Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jun 6.
Published in final edited form as: Transl Neurosci. 2012 Dec 1;3(4):355–372. doi: 10.2478/s13380-012-0044-7

PATHOLOGIES OF AXONAL TRANSPORT IN NEURODEGENERATIVE DISEASES

Xin-An Liu 1, Valerio Rizzo 1, Sathyanarayanan V Puthanveettil 1,*
PMCID: PMC3674637  NIHMSID: NIHMS471780  PMID: 23750323

Abstract

Gene products such as organelles, proteins and RNAs are actively transported to synaptic terminals for the remodeling of pre-existing neuronal connections and formation of new ones. Proteins described as molecular motors mediate this transport and utilize specialized cytoskeletal proteins that function as molecular tracks for the motor based transport of cargos. Molecular motors such as kinesins and dynein's move along microtubule tracks formed by tubulins whereas myosin motors utilize tracks formed by actin. Deficits in active transport of gene products have been implicated in a number of neurological disorders. We describe such disorders collectively as “transportopathies”. Here we review current knowledge of critical components of active transport and their relevance to neurodegenerative diseases.

Keywords: Axonal transport, Neurodegenerative diseases

Introduction

Neurons are highly polarized cells characterized by their unique morphology and compartmental specialization. Compartments such as cell body, axons, dendrites and synapses possess a unique stoichiometry of various gene products for carrying out specific biochemical functions. Synapses that connect two neurons are dynamic and mediate intercellular communication between neurons in the circuitry. Gene products such as RNA, proteins and organelles fundamental for neuronal survival, morphogenesis, function and plasticity are synthesized in the cell body and are transported through axons and delivered to synapses. For example, organelles such as mitochondria and synaptic vesicles, proteins such as ion channels and neurotropic factor receptors, and RNAs such as CaMKII and Arc are actively transported to synaptic terminals [1]. Most of the proteins necessary for the axon and synaptic terminals must be synthesized in the cell body and then transported down the axon [1]. An active transport mechanism is necessary not only to supply newly synthesized materials from soma; but also to transport of damaged organelles from the axon terminal to the cell body [2-4].

Active transport of gene products is a complex process and requires three critical components such as cytoskeletal tracks (formed by microtubules, MTs, and actin), molecular motors (kinesin, dynein, and myosin), and various cargos transported in neurons (Figure 1). Microtubules and neurofilaments are the fundamental longitudinal cytoskeletal filament in the axon and dendrites of neurons, while actin filaments are generally considered to form the major cytoskeletal architecture in the synaptic regions, such as presynaptic terminals and postsynaptic spines. Apart from motors, tracks and cargos, active transport also requires several scaffolding proteins as adaptors which facilitate transport of specific cargos [5-9].

Figure 1.

Figure 1

Components of the active transport machinery. Microtubule and actin forms the tracks for active transport. Kinesin, dynein and myosin are the ATP dependent molecular motors that transport various cargos. Pre and postsynaptic neurons and direction of transport and components of the transport machinery are shown.

1. Molecular motors in neuronal tracks

Microtubules (MT) are key determinants of neuronal polarity [10-13] and form the transport highways for cargo trafficking in axons and dendrites in neurons [6]. MT are formed from the association of dimers of α-tubulin and β-tubulin into protofilaments. The head to tail association of α- β heterodimers imparts polarity, β monomer pointing towards the plus end (faster growing end), α monomers are pointing towards minus (or slow growing end). Protein such as γ tubulin binds to the minus ends [14] whereas end binding proteins (EB) bind to the plus end of MT [15,16] and stabilize the ends. Several proteins such as microtubule associated proteins (MAP) binding to MT regulate their stability and interaction with motors [17-21].

A. Kinesin- and dynein-driven transports on microtubules

Active transport of gene products is mediated by three classes of molecular motor proteins: kinesin, dynein and myosin. Anterograde axonal transport (from cell body to synaptic terminals) of cargos is mediated by kinesin proteins, whereas retrograde transport (from synaptic terminals to cell body) use the dynein-dynactin system. Both kinesins and dyneins move along microtubules and require ATP for motility [22]. Kinesins were originally identified by Brady [23] and Vale et al., [24], and were found to be composed of two heavy chains (KHC) and two light chains (KLC). To date, more than 45 kinesins, which are classified into 14 classes, have been identified in mammals [1,25,26]. Kinesins mediate a number of important processes such as cell division, differentiation and development of the nervous system by transporting different gene products such as proteins, organelles and RNA (reviewed in [1,27-32]). Kinesin heavy chain, ApKHC1 and light chain ApKLC2 are upregulated during the formation of long-term memory in marine snail Aplysia. The coordinated regulation of axonal transport in pre and post-synaptic neurons has been identified to play a critical mediator of long-term learning-related plasticity [33].

Dynein protein was originally identified from cilia by Gibbons and Rowe [34] and from the nervous system by Burns and Pollard [35]. Dynein is a large complex and consists of two dynein heavy chains (DHC), two dynein intermediate chains (DIC), four dynein light intermediate chains (DLIC), and various dynein light chains (DLC) and move towards the minus end of MTs (reviewed in [22,29,30,36,37]). Dynein requires dynactin, which increases the motor efficiency of dynein and cross-links dynein for retrograde transport [38,39]. Like kinesins, dyneins are also involved in a number of functions such as cell division, retrograde transport of organelles and vesicles, and are important in the development of the nervous system (reviewed in [22,28,36,40,41]).

B. Myosin-driven transport on actin

Actin exists as globular monomer called G-actin and filamentous polymer called F-actin. In the presence of Mg2+, K+ or Na+ ions G-actin assembles into long, helical F-actin polymers. Actin filaments are generally considered to function as mediators of synapse dynamics and plasticity, and the predominant cytoskeletal element in dendritic spines [42]. Like microtubule tracks, actin filaments also have a polarity: the barbed end (the growing end) points to the plasma membrane in the presynaptic and postsynaptic regions. Actin organization and dynamics is important for neuronal morphology and function (reviewed in [43,44]). As in the case of microtubule binding proteins regulating MT, several actin binding proteins are known and regulate actin cytoskeleton [45].

The presence of myosin in the brain was discovered in the late 1960s [46,47]. Myosin superfamily motor proteins are classified into 18 classes and use the energy of ATP hydrolysis to generate force for movement along actin (reviewed in [48-52]. Myosins function as monomers [53], dimers [54] or oligomers [55] for cargo transport in different organs or cell types based on their different kinetic properties, structural adaptations and functional properties. In general, myosins are composed of three domains: motor domain or head, which is the most conserved, usually located at the amino terminus and which binds ATP and actin [56]; the head is mechanically connected to a second domain consisting of an extended α-helical ‘neck’, which contains numerous IQ motifs that bind light chains of calmodulin family. The C-terminal tail of unconventional myosins can include coiled-coil motifs for dimerization and a terminal globular cargo-binding domain, as well as domains that bind to membranes or to cargo receptors mediating specific interactions with vesicular or non-vesicular cargo [57]. The general myosin head-coiled coil-cargo binding domain architecture of many unconventional myosins facilitate the walking motions of the dimer along the actin filaments that result from vesicles or organelles attached at the cargo binding domain of the dimer and the ATPase hydrolysis [58].

Myosins contribute to three types of transport processes in neurons: recycling of receptors or other membrane components; dynamic tethering of vesicular components; and transport or tethering of protein translational machinery including mRNA. Myosin Vb transports recycling endosomes into the dendritic spines of hippocampal neurons in response to strong spine stimulation. These endosomes then serve as a source of AMPA receptors for insertion into the postsynaptic membrane to drive LTP [59]. Myosin Va serves as a point-to-point transporter to move tubules of ER into the dendritic spines of cerebellar Purkinje neurons, which is required for the local Ca2+ transients that drive LTD [60]. Myosin VI is the only known retrograde myosin, which walks toward the minus end of the actin filament and the dimerization of which appears to occur upon cargo binding [61]. Myosin Vb (MyoVb), a Ca2+-sensitive motor, conducts spine trafficking during long-term potentiation (LTP) of synaptic strength [59]. Myosin II, which is present presynaptically, is important for synaptic vesicle mobility at the Drosophila neuromuscular junction [62] and for memory storage in mouse [63,64].

C. Regulation of microtubule (MT) and actin dependent transport

Synaptic activity could play an important role in regulation of MT. It has been suggested that raising global electrical activity increases the number of MT invading dendritic spines in hippocampal cultures [65]. MT are generally considered to be the main tracks for transporting synaptic materials to and from synapses. Increased synaptic activity might reorganize microtubule to redirect the transport of synaptic proteins and organelles into spines [66]. Local regulation of microtubule stability and subsequent microtubule modifications might be the primary regulators of activity-induced synaptic changes [67,68]. Several post-translational modifications of microtubules that regulate its properties are known [69-75]. For example, Kinesin-1, which is abundant in axons, moves preferentially on acetylated microtubules [76], a post-translational modification of microtubules [77], instead of tyrosinated microtubules [78]. Microtubule modifications can be regulated by synaptic activity, including reduced motor protein mobility and cargo delivery into neurites [66].

Mammalian plus-end-tracking proteins (+TIP) localize to the ends of growing microtubules and regulate both the dynamic behavior of microtubules as well as the interactions of microtubules with other cellular components [16]. Mutations in the MT-associated protein Futsch, the fly homolog of microtubule-associated protein 1B (MAP1B), has been proved to disrupt the architecture and network of axonal MT that induce defects in axonal transport, causing progressive degeneration of neurons in the fly brain [79].

Actin cytoskeleton is also highly responsive to changes in electrical activity, which can be dynamically rearranged by depolarization [80]. In addition, actin polymerization is required for the maintenance of prolonged periods of synaptic activity during LTP [81]. Rho family proteins of GTPases have been shown to act as molecular switches that increase/decrease actin polymerization rates, crosslinking activities, or branch formation [82], which could be a cellular basis for impaired cognitive functions.

Apart from MT and actin cytoskeleton being covalently modified to regulate transport, molecular motors are also post-translationally modified. For example, kinesins are modified by phosphorylation and regulate its interaction with microtubules and cargos [83-85]. Active c-Jun N-terminal kinase (JNK) phosphorylates kinesin-1 heavy chains and inhibits kinesin-1 microtubule-binding activity [86]. The inflammatory cytokine tumor necrosis factor-alpha (TNF) stimulates phosphorylation of JNK and induces dissociation of KIF5B from tubulin in axons, it accordingly inhibits axonal transport of mitochondria and synaptophysin by reducing the mobile fraction [87]. JNK interacting proteins (JIP), which are scaffolding molecules for JNK signaling pathways, are illustrated to regulate Kinesin-1 mediated transport [88]: they activate Kinesin-1 in concert with FEZ1/UNC-76 [89]; release JIP-dependent cargo through activation of the JNK cascade, in turn inactivating Kinesin-1 [90]; and regulate the dynamics of neuronal microtubules by modulating phosphorylation's of MT-associated proteins (MAP) [91,92] and microtubule-destabilizing protein SCG10 [93]. The regulation of kinesin/cargo association through adaptor proteins is another major control point for regulating cargo-specific transport. Phosphorylation of huntingtin at S421 promotes recruitment of kinesin-1 to the dynactin complex on vesicles and MT and the following anterograde transport, which suggests that huntingtin phosphorylation can act as a molecular switch for anterograde/retrograde transport in neurons [94]. Actin and MT cytoskeleton also interact [95,96] and such interactions are important for axon guidance and specification [97,98].

2. Neuronal cargos

A wide variety of cargos such as organelles, cytoskeletal components, growth factors, trophic factors [99], synaptic vesicle precursors, neurotransmitter, signaling molecules, and mRNA etc., are actively transported from their sites of synthesis in the cell body through the axoplasm to intracellular target sites. Transported cargos can be mainly classified into three groups: proteins, RNA, and organelles. Specific sets of cargos are packaged and transported along axons to specific destinations for the establishment of neuronal connections and for the modifications of these connections during memory storage. Recognition, binding and unloading of cargos are important regulation mechanisms for neuronal traffic.

Proteins are transported in various membranous organelles and protein complexes, and mRNA are carried in large protein complexes [100]. RNA transport is an important and fundamental event for local protein synthesis, especially in neurons, while local protein synthesis is believed to contribute to synaptic plasticity that requires a rapid supply of new proteins to specific synaptic sites in response to appropriate stimuli and may also participate in long-lasting changes in synaptic strength [101-108]. A number of transported protein components including synaptic proteins have been identified [28,33,109]. Kinesin 1 family KIF5 motors can directly bind to and transport large RNase-sensitive granules, known as messenger ribonucleoprotein (mRNP) complexes, which contain mRNA and at least 40 RNA binding proteins [110]. However it remains to be determined how, when, and what mRNA and proteins are assembled into these complexes and transported down to the dendrites.

3. Pathologies of axonal transport in neurodegenerative diseases - “transportopathies”

As illustrated above, molecular motors regulate several vital processes in neurons. Disruption of axonal transport is a hallmark and precipitating factor of a wide variety of neurodegenerative diseases. Table 1 lists the involvement of active transport machinery in these disorders. They involve different components of the transport machinery and affect different regions of the brain.

Table 1.

The neurodegenerative disorders and the involvement of active transport machineries.

Neurodegenerative diseases Component of the transport machinery involved References
Alzheimer (AD) Tau and Aβ facilitate cargo release and disturbed neuronal trafficking [176,189]
PS1 Mutations increase KLC phosphorylation and cargo release [190]
Mitochondrial abnormalities lead to reduction in the number of microtubules [136]
Huntington's disease (HD) Mutant Htt causes defective anterograde mitochondrial movement [259]
Amyotrophic lateral sclerosis (ALS) Kinesin-associated protein 3 (KAP3) is sequestrated by misfolded SOD1 and results in axonal transport inhibition of ChAT [210]
Dynactin mutation in the p150 subunit [154,216-219]
Mutations in dynein-dynactin retrograde system protein genes [222-224]
KHC gene mutation [262]
Mislocalization and disruption of dynein function [214]
Frontotemporal dementia (FTD) Hyperphosphorylated tau deposition decrease binding of motor proteins to MT tracks [167,168,176]
Mutation of FUS, which is a DNA/RNA-binding protein involved in transport of mRNA molecules [263]
Hereditary spastic paraplegias (HSP) Mutations in the KIF5A in families with SPG10 reduce microtubule binding affinity [240]
SPG7 models exhibit impaired mitochondrial axonal transport [241]
HSAN II FAM134B mutations lead to KIF1A mutations [123]
Charcot-Marie-Tooth (CMT2) DYNC1H1 mutation impairs dynein heavy chain 1 involved retrograde axonal transport [264]
Mfn2 mutation produces a severe mitochondrial transport disorder [257]
Mutation in kinesin 5A [249]
KIF1Bβ mutation [130]
axonal CMT Expression of mutant HSPB1 decreased acetylated α-tubulin abundance and induced axonal transport deficits [251]
Mutations in GDAP1 alter the interaction between mitochondria and the microtubule cytoskeleton and affect mitochondrial axonal transport [252,253,255]
X-linked spinal and bulbar muscular atrophy (SBMA) Androgen receptor gene mutations with enlargement of the CAG repeat form aggregates that alter axonal trafficking [265-267]
Parkinson's disease (PD) Mutation and phosphorylation of α-synuclein reduces its axonal transport [268]
Mutations of parkin, PINK1, and DJ-1 lead to damage to mitochondria and perturb transport of mitochondria through axons [269,270]
Inhibition of complex I of the electron transport chain decreases anterograde and increases retrograde axonal transport of membranous vesicles [271]
Multiple sclerosis (MS) Ca2+, free radical production and mitochondrial dysfunction impair axonal transport [272]
Spinal muscular atrophy (SMA) SMN1 mutation leads to reduced transport of specific mRNAs within motor neurons [273,274]
polyglutamine disorder (polyQ) Polyglutamine proteins interrupt axonal transport and affect enzymatic activities involved in FAT regulation [275-277]
Prion diseases Intracerebral prion lead to axonal transport impairments which involves Rab7-mediated cargo attachment to the dynein-dynactin pathway [278]
Glaucoma Mitochondrial dysfunction and axonal transport failure induce retinal ganglion cell death [279]
Spinocerebellar ataxia type 5 (SCA5) SCA5 mutant spectrin impair axonal transport and induce neurodegeneration [280]
Traumatic brain injury (TBI) Extensive axonal injury result in the interruption of axonal transport and long-term accumulation of proteins [281]
Dystonia musculorum (dt) Microtubule network perturbation lead to axonal transport defects [282]
Tuberous sclerosis (TSC1) disease kinesin-related gene ATSV is characterized as candidate genes for TSC1 disease locus on chromosome 9q34 [283]

Here we discuss three different causes of axonal transport defects: gene mutation, oxidative stress and post-translational modifications of the transport machinery. We specifically focus on Alzheimer's disease (AD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) and briefly describe transport defects in other nervous system diseases with axonal pathologies.

