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










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(3): e2212507120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36626558

ABSTRACT

Intracellular cargos are often membrane-enclosed and transported by microtubule-based motors in the presence of microtubule-associated proteins (MAPs). Whereas increasing evidence reveals how MAPs impact the interactions between motors and microtubules, critical questions remain about the impact of the cargo membrane on transport. Here we combined in vitro optical trapping with theoretical approaches to determine the effect of a lipid cargo membrane on kinesin-based transport in the presence of MAP tau. Our results demonstrate that attaching kinesin to a fluid lipid membrane reduces the inhibitory effect of tau on kinesin. Moreover, adding cholesterol, which reduces kinesin diffusion in the cargo membrane, amplifies the inhibitory effect of tau on kinesin binding in a dosage-dependent manner. We propose that reduction of kinesin diffusion in the cargo membrane underlies the effect of cholesterol on kinesin binding in the presence of tau, and we provide a simple model for this proposed mechanism. Our study establishes a direct link between cargo membrane cholesterol and MAP-based regulation of kinesin-1. The cholesterol effects uncovered here may more broadly extend to other lipid alterations that impact motor diffusion in the cargo membrane, including those associated with aging and neurological diseases.


Subject(s)
Kinesins , Microtubule-Associated Proteins , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Biological Transport/physiology , Lipids
2.
Viruses ; 13(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34696376

ABSTRACT

Immature HIV virions harbor a lattice of Gag molecules with significant ordering in CA-NTD, CA-CTD and SP1 regions. This ordering plays a major role during HIV maturation. To test the condition in which the Gag lattice forms in vivo, we assembled virus like particles (VLPs) by expressing only HIV Gag in mammalian cells. Here we show that these VLPs incorporate a similar number of Gag molecules compared to immature HIV virions. However, within these VLPs, Gag molecules diffuse with a pseudo-diffusion rate of 10 nm2/s, this pseudo-diffusion is abrogated in the presence of melittin and is sensitive to mutations within the SP1 region. Using cryotomography, we show that unlike immature HIV virions, in the Gag lattice of VLPs the CA-CTD and SP1 regions are significantly less ordered. Our observations suggest that within immature HIV virions, other viral factors in addition to Gag, contribute to ordering in the CA-CTD and SP1 regions.


Subject(s)
HIV-1/chemistry , Virion/chemistry , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/metabolism , HIV Infections/virology , HIV-1/genetics , Humans , Models, Molecular , Mutation , Virion/genetics , Virus Assembly , gag Gene Products, Human Immunodeficiency Virus/genetics
4.
Sci Rep ; 10(1): 21877, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318562

ABSTRACT

SARS-CoV-2 virus is the causative agent of COVID-19. Here we demonstrate that non-infectious SARS-CoV-2 virus like particles (VLPs) can be assembled by co-expressing the viral proteins S, M and E in mammalian cells. The assembled SARS-CoV-2 VLPs possess S protein spikes on particle exterior, making them ideal for vaccine development. The particles range in shape from spherical to elongated with a characteristic size of 129 ± 32 nm. We further show that SARS-CoV-2 VLPs dried in ambient conditions can retain their structural integrity upon repeated scans with Atomic Force Microscopy up to a peak force of 1 nN.


Subject(s)
COVID-19/virology , SARS-CoV-2/physiology , Virion/metabolism , Virus Assembly , HEK293 Cells , Humans , Spike Glycoprotein, Coronavirus/metabolism , Viral Matrix Proteins/metabolism
5.
Biochem Biophys Res Commun ; 529(2): 303-305, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703427

ABSTRACT

Microtubules often form sparse networks in eukaryotic cells which simultaneously contribute to shape maintenance and help establish overall cell layout. It is therefore important to quantify not only how these filaments function individually but also as a coupled network. We have developed a straightforward approach to assemble such networks de novo and we now use it to measure microtubule rigidity within small networks under controlled conditions. Our results suggest that microtubule rigidity increases with the contour length of the filament both for single microtubules and within small microtubule networks.


