KIF1A

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 1482 Experts worldwide ranked by ideXlab platform

Nobutaka Hirokawa - One of the best experts on this subject based on the ideXlab platform.

  • the molecular motor KIF1A transports the trka neurotrophin receptor and is essential for sensory neuron survival and function
    Neuron, 2016
    Co-Authors: Yosuke Tanaka, Shinsuke Niwa, Atena Farkhondeh, Ruyun Zhou, Ming Dong, Nobutaka Hirokawa, Li Wang
    Abstract:

    Summary KIF1A is a major axonal transport motor protein, but its functional significance remains elusive. Here we show that KIF1A-haploinsufficient mice developed sensory neuropathy. We found progressive loss of TrkA(+) sensory neurons in KIF1A +/− dorsal root ganglia (DRGs). Moreover, axonal transport of TrkA was significantly disrupted in KIF1A +/− neurons. Live imaging and immunoprecipitation assays revealed that KIF1A bound to TrkA-containing vesicles through the adaptor GTP-Rab3, suggesting that TrkA is a cargo of the KIF1A motor. Physiological measurements revealed a weaker capsaicin response in KIF1A +/− DRG neurons. Moreover, these neurons were hyposensitive to nerve growth factor, which could explain the reduced neuronal survival and the functional deficiency of the pain receptor TRPV1. Because phosphatidylinositol 3-kinase (PI3K) signaling significantly rescued these phenotypes and also increased KIF1A mRNA, we propose that KIF1A is essential for the survival and function of sensory neurons because of the TrkA transport and its synergistic support of the NGF/TrkA/PI3K signaling pathway.

  • Intracellular transport, molecular motors, KIFs and related diseases
    BMC Genomics, 2014
    Co-Authors: Nobutaka Hirokawa
    Abstract:

    The intracellular transport is fundamental for cellular functions, morphogenesis and survival in general including neurons composed of a very long axon and dendrites. We discovered most of the kinesin superfamily motor proteins, KIFs, 45 genes in mammals, elucidated their molecular structures and functional roles by molecular cell biology, molecular genetics, biophysics and structural biology and successfully disclosed the mechanism of intracellular transport fundamental for neuronal functions. In the axon and dendrites KIFs transport their cargos such as synaptic vesicle precursors (KIF1A/KIF1Bbeta), mitochondria (KIF1Balpha/KIF5s), plasma membrane proteins (KIF3/KIF5s), NMDA glutamate receptors (KIF17), AMPA receptors (KIF5s) and mRNA with large protein complex (KIF5s). KIFs mainly recognize and bind their cargoes through adaptor protein complex and release them via phosphorylation of KIFs or GTP hydrolyses of cargo G-protein. Furthermore, using molecular genetics we successfully uncovered that KIFs play significant roles for fundamental physiological phenomena in development and functions of nervous system and that deletion of KIFs causes certain diseases by clarifying followings: 1) KIF1A/KIF1B beta hetero mice serve as a model for neuropathy, 2) KIF3 determines left/right asymmetry by generating cilia and nodal flow in the node of early embryos, 3) KIF17 plays a fundamental role on learning and memory by not only transporting NMDA glutamate receptor in dendrites but also controlling transcription and translation of KIF17 and NMDA receptor mRNAs by enhancing phosphorylated CREB, 4) KIF1A is essential for hippocampal synaptogenesis and learning enhancement in an enriched environment, 5) KIF2A is fundamental for brain wiring by controlling unnecessary elongation of growth cones by depolymerizing microtubules, 6) KIF4 plays a crucial role in the activity-dependent survival of postmitotic neurons in brain development by regulating poly(ADP-ribose) polymerase-1 activity, 7) KIF26A is essential for enteric neuronal development by regulating GDNF-Ret signaling, 8) KIF3 suppresses tumorigenesis by transporting beta-catenin from Golgi to plasma membrane for serving as cell-cell adhesion molecules, inhibiting its accumulation in the nucleus and suppressing hyper proliferation of progenitor cells, 9)KIF5A is essential for GABAa receptor transport and KIF5A deletion causes epilepsy, 10)KIF19A is a microtubule depolymerizing KIF for ciliary length control and its deletion causes female infertility and hydrocephalus based on affected fluid flows, 11)KIF13A transports serotonin receptors to plasma membranes and its deletion causes elevated–anxiety phenotypes. Thus, KIFs play significant roles not only at cellular level, but also in brain function and development. Further, their malfunctions cause diseases such as neuropathy, epilepsy, dementia, elevated anxiety, tumor, megacolon and hydrocephalus.