3a. Gene mutations and axonal transport defects

Mutations in motor proteins and other defects in intracellular transport have been described to cause neuropathologies [111]. Gene mutations that disrupt axonal transport are gaining attention as a cause of neuronal dysfunction in a variety of neurodegenerative motor neuron diseases in humans. Mutations in motor proteins, such as kinesins or dyneins, cause acute organelle jams in axons, preventing transport and recycling of synaptic metabolites. The immaculate connections (imac), a Drosophila Kinesin-3, are selectively required in motoneurons for transport of many synaptogenic cargos. In imac embryos, motoneuron axons extend properly and are guided to and arrest on the appropriate muscle fibers, but synapses cannot form. Loss of imac leads to a severe reduction of synaptic vesicles (SV) at terminals and an increase of SV stranded in the cell body [32]. Active zone proteins are greatly reduced, synaptic vesicles are absent, and the nerve endings do not mature into rounded boutons [112], which also illustrates that axonal transport is critically required for forming synapses. In addition, Kinesin-1 mutants or mutations in proteins associated with Kinesin-1 motors including kinesin light chain (KLC), the JNK scaffolding adaptor [88,113] and its associated MAPKKK/MAPKK/JNK signaling complex, Liprin-alpha [114] and Unc-76 [115] develop an SV transport phenotype. Mutations in Kinesin-3 prevent synapse formation at the Drosophila neuromuscular junction although axon outgrowth and guidance are normal. The phenotype illustrates the selectivity of this Kinesin-3 motor for synaptogenesis [112].

In mammalian cells, KIF5B (Kinesin-1 family) and KIF1B (Kinesin-3 family) are reported to mediate transport of mitochondria. Proteins milton and miro form an essential protein complex that links kinesin to mitochondria for light chain-independent, anterograde transport to synapses. The amino-terminus variant of Drosophila milton, milton-C, inhibits kinesin binding to milton and thereby prevents kinesin recruitment to mitochondria [116].

In 2003, it was reported that missense point mutations in the cytoplasmic dynein heavy chain result in late onset and progressive motor neuron degeneration (MND) in two lines of mice called Legs at odd angles (Loa/+) and Cramping 1 (Cra1/+) [117], which was also proved by Vallee et al. [118], while some other studies provide evidence that the primary pathology in Cra1/+ animals may be an early onset, non-progressive synaptic dysfunction that affects the neuromuscular junction [119] without motor neuron involvement [120]. A point mutation in dynein heavy chain gene leads to striatal function deficit and striatal atrophy, which supports a role for dynein dysfunction in the pathogenesis of neurodegenerative disorders of the basal ganglia, such as Perry syndrome and Huntington's disease (HD) [121].

Dynactin-1 mRNA is indicated to be down regulated in degenerating spinal motor neurons of autopsied patients with sporadic ALS [122]; KIF1A, was reported to be mutated in hereditary sensory and autonomic neuropathy type 2 (HSANII), which is a rare autosomal-recessive disorder characterized by peripheral nerve degeneration resulting in a severe distal sensory loss [123], the causative mutation in the motor domain of KIF1A is also implicated in hereditary spastic paraplegias (HSP) patients [124]. Mutations in the KIF5A gene can also be associated with adult onset of autosomal dominant hereditary spastic paraplegia (AD HSP) [125]. Mutations in the KIF7 gene were identified to cause modified microtubule stability and growth direction, which is an underlying disease mechanism contributing to Joubert syndrome (JBTS) [126]. De novo truncating mutation in Kinesin17 is associated with schizophrenia [127] and KIF21A mutation is involved in congenital fibrosis of the extra-ocular muscles type 1 and 3 [128,129]. Charcot-Marie-Tooth disease type 2A (CMT2A) patients contain a loss-of-function mutation in the motor domain of the KIF1B gene [130].

3b. Mitochondrial oxidative stress and axonal transport defects

Oxidative stress is regulated by the levels of reactive oxygen species (ROS) that includes superoxides, hydroxyl radical, and hydrogen peroxide. Oxidative stress, which induce mitochondrial injury, impairs axonal transport rates in mice [131,132] and would result in an accumulation of axonal constituents delivered by fast and slow axonal transport [133,134], disorders of synaptic transmission and synapse degeneration. Oxidative stress may also disrupt the MAP:tubulin ratio and thus result in disruption of neuronal intracellular transport [135]. Hirai et al. suggested that mitochondrial abnormalities might be part of the spectrum of chronic oxidative stress of AD [136]. Accumulation of mitochondrial DNA was found in the neurons vulnerable to death in AD and attributed it to a possible reduction in the number of microtubules, which would result in diminished vesicular transport and mitochondrial turnover [136]. Reduction of mitochondrial superoxide significantly prevented the deficits in axonal transport rates in Tg2576 mice, indicating that ROS of mitochondrial origin are a key factor in transducing the impact of Aβ on neuronal physiology in AD mice model [137]. Axonal transport deficits and accumulation of depolarized mitochondria could be concurrent with axonal neuropathy and without increased reactive oxygen species production [138]. Mitochondrial dysfunction, such as altered Ca2+ homeostasis and increase in reactive oxygen species [139], have been reported in both in vitro and in vivo models of ALS [140]. Mitochondria accumulation was also shown in the axons of spinal motor neurons in sporadic ALS (SALS) patients, suggesting the deficit in mitochondria metabolism due to impaired axonal transport in ALS [141,142].

The autophagosomes are membraneous cargos that move bidirectionally along microtubules and are transported by kinesin and dynein/dynactin complex, which accumulate in motor neurons with altered axonal transport [143-144]. The dysfunction of autophagy-lysosome pathway that is responsible for recycling of intracellular contents, have been described to cause neurodegeneration [145,146]. Abnormal intraneuronal accumulation of autophagosomes occurs in AD [147,148], Parkinson's disease (PD) [149], HD [150,151], and in ALS patients [152] as well as the mutant dynein model (Loa mouse) [153] and the mutant dynactin-1 mouse model [154].

3c. Post translational changes of the transport machinery

3C1. Alzheimer's disease (AD)

Widespread synaptic and neuronal loss and the pathological accumulation of amyloid-beta peptide (Aβ) in senile plaques, as well as hyperphosphorylated tau in neurofibrillary tangles (NFT) are the pathological hallmarks of AD, the leading cause of dementia among the elderly [155]. Axonal transport deficits may represent an early step in AD pathogenesis because the reduced axonal transport was observed before the apparent AD hallmarks.

Tau, axonal transport and AD

Tau, a major microtubule-associated protein that plays an important role in the outgrowth of neuronal processes and the development of neuronal polarity, promotes microtubule assembly, stabilizes microtubules, and affects the dynamics of microtubules in neurons [156,157]. Tau is hyperphosphorylated, which loses its capability to bind with microtubules, accumulates in neurons, and forms paired helical filaments in AD [158]. It has been reported that Aβ, which is produced by putative intramembranous processing of β-amyloid precursor protein (APP) at the proposed active site of the Ɣ-secretase/PS1 aspartyl protease, is a critical factor for hyperphosphorylation of tau in AD neurons [159], while increasing evidence suggests that hyperphosphorylated tau is critically involved in AD pathogenesis, particularly in impairing axonal transport of APP and subcellular organelles including mitochondria in neurons affected by AD [160-162]. Various intronic and exonic pathogenic mutations of TAU gene provided unequivocal proof that tau abnormalities alone are sufficient to cause neurodegeneration [163].

As kinesin and dynein motor proteins transport cellular cargoes toward opposite ends of microtubule tracks, which are abundantly decorated with microtubule-associated proteins (MAP) such as tau in neurons, long-distance trafficking uses mainly the axonal microtubule highway and tau does (de)stabilize this network. Therefore, it is no surprise that a deregulation of its expression and/or phosphorylation level can lead to defects in axonal transport such as found in the early stages of AD [164] or even at the later stages of the disease [165]. Tau expression in stably transfected CHO cells and differentiated neuroblastoma N2a cells dramatically alters the distribution of various organelles, including mitochondria and endoplasmic reticulum, known to be transported via microtubule-dependent motor proteins. These effects were caused by tau's binding to microtubules and slowing down intracellular transport by preferential impairment of plus-end–directed transport mediated by kinesin motor proteins [166]. Axonal transport defects have also been reproduced in wild-type tau transgenic mice [167] and in K369I mutant tau K3 mice [168]. Mice transgenic for human four-repeat tau [169,170] and neurofilament [171,172] have been suggested to indicate disturbed axonal transport. Transgenic (Tg) mice expressing the longest human tau isoform (T40) with R406W mutation developed retarded axonal transport, which indicate that R406W mutation causes reduced binding of the mutant tau to microtubules, resulting in slower axonal transport [173]. Tg mice expressing the FTDP-17 human P301L mutant tau cause neurodegeneration by disrupting axonal transport since impairments in axonal transport occurred earlier, whereas motor deficits subsequently developed [174]. Reduced axonal transport and increased excitotoxic retinal ganglion cell degeneration were also revealed in mice transgenic for human mutant P301S tau [175]. Dixit et al. suggested that kinesin was inhibited at about a tenth of the tau concentration that inhibited dynein; higher tau concentration at the synapse would facilitate cargo release, and the microtubule-binding domain of tau was sufficient to inhibit motor activity, which suggests tau can spatially regulate the balance of microtubule-dependent axonal transport in the neuron by locally modulating motor function [176]. On the other hand, kinesin-1 deficient mice (KLC1-/-) revealed that axonal transport defects could initiate biochemical changes that induce activation of axonal c-jun N-terminal stress kinase pathways leading to abnormal tau hyperphosphorylation [177].

Several studies have suggested a critical role for tau in axonal transport and in AD [160,178-180]. In drosophila and mouse models of tauopathies reductions in axonal transport can exacerbate human tau protein hyperphosphorylation, formation of insoluble aggregates and tau-dependent neurodegeneration [181]. However, Yuan et al. showed that global axonal transport rates of slow and fast transport cargoes in axons are not significantly impaired when tau expression is eliminated or increased, which suggest that tau is not essential for axonal transport [182]. Although tau did not affect axonal transport under baseline untreated conditions, partial tau reduction can prevent Aβ-induced axonal transport defects in hAPP mice [160]. Thus, whether and how tau affects axonal transport and its physiological functions are still poorly understood.

Aβ, axonal transport and AD

APP is axonally transported by binding to the motor protein kinesin-1 on microtubules [183]. Reductions in microtubule-dependent transport may stimulate proteolytic processing of beta-amyloid precursor protein, resulting in the development of senile plaques and AD [184]. The axonal swellings and varicosities were found to contain abnormal accumulations of transport cargos, APP, and Aβ, and such transport deficits appear to enhance APP processing and local Aβ production. Additionally, it was reported that impairing axonal transport by reducing the dosage of a kinesin molecular motor protein enhanced the frequency of axonal defects and increased amyloid-beta peptide levels and amyloid deposition. In aged APP/PS1 mice, axonal swellings as well as amyloid plaques, were found in dorsal funiculus, a region with strong APP expression in axonal processes. The observations that spheroids, the most prominent evidence for axonopathy, were immunoreactive with axonal markers such as APP, neurofilament subunits and ubiquitin, instead of tau or phosphorylated tau, add significant evidence to the assumption that toxic Aβ peptides play a role in the mediation of disturbed neuronal trafficking [185]. Early axonopathy and transport deficits in APP-transgenic mice, could be fostered by a reduction in the amount of the kinesin light-chain (KLC1) by crossing APP-transgenic with KLC-1 knockout mice [184]. Hiruma has found that Aβ exerts its inhibitory effect on axonal transport via actin polymerization and aggregation in cultured rat hippocampal neurons [186]. Furthermore, Aβ could likely act through GSK3 to impair mitochondrial transport, and the impairment can be alleviated by PKA activation, without affecting actin polymerization and mitochondria membrane polarization [187].

Soluble oligomers of the amyloid-β peptide (AβO), recognized as the proximal neurotoxins in AD pathology, can induce disruption of organelle transport in primary hippocampal neurons by a mechanism that is initiated by NMDAR and mediated by GSK-3β leading to microtubule destabilization [188]. Real-time analysis of vesicle mobility in isolated axoplasms perfused with AβO, instead of unaggregated Aβ or fibrillar Aβ, showed bidirectional axonal transport inhibition as a consequence of endogenous casein kinase 2 (CK2) activation. Both AβO and CK2 treatment of axoplasm led to increased phosphorylation of kinesin-1 light chains and subsequent release of kinesin from its cargoes, which cause deficiencies in fast axonal transport in AD [189].

Other factors involved in transport defects observed in AD

Mutations in presenilin 1 (PS1), which is associated with early-onset familial Alzheimer's disease (FAD), have been implicated to be involved in kinesin-based axonal transport due to an interaction with glycogen synthase kinase 3β (GSK3β). The relative levels of GSK3β activity were increased either in the presence of mutant PS1 or in the absence of PS1, leading to increased kinesin light chain phosphorylation, the release of kinesin-1 from membrane-bound organelles (MBO) and reduced fast anterograde axonal transport [190]. It is also reported that defects in anterograde fast axonal transport and motor neuron deficits underlie PS-1-mediated neurodegeneration in transgenic mice expressing familial AD linked mutant PS-1 through a mechanism involving impairments in neurotrophin signaling and synaptic dysfunction [190-192].