Subject(s)
Cytoskeleton/chemistry , Microtubules/chemistry , Animals , Biomechanical Phenomena , Elastic Modulus , Swine
6.
Nat Struct Mol Biol ; 27(4): 392-399, 2020 04.
Article in English | MEDLINE | ID: mdl-32251413

ABSTRACT

The endosomal sorting complexes required for transport (ESCRTs) mediate diverse membrane remodeling events. These typically require ESCRT-III proteins to stabilize negatively curved membranes; however, recent work has indicated that certain ESCRT-IIIs also participate in positive-curvature membrane-shaping reactions. ESCRT-IIIs polymerize into membrane-binding filaments, but the structural basis for negative versus positive membrane remodeling by these proteins remains poorly understood. To learn how certain ESCRT-IIIs shape positively curved membranes, we determined structures of human membrane-bound CHMP1B-only, membrane-bound CHMP1B + IST1, and IST1-only filaments by cryo-EM. Our structures show how CHMP1B first polymerizes into a single-stranded helical filament, shaping membranes into moderate-curvature tubules. Subsequently, IST1 assembles a second strand on CHMP1B, further constricting the membrane tube and reducing its diameter nearly to the fission point. Each step of constriction thins the underlying bilayer, lowering the barrier to membrane fission. Our structures reveal how a two-component, sequential polymerization mechanism drives membrane tubulation, constriction and bilayer thinning.


Subject(s)
Cell Membrane/ultrastructure , Endosomal Sorting Complexes Required for Transport/ultrastructure , Oncogene Proteins/ultrastructure , Cell Membrane/chemistry , Cell Membrane/genetics , Cytokinesis/genetics , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/chemistry , Endosomes/genetics , Endosomes/ultrastructure , Humans , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Polymerization , Protein Conformation
7.
PLoS Pathog ; 16(1): e1007985, 2020 01.
Article in English | MEDLINE | ID: mdl-31995633

ABSTRACT

Axonal sorting, the controlled passage of specific cargoes from the cell soma into the axon compartment, is critical for establishing and maintaining the polarity of mature neurons. To delineate axonal sorting events, we took advantage of two neuroinvasive alpha-herpesviruses. Human herpes simplex virus 1 (HSV-1) and pseudorabies virus of swine (PRV; suid herpesvirus 1) have evolved as robust cargo of axonal sorting and transport mechanisms. For efficient axonal sorting and subsequent egress from axons and presynaptic termini, progeny capsids depend on three viral membrane proteins (Us7 (gI), Us8 (gE), and Us9), which engage axon-directed kinesin motors. We present evidence that Us7-9 of the veterinary pathogen pseudorabies virus (PRV) form a tripartite complex to recruit Kif1a, a kinesin-3 motor. Based on multi-channel super-resolution and live TIRF microscopy, complex formation and motor recruitment occurs at the trans-Golgi network. Subsequently, progeny virus particles enter axons as enveloped capsids in a transport vesicle. Artificial recruitment of Kif1a using a drug-inducible heterodimerization system was sufficient to rescue axonal sorting and anterograde spread of PRV mutants devoid of Us7-9. Importantly, biophysical evidence suggests that Us9 is able to increase the velocity of Kif1a, a previously undescribed phenomenon. In addition to elucidating mechanisms governing axonal sorting, our results provide further insight into the composition of neuronal transport systems used by alpha-herpesviruses, which will be critical for both inhibiting the spread of infection and the safety of herpesvirus-based oncolytic therapies.


Subject(s)
Axons/virology , Capsid/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Suid/metabolism , Kinesins/metabolism , Pseudorabies/metabolism , Animals , Axonal Transport , Axons/metabolism , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Suid/genetics , Host-Pathogen Interactions , Humans , Kinesins/genetics , Protein Binding , Pseudorabies/genetics , Pseudorabies/virology , Swine , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , trans-Golgi Network/metabolism , trans-Golgi Network/virology
8.
Nat Cell Biol ; 21(9): 1078-1085, 2019 09.
Article in English | MEDLINE | ID: mdl-31481790