  • Regulation of NMDA Receptor Transport: A KIF17–Cargo Binding/Releasing Underlies Synaptic Plasticity and Memory In Vivo
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012
    Co-Authors: Xiling Yin, Yosuke Takei, Xue Feng, Nobutaka Hirokawa
    Abstract:

    Regulation of NMDA receptor trafficking is crucial to modulate neuronal communication. Ca 2+ /calmodulin-dependent protein kinase phosphorylates the tail domain of KIF17, a member of the kinesin superfamily, to control NMDA receptor subunit 2B (GluN2B) transport by changing the KIF17–cargo interaction in vitro . However, the mechanisms of regulation of GluN2B transport in vivo and its physiological significance are unknown. We generated transgenic mice carrying wild-type KIF17 ( TgS ), or KIF17 with S1029A ( TgA ) or S1029D ( TgD ) phosphomimic mutations in kif17 −/− background. TgA/kif17 −/− and TgD/kif17 −/− mice exhibited reductions in synaptic NMDA receptors because of their inability to load/unload GluN2B onto/from KIF17, leading to impaired neuronal plasticity, CREB activation, and spatial memory. Expression of GFP-KIF17 in TgS/kif17 −/− mouse neurons rescued the synaptic and behavioral defects of kif17 −/− mice. These results suggest that phosphorylation-based regulation of NMDA receptor transport is critical for learning and memory in vivo .

  • regulation of nmda receptor transport a kif17 cargo binding releasing underlies synaptic plasticity and memory in vivo
    The Journal of Neuroscience, 2012
    Co-Authors: Xiling Yin, Yosuke Takei, Nobutaka Hirokawa, Xue Feng
    Abstract:

    Regulation of NMDA receptor trafficking is crucial to modulate neuronal communication. Ca 2+ /calmodulin-dependent protein kinase phosphorylates the tail domain of KIF17, a member of the kinesin superfamily, to control NMDA receptor subunit 2B (GluN2B) transport by changing the KIF17–cargo interaction in vitro . However, the mechanisms of regulation of GluN2B transport in vivo and its physiological significance are unknown. We generated transgenic mice carrying wild-type KIF17 ( TgS ), or KIF17 with S1029A ( TgA ) or S1029D ( TgD ) phosphomimic mutations in kif17 −/− background. TgA/kif17 −/− and TgD/kif17 −/− mice exhibited reductions in synaptic NMDA receptors because of their inability to load/unload GluN2B onto/from KIF17, leading to impaired neuronal plasticity, CREB activation, and spatial memory. Expression of GFP-KIF17 in TgS/kif17 −/− mouse neurons rescued the synaptic and behavioral defects of kif17 −/− mice. These results suggest that phosphorylation-based regulation of NMDA receptor transport is critical for learning and memory in vivo .

  • Motor protein KIF1A is essential for hippocampal synaptogenesis and learning enhancement in an enriched environment.
    Neuron, 2012
    Co-Authors: Makoto Kondo, Yosuke Takei, Nobutaka Hirokawa
    Abstract:

    Environmental enrichment causes a variety of effects on brain structure and function. Brain-derived neurotrophic factor (BDNF) plays an important role in enrichment-induced neuronal changes; however, the precise mechanism underlying these effects remains uncertain. In this study, a specific upregulation of kinesin superfamily motor protein 1A (KIF1A) was observed in the hippocampi of mice kept in an enriched environment and, in hippocampal neurons in vitro, BDNF increased the levels of KIF1A and of KIF1A-mediated cargo transport. Analysis of Bdnf(+/-) and KIF1A(+/-) mice revealed that a lack of KIF1A upregulation resulted in a loss of enrichment-induced hippocampal synaptogenesis and learning enhancement. Meanwhile, KIF1A overexpression promoted synaptogenesis via the formation of presynaptic boutons. These findings demonstrate that KIF1A is indispensable for BDNF-mediated hippocampal synaptogenesis and learning enhancement induced by enrichment. This is a new molecular motor-mediated presynaptic mechanism underlying experience-dependent neuroplasticity.

Kristen J Verhey - One of the best experts on this subject based on the ideXlab platform.