The epsilon 4 allele (e4) of the human apolipoprotein E gene (ApoE4) constitutes an important genetic risk factor for AD. Transgenic mice expressing human ApoE4 in neurons developed axonopathy and impairment of axonal transport. In ApoE4 transgenic mice, axonal dilatations with accumulation of synaptophysin, neurofilaments, mitochondria, and vesicles were observed, suggesting impairment of axonal transport [193].

3C2. Frontotemporal dementia (FTD)

Frontotemporal dementia (FTD) is the clinical syndrome caused by degeneration of the frontal lobe of the brain and is the second most common pre-senile dementia after AD among people under the age of 65 [194]. About 50% of FTD patients have an associated family history. Several genes such as microtubule-associated protein tau (MAPT), progranulin (PGRN), chromatin modifying protein 2B (CHMP2B), and fused in sarcoma (FUS), have been described to be associated with FTD. Both MAPT and PGRN are pathogenic genes located on chromosome 17 associated with FTDP phenotype [195]. FTD frequently presents with tau-containing lesions histopathologically, which result from mutations in the MAPT gene. The identification of early and progressive axonal swellings [196] and impaired Rab7 recruitment to endosomes [197] in CHMP2Bintron 5 mice suggested a potential mechanism for impaired axonal transport.

The transgenic mouse model (K3) which expresses human tau carrying the FTD mutation K369I showed early-onset memory impairment, amyotrophy in the absence of overt neurodegeneration, which may result from impaired transport in sciatic nerves, and particularly revealed an early-onset motor phenotype that reproduces parkinsonism with tremor, bradykinesia, abnormal gait, and postural instability. The functional impairment of K3 mice is accompanied by progressive morphological changes including axonal swellings and spheroids that are histopathological correlates of disrupted axonal transport [184]. The deposition of hyperphosphorylated tau in K3 mice selectively impaired kinesin-driven anterograde transport of identified cargos such as TH-containing vesicles and mitochondria [168] by decreasing binding of motor proteins to MT tracks [167,176]. The kinesin motor complex formation is also disturbed by tau in K3 mice with the mechanism that hyperphosphorylated tau interacts with c-Jun N-terminal kinase-interacting protein 1 (JIP1), which is associated with the kinesin motor protein complex. Because JIP1 is involved in regulating cargo binding to kinesin motors, these findings partly explain how hyperphosphorylated tau mediates impaired axonal transport in FTD [198]. Moreover, it has been identified that N-terminal projection domain of tau binds to the C-terminus of the p150 subunit of the dynactin complex and the attachment of the dynactin complex to microtubules is enhanced by tau. Mutations of the N-terminus arginine residue of tau, found in patients with FTDP-17, wreck its binding to dynactin, which is abnormally distributed in the retinal ganglion cell axons of transgenic mice expressing human tau with a mutation in the microtubule-binding domain [199,200].

The increased ratio of 4 repeat over 3 repeat tau isoforms is associated with neurodegeneration in inherited forms of FTD. Tau overexpression that diminishes axonal transport in several models has been well studied. Furthermore, Hyman et al. found that both 3 repeat and 4 repeat tau change normal mitochondrial distribution within the cell body and reduce mitochondrial localization to axons; 4 repeat tau has a greater effect than 3 repeat tau, while 3 repeat tau has a slightly stronger effect on retrograde and anterograde axon transport dynamics [201]. The effect of tau/PTL-1 (protein with tau-like repeats) on the transport characteristics of the major axonal transporter kinesin-3 KIF1A/UNC-104 was also proved in the nervous system of Caenorhabditis elegans [202].

Recently, Coleman et al. have generated a novel knock-in mouse model of an inherited tauopathy, FTD with parkinsonism linked to TAU mutations on chromosome 17 (FTDP-17T). The engineered mice with a mutation in the endogenous MAPT gene that is homologous with the common P301L MAPT mutation found in patients with FTDP-17T, mimicking the human disease situation, revealed reduced tau phosphorylation and reduced MT association of tau, intriguing age-dependent changes in axonal transport of mitochondria, and increased spontaneous locomotor activity in old age [203].

3C3. Motor neuron diseases: Amyotrophic lateral sclerosis (ALS)

Amyotrophic lateral sclerosis (ALS) is a fatal and progressive neurodegenerative disease with the selective death of motor neurons in the brain, spinal cord and brainstem as a main pathological feature which leads to muscle weakness and paralysis [204]. ALS leads specifically to muscle weakness and atrophy of limb and respiratory muscles. The molecular basis of this disease is still poorly understood and several hypotheses for its etiology is under debate [205]. Axonal transport derangement is advocated as the early molecular event and a key event of pathogenic mechanisms leading to neurodegeneration in ALS.

Five Mendalian gene defects have been reported to cause ALS, in which copper-zinc superoxide dismutase type 1 (SOD1) gene mutations [206,207] have been demonstrated to be the most prevalent. Transgenic mice with mutant SOD1 show slowing of axonal transport early in the disease course [208], Cu/Zn SOD1(G93A and G37R) transgenic mice develop phenotypical hallmarks of ALS and therefore serve as an established model to study the molecular mechanisms underlying this disease. Both slow and fast anterograde transport are slowed in transgenic G93A and G37R ALS mice prior to disease onset and is exacerbated as the disease progresses [208,209]. Kinesin-associated protein 3 (KAP3) sequestration by misfolded SOD1 species and the resultant inhibition of the axonal transport of choline acetyltransferase (ChAT) play a role in the dysfunction of ALS [210], and the reduction in the levels of KAP3 has been linked to increased survival in ALS patients [211], While an early upregulation of the kinesin superfamily motor protein KIF1A was detected in spinal motor neurons in SOD mutant mice [212]. Retrograde transport is also disrupted in ALS mice [213]. The decreased retrograde transport was described in G93A SOD1 mice at an early stage of disease, coincident with neuromuscular junction degeneration and muscle weakness [140,214,215].

Reduction of retrograde transport in ALS mice has been attributed to the mislocalization and disruption of dynein function [214]. Mutations in the p150 subunit of dynactin is involved in ALS [154,216-219] and a dynein mutation which attenuates motor neuron degeneration in SOD1(G93A) mice has also been described [220,221]. Mutations in dynein-dynactin retrograde system protein genes has been demonstrated to be linked to neuronal degenerations in ALS mouse model [222,223] and Drosophila melanogaster [224], as well as distal spinal and bulbar muscular atrophy (dSBMA) [225]. Studies using autopsy material and spinal cord tissue from ALS animal models suggest that damage to the axonal cytoskeleton due to alterations of neurofilament [226] and of molecular motor proteins such as dynein and kinesin contribute to spheroid formation.

Densely accumulated neurofilaments can be seen in axons in close proximity with vacuoles and at many other proximal axon sites without vacuoles in human SOD1 transgenic mice, suggesting that axonal transport begins to fail before the onset of rapid declining (RD) stage [215,227]. In SOD (G93A) model of ALS, transport deficits are detected soon after birth, months before the onset of axon degeneration, suggesting axons can survive despite long-lasting transport deficits. On the other hand, in SOD (G85R) model of ALS motor axons degenerate, but transport is unaffected. This finding suggests that transport deficits are not necessary for axon degeneration. Additionally, axons show chronic transport deficits, but survive in mice that overexpress wild-type SOD1, indicating that axon transport deficits are not sufficient to cause immediate degeneration in this ALS model [228]. Cytoskeletal abnormalities with accumulation of ubiquinated inclusions in the anterior horn cells are a pathological hallmark of both familial and sporadic ALS and of mouse models for ALS. Further, it was reported that p38 mitogen-activated protein kinase (p38MAPK) is activated in ALS patients and mutant SOD1 transgenic mice [229,230], and biochemical studies revealed that KHC are phosphorylated by p38, which inhibited conventional kinesin-based motility, indicating that mutant SOD1 impairs anterograde fast axonal transport by activation of p38 MAPK [231].

High levels of intracellular calcium observed in ALS model neurons may interfere with mitochondria movement toward the extremities of long axons causing retrograde degeneration because microtubule transported mitochondria can be stopped at high calcium concentration sites such as the neuromuscular junction [232]. Ca2+-mediated glutamate excitotoxicity has been put forward as one of the potential mechanisms for the motor neuron-selective death in ALS [233]. It was also reported that mutant SOD1 increase the formation of damaging hydroxyl radicals and peroxynitrite derivatives [234], which inhibit the mitochondrial electron-transport chain [235]. Intracellular free radical species inhibit the activities of specific mitochondria enzymes. The mitochondria accumulate in proximal axons of the anterior horn neurons in ALS patients because of blocking of axonal trafficking into proximal neurons [141].

Hereditary spastic paraplegias (HSP)

HSP comprise a heterogeneous group of genetic neurodegenerative disorders characterized by progressive spasticity and weakness of lower limbs due to retrograde degeneration of the corticospinal tracts and posterior columns, which is often accompanied by brisk reflexes, extensor plantar reflexes, and urinary urgency [236,237]. Genetic loci for HSP are designated SPG for “spastic gait” followed consecutively by the locus number, which is assigned in order of its discovery. The identification of genes that are implicated in HSP has shown that the largest group of HSP proteins are either known or thought to be involved in the intracellular trafficking, including KHC gene, KIF5A, directly implicating axonal transport impairments to HSP pathogenesis in SPG10 [238,239]. SPG10, which causes dominant forms and originates in point mutations in the neuronal KINESIN-1 gene (KIF5A), reduces microtubule binding affinity of KIF5A that acts in a dominant negative manner by competing with wild-type motors for cargo binding, impairs motor-based transport underlying HSP [240]. Mouse models of SPG7 [241], which is associated with mutations in the mitochondrial ATPase paraplegin, and SPG4 [242-244], which results from mutations in microtubule severing protein spastin, provide in vivo evidence for axonal transport impairment in HSP. Mutations in REEP1 were recently associated with a pure dominant Hereditary spastic paraplegias (HSP), SPG31 [245] and direct evidence for axonal transport defects in a novel mouse model of mutant spastin-induced HSP and human HSP patients have also been produced [246,247].

Charcot-Marie-Tooth (CMT) disease

Charcot-Marie-Tooth (CMT) disease is the most common inherited disorder of the peripheral nervous system. Two classes of CMT have been differentiated until now: demyelinating forms of CMT (CMT1), in which nerve conduction velocities are decreased, and the axonal CMT2 forms, in which nerve conduction velocities are preserved [248]. Mutation in Kif5A causes hereditary spastic paraparesis type 10 [125] and axonal Charcot-Marie-Tooth type 2 disease (CMT2) [249]. An autosomal dominant mutation in DYNC1H1 was identified in a family with CMT2 which was also proved to cause severe intellectual disability with neuronal migration defects [250]. Mutations in the 27-kDa small heat-shock protein gene (HSPB1) cause axonal CMT or distal hereditary motor neuropathy (distal HMN). Expression of mutant HSPB1 decreased acetylated α-tubulin abundance induced severe axonal transport deficits [251].

Mutations in GDAP1 (ganglioside-induced differentiation-associated protein-1), which alter the interaction between mitochondria and the microtubule cytoskeleton and affect mitochondrial axonal transport and movement [252,253], are described as the cause of the inherited human neuropathy CMT [254,255] disease: autosomal recessive demyelinating CMT4A [256], autosomal recessive axonal CMT2K or dominant axonal CMT2K. Defects in axonal transport due to a mutation in the motor protein KIF1Bβ, has been described to be responsible for axonopathy in the CMT type 2A [130].

Mitofusins (Mfn1 and Mfn2) are outer mitochondrial membrane proteins involved in regulating mitochondrial dynamics. Mutations in MFN2 alone cause CMT type 2A for the reason that Mfn2 is a key component of the linker/adaptor complex between mitochondria and kinesin/microtubules, the deficit of which produce a severe mitochondrial transport disorder in dorsal root ganglion neurons [257].

Other motor neuron diseases

Dysfunction of dynactin-mediated transport is also documented to lead human motor neuron disease [219]. Mutations in anterograde axonal transport proteins, including KIF1B, lead to slow progressive motor neuronopathy [130]. These studies indicate the potential generality of the link between retrograde and anterograde axonal transport deficits and motor neuron degeneration [117].

Huntington's disease (HD)

Huntington's disease (HD) is one of an increasing number of human neurodegenerative disorders caused by a CAG/polyglutamine-repeat expansion. Mice models of HD (transgenic for exon 1 of the human HD gene carrying (CAG)115 to (CAG)156 repeat expansions), develop characteristic morphological changes within neurons, which are strikingly similar to abnormalities observed in biopsy material from HD patients [258]. Mutant huntingtin (Htt) interacts with mitochondrial protein Drp1, leading to impairment of mitochondrial biogenesis, defective axonal transport and synaptic degeneration in HD [259]. Htt activates axonal c-Jun N-terminal kinase (JNK3) that phosphorylate kinesin 1. Phosphorylated kinesin 1 has reduced affinity for MT leading to impaired transport [260]. Disruption of axonal transport is also described in Drosophila model for HD [261].

4. Conclusion

Studies described in this review establish a major role of axonal transport defects in several neurodegenerative diseases. These defects are caused by genetic mutations, oxidative stress and post-translational modifications of the transport machinery. However, detailed experiments are necessary to elucidate the causality of these defects as underlying mechanisms for neurodegeneration. Though we understand aspects of the major players of active transport and their physiological functions, details of the spatial and temporal regulation of transport and various cargos transported in the normal or pathological state of neuron are poorly defined. Thus, dissecting the molecular mechanisms of axonal transport and its regulation in different parts of the brain during development and maturation of the nervous system and their role in neurodegenerative diseases is required to understand cell biology and biochemistry of active transport process. A systems level understanding of the axonal transport is critical in identifying novel molecular targets for therapeutic development in treating these disorders.

Acknowledgements

We thank Nancy Norton for helpful comments and funding support from the Alzheimer's Drug Discovery Foundation and Margaret Q. Landenberger Research Foundation.

