p80 and/or NuMA induced abnormal mitotic phenotypes in cultured mouse embryonic fibroblasts and aberrant neurogenesis and neuronal migration in the mouse embryonic brain. Importantly, these results were confirmed in p80-mutant harboring patient-derived induced pluripotent stem cells and brain organoids. Taken together, our findings provide valuable insights into the pathogenesis of severe microlissencephaly, in which p80 and NuMA delineate a common pathway for neurogenesis and neuronal migration via MT organization at the centrosome/spindle pole.


Subject(s)
Adenosine Triphosphatases/metabolism , Fibroblasts/physiology , Induced Pluripotent Stem Cells/physiology , Katanin/metabolism , Microtubules/metabolism , Nervous System Malformations/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , Adenosine Triphosphatases/genetics , Animals , Cell Cycle Proteins , Dyneins/metabolism , HeLa Cells , Humans , Katanin/genetics , Mice , Mice, Inbred Strains , Mitosis/genetics , Mutation/genetics , Nervous System Malformations/genetics , Neurogenesis/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/genetics
20.
Nat Nanotechnol ; 12(3): 233-237, 2017 03.
Article in English | MEDLINE | ID: mdl-27842063

ABSTRACT

Biomolecular motors such as myosin, kinesin and dynein are protein machines that can drive directional movement along cytoskeletal tracks and have the potential to be used as molecule-sized actuators. Although control of the velocity and directionality of biomolecular motors has been achieved, the design and construction of novel biomolecular motors remains a challenge. Here we show that naturally occurring protein building blocks from different cytoskeletal systems can be combined to create a new series of biomolecular motors. We show that the hybrid motors-combinations of a motor core derived from the microtubule-based dynein motor and non-motor actin-binding proteins-robustly drive the sliding movement of an actin filament. Furthermore, the direction of actin movement can be reversed by simply changing the geometric arrangement of these building blocks. Our synthetic strategy provides an approach to fabricating biomolecular machines that work along artificial tracks at nanoscale dimensions.


Subject(s)
Cytoskeleton/chemistry , Dyneins/chemistry , Microfilament Proteins/chemistry , Cytoskeleton/genetics , Dyneins/genetics , Humans , Microfilament Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...