ABSTRACT

Tau is an abundant microtubule-associated protein in neurons. Tau aggregation into insoluble fibrils is a hallmark of Alzheimer's disease and other types of dementia1, yet the physiological state of tau molecules within cells remains unclear. Using single-molecule imaging, we directly observe that the microtubule lattice regulates reversible tau self-association, leading to localized, dynamic condensation of tau molecules on the microtubule surface. Tau condensates form selectively permissible barriers, spatially regulating the activity of microtubule-severing enzymes and the movement of molecular motors through their boundaries. We propose that reversible self-association of tau molecules, gated by the microtubule lattice, is an important mechanism of the biological functions of tau, and that oligomerization of tau is a common property shared between the physiological and disease-associated forms of the molecule.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spastin/metabolism , tau Proteins/metabolism , Animals , Mice , Neuroimaging/methods , Neurons/metabolism , Swine
9.
Proc Natl Acad Sci U S A ; 115(3): 537-542, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29295928

ABSTRACT

The eukaryotic cell's microtubule cytoskeleton is a complex 3D filament network. Microtubules cross at a wide variety of separation distances and angles. Prior studies in vivo and in vitro suggest that cargo transport is affected by intersection geometry. However, geometric complexity is not yet widely appreciated as a regulatory factor in its own right, and mechanisms that underlie this mode of regulation are not well understood. We have used our recently reported 3D microtubule manipulation system to build filament crossings de novo in a purified in vitro environment and used them to assay kinesin-1-driven model cargo navigation. We found that 3D microtubule network geometry indeed significantly influences cargo routing, and in particular that it is possible to bias a cargo to pass or switch just by changing either filament spacing or angle. Furthermore, we captured our experimental results in a model which accounts for full 3D geometry, stochastic motion of the cargo and associated motors, as well as motor force production and force-dependent behavior. We used a combination of experimental and theoretical analysis to establish the detailed mechanisms underlying cargo navigation at microtubule crossings.


Subject(s)
Microtubules/chemistry , Microtubules/metabolism , Biological Transport , Cytoskeleton/metabolism , Humans , Imaging, Three-Dimensional , Kinesins/chemistry , Kinesins/metabolism , Kinetics , Models, Biological , Models, Theoretical , Protein Binding
10.
Traffic ; 18(10): 658-671, 2017 10.
Article in English | MEDLINE | ID: mdl-28731566

ABSTRACT

The kinesin family proteins are often studied as prototypical molecular motors; a deeper understanding of them can illuminate regulation of intracellular transport. It is typically assumed that they function identically. Here we find that this assumption of homogeneous function appears incorrect: variation among motors' velocities in vivo and in vitro is larger than the stochastic variation expected for an ensemble of "identical" motors. When moving on microtubules, slow and fast motors are persistently slow, and fast, respectively. We develop theory that provides quantitative criteria to determine whether the observed single-molecule variation is too large to be generated from an ensemble of identical molecules. To analyze such heterogeneity, we group traces into homogeneous sub-ensembles. Motility studies varying the temperature, pH and glycerol concentration suggest at least 2 distinct functional states that are independently affected by external conditions. We end by investigating the functional ramifications of such heterogeneity through Monte-Carlo multi-motor simulations.


Subject(s)
Drosophila Proteins/metabolism , Kinesins/metabolism , Molecular Dynamics Simulation , Animals , Cell Line, Tumor , Drosophila , Drosophila Proteins/chemistry , Humans , Kinesins/chemistry , Motion , Protein Domains
11.
J Biol Chem ; 292(29): 12245-12255, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28576829