References

  • 1.Hirokawa N, Niwa S, Tanaka Y. Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron. 2010;68:610–638. doi: 10.1016/j.neuron.2010.09.039. [DOI] [PubMed] [Google Scholar]
  • 2.Chao MV. Retrograde transport redux. Neuron. 2003;39:1–2. doi: 10.1016/s0896-6273(03)00401-x. [DOI] [PubMed] [Google Scholar]
  • 3.Ikenaka K, Katsuno M, Kawai K, Ishigaki S, Tanaka F, Sobue G. Disruption of axonal transport in motor neuron diseases. Int. J. Mol. Sci. 2012;13:1225–1238. doi: 10.3390/ijms13011225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hollenbeck PJ, Saxton WM. The axonal transport of mitochondria. J. Cell Sci. 2005;118:5411–5419. doi: 10.1242/jcs.02745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Caviston JP, Holzbaur EL. Microtubule motors at the intersection of trafficking and transport. Trends Cell Biol. 2006;16:530–537. doi: 10.1016/j.tcb.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 6.Hirokawa N, Takemura R. Molecular motors and mechanisms of directional transport in neurons. Nat. Rev. Neurosci. 2005;6:201–214. doi: 10.1038/nrn1624. [DOI] [PubMed] [Google Scholar]
  • 7.Verhey KJ, Rapoport TA. Kinesin carries the signal. Trends Biochem. Sci. 2001;26:545–550. doi: 10.1016/s0968-0004(01)01931-4. [DOI] [PubMed] [Google Scholar]
  • 8.Gunawardena S, Goldstein LS. Cargo-carrying motor vehicles on the neuronal highway: transport pathways and neurodegenerative disease. J. Neurobiol. 2004;58:258–271. doi: 10.1002/neu.10319. [DOI] [PubMed] [Google Scholar]
  • 9.Kardon JR, Vale RD. Regulators of the cytoplasmic dynein motor. Nat. Rev. Mol. Cell. Biol. 2009;10:854–865. doi: 10.1038/nrm2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kapitein LC, Hoogenraad CC. Which way to go? Cytoskeletal organization and polarized transport in neurons. Mol. Cell. Neurosci. 2011;46:9–20. doi: 10.1016/j.mcn.2010.08.015. [DOI] [PubMed] [Google Scholar]
  • 11.Conde C, Caceres A. Microtubule assembly, organization and dynamics in axons and dendrites. Nat. Rev. Neurosci. 2009;10:319–332. doi: 10.1038/nrn2631. [DOI] [PubMed] [Google Scholar]
  • 12.Signor D, Scholey JM. Microtubule-based transport along axons, dendrites and axonemes. Essays Biochem. 2000;35:89–102. doi: 10.1042/bse0350089. [DOI] [PubMed] [Google Scholar]
  • 13.Black MM, Baas PW. The basis of polarity in neurons. Trends Neurosci. 1989;12:211–214. doi: 10.1016/0166-2236(89)90124-0. [DOI] [PubMed] [Google Scholar]
  • 14.Zheng Y, Wong ML, Alberts B, Mitchison T. Nucleation of microtubule assembly by a gamma-tubulin-containing ring complex. Nature. 1995;378:578–583. doi: 10.1038/378578a0. [DOI] [PubMed] [Google Scholar]
  • 15.Schuyler SC, Pellman D. Microtubule “plus-end-tracking proteins”: The end is just the beginning. Cell. 2001;105:421–424. doi: 10.1016/s0092-8674(01)00364-6. [DOI] [PubMed] [Google Scholar]
  • 16.Galjart N. Plus-end-tracking proteins and their interactions at microtubule ends. Curr. Biol. 2010;20:R528–537. doi: 10.1016/j.cub.2010.05.022. [DOI] [PubMed] [Google Scholar]
  • 17.Dehmelt L, Halpain S. The MAP2/Tau family of microtubule-associated proteins. Genome Biol. 2005;6:204. doi: 10.1186/gb-2004-6-1-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Halpain S, Dehmelt L. The MAP1 family of microtubule-associated proteins. Genome Biol. 2006;7:224. doi: 10.1186/gb-2006-7-6-224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Caceres A, Kosik KS. Inhibition of neurite polarity by tau antisense oligonucleotides in primary cerebellar neurons. Nature. 1990;343:461–463. doi: 10.1038/343461a0. [DOI] [PubMed] [Google Scholar]
  • 20.Caceres A, Mautino J, Kosik KS. Suppression of MAP2 in cultured cerebellar macroneurons inhibits minor neurite formation. Neuron. 1992;9:607–618. doi: 10.1016/0896-6273(92)90025-9. [DOI] [PubMed] [Google Scholar]
  • 21.Harada A, Oguchi K, Okabe S, Kuno J, Terada S, Ohshima T, et al. Altered microtubule organization in small-calibre axons of mice lacking tau protein. Nature. 1994;369:488–491. doi: 10.1038/369488a0. [DOI] [PubMed] [Google Scholar]
  • 22.Hirokawa N. Kinesin and dynein superfamily proteins and the mechanism of organelle transport. Science. 1998;279:519–526. doi: 10.1126/science.279.5350.519. [DOI] [PubMed] [Google Scholar]
  • 23.Brady ST. A novel brain ATPase with properties expected for the fast axonal transport motor. Nature. 1985;317:73–75. doi: 10.1038/317073a0. [DOI] [PubMed] [Google Scholar]
  • 24.Vale RD, Reese TS, Sheetz MP. Identification of a novel force-generating protein, kinesin, involved in microtubule-based motility. Cell. 1985;42:39–50. doi: 10.1016/s0092-8674(85)80099-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Aizawa H, Sekine Y, Takemura R, Zhang Z, Nangaku M, Hirokawa N. Kinesin family in murine central nervous system. J. Cell Biol. 1992;119:1287–1296. doi: 10.1083/jcb.119.5.1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lawrence CJ, Dawe RK, Christie KR, Cleveland DW, Dawson SC, Endow SA, et al. A standardized kinesin nomenclature. J. Cell Biol. 2004;167:19–22. doi: 10.1083/jcb.200408113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Brady ST. Molecular motors in the nervous system. Neuron. 1991;7:521–533. doi: 10.1016/0896-6273(91)90365-7. [DOI] [PubMed] [Google Scholar]
  • 28.Goldstein LS, Yang Z. Microtubule-based transport systems in neurons: the roles of kinesins and dyneins. Annu. Rev. Neurosci. 2000;23:39–71. doi: 10.1146/annurev.neuro.23.1.39. [DOI] [PubMed] [Google Scholar]
  • 29.Goldstein LS. Molecular motors: from one motor many tails to one motor many tales. Trends Cell Biol. 2001;11:477–482. doi: 10.1016/s0962-8924(01)02143-2. [DOI] [PubMed] [Google Scholar]
  • 30.Goldstein LS. Kinesin molecular motors: transport pathways, receptors, and human disease. Proc. Natl. Acad. Sci. USA. 2001;98:6999–7003. doi: 10.1073/pnas.111145298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hirokawa N. mRNA transport in dendrites: RNA granules, motors, and tracks. J. Neurosci. 2006;26:7139–7142. doi: 10.1523/JNEUROSCI.1821-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Goldstein AY, Wang X, Schwarz TL. Axonal transport and the delivery of pre-synaptic components. Curr. Opin. Neurobiol. 2008;18:495–503. doi: 10.1016/j.conb.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Puthanveettil SV, Monje FJ, Miniaci MC, Choi YB, Karl KA, Khandros E, et al. A new component in synaptic plasticity: upregulation of kinesin in the neurons of the gill-withdrawal reflex. Cell. 2008;135:960–973. doi: 10.1016/j.cell.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gibbons IR, Rowe AJ. Dynein: a protein with adenosine triphosphatase activity from cilia. Science. 1965;149:424–426. doi: 10.1126/science.149.3682.424. [DOI] [PubMed] [Google Scholar]
  • 35.Burns RG, Pollard TD. A dynein-like protein from brain. FEBS Lett. 1974;40:274–280. doi: 10.1016/0014-5793(74)80243-7. [DOI] [PubMed] [Google Scholar]
  • 36.Vallee RB, Shpetner HS, Paschal BM. The role of dynein in retrograde axonal transport. Trends Neurosci. 1989;12:66–70. doi: 10.1016/0166-2236(89)90138-0. [DOI] [PubMed] [Google Scholar]
  • 37.Vale RD. The molecular motor toolbox for intracellular transport. Cell. 2003;112:467–480. doi: 10.1016/s0092-8674(03)00111-9. [DOI] [PubMed] [Google Scholar]
  • 38.McGrath JL. Dynein motility: four heads are better than two. Curr. Biol. 2005;15:R970–972. doi: 10.1016/j.cub.2005.11.021. [DOI] [PubMed] [Google Scholar]
  • 39.King SJ, Schroer TA. Dynactin increases the processivity of the cytoplasmic dynein motor. Nat. Cell Biol. 2000;2:20–24. doi: 10.1038/71338. [DOI] [PubMed] [Google Scholar]
  • 40.Susalka SJ, Pfister KK. Cytoplasmic dynein subunit heterogeneity: implications for axonal transport. J. Neurocytol. 2000;29:819–829. doi: 10.1023/a:1010995408343. [DOI] [PubMed] [Google Scholar]
  • 41.Vallee RB, Williams JC, Varma D, Barnhart LE. Dynein: An ancient motor protein involved in multiple modes of transport. J. Neurobiol. 2004;58:189–200. doi: 10.1002/neu.10314. [DOI] [PubMed] [Google Scholar]
  • 42.Fifkova E, Delay RJ. Cytoplasmic actin in neuronal processes as a possible mediator of synaptic plasticity. J. Cell Biol. 1982;95:345–350. doi: 10.1083/jcb.95.1.345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Luo L. Actin cytoskeleton regulation in neuronal morphogenesis and structural plasticity. Annu. Rev. Cell. Dev. Biol. 2002;18:601–635. doi: 10.1146/annurev.cellbio.18.031802.150501. [DOI] [PubMed] [Google Scholar]
  • 44.Hotulainen P, Hoogenraad CC. Actin in dendritic spines: connecting dynamics to function. J. Cell Biol. 2010;189:619–629. doi: 10.1083/jcb.201003008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.dos Remedios CG, Chhabra D, Kekic M, Dedova IV, Tsubakihara M, Berry DA, et al. Actin binding proteins: regulation of cytoskeletal microfilaments. Physiol. Rev. 2003;83:433–473. doi: 10.1152/physrev.00026.2002. [DOI] [PubMed] [Google Scholar]
  • 46.Puszkin S, Berl S, Puszkin E, Clarke DD. Actomyosin-like protein isolated from mammalian brain. Science. 1968;161:170–171. doi: 10.1126/science.161.3837.170. [DOI] [PubMed] [Google Scholar]
  • 47.Puszkin S, Nicklas WJ, Berl S. Actomyosin-like protein in brain: subcellular distribution. J. Neurochem. 1972;19:1319–1333. doi: 10.1111/j.1471-4159.1972.tb01457.x. [DOI] [PubMed] [Google Scholar]
  • 48.Bridgman PC. Myosin-dependent transport in neurons. J. Neurobiol. 2004;58:164–174. doi: 10.1002/neu.10320. [DOI] [PubMed] [Google Scholar]
  • 49.Bridgman PC, Elkin LL. Axonal myosins. J. Neurocytol. 2000;29:831–841. doi: 10.1023/a:1010947525181. [DOI] [PubMed] [Google Scholar]
  • 50.Sellers JR. Myosins: a diverse superfamily. Biochim. Biophys. Acta. 2000;1496:3–22. doi: 10.1016/s0167-4889(00)00005-7. [DOI] [PubMed] [Google Scholar]
  • 51.Titus MA. Myosins. Curr. Opin. Cell Biol. 1993;5:77–81. doi: 10.1016/s0955-0674(05)80011-0. [DOI] [PubMed] [Google Scholar]
  • 52.Foth BJ, Goedecke MC, Soldati D. New insights into myosin evolution and classification. Proc. Natl. Acad. Sci. USA. 2006;103:3681–3686. doi: 10.1073/pnas.0506307103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dunn BD, Sakamoto T, Hong MS, Sellers JR, Takizawa PA. Myo4p is a monomeric myosin with motility uniquely adapted to transport mRNA. J. Cell Biol. 2007;178:1193–1206. doi: 10.1083/jcb.200707080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Harrington WF, Burke M. Geometry of the myosin dimer in high-salt media. I. Association behavior of rod segments from myosin. Biochemistry. 1972;11:1448–1455. doi: 10.1021/bi00758a019. [DOI] [PubMed] [Google Scholar]
  • 55.Saitoh T, Takemura S, Ueda K, Hosoya H, Nagayama M, Haga H, et al. Differential localization of non-muscle myosin II isoforms and phosphorylated regulatory light chains in human MRC-5 fibroblasts. FEBS Lett. 2001;509:365–369. doi: 10.1016/s0014-5793(01)03186-6. [DOI] [PubMed] [Google Scholar]
  • 56.Vibert P, Cohen C. Domains, motions and regulation in the myosin head. J. Muscle Res. Cell. Motil. 1988;9:296–305. doi: 10.1007/BF01773873. [DOI] [PubMed] [Google Scholar]
  • 57.Krendel M, Mooseker MS. Myosins: tails (and heads) of functional diversity. Physiology (Bethesda) 2005;20:239–251. doi: 10.1152/physiol.00014.2005. [DOI] [PubMed] [Google Scholar]
  • 58.Syamaladevi DP, Spudich JA, Sowdhamini R. Structural and functional insights on the Myosin superfamily. Bioinform. Biol. Insights. 2012;6:11–21. doi: 10.4137/BBI.S8451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Z, Edwards JG, Riley N, Provance DW, Jr., Karcher R, Li XD, et al. Myosin Vb mobilizes recycling endosomes and AMPA receptors for postsynaptic plasticity. Cell. 2008;135:535–548. doi: 10.1016/j.cell.2008.09.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wagner W, Brenowitz SD, Hammer JA., 3rd Myosin-Va transports the endoplasmic reticulum into the dendritic spines of Purkinje neurons. Nat. Cell Biol. 2011;13:40–48. doi: 10.1038/ncb2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Phichith D, Travaglia M, Yang Z, Liu X, Zong AB, Safer D, et al. Cargo binding induces dimerization of myosin VI. Proc. Natl. Acad. Sci. USA. 2009;106:17320–17324. doi: 10.1073/pnas.0909748106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Seabrooke S, Qiu X, Stewart BA. Nonmuscle Myosin II helps regulate synaptic vesicle mobility at the Drosophila neuromuscular junction. BMC Neurosci. 2010;11:37. doi: 10.1186/1471-2202-11-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gavin CF, Rubio MD, Young E, Miller C, Rumbaugh G. Myosin II motor activity in the lateral amygdala is required for fear memory consolidation. Learn. Mem. 2011;19:9–14. doi: 10.1101/lm.024042.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Rex CS, Gavin CF, Rubio MD, Kramar EA, Chen LY, Jia Y, et al. Myosin IIb regulates actin dynamics during synaptic plasticity and memory formation. Neuron. 2010;67:603–617. doi: 10.1016/j.neuron.2010.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hu X, Viesselmann C, Nam S, Merriam E, Dent EW. Activity-dependent dynamic microtubule invasion of dendritic spines. J. Neurosci. 2008;28:13094–13105. doi: 10.1523/JNEUROSCI.3074-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Maas C, Belgardt D, Lee HK, Heisler FF, Lappe-Siefke C, Magiera MM, et al. Synaptic activation modifies microtubules underlying transport of postsynaptic cargo. Proc. Natl. Acad. Sci. USA. 2009;106:8731–8736. doi: 10.1073/pnas.0812391106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hoogenraad CC, Bradke F. Control of neuronal polarity and plasticity--a renaissance for microtubules? Trends Cell Biol. 2009;19:669–676. doi: 10.1016/j.tcb.2009.08.006. [DOI] [PubMed] [Google Scholar]