  • Pathogenic mutations in the kinesin-3 motor KIF1A diminish force generation and movement through allosteric mechanisms.
    The Journal of cell biology, 2021
    Co-Authors: Breane G. Budaitis, Kristen J Verhey, Lu Rao, Shashank Jariwala, David Sept, Yang Yue, Arne Gennerich
    Abstract:

    The kinesin-3 motor KIF1A functions in neurons, where its fast and superprocessive motility facilitates long-distance transport, but little is known about its force-generating properties. Using optical tweezers, we demonstrate that KIF1A stalls at an opposing load of ~3 pN but more frequently detaches at lower forces. KIF1A rapidly reattaches to the microtubule to resume motion due to its class-specific K-loop, resulting in a unique clustering of force generation events. To test the importance of neck linker docking in KIF1A force generation, we introduced mutations linked to human neurodevelopmental disorders. Molecular dynamics simulations predict that V8M and Y89D mutations impair neck linker docking. Indeed, both mutations dramatically reduce the force generation of KIF1A but not the motor's ability to rapidly reattach to the microtubule. Although both mutations relieve autoinhibition of the full-length motor, the mutant motors display decreased velocities, run lengths, and landing rates and delayed cargo transport in cells. These results advance our understanding of how mutations in KIF1A can manifest in disease.

  • GSK3β Impairs KIF1A Transport in a Cellular Model of Alzheimer's Disease but Does Not Regulate Motor Motility at S402.
    eNeuro, 2020
    Co-Authors: Kathlyn J Gan, Kristen J Verhey, Breane G. Budaitis, A Akram, T L Blasius, E M Ramser, D R Gabrych, Michael A. Silverman
    Abstract:

    Impairment of axonal transport is an early pathologic event that precedes neurotoxicity in Alzheimer's disease (AD). Soluble amyloid-β oligomers (AβOs), a causative agent of AD, activate intracellular signaling cascades that trigger phosphorylation of many target proteins, including tau, resulting in microtubule destabilization and transport impairment. Here, we investigated how KIF1A, a kinesin-3 family motor protein required for the transport of neurotrophic factors, is impaired in mouse hippocampal neurons treated with AβOs. By live cell imaging, we observed that AβOs inhibit transport of KIF1A-GFP similarly in wild-type and tau knock-out neurons, indicating that tau is not required for this effect. Pharmacological inhibition of glycogen synthase kinase 3β (GSK3β), a kinase overactivated in AD, prevented the transport defects. By mass spectrometry on KIF1A immunoprecipitated from transgenic AD mouse brain, we detected phosphorylation at S402, which conforms to a highly conserved GSK3β consensus site. We confirmed that this site is phosphorylated by GSK3β in vitro Finally, we tested whether a phosphomimic of S402 could modulate KIF1A motility in control and AβO-treated mouse neurons and in a Golgi dispersion assay devoid of endogenous KIF1A. In both systems, transport driven by mutant motors was similar to that of WT motors. In conclusion, GSK3β impairs KIF1A transport but does not regulate motor motility at S402. Further studies are required to determine the specific phosphorylation sites on KIF1A that regulate its cargo binding and/or motility in physiological and disease states.

  • gsk3β impairs KIF1A transport in a cellular model of alzheimer s disease but does not regulate motor motility at s402
    eNeuro, 2020
    Co-Authors: Kathlyn J Gan, Kristen J Verhey, Breane G. Budaitis, A Akram, T L Blasius, E M Ramser, D R Gabrych, Michael A. Silverman
    Abstract:

    Impairment of axonal transport is an early pathological event that precedes neurotoxicity in Alzheimer’s disease (AD). Soluble amyloid-β oligomers (AβOs), a causative agent of AD, activate intracellular signaling cascades that trigger phosphorylation of many target proteins, including tau, resulting in microtubule destabilization and transport impairment. Here, we investigated how KIF1A, a kinesin-3 family motor protein required for the transport of neurotrophic factors, is impaired in mouse hippocampal neurons treated with AβOs. By live cell imaging, we observed that AβOs inhibit transport of KIF1A-GFP similarly in wildtype and tau knockout neurons, indicating that tau is not required for this effect. Pharmacological inhibition of glycogen synthase kinase 3β (GSK3β), a kinase overactivated in AD, prevented the transport defects. By mass spectrometry on KIF1A immunoprecipitated from transgenic AD mouse brain, we detected phosphorylation at Ser 402, which conforms to a highly conserved GSK3β consensus site, and confirmed that this site is phosphorylated by GSK3β in vitro. Finally, we tested whether a phosphomimic of S402 could modulate KIF1A motility in control and AβO-treated mouse neurons and in a Golgi dispersion assay devoid of endogenous KIF1A. In both systems, transport driven by mutant motors was similar to that of wildtype motors. In conclusion, GSK3β impairs KIF1A transport but does not regulate motor motility at S402. Further studies are required to determine the specific phosphorylation sites on KIF1A that regulate its cargo binding and/or motility in physiological and disease states. SIGNIFICANCE STATEMENT Axonal transport of proteins and organelles is required for neuronal function and survival and is impaired in Alzheimer’s disease (AD). Pathogenic mechanisms that directly impact motor protein motility prior to neuronal toxicity have not been widely investigated. Here, we show that KIF1A, the primary kinesin motor required for transport of neurotrophic factors, is impaired in mouse neurons treated with amyloid-β oligomers, a causative agent of AD. Inhibition of GSK3β, a kinase overactivated in AD, prevents these defects. We detected phosphorylation of S402, a highly conserved GSK3β consensus site, in KIF1A immunoprecipitated from AD mouse brain. However, a phosphomimic of S402 did not modulate KIF1A motility in cell-based assays. Thus, GSK3β impairs KIF1A transport but likely not through phosphorylation at S402.

  • Pathogenic Mutations in the Kinesin-3 Motor KIF1A Diminish Force Generation and Movement Through Allosteric Mechanisms
    2020
    Co-Authors: Breane G. Budaitis, Kristen J Verhey, Lu Rao, Shashank Jariwala, David Sept, Arne Gennerich
    Abstract:

    The kinesin-3 motor KIF1A functions in neurons where its fast and superprocessive motility is thought to be critical for long-distance transport. However, little is known about the force-generating properties of kinesin-3 motors. Using optical tweezers, we demonstrate that KIF1A and its C. elegans homolog UNC-104 undergo force-dependent detachments at ~3 pN and then rapidly reattach to the microtubule to resume motion, resulting in a sawtooth pattern of clustered force generation events that is unique among the kinesin superfamily. Whereas UNC-104 motors stall before detaching, KIF1A motors do not. To examine the mechanism of KIF1A force generation, we introduced mutations linked to human neurodevelopmental disorders, V8M and Y89D, based on their location in structural elements required for force generation in kinesin-1. Molecular dynamics simulations predict that the V8M and Y89D mutations impair docking of the N-terminal ({beta}9) or C-terminal ({beta}10) portions of the neck linker, respectively, to the KIF1A motor domain. Indeed, both mutations dramatically impair force generation of KIF1A but not the motors ability to rapidly reattach to the microtubule track. Homodimeric and heterodimeric mutant motors also display decreased velocities, run lengths, and landing rates and homodimeric Y89D motors exhibit a higher frequency of non-productive, diffusive events along the microtubule. In cells, cargo transport by the mutant motors is delayed. Our work demonstrates the importance of the neck linker in the force generation of kinesin-3 motors and advances our understanding of how mutations in the kinesin motor domain can manifest in disease.

  • Expansion of the phenotypic spectrum of de novo missense variants in kinesin family member 1A (KIF1A )
    Human mutation, 2020
    Co-Authors: Simranpreet Kaur, Kristen J Verhey, Breane G. Budaitis, Yang Yue, Nicole J. Van Bergen, Cameron J. Nowell, Carolyn Ellaway, Nicola Brunetti-pierri, Gerarda Cappuccio, Irene Bruno
    Abstract:

    Defects in the motor domain of kinesin family member 1A (KIF1A), a neuron-specific ATP-dependent anterograde axonal transporter of synaptic cargo, are well-recognized to cause a spectrum of neurological conditions, commonly known as KIF1A-associated neurological disorders (KAND). Here, we report one mutation-negative female with classic Rett syndrome (RTT) harboring a de novo heterozygous novel variant [NP_001230937.1:p.(Asp248Glu)] in the highly conserved motor domain of KIF1A. In addition, three individuals with severe neurodevelopmental disorder along with clinical features overlapping with KAND are also reported carrying de novo heterozygous novel [NP_001230937.1:p.(Cys92Arg) and p.(Pro305Leu)] or previously reported [NP_001230937.1:p.(Thr99Met)] variants in KIF1A. In silico tools predicted these variants to be likely pathogenic, and 3D molecular modeling predicted defective ATP hydrolysis and/or microtubule binding. Using the neurite tip accumulation assay, we demonstrated that all novel KIF1A variants significantly reduced the ability of the motor domain of KIF1A to accumulate along the neurite lengths of differentiated SH-SY5Y cells. In vitro microtubule gliding assays showed significantly reduced velocities for the variant p.(Asp248Glu) and reduced microtubule binding for the p.(Cys92Arg) and p.(Pro305Leu) variants, suggesting a decreased ability of KIF1A to move along microtubules. Thus, this study further expanded the phenotypic characteristics of KAND individuals with pathogenic variants in the KIF1A motor domain to include clinical features commonly seen in RTT individuals.

Daniel A Colonramos - One of the best experts on this subject based on the ideXlab platform.

  • KIF1A unc 104 transports atg 9 to regulate neurodevelopment and autophagy at synapses
    Developmental Cell, 2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, David H. Hall, Daniel A Colonramos
    Abstract:

    Autophagy is a cellular degradation process important for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in Caenorhabditis elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determine, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosome biogenesis in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.

  • KIF1A unc 104 transports atg 9 to regulate neurodevelopment and autophagy at synapses
    bioRxiv, 2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, Daniel A Colonramos
    Abstract:

    Autophagy is a cellular degradation process essential for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in C. elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determined, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell-autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosomes in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.

Lynn W Enquist - One of the best experts on this subject based on the ideXlab platform.

  • Pseudorabies Virus Infection Accelerates Degradation of the Kinesin-3 Motor KIF1A.
    Journal of virology, 2020
    Co-Authors: Hao Huang, Orkide O Koyuncu, Lynn W Enquist
    Abstract:

    Alphaherpesviruses, including pseudorabies virus (PRV), are neuroinvasive pathogens that establish lifelong latency in peripheral ganglia following the initial infection at mucosal surfaces. The establishment of latent infection and subsequent reactivations, during which newly assembled virions are sorted into and transported anterogradely inside axons to the initial mucosal site of infection, rely on axonal bidirectional transport mediated by microtubule-based motors. Previous studies using cultured peripheral nervous system (PNS) neurons have demonstrated that KIF1A, a kinesin-3 motor, mediates the efficient axonal sorting and transport of newly assembled PRV virions. Here we report that KIF1A, unlike other axonal kinesins, is an intrinsically unstable protein prone to proteasomal degradation. Interestingly, PRV infection of neuronal cells leads not only to a nonspecific depletion of KIF1A mRNA but also to an accelerated proteasomal degradation of KIF1A proteins, leading to a near depletion of KIF1A protein late in infection. Using a series of PRV mutants deficient in axonal sorting and anterograde spread, we identified the PRV US9/gE/gI protein complex as a viral factor facilitating the proteasomal degradation of KIF1A proteins. Moreover, by using compartmented neuronal cultures that fluidically and physically separate axons from cell bodies, we found that the proteasomal degradation of KIF1A occurs in axons during infection. We propose that the PRV anterograde sorting complex, gE/gI/US9, recruits KIF1A to viral transport vesicles for axonal sorting and transport and eventually accelerates the proteasomal degradation of KIF1A in axons.IMPORTANCE Pseudorabies virus (PRV) is an alphaherpesvirus related to human pathogens herpes simplex viruses 1 and 2 and varicella-zoster virus. Alphaherpesviruses are neuroinvasive pathogens that establish lifelong latent infections in the host peripheral nervous system (PNS). Following reactivation from latency, infection spreads from the PNS back via axons to the peripheral mucosal tissues, a process mediated by kinesin motors. Here, we unveil and characterize the underlying mechanisms for a PRV-induced, accelerated degradation of KIF1A, a kinesin-3 motor promoting the sorting and transport of PRV virions in axons. We show that PRV infection disrupts the synthesis of KIF1A and simultaneously promotes the degradation of intrinsically unstable KIF1A proteins by proteasomes in axons. Our work implies that the timing of motor reduction after reactivation would be critical because progeny particles would have a limited time window for sorting into and transport in axons for further host-to-host spread.