ABSTRACT

Cytoplasmic dynein is the primary minus-end-directed microtubule motor protein in animal cells, performing a wide range of motile activities, including transport of vesicular cargos, mRNAs, viruses, and proteins. Lissencephaly-1 (LIS1) is a highly conserved dynein-regulatory factor that binds directly to the dynein motor domain, uncoupling the enzymatic and mechanical cycles of the motor and stalling dynein on the microtubule track. Dynactin, another ubiquitous dynein-regulatory factor, releases dynein from an autoinhibited state, leading to a dramatic increase in fast, processive dynein motility. How these opposing activities are integrated to control dynein motility is unknown. Here, we used fluorescence single-molecule microscopy to study the interaction of LIS1 with the processive dynein-dynactin-BicD2N (DDB) complex. Surprisingly, in contrast to the prevailing model for LIS1 function established in the context of dynein alone, we found that binding of LIS1 to DDB does not strongly disrupt processive motility. Motile DDB complexes bound up to two LIS1 dimers, and mutational analysis suggested that LIS1 binds directly to the dynein motor domains during DDB movement. Interestingly, LIS1 enhanced DDB velocity in a concentration-dependent manner, in contrast to observations of the effect of LIS1 on the motility of isolated dynein. Thus, LIS1 exerts concentration-dependent effects on dynein motility and can synergize with dynactin to enhance processive dynein movement. Our results suggest that the effect of LIS1 on dynein motility depends on both LIS1 concentration and the presence of other regulatory factors such as dynactin and may provide new insights into the mechanism of LIS1 haploinsufficiency in the neurodevelopmental disorder lissencephaly.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Dynactin Complex/metabolism , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Dimerization , Dyneins/chemistry , Humans , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/enzymology , Mutagenesis, Site-Directed , Mutation , Nerve Tissue Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Rats , Recombinant Fusion Proteins/metabolism , Sf9 Cells , Spodoptera , Sus scrofa , Tubulin/metabolism
13.
Biophys J ; 111(6): 1287-1294, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27653487

ABSTRACT

Cytoplasmic dynein and kinesin are both microtubule-based molecular motors but are structurally and evolutionarily unrelated. Under standard conditions, both move with comparable unloaded velocities toward either the microtubule minus (dynein) or plus (most kinesins) end. This similarity is important because it is often implicitly incorporated into models that examine the balance of cargo fluxes in cells and into models of the bidirectional motility of individual cargos. We examined whether this similarity is a robust feature, and specifically whether it persists across the biologically relevant temperature range. The velocity of mammalian cytoplasmic dynein, but not of mammalian kinesin-1, exhibited a break from simple Arrhenius behavior below 15°C-just above the restrictive temperature of mammalian fast axonal transport. In contrast, the velocity of yeast cytoplasmic dynein showed a break from Arrhenius behavior at a lower temperature (∼8°C). Our studies implicate cytoplasmic dynein as a more thermally tunable motor and therefore a potential thermal regulator of microtubule-based transport. Our theoretical analysis further suggests that motor velocity changes can lead to qualitative changes in individual cargo motion and hence net intracellular cargo fluxes. We propose that temperature can potentially be used as a noninvasive probe of intracellular transport.


Subject(s)
Biological Transport , Cytoplasmic Dyneins/chemistry , Kinesins/chemistry , Temperature , Animals , Biological Transport/physiology , Computer Simulation , Cytoplasmic Dyneins/metabolism , Kinesins/metabolism , Models, Molecular , Optical Tweezers , Rats , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Stochastic Processes
14.
J Virol ; 90(21): 9997-10006, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27581973

ABSTRACT

Alphaherpesviruses such as herpes simplex virus and pseudorabies virus (PRV) are neuroinvasive double-stranded DNA (dsDNA) viruses that establish lifelong latency in peripheral nervous system (PNS) neurons of their native hosts. Following reactivation, infection can spread back to the initial mucosal site of infection or, in rare cases, to the central nervous system, with usually serious outcomes. During entry and egress, viral capsids depend on microtubule-based molecular motors for efficient and fast transport. In axons of PNS neurons, cytoplasmic dynein provides force for retrograde movements toward the soma, and kinesins move cargo in the opposite, anterograde direction. The dynamic properties of virus particles in cells can be imaged by fluorescent protein fusions to the small capsid protein VP26, which are incorporated into capsids. However, single-color fluorescent protein tags fail to distinguish the virus inoculum from progeny. Therefore, we established a dual-color system by growing a recombinant PRV expressing a red fluorescent VP26 fusion (PRV180) on a stable cell line expressing a green VP26 fusion (PK15-mNG-VP26). The resulting dual-color virus preparation (PRV180G) contains capsids tagged with both red and green fluorescent proteins, and 97% of particles contain detectable levels of mNeonGreen (mNG)-tagged VP26. After replication in neuronal cells, all PRV180G progeny exclusively contain monomeric red fluorescent protein (mRFP)-VP26-tagged capsids. We used PRV180G for an analysis of axonal capsid transport dynamics in PNS neurons. Fast dual-color total internal reflection fluorescence (TIRF) microscopy, single-particle tracking, and motility analyses reveal robust, bidirectional capsid motility mediated by cytoplasmic dynein and kinesin during entry, whereas egressing progeny particles are transported exclusively by kinesins. IMPORTANCE Alphaherpesviruses are neuroinvasive viruses that infect the peripheral nervous system (PNS) of infected hosts as an integral part of their life cycle. Establishment of a quiescent or latent infection in PNS neurons is a hallmark of most alphaherpesviruses. Spread of infection to the central nervous system is surprisingly rare in natural hosts but can be fatal. Pseudorabies virus (PRV) is a broad-host-range swine alphaherpesvirus that enters neuronal cells and utilizes intracellular transport processes to establish infection and to spread between cells. By using a virus preparation with fluorescent viral capsids that change color depending on the stage of the infectious cycle, we find that during entry, axons of PNS neurons support robust, bidirectional capsid motility, similar to cellular cargo, toward the cell body. In contrast, progeny particles appear to be transported unidirectionally by kinesin motors toward distal egress sites.