  • 68.Holzbaur EL, Scherer SS. Microtubules, axonal transport, and neuropathy. N. Engl. J. Med. 2011;365:2330–2332. doi: 10.1056/NEJMcibr1112481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cambray-Deakin MA, Burgoyne RD. Posttranslational modifications of alpha-tubulin: acetylated and detyrosinated forms in axons of rat cerebellum. J. Cell Biol. 1987;104:1569–1574. doi: 10.1083/jcb.104.6.1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Audebert S, Koulakoff A, Berwald-Netter Y, Gros F, Denoulet P, Edde B. Developmental regulation of polyglutamylated alpha- and beta-tubulin in mouse brain neurons. J. Cell Sci. 1994;107:2313–2322. doi: 10.1242/jcs.107.8.2313. [DOI] [PubMed] [Google Scholar]
  • 71.Mansfield SG, Gordon-Weeks PR. Dynamic post-translational modification of tubulin in rat cerebral cortical neurons extending neurites in culture: effects of taxol. J. Neurocytol. 1991;20:654–666. doi: 10.1007/BF01187067. [DOI] [PubMed] [Google Scholar]
  • 72.Billingsley ML, Kincaid RL. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. Biochem. J. 1997;323:577–591. doi: 10.1042/bj3230577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Perdiz D, Mackeh R, Pous C, Baillet A. The ins and outs of tubulin acetylation: more than just a post-translational modification? Cell. Signal. 2011;23:763–771. doi: 10.1016/j.cellsig.2010.10.014. [DOI] [PubMed] [Google Scholar]
  • 74.Janke C, Kneussel M. Tubulin post-translational modifications: encoding functions on the neuronal microtubule cytoskeleton. Trends Neurosci. 2010;33:362–372. doi: 10.1016/j.tins.2010.05.001. [DOI] [PubMed] [Google Scholar]
  • 75.Fukushima N, Furuta D, Hidaka Y, Moriyama R, Tsujiuchi T. Post-translational modifications of tubulin in the nervous system. J. Neurochem. 2009;109:683–693. doi: 10.1111/j.1471-4159.2009.06013.x. [DOI] [PubMed] [Google Scholar]
  • 76.Reed NA, Cai D, Blasius TL, Jih GT, Meyhofer E, Gaertig J, et al. Microtubule acetylation promotes kinesin-1 binding and transport. Curr. Biol. 2006;16:2166–2172. doi: 10.1016/j.cub.2006.09.014. [DOI] [PubMed] [Google Scholar]
  • 77.Westermann S, Weber K. Post-translational modifications regulate microtubule function. Nat. Rev. Mol. Cell Biol. 2003;4:938–947. doi: 10.1038/nrm1260. [DOI] [PubMed] [Google Scholar]
  • 78.Konishi Y, Setou M. Tubulin tyrosination navigates the kinesin-1 motor domain to axons. Nat. Neurosci. 2009;12:559–567. doi: 10.1038/nn.2314. [DOI] [PubMed] [Google Scholar]
  • 79.Bettencourt da Cruz A, Schwarzel M, Schulze S, Niyyati M, Heisenberg M, Kretzschmar D. Disruption of the MAP1B-related protein FUTSCH leads to changes in the neuronal cytoskeleton, axonal transport defects, and progressive neurodegeneration in Drosophila. Mol. Biol. Cell. 2005;16:2433–2442. doi: 10.1091/mbc.E04-11-1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fischer M, Kaech S, Knutti D, Matus A. Rapid actin-based plasticity in dendritic spines. Neuron. 1998;20:847–854. doi: 10.1016/s0896-6273(00)80467-5. [DOI] [PubMed] [Google Scholar]
  • 81.Matsuzaki M, Honkura N, Ellis-Davies GC, Kasai H. Structural basis of long-term potentiation in single dendritic spines. Nature. 2004;429:761–766. doi: 10.1038/nature02617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Luo L, Hensch TK, Ackerman L, Barbel S, Jan LY, Jan YN. Differential effects of the Rac GTPase on Purkinje cell axons and dendritic trunks and spines. Nature. 1996;379:837–840. doi: 10.1038/379837a0. [DOI] [PubMed] [Google Scholar]
  • 83.McIlvain JM, Jr., Burkhardt JK, Hamm-Alvarez S, Argon Y, Sheetz MP. Regulation of kinesin activity by phosphorylation of kinesin-associated proteins. J. Biol. Chem. 1994;269:19176–19182. [PubMed] [Google Scholar]
  • 84.Lindesmith L, McIlvain JM, Jr., Argon Y, Sheetz MP. Phosphotransferases associated with the regulation of kinesin motor activity. J. Biol. Chem. 1997;272:22929–22933. doi: 10.1074/jbc.272.36.22929. [DOI] [PubMed] [Google Scholar]
  • 85.Sheetz MP. Motor and cargo interactions. Eur. J. Biochem. 1999;262:19–25. doi: 10.1046/j.1432-1327.1999.00340.x. [DOI] [PubMed] [Google Scholar]
  • 86.Morfini G, Pigino G, Szebenyi G, You Y, Pollema S, Brady ST. JNK mediates pathogenic effects of polyglutamine-expanded androgen receptor on fast axonal transport. Nat. Neurosci. 2006;9:907–916. doi: 10.1038/nn1717. [DOI] [PubMed] [Google Scholar]
  • 87.Stagi M, Gorlovoy P, Larionov S, Takahashi K, Neumann H. Unloading kinesin transported cargoes from the tubulin track via the inflammatory c-Jun N-terminal kinase pathway. FASEB J. 2006;20:2573–2575. doi: 10.1096/fj.06-6679fje. [DOI] [PubMed] [Google Scholar]
  • 88.Koushika SP. “JIP”ing along the axon: the complex roles of JIPs in axonal transport. Bioessays. 2008;30:10–14. doi: 10.1002/bies.20695. [DOI] [PubMed] [Google Scholar]
  • 89.Blasius TL, Cai D, Jih GT, Toret CP, Verhey KJ. Two binding partners cooperate to activate the molecular motor Kinesin-1. J. Cell Biol. 2007;176:11–17. doi: 10.1083/jcb.200605099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Horiuchi D, Collins CA, Bhat P, Barkus RV, Diantonio A, Saxton WM. Control of a kinesin-cargo linkage mechanism by JNK pathway kinases. Curr. Biol. 2007;17:1313–1317. doi: 10.1016/j.cub.2007.06.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chang L, Jones Y, Ellisman MH, Goldstein LS, Karin M. JNK1 is required for maintenance of neuronal microtubules and controls phosphorylation of microtubule-associated proteins. Dev. Cell. 2003;4:521–533. doi: 10.1016/s1534-5807(03)00094-7. [DOI] [PubMed] [Google Scholar]
  • 92.Reynolds CH, Utton MA, Gibb GM, Yates A, Anderton BH. Stress-activated protein kinase/c-jun N-terminal kinase phosphorylates tau protein. J. Neurochem. 1997;68:1736–1744. doi: 10.1046/j.1471-4159.1997.68041736.x. [DOI] [PubMed] [Google Scholar]
  • 93.Tararuk T, Ostman N, Li W, Bjorkblom B, Padzik A, Zdrojewska J, et al. JNK1 phosphorylation of SCG10 determines microtubule dynamics and axodendritic length. J. Cell Biol. 2006;173:265–277. doi: 10.1083/jcb.200511055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Colin E, Zala D, Liot G, Rangone H, Borrell-Pages M, Li XJ, et al. Huntingtin phosphorylation acts as a molecular switch for anterograde/retrograde transport in neurons. EMBO J. 2008;27:2124–2134. doi: 10.1038/emboj.2008.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schaefer AW, Schoonderwoert VT, Ji L, Mederios N, Danuser G, Forscher P. Coordination of actin filament and microtubule dynamics during neurite outgrowth. Dev. Cell. 2008;15:146–162. doi: 10.1016/j.devcel.2008.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Burnette DT, Ji L, Schaefer AW, Medeiros NA, Danuser G, Forscher P. Myosin II activity facilitates microtubule bundling in the neuronal growth cone neck. Dev. Cell. 2008;15:163–169. doi: 10.1016/j.devcel.2008.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Arimura N, Kaibuchi K. Neuronal polarity: from extracellular signals to intracellular mechanisms. Nat. Rev. Neurosci. 2007;8:194–205. doi: 10.1038/nrn2056. [DOI] [PubMed] [Google Scholar]
  • 98.Dent EW, Gertler FB. Cytoskeletal dynamics and transport in growth cone motility and axon guidance. Neuron. 2003;40:209–227. doi: 10.1016/s0896-6273(03)00633-0. [DOI] [PubMed] [Google Scholar]
  • 99.Mufson EJ, Kroin JS, Sendera TJ, Sobreviela T. Distribution and retrograde transport of trophic factors in the central nervous system: functional implications for the treatment of neurodegenerative diseases. Prog. Neurobiol. 1999;57:451–484. doi: 10.1016/s0301-0082(98)00059-8. [DOI] [PubMed] [Google Scholar]
  • 100.Hirokawa N, Noda Y. Intracellular transport and kinesin superfamily proteins, KIFs: structure, function, and dynamics. Physiol. Rev. 2008;88:1089–1118. doi: 10.1152/physrev.00023.2007. [DOI] [PubMed] [Google Scholar]
  • 101.Kandel ER. The molecular biology of memory storage: a dialogue between genes and synapses. Science. 2001;294:1030–1038. doi: 10.1126/science.1067020. [DOI] [PubMed] [Google Scholar]
  • 102.Kiebler MA, DesGroseillers L. Molecular insights into mRNA transport and local translation in the mammalian nervous system. Neuron. 2000;25:19–28. doi: 10.1016/s0896-6273(00)80868-5. [DOI] [PubMed] [Google Scholar]
  • 103.Martin KC, Casadio A, Zhu H, Yaping E, Rose JC, Chen M, et al. Synapse-specific, long-term facilitation of aplysia sensory to motor synapses: a function for local protein synthesis in memory storage. Cell. 1997;91:927–938. doi: 10.1016/s0092-8674(00)80484-5. [DOI] [PubMed] [Google Scholar]
  • 104.Si K, Giustetto M, Etkin A, Hsu R, Janisiewicz AM, Miniaci MC, et al. A neuronal isoform of CPEB regulates local protein synthesis and stabilizes synapse-specific long-term facilitation in aplysia. Cell. 2003;115:893–904. doi: 10.1016/s0092-8674(03)01021-3. [DOI] [PubMed] [Google Scholar]
  • 105.Martin KC, Ephrussi A. mRNA localization: gene expression in the spatial dimension. Cell. 2009;136:719–730. doi: 10.1016/j.cell.2009.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Tubing F, Vendra G, Mikl M, Macchi P, Thomas S, Kiebler MA. Dendritically localized transcripts are sorted into distinct ribonucleoprotein particles that display fast directional motility along dendrites of hippocampal neurons. J. Neurosci. 2010;30:4160–4170. doi: 10.1523/JNEUROSCI.3537-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lyles V, Zhao Y, Martin KC. Synapse formation and mRNA localization in cultured Aplysia neurons. Neuron. 2006;49:349–356. doi: 10.1016/j.neuron.2005.12.029. [DOI] [PubMed] [Google Scholar]
  • 108.Raymond CR, Thompson VL, Tate WP, Abraham WC. Metabotropic glutamate receptors trigger homosynaptic protein synthesis to prolong long-term potentiation. J. Neurosci. 2000;20:969–976. doi: 10.1523/JNEUROSCI.20-03-00969.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Miki H, Okada Y, Hirokawa N. Analysis of the kinesin superfamily: insights into structure and function. Trends Cell Biol. 2005;15:467–476. doi: 10.1016/j.tcb.2005.07.006. [DOI] [PubMed] [Google Scholar]
  • 110.Kanai Y, Dohmae N, Hirokawa N. Kinesin transports RNA: isolation and characterization of an RNA-transporting granule. Neuron. 2004;43:513–525. doi: 10.1016/j.neuron.2004.07.022. [DOI] [PubMed] [Google Scholar]
  • 111.De Vos KJ, Grierson AJ, Ackerley S, Miller CC. Role of axonal transport in neurodegenerative diseases. Annu. Rev. Neurosci. 2008;31:151–173. doi: 10.1146/annurev.neuro.31.061307.090711. [DOI] [PubMed] [Google Scholar]
  • 112.Pack-Chung E, Kurshan PT, Dickman DK, Schwarz TL. A Drosophila kinesin required for synaptic bouton formation and synaptic vesicle transport. Nat. Neurosci. 2007;10:980–989. doi: 10.1038/nn1936. [DOI] [PubMed] [Google Scholar]
  • 113.Horiuchi D, Barkus RV, Pilling AD, Gassman A, Saxton WM. APLIP1, a kinesin binding JIP-1/JNK scaffold protein, influences the axonal transport of both vesicles and mitochondria in Drosophila. Curr. Biol. 2005;15:2137–2141. doi: 10.1016/j.cub.2005.10.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Miller KE, DeProto J, Kaufmann N, Patel BN, Duckworth A, Van Vactor D. Direct observation demonstrates that Liprin-alpha is required for trafficking of synaptic vesicles. Curr. Biol. 2005;15:684–689. doi: 10.1016/j.cub.2005.02.061. [DOI] [PubMed] [Google Scholar]
  • 115.Gindhart JG, Chen J, Faulkner M, Gandhi R, Doerner K, Wisniewski T, et al. The kinesin-associated protein UNC-76 is required for axonal transport in the Drosophila nervous system. Mol. Biol. Cell. 2003;14:3356–3365. doi: 10.1091/mbc.E02-12-0800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Glater EE, Megeath LJ, Stowers RS, Schwarz TL. Axonal transport of mitochondria requires milton to recruit kinesin heavy chain and is light chain independent. J. Cell Biol. 2006;173:545–557. doi: 10.1083/jcb.200601067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hafezparast M, Klocke R, Ruhrberg C, Marquardt A, Ahmad-Annuar A, Bowen S, et al. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science. 2003;300:808–812. doi: 10.1126/science.1083129. [DOI] [PubMed] [Google Scholar]
  • 118.Ori-McKenney KM, Xu J, Gross SP, Vallee RB. A cytoplasmic dynein tail mutation impairs motor processivity. Nat. Cell Biol. 2010;12:1228–1234. doi: 10.1038/ncb2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Courchesne SL, Pazyra-Murphy MF, Lee DJ, Segal RA. Neuromuscular junction defects in mice with mutation of dynein heavy chain 1. PLoS One. 2011;6:e16753. doi: 10.1371/journal.pone.0016753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ilieva HS, Yamanaka K, Malkmus S, Kakinohana O, Yaksh T, Marsala M, et al. Mutant dynein (Loa) triggers proprioceptive axon loss that extends survival only in the SOD1 ALS model with highest motor neuron death. Proc. Natl. Acad. Sci. USA. 2008;105:12599–12604. doi: 10.1073/pnas.0805422105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Braunstein KE, Eschbach J, Rona-Voros K, Soylu R, Mikrouli E, Larmet Y, et al. A point mutation in the dynein heavy chain gene leads to striatal atrophy and compromises neurite outgrowth of striatal neurons. Hum. Mol. Genet. 2010;19:4385–4398. doi: 10.1093/hmg/ddq361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Jiang YM, Yamamoto M, Kobayashi Y, Yoshihara T, Liang Y, Terao S, et al. Gene expression profile of spinal motor neurons in sporadic amyotrophic lateral sclerosis. Ann. Neurol. 2005;57:236–251. doi: 10.1002/ana.20379. [DOI] [PubMed] [Google Scholar]