  • pseudorabies virus infection accelerates degradation of the kinesin 3 motor KIF1A
    bioRxiv, 2019
    Co-Authors: Hao Huang, Orkide O Koyuncu, Lynn W Enquist
    Abstract:

    Alphaherpesviruses, including pseudorabies virus (PRV), are neuroinvasive pathogens that establish life-long latency in peripheral ganglia following the initial infection at mucosal surfaces. The establishment of latent infection and the subsequent reactivations during which newly-assembled virions are sorted into and transported anterogradely inside axons to the initial mucosal site of infection, rely on axonal bidirectional transport mediated by microtubule-based motors. Previous studies using cultured peripheral nervous system (PNS) neurons have demonstrated that KIF1A, a kinesin-3 motor, mediates the efficient axonal sorting and transport of newly-assembled PRV virions. In this study, we report that KIF1A, unlike other axonal kinesins, is an intrinsically unstable protein prone to proteasomal degradation. Interestingly, PRV infection of neuronal cells leads not only to a non-specific depletion of KIF1A mRNA, but also to an accelerated proteasomal degradation of KIF1A proteins, leading to a near depletion of KIF1A protein late in infection. Using a series of PRV mutants deficient in axonal sorting and anterograde spread, we identified the PRV US9/gE/gI protein complex as a viral factor facilitating the proteasomal degradation of KIF1A proteins. Moreover, by using compartmented neuronal cultures that fluidically and physically separate axons from cell bodies, we found that the proteasomal degradation of KIF1A occurs in axons during infection. We propose that PRV anterograde sorting complex, gE/gI/US9, recruits KIF1A to viral transport vesicles for axonal sorting and transport, and eventually accelerates the proteasomal degradation of KIF1A in axons.

  • Pseudorabies Virus Infection Accelerates Degradation of the Kinesin-3 Motor KIF1A
    2019
    Co-Authors: Hao Huang, Orkide O Koyuncu, Lynn W Enquist
    Abstract:

    AbstractAlphaherpesviruses, including pseudorabies virus (PRV), are neuroinvasive pathogens that establish life-long latency in peripheral ganglia following the initial infection at mucosal surfaces. The establishment of latent infection and the subsequent reactivations during which newly-assembled virions are sorted into and transported anterogradely inside axons to the initial mucosal site of infection, rely on axonal bidirectional transport mediated by microtubule-based motors. Previous studies using cultured peripheral nervous system (PNS) neurons have demonstrated that KIF1A, a kinesin-3 motor, mediates the efficient axonal sorting and transport of newly-assembled PRV virions. In this study, we report that KIF1A, unlike other axonal kinesins, is an intrinsically unstable protein prone to proteasomal degradation. Interestingly, PRV infection of neuronal cells leads not only to a non-specific depletion of KIF1A mRNA, but also to an accelerated proteasomal degradation of KIF1A proteins, leading to a near depletion of KIF1A protein late in infection. Using a series of PRV mutants deficient in axonal sorting and anterograde spread, we identified the PRV US9/gE/gI protein complex as a viral factor facilitating the proteasomal degradation of KIF1A proteins. Moreover, by using compartmented neuronal cultures that fluidically and physically separate axons from cell bodies, we found that the proteasomal degradation of KIF1A occurs in axons during infection. We propose that PRV anterograde sorting complex, gE/gI/US9, recruits KIF1A to viral transport vesicles for axonal sorting and transport, and eventually accelerates the proteasomal degradation of KIF1A in axons.ImportancePseudorabies virus (PRV) is an alphaherpesvirus related to human pathogens herpes simplex virus −1, −2 and varicella zoster virus. Alphaherpesviruses are neuroinvasive pathogens that establish life-long latent infections in the host peripheral nervous system (PNS). Following reactivation from latency, infection spreads from the PNS back via axons to the peripheral mucosal tissues, a process mediated by kinesin motors. Here, we unveil and characterize the underlying mechanisms for a PRV-induced, accelerated degradation of KIF1A, a kinesin-3 motor promoting the sorting and transport of PRV virions in axons. We show that PRV infection disrupts the synthesis of KIF1A, and simultaneously promotes the degradation of intrinsically unstable KIF1A proteins by proteasomes in axons. Our work implies that the timing of motor reduction after reactivation would be critical because progeny particles would have a limited time window for sorting into and transport in axons for further host-to-host spread.