15.
Biophys J ; 108(6): 1480-1483, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25809260

ABSTRACT

Intracellular transport of cargos along microtubules is often complicated by the topology of the underlying filament network. The fundamental building blocks for this complex arrangement are filament intersections. The navigation of cargos across microtubule intersections remains poorly understood. Here, we demonstrate that kinesin-driven cargos are engaged in a tug-of-war at microtubule intersections. Tug-of-war events result in long pauses that can last from a few seconds to several minutes. We demonstrate that the extent of the tug-of-war and the duration of pauses change with the number of motors on the cargo and can be regulated by ionic strength. We also show that dwell times at intersections depend on the angle between crossing microtubules. Our data suggest that local microtubule geometry can regulate microtubule-based transport.


Subject(s)
Biological Transport , Kinesins/metabolism , Microtubules/metabolism , Animals , Escherichia coli , Kinetics , Melanophores/physiology , Video Recording , Xenopus
16.
Proc Natl Acad Sci U S A ; 110(47): 18880-5, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24191039

ABSTRACT

Two merotriterpenoid hydroquinone sulfates designated adociasulfate-13 (1) and adociasulfate-14 (2) were purified from Cladocroce aculeata (Chalinidae) along with adociasulfate-8. All three compounds were found to inhibit microtubule-stimulated ATPase activity of kinesin at 15 µM by blocking both the binding of microtubules and the processive motion of kinesin along microtubules. These findings directly show that substitution of the 5'-sulfate in 1 for a glycolic acid moiety in 2 maintains kinesin inhibition. Nomarski imaging and bead diffusion assays in the presence of adociasulfates showed no signs of either free-floating or bead-bound adociasulfate aggregates. Single-molecule biophysical experiments also suggest that inhibition of kinesin activity does not involve adociasulfate aggregation. Furthermore, both mitotic and nonmitotic kinesins are inhibited by adociasulfates to a significantly different extent. We also report evidence that microtubule binding of nonkinesin microtubule binding domains may be affected by adociasulfates.


Subject(s)
Drug Discovery/trends , Hydroquinones/pharmacology , Kinesins/antagonists & inhibitors , Porifera/chemistry , Sulfuric Acid Esters/pharmacology , Triterpenes/pharmacology , Animals , Biophysics , Cell Membrane Permeability/physiology , Drug Discovery/methods , Humans , Hydroquinones/metabolism , Molecular Structure , Protein Binding , Spectrophotometry , Sulfuric Acid Esters/metabolism , Triterpenes/metabolism
17.
J Vis Exp ; (74)2013 Apr 22.
Article in English | MEDLINE | ID: mdl-23629580

ABSTRACT

High resolution microscope systems with optical traps allow for precise manipulation of various refractive objects, such as dielectric beads (1) or cellular organelles (2,3), as well as for high spatial and temporal resolution readout of their position relative to the center of the trap. The system described herein has one such "traditional" trap operating at 980 nm. It additionally provides a second optical trapping system that uses a commercially available holographic package to simultaneously create and manipulate complex trapping patterns in the field of view of the microscope (4,5) at a wavelength of 1,064 nm. The combination of the two systems allows for the manipulation of multiple refractive objects at the same time while simultaneously conducting high speed and high resolution measurements of motion and force production at nanometer and piconewton scale.