  • 123.Riviere JB, Ramalingam S, Lavastre V, Shekarabi M, Holbert S, Lafontaine J, et al. KIF1A, an axonal transporter of synaptic vesicles, is mutated in hereditary sensory and autonomic neuropathy type 2. Am. J. Hum. Genet. 2011;89:219–230. doi: 10.1016/j.ajhg.2011.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Erlich Y, Edvardson S, Hodges E, Zenvirt S, Thekkat P, Shaag A, et al. Exome sequencing and disease-network analysis of a single family implicate a mutation in KIF1A in hereditary spastic paraparesis. Genome Res. 2011;21:658–664. doi: 10.1101/gr.117143.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Blair MA, Ma S, Hedera P. Mutation in KIF5A can also cause adult-onset hereditary spastic paraplegia. Neurogenetics. 2006;7:47–50. doi: 10.1007/s10048-005-0027-8. [DOI] [PubMed] [Google Scholar]
  • 126.Dafinger C, Liebau MC, Elsayed SM, Hellenbroich Y, Boltshauser E, Korenke GC, et al. Mutations in KIF7 link Joubert syndrome with Sonic Hedgehog signaling and microtubule dynamics. J. Clin. Invest. 2011;121:2662–2667. doi: 10.1172/JCI43639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tarabeux J, Champagne N, Brustein E, Hamdan FF, Gauthier J, Lapointe M, et al. De novo truncating mutation in Kinesin 17 associated with schizophrenia. Biol. Psychiatry. 2010;68:649–656. doi: 10.1016/j.biopsych.2010.04.018. [DOI] [PubMed] [Google Scholar]
  • 128.Lu S, Zhao C, Zhao K, Li N, Larsson C. Novel and recurrent KIF21A mutations in congenital fibrosis of the extraocular muscles type 1 and 3. Arch. Ophthalmol. 2008;126:388–394. doi: 10.1001/archopht.126.3.388. [DOI] [PubMed] [Google Scholar]
  • 129.Khan AO, Khalil DS, Al Sharif LJ, Al-Ghadhfan FE, Al Tassan NA. Germline mosaicism for KIF21A mutation (p.R954L) mimicking recessive inheritance for congenital fibrosis of the extraocular muscles. Ophthalmology. 2010;117:154–158. doi: 10.1016/j.ophtha.2009.06.029. [DOI] [PubMed] [Google Scholar]
  • 130.Zhao C, Takita J, Tanaka Y, Setou M, Nakagawa T, Takeda S, et al. Charcot-Marie-Tooth disease type 2A caused by mutation in a microtubule motor KIF1Bbeta. Cell. 2001;105:587–597. doi: 10.1016/s0092-8674(01)00363-4. [DOI] [PubMed] [Google Scholar]
  • 131.Sharma R, Buras E, Terashima T, Serrano F, Massaad CA, Hu L, et al. Hyperglycemia induces oxidative stress and impairs axonal transport rates in mice. PLoS One. 2010;5:e13463. doi: 10.1371/journal.pone.0013463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Haider L, Fischer MT, Frischer JM, Bauer J, Hoftberger R, Botond G, et al. Oxidative damage in multiple sclerosis lesions. Brain. 2011;134:1914–1924. doi: 10.1093/brain/awr128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wilkinson AE, Bridges LR, Sivaloganathan S. Correlation of survival time with size of axonal swellings in diffuse axonal injury. Acta Neuropathol. 1999;98:197–202. doi: 10.1007/s004010051069. [DOI] [PubMed] [Google Scholar]
  • 134.Roediger B, Armati PJ. Oxidative stress induces axonal beading in cultured human brain tissue. Neurobiol. Dis. 2003;13:222–229. doi: 10.1016/s0969-9961(03)00038-x. [DOI] [PubMed] [Google Scholar]
  • 135.Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J. Cell Biol. 2002;156:1051–1063. doi: 10.1083/jcb.200108057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, et al. Mitochondrial abnormalities in Alzheimer's disease. J. Neurosci. 2001;21:3017–3023. doi: 10.1523/JNEUROSCI.21-09-03017.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Massaad CA, Amin SK, Hu L, Mei Y, Klann E, Pautler RG. Mitochondrial superoxide contributes to blood flow and axonal transport deficits in the Tg2576 mouse model of Alzheimer's disease. PLoS One. 2010;5:e10561. doi: 10.1371/journal.pone.0010561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Shidara Y, Hollenbeck PJ. Defects in mitochondrial axonal transport and membrane potential without increased reactive oxygen species production in a Drosophila model of Friedreich ataxia. J. Neurosci. 2010;30:11369–11378. doi: 10.1523/JNEUROSCI.0529-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Green DR, Reed JC. Mitochondria and apoptosis. Science. 1998;281:1309–1312. doi: 10.1126/science.281.5381.1309. [DOI] [PubMed] [Google Scholar]
  • 140.Magrané J, Manfredi G. Mitochondrial function, morphology, and axonal transport in amyotrophic lateral sclerosis. Antioxid. Redox Signal. 2009;11:1615–1626. doi: 10.1089/ars.2009.2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sasaki S, Iwata M. Impairment of fast axonal transport in the proximal axons of anterior horn neurons in amyotrophic lateral sclerosis. Neurology. 1996;47:535–540. doi: 10.1212/wnl.47.2.535. [DOI] [PubMed] [Google Scholar]
  • 142.Sasaki S, Iwata M. Mitochondrial alterations in the spinal cord of patients with sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2007;66:10–16. doi: 10.1097/nen.0b013e31802c396b. [DOI] [PubMed] [Google Scholar]
  • 143.Yue Z, Wang QJ, Komatsu M. Neuronal autophagy: going the distance to the axon. Autophagy. 2008;4:94–96. doi: 10.4161/auto.5202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Katsumata K, Nishiyama J, Inoue T, Mizushima N, Takeda J, Yuzaki M. Dynein- and activity-dependent retrograde transport of autophagosomes in neuronal axons. Autophagy. 2010;6:378–385. doi: 10.4161/auto.6.3.11262. [DOI] [PubMed] [Google Scholar]
  • 145.Harris H, Rubinsztein DC. Control of autophagy as a therapy for neurodegenerative disease. Nat. Rev. Neurol. 2012;8:108–117. doi: 10.1038/nrneurol.2011.200. [DOI] [PubMed] [Google Scholar]
  • 146.Nixon RA. Autophagy in neurodegenerative disease: friend, foe or turncoat? Trends Neurosci. 2006;29:528–535. doi: 10.1016/j.tins.2006.07.003. [DOI] [PubMed] [Google Scholar]
  • 147.Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH, et al. Macroautophagy - a novel beta-amyloid peptide-generating pathway activated in Alzheimer's disease. J. Cell Biol. 2005;171:87–98. doi: 10.1083/jcb.200505082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J. Cell Sci. 2007;120:4081–4091. doi: 10.1242/jcs.019265. [DOI] [PubMed] [Google Scholar]
  • 149.Chu CT. Tickled PINK1: mitochondrial homeostasis and autophagy in recessive Parkinsonism. Biochim. Biophys. Acta. 2010;1802:20–28. doi: 10.1016/j.bbadis.2009.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sapp E, Schwarz C, Chase K, Bhide PG, Young AB, Penney J, et al. Huntingtin localization in brains of normal and Huntington's disease patients. Ann. Neurol. 1997;42:604–612. doi: 10.1002/ana.410420411. [DOI] [PubMed] [Google Scholar]
  • 151.Martinez-Vicente M, Talloczy Z, Wong E, Tang G, Koga H, Kaushik S, et al. Cargo recognition failure is responsible for inefficient autophagy in Huntington's disease. Nat. Neurosci. 2010;13:567–576. doi: 10.1038/nn.2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Sasaki S. Autophagy in spinal cord motor neurons in sporadic amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2011;70:349–359. doi: 10.1097/NEN.0b013e3182160690. [DOI] [PubMed] [Google Scholar]
  • 153.Ravikumar B, Acevedo-Arozena A, Imarisio S, Berger Z, Vacher C, O'Kane CJ, et al. Dynein mutations impair autophagic clearance of aggregate-prone proteins. Nat. Genet. 2005;37:771–776. doi: 10.1038/ng1591. [DOI] [PubMed] [Google Scholar]
  • 154.Laird FM, Farah MH, Ackerley S, Hoke A, Maragakis N, Rothstein JD, et al. Motor neuron disease occurring in a mutant dynactin mouse model is characterized by defects in vesicular trafficking. J. Neurosci. 2008;28:1997–2005. doi: 10.1523/JNEUROSCI.4231-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Mattson MP. Pathways towards and away from Alzheimer's disease. Nature. 2004;430:631–639. doi: 10.1038/nature02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 2001;24:1121–1159. doi: 10.1146/annurev.neuro.24.1.1121. [DOI] [PubMed] [Google Scholar]
  • 157.Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron. 2011;70:410–426. doi: 10.1016/j.neuron.2011.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wang JZ, Liu F. Microtubule-associated protein tau in development, degeneration and protection of neurons. Prog. Neurobiol. 2008;85:148–175. doi: 10.1016/j.pneurobio.2008.03.002. [DOI] [PubMed] [Google Scholar]
  • 159.Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  • 160.Vossel KA, Zhang K, Brodbeck J, Daub AC, Sharma P, Finkbeiner S, et al. Tau reduction prevents amyloid beta-induced defects in axonal transport. Science. 2010;330:198. doi: 10.1126/science.1194653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ittner LM, Gotz J. Amyloid-beta and tau - a toxic pas de deux in Alzheimer's disease. Nat. Rev. Neurosci. 2011;12:65–72. doi: 10.1038/nrn2967. [DOI] [PubMed] [Google Scholar]
  • 162.Reddy PH. Abnormal tau, mitochondrial dysfunction, impaired axonal transport of mitochondria, and synaptic deprivation in Alzheimer's disease. Brain. Res. 2011;1415:136–148. doi: 10.1016/j.brainres.2011.07.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Spillantini MG, Murrell JR, Goedert M, Farlow MR, Klug A, Ghetti B. Mutation in the tau gene in familial multiple system tauopathy with presenile dementia. Proc. Natl. Acad. Sci. USA. 1998;95:7737–7741. doi: 10.1073/pnas.95.13.7737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Cash AD, Aliev G, Siedlak SL, Nunomura A, Fujioka H, Zhu X, et al. Microtubule reduction in Alzheimer's disease and aging is independent of tau filament formation. Am. J. Pathol. 2003;162:1623–1627. doi: 10.1016/s0002-9440(10)64296-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Khatoon S, Grundke-Iqbal I, Iqbal K. Brain levels of microtubule-associated protein tau are elevated in Alzheimer's disease: a radioimmuno-slot-blot assay for nanograms of the protein. J. Neurochem. 1992;59:750–753. doi: 10.1111/j.1471-4159.1992.tb09432.x. [DOI] [PubMed] [Google Scholar]
  • 166.Ebneth A, Godemann R, Stamer K, Illenberger S, Trinczek B, Mandelkow E. Overexpression of tau protein inhibits kinesin-dependent trafficking of vesicles, mitochondria, and endoplasmic reticulum: implications for Alzheimer's disease. J. Cell Biol. 1998;143:777–794. doi: 10.1083/jcb.143.3.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ishihara T, Hong M, Zhang B, Nakagawa Y, Lee MK, Trojanowski JQ, et al. Age-dependent emergence and progression of a tauopathy in transgenic mice overexpressing the shortest human tau isoform. Neuron. 1999;24:751–762. doi: 10.1016/s0896-6273(00)81127-7. [DOI] [PubMed] [Google Scholar]
  • 168.Ittner LM, Fath T, Ke YD, Bi M, van Eersel J, Li KM, et al. Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia. Proc. Natl. Acad. Sci. USA. 2008;105:15997–16002. doi: 10.1073/pnas.0808084105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Probst A, Gotz J, Wiederhold KH, Tolnay M, Mistl C, Jaton AL, et al. Axonopathy and amyotrophy in mice transgenic for human four-repeat tau protein. Acta Neuropathol. 2000;99:469–481. doi: 10.1007/s004010051148. [DOI] [PubMed] [Google Scholar]
  • 170.Spittaels K, Van den Haute C, Van Dorpe J, Bruynseels K, Vandezande K, Laenen I, et al. Prominent axonopathy in the brain and spinal cord of transgenic mice overexpressing four-repeat human tau protein. Am. J. Pathol. 1999;155:2153–2165. doi: 10.1016/S0002-9440(10)65533-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Cote F, Collard JF, Julien JP. Progressive neuronopathy in transgenic mice expressing the human neurofilament heavy gene: a mouse model of amyotrophic lateral sclerosis. Cell. 1993;73:35–46. doi: 10.1016/0092-8674(93)90158-m. [DOI] [PubMed] [Google Scholar]
  • 172.Xu Z, Cork LC, Griffin JW, Cleveland DW. Increased expression of neurofilament subunit NF-L produces morphological alterations that resemble the pathology of human motor neuron disease. Cell. 1993;73:23–33. doi: 10.1016/0092-8674(93)90157-l. [DOI] [PubMed] [Google Scholar]
  • 173.Zhang B, Higuchi M, Yoshiyama Y, Ishihara T, Forman MS, Martinez D, et al. Retarded axonal transport of R406W mutant tau in transgenic mice with a neurodegenerative tauopathy. J. Neurosci. 2004;24:4657–4667. doi: 10.1523/JNEUROSCI.0797-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Higuchi M, Zhang B, Forman MS, Yoshiyama Y, Trojanowski JQ, Lee VM. Axonal degeneration induced by targeted expression of mutant human tau in oligodendrocytes of transgenic mice that model glial tauopathies. J. Neurosci. 2005;25:9434–9443. doi: 10.1523/JNEUROSCI.2691-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Bull ND, Guidi A, Goedert M, Martin KR, Spillantini MG. Reduced axonal transport and increased excitotoxic retinal ganglion cell degeneration in mice transgenic for human mutant P301S tau. PLoS One. 2012;7:e34724. doi: 10.1371/journal.pone.0034724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Dixit R, Ross JL, Goldman YE, Holzbaur EL. Differential regulation of dynein and kinesin motor proteins by tau. Science. 2008;319:1086–1089. doi: 10.1126/science.1152993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Falzone TL, Stokin GB, Lillo C, Rodrigues EM, Westerman EL, Williams DS, et al. Axonal stress kinase activation and tau misbehavior induced by kinesin-1 transport defects. J. Neurosci. 2009;29:5758–5767. doi: 10.1523/JNEUROSCI.0780-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–481. doi: 10.1126/science.1113694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Lewis J, Dickson DW, Lin WL, Chisholm L, Corral A, Jones G, et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science. 2001;293:1487–1491. doi: 10.1126/science.1058189. [DOI] [PubMed] [Google Scholar]