  • Glycoproteins gE and gI are required for efficient KIF1A-dependent anterograde axonal transport of alphaherpesvirus particles in neurons
    Journal of virology, 2013
    Co-Authors: Radomir Kratchmarov, Tal Kramer, Todd M. Greco, Matthew P. Taylor, Ileana M. Cristea, Lynn W Enquist
    Abstract:

    Alphaherpesviruses, including pseudorabies virus (PRV), spread directionally within the nervous systems of their mammalian hosts. Three viral membrane proteins are required for efficient anterograde-directed spread of infection in neurons, including Us9 and a heterodimer composed of the glycoproteins gE and gI. We previously demonstrated that the kinesin-3 motor KIF1A mediates anterograde-directed transport of viral particles in axons of cultured peripheral nervous system (PNS) neurons. The PRV Us9 protein copurifies with KIF1A, recruiting the motor to transport vesicles, but at least one unidentified additional viral protein is necessary for this interaction. Here we show that gE/gI are required for efficient anterograde transport of viral particles in axons by mediating the interaction between Us9 and KIF1A. In the absence of gE/gI, viral particles containing green fluorescent protein (GFP)-tagged Us9 are assembled in the cell body but are not sorted efficiently into axons. Importantly, we found that gE/gI are necessary for efficient copurification of KIF1A with Us9, especially at early times after infection. We also constructed a PRV recombinant that expresses a functional gE-GFP fusion protein and used affinity purification coupled with mass spectrometry to identify gE-interacting proteins. Several viral and host proteins were found to associate with gE-GFP. Importantly, both gI and Us9, but not KIF1A, copurified with gE-GFP. We propose that gE/gI are required for efficient KIF1A-mediated anterograde transport of viral particles because they indirectly facilitate or stabilize the interaction between Us9 and KIF1A.

  • Kinesin-3 mediates axonal sorting and directional transport of alphaherpesvirus particles in neurons.
    Cell host & microbe, 2012
    Co-Authors: Tal Kramer, Todd M. Greco, Matthew P. Taylor, Ileana M. Cristea, Anthony E. Ambrosini, Lynn W Enquist
    Abstract:

    During infection of the nervous system, alphaherpesviruses-including pseudorabies virus (PRV)-use retrograde axonal transport to travel toward the neuronal cell body and anterograde transport to traffic back to the cell periphery upon reactivation from latency. The PRV protein Us9 plays an essential but unknown role in anterograde viral spread. To determine Us9 function, we identified viral and host proteins that interact with Us9 and explored the role of KIF1A, a microtubule-dependent kinesin-3 motor involved in axonal sorting and transport. Viral particles are cotransported with KIF1A in axons of primary rat superior cervical ganglion neurons, and overexpression or disruption of KIF1A function, respectively, increases and reduces anterograde capsid transport. Us9 and KIF1A interact early during infection with the aid of additional viral protein(s) but exhibit diminished binding at later stages, when capsids typically stall in axons. Thus, alphaherpesviruses repurpose the axonal transport and sorting pathway to spread within their hosts.

Andrea K.h. Stavoe - One of the best experts on this subject based on the ideXlab platform.

  • KIF1A unc 104 transports atg 9 to regulate neurodevelopment and autophagy at synapses
    Developmental Cell, 2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, David H. Hall, Daniel A Colonramos
    Abstract:

    Autophagy is a cellular degradation process important for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in Caenorhabditis elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determine, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosome biogenesis in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.

  • KIF1A/UNC-104 Transports ATG-9 to Regulate Neurodevelopment and Autophagy at Synapses.
    Developmental cell, 2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, David H. Hall, Daniel A. Colón-ramos
    Abstract:

    Autophagy is a cellular degradation process important for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in Caenorhabditis elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determine, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosome biogenesis in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.

  • KIF1A/UNC-104 transports ATG-9 to regulate neurodevelopment and autophagy at synapses
    2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, Daniel A. Colón-ramos
    Abstract:

    Autophagy is a cellular degradation process essential for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in C. elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determined, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell-autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosomes in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.

  • KIF1A unc 104 transports atg 9 to regulate neurodevelopment and autophagy at synapses
    bioRxiv, 2016
    Co-Authors: Andrea K.h. Stavoe, Sarah E. Hill, Daniel A Colonramos
    Abstract:

    Autophagy is a cellular degradation process essential for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in C. elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determined, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell-autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosomes in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.