Subject(s)
Holography/instrumentation , Microscopy/instrumentation , Optics and Photonics/instrumentation , Holography/methods , Microscopy/methods , Optics and Photonics/methods
18.
Proc Natl Acad Sci U S A ; 108(47): 18960-5, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22084076

ABSTRACT

Intracellular transport via the microtubule motors kinesin and dynein plays an important role in maintaining cell structure and function. Often, multiple kinesin or dynein motors move the same cargo. Their collective function depends critically on the single motors' detachment kinetics under load, which we experimentally measure here. This experimental constraint--combined with other experimentally determined parameters--is then incorporated into theoretical stochastic and mean-field models. Comparison of modeling results and in vitro data shows good agreement for the stochastic, but not mean-field, model. Many cargos in vivo move bidirectionally, frequently reversing course. Because both kinesin and dynein are present on the cargos, one popular hypothesis explaining the frequent reversals is that the opposite-polarity motors engage in unregulated stochastic tugs-of-war. Then, the cargos' motion can be explained entirely by the outcome of these opposite-motor competitions. Here, we use fully calibrated stochastic and mean-field models to test the tug-of-war hypothesis. Neither model agrees well with our in vivo data, suggesting that, in addition to inevitable tugs-of-war between opposite motors, there is an additional level of regulation not included in the models.


Subject(s)
Lipid Metabolism , Models, Biological , Molecular Motor Proteins/metabolism , Stochastic Processes , Biological Transport/physiology , Computer Simulation , Kinetics
19.
J Cell Biol ; 195(2): 193-201, 2011 Oct 17.
Article in English | MEDLINE | ID: mdl-22006948

ABSTRACT

The specific physiological roles of dynein regulatory factors remain poorly understood as a result of their functional complexity and the interdependence of dynein and kinesin motor activities. We used a novel approach to overcome these challenges, combining acute in vivo inhibition with automated high temporal and spatial resolution particle tracking. Acute dynein inhibition in nonneuronal cells caused an immediate dispersal of diverse forms of cargo, resulting from a sharp decrease in microtubule minus-end run length followed by a gradual decrease in plus-end runs. Acute LIS1 inhibition or LIS1 RNA interference had little effect on lysosomes/late endosomes but severely inhibited axonal transport of large, but not small, vesicular structures. Our acute inhibition results argue against direct mechanical activation of opposite-directed motors and offer a novel approach of potential broad utility in the study of motor protein function in vivo. Our data also reveal a specific but cell type-restricted role for LIS1 in large vesicular transport and provide the first quantitative support for a general role for LIS1 in high-load dynein functions.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Axonal Transport , Microtubule-Associated Proteins/physiology , Molecular Motor Proteins/physiology , Dyneins , Endosomes , Humans , Kinesins , Lysosomes , Microscopy , Microtubules
20.
Cell ; 141(2): 304-14, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20403325

ABSTRACT

Cytoplasmic dynein is responsible for many aspects of cellular and subcellular movement. LIS1, NudE, and NudEL are dynein interactors initially implicated in brain developmental disease but now known to be required in cell migration, nuclear, centrosomal, and microtubule transport, mitosis, and growth cone motility. Identification of a specific role for these proteins in cytoplasmic dynein motor regulation has remained elusive. We find that NudE stably recruits LIS1 to the dynein holoenzyme molecule, where LIS1 interacts with the motor domain during the prepowerstroke state of the dynein crossbridge cycle. NudE abrogates dynein force production, whereas LIS1 alone or with NudE induces a persistent-force dynein state that improves ensemble function of multiple dyneins for transport under high-load conditions. These results likely explain the requirement for LIS1 and NudE in the transport of nuclei, centrosomes, chromosomes, and the microtubule cytoskeleton as well as the particular sensitivity of migrating neurons to reduced LIS1 expression.


Subject(s)
Carrier Proteins/metabolism , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Algorithms , Animals , Cattle , Chlorocebus aethiops , Humans , Kinesins/metabolism , Lissencephaly/metabolism , Rats , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...