  • 180.Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science. 2007;316:750–754. doi: 10.1126/science.1141736. [DOI] [PubMed] [Google Scholar]
  • 181.Falzone TL, Gunawardena S, McCleary D, Reis GF, Goldstein LS. Kinesin-1 transport reductions enhance human tau hyperphosphorylation, aggregation and neurodegeneration in animal models of tauopathies. Hum. Mol. Genet. 2010;19:4399–4408. doi: 10.1093/hmg/ddq363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Yuan A, Kumar A, Peterhoff C, Duff K, Nixon RA. Axonal transport rates in vivo are unaffected by tau deletion or overexpression in mice. J. Neurosci. 2008;28:1682–1687. doi: 10.1523/JNEUROSCI.5242-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Kamal A, Stokin GB, Yang Z, Xia CH, Goldstein LS. Axonal transport of amyloid precursor protein is mediated by direct binding to the kinesin light chain subunit of kinesin-I. Neuron. 2000;28:449–459. doi: 10.1016/s0896-6273(00)00124-0. [DOI] [PubMed] [Google Scholar]
  • 184.Stokin GB, Lillo C, Falzone TL, Brusch RG, Rockenstein E, Mount SL, et al. Axonopathy and transport deficits early in the pathogenesis of Alzheimer's disease. Science. 2005;307:1282–1288. doi: 10.1126/science.1105681. [DOI] [PubMed] [Google Scholar]
  • 185.Wirths O, Weis J, Szczygielski J, Multhaup G, Bayer TA. Axonopathy in an APP/PS1 transgenic mouse model of Alzheimer's disease. Acta Neuropathol. 2006;111:312–319. doi: 10.1007/s00401-006-0041-4. [DOI] [PubMed] [Google Scholar]
  • 186.Hiruma H, Katakura T, Takahashi S, Ichikawa T, Kawakami T. Glutamate and amyloid beta-protein rapidly inhibit fast axonal transport in cultured rat hippocampal neurons by different mechanisms. J. Neurosci. 2003;23:8967–8977. doi: 10.1523/JNEUROSCI.23-26-08967.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Rui Y, Tiwari P, Xie Z, Zheng JQ. Acute impairment of mitochondrial trafficking by beta-amyloid peptides in hippocampal neurons. J. Neurosci. 2006;26:10480–10487. doi: 10.1523/JNEUROSCI.3231-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Decker H, Lo KY, Unger SM, Ferreira ST, Silverman MA. Amyloid-beta peptide oligomers disrupt axonal transport through an NMDA receptor-dependent mechanism that is mediated by glycogen synthase kinase 3beta in primary cultured hippocampal neurons. J. Neurosci. 2010;30:9166–9171. doi: 10.1523/JNEUROSCI.1074-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Pigino G, Morfini G, Atagi Y, Deshpande A, Yu C, Jungbauer L, et al. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal amyloid beta. Proc. Natl. Acad. Sci. USA. 2009;106:5907–5912. doi: 10.1073/pnas.0901229106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Pigino G, Morfini G, Pelsman A, Mattson MP, Brady ST, Busciglio J. Alzheimer's presenilin 1 mutations impair kinesin-based axonal transport. J. Neurosci. 2003;23:4499–4508. doi: 10.1523/JNEUROSCI.23-11-04499.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Lazarov O, Morfini GA, Pigino G, Gadadhar A, Chen X, Robinson J, et al. Impairments in fast axonal transport and motor neuron deficits in transgenic mice expressing familial Alzheimer's disease-linked mutant presenilin 1. J. Neurosci. 2007;27:7011–7020. doi: 10.1523/JNEUROSCI.4272-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Cai D, Leem JY, Greenfield JP, Wang P, Kim BS, Wang R, et al. Presenilin-1 regulates intracellular trafficking and cell surface delivery of beta-amyloid precursor protein. J. Biol. Chem. 2003;278:3446–3454. doi: 10.1074/jbc.M209065200. [DOI] [PubMed] [Google Scholar]
  • 193.Tesseur I, Van Dorpe J, Bruynseels K, Bronfman F, Sciot R, Van Lommel A, et al. Prominent axonopathy and disruption of axonal transport in transgenic mice expressing human apolipoprotein E4 in neurons of brain and spinal cord. Am. J. Pathol. 2000;157:1495–1510. doi: 10.1016/S0002-9440(10)64788-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Haberland C. Frontotemporal dementia or frontotemporal lobar degeneration -overview of a group of proteinopathies. Ideggyogy Sz. 2010;63:87–93. [PubMed] [Google Scholar]
  • 195.Fujioka S, Wszolek ZK. Clinical aspects of familial forms of frontotemporal dementia associated with parkinsonism. J. Mol. Neurosci. 2011;45:359–365. doi: 10.1007/s12031-011-9568-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Ghazi-Noori S, Froud KE, Mizielinska S, Powell C, Smidak M, Fernandez de Marco M, et al. Progressive neuronal inclusion formation and axonal degeneration in CHMP2B mutant transgenic mice. Brain. 2012;135:819–832. doi: 10.1093/brain/aws006. [DOI] [PubMed] [Google Scholar]
  • 197.Urwin H, Authier A, Nielsen JE, Metcalf D, Powell C, Froud K, et al. Disruption of endocytic trafficking in frontotemporal dementia with CHMP2B mutations. Hum. Mol. Genet. 2010;19:2228–2238. doi: 10.1093/hmg/ddq100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ittner LM, Ke YD, Gotz J. Phosphorylated Tau interacts with c-Jun N-terminal kinase-interacting protein 1 (JIP1) in Alzheimer disease. J. Biol. Chem. 2009;284:20909–20916. doi: 10.1074/jbc.M109.014472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Magnani E, Fan J, Gasparini L, Golding M, Williams M, Schiavo G, et al. Interaction of tau protein with the dynactin complex. EMBO J. 2007;26:4546–4554. doi: 10.1038/sj.emboj.7601878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hong M, Zhukareva V, Vogelsberg-Ragaglia V, Wszolek Z, Reed L, Miller BI, et al. Mutation-specific functional impairments in distinct tau isoforms of hereditary FTDP-17. Science. 1998;282:1914–1917. doi: 10.1126/science.282.5395.1914. [DOI] [PubMed] [Google Scholar]
  • 201.Stoothoff W, Jones PB, Spires-Jones TL, Joyner D, Chhabra E, Bercury K, et al. Differential effect of three-repeat and four-repeat tau on mitochondrial axonal transport. J. Neurochem. 2009;111:417–427. doi: 10.1111/j.1471-4159.2009.06316.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Tien NW, Wu GH, Hsu CC, Chang CY, Wagner OI. Tau/PTL-1 associates with kinesin-3 KIF1A/UNC-104 and affects the motor's motility characteristics in C. elegans neurons. Neurobiol. Dis. 2011;43:495–506. doi: 10.1016/j.nbd.2011.04.023. [DOI] [PubMed] [Google Scholar]
  • 203.Gilley J, Seereeram A, Ando K, Mosely S, Andrews S, Kerschensteiner M, et al. Age-dependent axonal transport and locomotor changes and tau hypophosphorylation in a “P301L” tau knockin mouse. Neurobiol. Aging. 2012;33:621.e1–621.e15. doi: 10.1016/j.neurobiolaging.2011.02.014. [DOI] [PubMed] [Google Scholar]
  • 204.Mulder DW. Clinical limits of amyotrophic lateral sclerosis. Adv. Neurol. 1982;36:15–22. [PubMed] [Google Scholar]
  • 205.Rowland LP, Shneider NA. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2001;344:1688–1700. doi: 10.1056/NEJM200105313442207. [DOI] [PubMed] [Google Scholar]
  • 206.Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62. doi: 10.1038/362059a0. [DOI] [PubMed] [Google Scholar]
  • 207.Zhang B, Tu P, Abtahian F, Trojanowski JQ, Lee VM. Neurofilaments and orthograde transport are reduced in ventral root axons of transgenic mice that express human SOD1 with a G93A mutation. J. Cell Biol. 1997;139:1307–1315. doi: 10.1083/jcb.139.5.1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Williamson TL, Cleveland DW. Slowing of axonal transport is a very early event in the toxicity of ALS-linked SOD1 mutants to motor neurons. Nat. Neurosci. 1999;2:50–56. doi: 10.1038/4553. [DOI] [PubMed] [Google Scholar]
  • 209.Borchelt DR, Wong PC, Becher MW, Pardo CA, Lee MK, Xu ZS, et al. Axonal transport of mutant superoxide dismutase 1 and focal axonal abnormalities in the proximal axons of transgenic mice. Neurobiol. Dis. 1998;5:27–35. doi: 10.1006/nbdi.1998.0178. [DOI] [PubMed] [Google Scholar]
  • 210.Tateno M, Kato S, Sakurai T, Nukina N, Takahashi R, Araki T. Mutant SOD1 impairs axonal transport of choline acetyltransferase and acetylcholine release by sequestering KAP3. Hum. Mol. Genet. 2009;18:942–955. doi: 10.1093/hmg/ddn422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Landers JE, Melki J, Meininger V, Glass JD, van den Berg LH, van Es MA, et al. Reduced expression of the Kinesin-Associated Protein 3 (KIFAP3) gene increases survival in sporadic amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA. 2009;106:9004–9009. doi: 10.1073/pnas.0812937106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Dupuis L, de Tapia M, Rene F, Lutz-Bucher B, Gordon JW, Mercken L, et al. Differential screening of mutated SOD1 transgenic mice reveals early up-regulation of a fast axonal transport component in spinal cord motor neurons. Neurobiol. Dis. 2000;7:274–285. doi: 10.1006/nbdi.2000.0292. [DOI] [PubMed] [Google Scholar]
  • 213.Murakami T, Nagano I, Hayashi T, Manabe Y, Shoji M, Setoguchi Y, et al. Impaired retrograde axonal transport of adenovirus-mediated E. coli LacZ gene in the mice carrying mutant SOD1 gene. Neurosci. Lett. 2001;308:149–152. doi: 10.1016/s0304-3940(01)02036-5. [DOI] [PubMed] [Google Scholar]
  • 214.Ligon LA, LaMonte BH, Wallace KE, Weber N, Kalb RG, Holzbaur EL. Mutant superoxide dismutase disrupts cytoplasmic dynein in motor neurons. Neuroreport. 2005;16:533–536. doi: 10.1097/00001756-200504250-00002. [DOI] [PubMed] [Google Scholar]
  • 215.Bilsland LG, Sahai E, Kelly G, Golding M, Greensmith L, Schiavo G. Deficits in axonal transport precede ALS symptoms in vivo. Proc. Natl. Acad. Sci. USA. 2010;107:20523–20528. doi: 10.1073/pnas.1006869107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Munch C, Sedlmeier R, Meyer T, Homberg V, Sperfeld AD, Kurt A, et al. Point mutations of the p150 subunit of dynactin (DCTN1) gene in ALS. Neurology. 2004;63:724–726. doi: 10.1212/01.wnl.0000134608.83927.b1. [DOI] [PubMed] [Google Scholar]
  • 217.Moore JK, Sept D, Cooper JA. Neurodegeneration mutations in dynactin impair dynein-dependent nuclear migration. Proc. Natl. Acad. Sci. USA. 2009;106:5147–5152. doi: 10.1073/pnas.0810828106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Pasinelli P, Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nat. Rev. Neurosci. 2006;7:710–723. doi: 10.1038/nrn1971. [DOI] [PubMed] [Google Scholar]
  • 219.Puls I, Jonnakuty C, LaMonte BH, Holzbaur EL, Tokito M, Mann E, et al. Mutant dynactin in motor neuron disease. Nat. Genet. 2003;33:455–456. doi: 10.1038/ng1123. [DOI] [PubMed] [Google Scholar]
  • 220.Teuchert M, Fischer D, Schwalenstoecker B, Habisch HJ, Bockers TM, Ludolph AC. A dynein mutation attenuates motor neuron degeneration in SOD1(G93A) mice. Exp. Neurol. 2006;198:271–274. doi: 10.1016/j.expneurol.2005.12.005. [DOI] [PubMed] [Google Scholar]
  • 221.Kieran D, Hafezparast M, Bohnert S, Dick JR, Martin J, Schiavo G, et al. A mutation in dynein rescues axonal transport defects and extends the life span of ALS mice. J. Cell Biol. 2005;169:561–567. doi: 10.1083/jcb.200501085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Teuling E, van Dis V, Wulf PS, Haasdijk ED, Akhmanova A, Hoogenraad CC, et al. A novel mouse model with impaired dynein/dynactin function develops amyotrophic lateral sclerosis (ALS)-like features in motor neurons and improves lifespan in SOD1-ALS mice. Hum. Mol. Genet. 2008;17:2849–2862. doi: 10.1093/hmg/ddn182. [DOI] [PubMed] [Google Scholar]
  • 223.LaMonte BH, Wallace KE, Holloway BA, Shelly SS, Ascano J, Tokito M, et al. Disruption of dynein/dynactin inhibits axonal transport in motor neurons causing late-onset progressive degeneration. Neuron. 2002;34:715–727. doi: 10.1016/s0896-6273(02)00696-7. [DOI] [PubMed] [Google Scholar]
  • 224.Gepner J, Li M, Ludmann S, Kortas C, Boylan K, Iyadurai SJ, et al. Cytoplasmic dynein function is essential in Drosophila melanogaster. Genetics. 1996;142:865–878. doi: 10.1093/genetics/142.3.865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Chevalier-Larsen ES, Wallace KE, Pennise CR, Holzbaur EL. Lysosomal proliferation and distal degeneration in motor neurons expressing the G59S mutation in the p150Glued subunit of dynactin. Hum. Mol. Genet. 2008;17:1946–1955. doi: 10.1093/hmg/ddn092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Robertson J, Doroudchi MM, Nguyen MD, Durham HD, Strong MJ, Shaw G, et al. A neurotoxic peripherin splice variant in a mouse model of ALS. J. Cell Biol. 2003;160:939–949. doi: 10.1083/jcb.200205027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kong J, Xu Z. Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J. Neurosci. 1998;18:3241–3250. doi: 10.1523/JNEUROSCI.18-09-03241.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Marinkovic P, Reuter MS, Brill MS, Godinho L, Kerschensteiner M, Misgeld T. Axonal transport deficits and degeneration can evolve independently in mouse models of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA. 2012;109:4296–4301. doi: 10.1073/pnas.1200658109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Bendotti C, Atzori C, Piva R, Tortarolo M, Strong MJ, DeBiasi S, Migheli A. Activated p38MAPK is a novel component of the intracellular inclusions found in human amyotrophic lateral sclerosis and mutant SOD1 transgenic mice. J. Neuropathol. Exp. Neurol. 2004;63:113–119. doi: 10.1093/jnen/63.2.113. [DOI] [PubMed] [Google Scholar]
  • 230.Tortarolo M, Veglianese P, Calvaresi N, Botturi A, Rossi C, Giorgini A, et al. Persistent activation of p38 mitogen-activated protein kinase in a mouse model of familial amyotrophic lateral sclerosis correlates with disease progression. Mol. Cell. Neurosci. 2003;23:180–192. doi: 10.1016/s1044-7431(03)00022-8. [DOI] [PubMed] [Google Scholar]
  • 231.Morfini GA, Burns M, Binder LI, Kanaan NM, LaPointe N, Bosco DA, et al. Axonal transport defects in neurodegenerative diseases. J. Neurosci. 2009;29:12776–12786. doi: 10.1523/JNEUROSCI.3463-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Pizzuti A, Petrucci S. Mitochondrial disfunction as a cause of ALS. Arch. Ital. Biol. 2011;149:113–119. doi: 10.4449/aib.v149i1.1266. [DOI] [PubMed] [Google Scholar]
  • 233.Rothstein JD. Excitotoxicity hypothesis. Neurology. 1996;47:S19–25. doi: 10.1212/wnl.47.4_suppl_2.19s. discussion S26. [DOI] [PubMed] [Google Scholar]
  • 234.Wiedau-Pazos M, Goto JJ, Rabizadeh S, Gralla EB, Roe JA, Lee MK, et al. Altered reactivity of superoxide dismutase in familial amyotrophic lateral sclerosis. Science. 1996;271:515–518. doi: 10.1126/science.271.5248.515. [DOI] [PubMed] [Google Scholar]
  • 235.Zhang Y, Marcillat O, Giulivi C, Ernster L, Davies KJ. The oxidative inactivation of mitochondrial electron transport chain components and ATPase. J. Biol. Chem. 1990;265:16330–16336. [PubMed] [Google Scholar]
  • 236.Salinas S, Proukakis C, Crosby A, Warner TT. Hereditary spastic paraplegia: clinical features and pathogenetic mechanisms. Lancet Neurol. 2008;7:1127–1138. doi: 10.1016/S1474-4422(08)70258-8. [DOI] [PubMed] [Google Scholar]
  • 237.Blackstone C, O'Kane CJ, Reid E. Hereditary spastic paraplegias: membrane traffic and the motor pathway. Nat. Rev. Neurosci. 2011;12:31–42. doi: 10.1038/nrn2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Reid E, Kloos M, Ashley-Koch A, Hughes L, Bevan S, Svenson IK, et al. A kinesin heavy chain (KIF5A) mutation in hereditary spastic paraplegia (SPG10) Am. J. Hum. Genet. 2002;71:1189–1194. doi: 10.1086/344210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Fichera M, Lo Giudice M, Falco M, Sturnio M, Amata S, Calabrese O, et al. Evidence of kinesin heavy chain (KIF5A) involvement in pure hereditary spastic paraplegia. Neurology. 2004;63:1108–1110. doi: 10.1212/01.wnl.0000138731.60693.d2. [DOI] [PubMed] [Google Scholar]
  • 240.Ebbing B, Mann K, Starosta A, Jaud J, Schols L, Schule R, et al. Effect of spastic paraplegia mutations in KIF5A kinesin on transport activity. Hum. Mol. Genet. 2008;17:1245–1252. doi: 10.1093/hmg/ddn014. [DOI] [PubMed] [Google Scholar]
  • 241.Ferreirinha F, Quattrini A, Pirozzi M, Valsecchi V, Dina G, Broccoli V, et al. Axonal degeneration in paraplegin-deficient mice is associated with abnormal mitochondria and impairment of axonal transport. J. Clin. Invest. 2004;113:231–242. doi: 10.1172/JCI20138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Baas PW, Karabay A, Qiang L. Microtubules cut and run. Trends Cell Biol. 2005;15:518–524. doi: 10.1016/j.tcb.2005.08.004. [DOI] [PubMed] [Google Scholar]
  • 243.Zhao X, Alvarado D, Rainier S, Lemons R, Hedera P, Weber CH, et al. Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat. Genet. 2001;29:326–331. doi: 10.1038/ng758. [DOI] [PubMed] [Google Scholar]
  • 244.Hazan J, Fonknechten N, Mavel D, Paternotte C, Samson D, Artiguenave F, et al. Spastin, a new AAA protein, is altered in the most frequent form of autosomal dominant spastic paraplegia. Nat. Genet. 1999;23:296–303. doi: 10.1038/15472. [DOI] [PubMed] [Google Scholar]
  • 245.Goizet C, Depienne C, Benard G, Boukhris A, Mundwiller E, Sole G, et al. REEP1 mutations in SPG31: frequency, mutational spectrum, and potential association with mitochondrial morpho-functional dysfunction. Hum. Mutat. 2011;32:1118–1127. doi: 10.1002/humu.21542. [DOI] [PubMed] [Google Scholar]
  • 246.Kasher PR, De Vos KJ, Wharton SB, Manser C, Bennett EJ, Bingley M, et al. Direct evidence for axonal transport defects in a novel mouse model of mutant spastin-induced hereditary spastic paraplegia (HSP) and human HSP patients. J. Neurochem. 2009;110:34–44. doi: 10.1111/j.1471-4159.2009.06104.x. [DOI] [PubMed] [Google Scholar]
  • 247.Tarrade A, Fassier C, Courageot S, Charvin D, Vitte J, Peris L, et al. A mutation of spastin is responsible for swellings and impairment of transport in a region of axon characterized by changes in microtubule composition. Hum. Mol. Genet. 2006;15:3544–3558. doi: 10.1093/hmg/ddl431. [DOI] [PubMed] [Google Scholar]
  • 248.Zuchner S, Vance JM. Mechanisms of disease: a molecular genetic update on hereditary axonal neuropathies. Nat. Clin. Pract. Neurol. 2006;2:45–53. doi: 10.1038/ncpneuro0071. [DOI] [PubMed] [Google Scholar]
  • 249.Crimella C, Baschirotto C, Arnoldi A, Tonelli A, Tenderini E, Airoldi G, et al. Mutations in the motor and stalk domains of KIF5A in spastic paraplegia type 10 and in axonal Charcot-Marie-Tooth type 2. Clin. Genet. 2011 doi: 10.1111/j.1399-0004.2011.01717.x. [DOI] [PubMed] [Google Scholar]
  • 250.Willemsen MH, Vissers LE, Willemsen MA, van Bon BW, Kroes T, de Ligt J, et al. Mutations in DYNC1H1 cause severe intellectual disability with neuronal migration defects. J. Med. Genet. 2012;49:179–183. doi: 10.1136/jmedgenet-2011-100542. [DOI] [PubMed] [Google Scholar]
  • 251.d'Ydewalle C, Krishnan J, Chiheb DM, Van Damme P, Irobi J, Kozikowski AP, et al. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat. Med. 2011;17:968–974. doi: 10.1038/nm.2396. [DOI] [PubMed] [Google Scholar]
  • 252.Estela A, Pla-Martin D, Sanchez-Piris M, Sesaki H, Palau F. Charcot- Marie-Tooth-related gene GDAP1 complements cell cycle delay at G2/M phase in Saccharomyces cerevisiae fis1 gene-defective cells. J. Biol. Chem. 2011;286:36777–36786. doi: 10.1074/jbc.M111.260042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Cassereau J, Chevrollier A, Gueguen N, Desquiret V, Verny C, Nicolas G, et al. Mitochondrial dysfunction and pathophysiology of Charcot-Marie-Tooth disease involving GDAP1 mutations. Exp. Neurol. 2011;227:31–41. doi: 10.1016/j.expneurol.2010.09.006. [DOI] [PubMed] [Google Scholar]
  • 254.Warren G, Wickner W. Organelle inheritance. Cell. 1996;84:395–400. doi: 10.1016/s0092-8674(00)81284-2. [DOI] [PubMed] [Google Scholar]
  • 255.Kabzinska D, Niemann A, Drac H, Huber N, Potulska-Chromik A, Hausmanowa-Petrusewicz I, et al. A new missense GDAP1 mutation disturbing targeting to the mitochondrial membrane causes a severe form of AR-CMT2C disease. Neurogenetics. 2011;12:145–153. doi: 10.1007/s10048-011-0276-7. [DOI] [PubMed] [Google Scholar]
  • 256.Baxter RV, Ben Othmane K, Rochelle JM, Stajich JE, Hulette C, Dew-Knight S, et al. Ganglioside-induced differentiation-associated protein-1 is mutant in Charcot-Marie-Tooth disease type 4A/8q21. Nat. Genet. 2002;30:21–22. doi: 10.1038/ng796. [DOI] [PubMed] [Google Scholar]
  • 257.Misko A, Jiang S, Wegorzewska I, Milbrandt J, Baloh RH. Mitofusin 2 is necessary for transport of axonal mitochondria and interacts with the Miro/Milton complex. J. Neurosci. 2010;30:4232–4240. doi: 10.1523/JNEUROSCI.6248-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Davies SW, Turmaine M, Cozens BA, DiFiglia M, Sharp AH, Ross CA, et al. Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell. 1997;90:537–548. doi: 10.1016/s0092-8674(00)80513-9. [DOI] [PubMed] [Google Scholar]
  • 259.Shirendeb UP, Calkins MJ, Manczak M, Anekonda V, Dufour B, McBride JL, et al. Mutant huntingtin's interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington's disease. Hum. Mol. Genet. 2012;21:406–420. doi: 10.1093/hmg/ddr475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Morfini GA, You YM, Pollema SL, Kaminska A, Liu K, Yoshioka K, et al. Pathogenic huntingtin inhibits fast axonal transport by activating JNK3 and phosphorylating kinesin. Nat. Neurosci. 2009;12:864–871. doi: 10.1038/nn.2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Gunawardena S, Her LS, Brusch RG, Laymon RA, Niesman IR, Gordesky-Gold B, et al. Disruption of axonal transport by loss of huntingtin or expression of pathogenic polyQ proteins in Drosophila. Neuron. 2003;40:25–40. doi: 10.1016/s0896-6273(03)00594-4. [DOI] [PubMed] [Google Scholar]
  • 262.Hurd DD, Saxton WM. Kinesin mutations cause motor neuron disease phenotypes by disrupting fast axonal transport in Drosophila. Genetics. 1996;144:1075–1085. doi: 10.1093/genetics/144.3.1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Ticozzi N, Ratti A, Silani V. Protein aggregation and defective RNA metabolism as mechanisms for motor neuron damage. CNS Neurol. Disord. Drug Targets. 2010;9:285–296. doi: 10.2174/187152710791292585. [DOI] [PubMed] [Google Scholar]
  • 264.Weedon MN, Hastings R, Caswell R, Xie W, Paszkiewicz K, Antoniadi T, et al. Exome sequencing identifies a DYNC1H1 mutation in a large pedigree with dominant axonal Charcot-Marie-Tooth disease. Am. J. Hum. Genet. 2011;89:308–312. doi: 10.1016/j.ajhg.2011.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Piccioni F, Pinton P, Simeoni S, Pozzi P, Fascio U, Vismara G, et al. Androgen receptor with elongated polyglutamine tract forms aggregates that alter axonal trafficking and mitochondrial distribution in motor neuronal processes. FASEB J. 2002;16:1418–1420. doi: 10.1096/fj.01-1035fje. [DOI] [PubMed] [Google Scholar]
  • 266.Kemp MQ, Poort JL, Baqri RM, Lieberman AP, Breedlove SM, Miller KE, et al. Impaired motoneuronal retrograde transport in two models of SBMA implicates two sites of androgen action. Hum. Mol. Genet. 2011;20:4475–4490. doi: 10.1093/hmg/ddr380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.La Spada AR, Wilson EM, Lubahn DB, Harding AE, Fischbeck KH. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature. 1991;352:77–79. doi: 10.1038/352077a0. [DOI] [PubMed] [Google Scholar]
  • 268.Saha AR, Hill J, Utton MA, Asuni AA, Ackerley S, Grierson AJ, et al. Parkinson's disease alpha-synuclein mutations exhibit defective axonal transport in cultured neurons. J. Cell Sci. 2004;117:1017–1024. doi: 10.1242/jcs.00967. [DOI] [PubMed] [Google Scholar]
  • 269.Abou-Sleiman PM, Muqit MM, Wood NW. Expanding insights of mitochondrial dysfunction in Parkinson's disease. Nat. Rev. Neurosci. 2006;7:207–219. doi: 10.1038/nrn1868. [DOI] [PubMed] [Google Scholar]
  • 270.Miller KE, Sheetz MP. Axonal mitochondrial transport and potential are correlated. J. Cell Sci. 2004;117:2791–2804. doi: 10.1242/jcs.01130. [DOI] [PubMed] [Google Scholar]
  • 271.Morfini G, Pigino G, Opalach K, Serulle Y, Moreira JE, Sugimori M, et al. 1-Methyl-4-phenylpyridinium affects fast axonal transport by activation of caspase and protein kinase C. Proc. Natl. Acad. Sci. USA. 2007;104:2442–2447. doi: 10.1073/pnas.0611231104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Su KG, Banker G, Bourdette D, Forte M. Axonal degeneration in multiple sclerosis: the mitochondrial hypothesis. Curr. Neurol. Neurosci. Rep. 2009;9:411–417. doi: 10.1007/s11910-009-0060-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Pagliardini S, Giavazzi A, Setola V, Lizier C, Di Luca M, DeBiasi S, et al. Subcellular localization and axonal transport of the survival motor neuron (SMN) protein in the developing rat spinal cord. Hum. Mol. Genet. 2000;9:47–56. doi: 10.1093/hmg/9.1.47. [DOI] [PubMed] [Google Scholar]
  • 274.Fallini C, Bassell GJ, Rossoll W. Spinal muscular atrophy: The role of SMN in axonal mRNA regulation. Brain Res. 2012;1462:81–92. doi: 10.1016/j.brainres.2012.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Morfini G, Pigino G, Brady ST. Polyglutamine expansion diseases: failing to deliver. Trends Mol. Med. 2005;11:64–70. doi: 10.1016/j.molmed.2004.12.002. [DOI] [PubMed] [Google Scholar]
  • 276.Takahashi T, Katada S, Onodera O. Polyglutamine diseases: where does toxicity come from? What is toxicity? Where are we going? J. Mol. Cell. Biol. 2010;2:180–191. doi: 10.1093/jmcb/mjq005. [DOI] [PubMed] [Google Scholar]
  • 277.Feany MB, La Spada AR. Polyglutamines stop traffic: axonal transport as a common target in neurodegenerative diseases. Neuron. 2003;40:1–2. doi: 10.1016/s0896-6273(03)00600-7. [DOI] [PubMed] [Google Scholar]
  • 278.Ermolayev V, Cathomen T, Merk J, Friedrich M, Hartig W, Harms GS, et al. Impaired axonal transport in motor neurons correlates with clinical prion disease. PLoS Pathog. 2009;5:e1000558. doi: 10.1371/journal.ppat.1000558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Almasieh M, Wilson AM, Morquette B, Cueva Vargas JL, Di Polo A. The molecular basis of retinal ganglion cell death in glaucoma. Prog. Retin. Eye Res. 2012;31:152–181. doi: 10.1016/j.preteyeres.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • 280.Lorenzo DN, Li MG, Mische SE, Armbrust KR, Ranum LP, Hays TS. Spectrin mutations that cause spinocerebellar ataxia type 5 impair axonal transport and induce neurodegeneration in Drosophila. J. Cell Biol. 2010;189:143–158. doi: 10.1083/jcb.200905158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Smith DH, Uryu K, Saatman KE, Trojanowski JQ, McIntosh TK. Protein accumulation in traumatic brain injury. Neuromolecular Med. 2003;4:59–72. doi: 10.1385/NMM:4:1-2:59. [DOI] [PubMed] [Google Scholar]
  • 282.Bernier G, Kothary R. Prenatal onset of axonopathy in Dystonia musculorum mice. Dev. Genet. 1998;22:160–168. doi: 10.1002/(SICI)1520-6408(1998)22:2<160::AID-DVG5>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 283.Furlong RA, Zhou CY, Ferguson-Smith MA, Affara NA. Characterization of a kinesin-related gene ATSV, within the tuberous sclerosis locus (TSC1) candidate region on chromosome 9Q34. Genomics. 1996;33:421–429. doi: 10.1006/geno.1996.0217. [DOI] [PubMed] [Google Scholar]

RESOURCES