You are on page 1of 16

Pharmacology & Therapeutics 130 (2011) 348–363

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / p h a r m t h e r a

Associate editor: N. Frossard

Cytoplasmic dynein in neurodegeneration


Judith Eschbach a,b,c, Luc Dupuis a,b,⁎
a
Inserm U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, F-67085, France
b
Université de Strasbourg, Faculté de Médecine, UMRS692, Strasbourg, F-67085, France
c
Department of Neurology, University of Ulm, Germany

a r t i c l e i n f o a b s t r a c t

Keywords: Cytoplasmic dynein 1 (later referred to as dynein) is the major molecular motor moving cargoes such as
Axonal transport mitochondria, organelles and proteins towards the minus end of microtubules. Dynein is involved in multiple
Microtubules basic cellular functions, such as mitosis, autophagy and structure of endoplasmic reticulum and Golgi, but also
Molecular motor in neuron specific functions in particular retrograde axonal transport. Dynein is regulated by a number of
Amyotrophic lateral sclerosis protein complexes, notably by dynactin. Several studies have supported indirectly the involvement of dynein
Alzheimer's disease
in neurodegeneration associated with Alzheimer's disease, Parkinson's disease, Huntington's disease and
Huntington's disease
motor neuron diseases. First, axonal transport disruption represents a common feature occurring in
neurodegenerative diseases. Second, a number of dynein-dependent processes, including autophagy or
clearance of aggregation-prone proteins, are found defective in most of these diseases. Third, a number of
mutant genes in various neurodegenerative diseases are involved in the regulation of dynein transport. This
includes notably mutations in the P150Glued subunit of dynactin that are found in Perry syndrome and motor
neuron diseases. Interestingly, gene products that are mutant in Huntington's disease, Parkinson's disease,
motor neuron disease or spino-cerebellar ataxia are also involved in the regulation of dynein motor activity or
of cargo binding. Despite a constellation of indirect evidence, direct links between the motor itself and
neurodegeneration are few, and this might be due to the requirement of fully active dynein for development.
Here, we critially review the evidence of dynein involvement in different neurodegenerative diseases and
discuss potential underlying mechanisms.
© 2011 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348
2. Cytoplasmic dynein: structure and function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349
3. Potential consequences of dynein dysfunction and their links to neurodegeneration . . . . . . . . . . . . 349
350
4. Cytoplasmic dynein in motor neuron diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349
351
5. Cytoplasmic dynein in basal ganglia degeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350
353
6. Cytoplasmic dynein in Alzheimer's disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350
355
7. Cytoplasmic dynein in other neurodegenerative diseases . . . . . . . . . . . . . . . . . . . . . . . . . 350
356
8. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
357
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
358
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 352
358

1. Introduction
Abbreviations: AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; AR,
androgen receptor; CMT, Charcot–Marie–Tooth disease; FAT, fast axonal transport;
FTD, fronto-temporal dementia; HAP-1, huntingtin associated protein 1; HD, Hunting- Neurodegenerative diseases (NDDs) are a large and diverse group
ton's disease; HTT, huntingtin; MND, motor neuron disease; NDD, Neurodegenerative of pathologies characterized by the degeneration of a subset of
disease; PD, Parkinson's disease; SBMA, spinal and bulbar muscular atrophy (Kennedy's neurons. NDDs include very frequent pathologies of the elderly, such
disease); SCA, spino-cerebellar ataxia; SMA, spinal muscular atrophy.
⁎ Corresponding author at: INSERM U692, Faculté de Médecine, bat 3, 8e étage, 11 rue
as Alzheimer's disease (AD) which concerns about 20 to 25 million
Humann, STRASBOURG, F-67085, France. Tel.: +33 3 68853091; fax: +33 3 68853065. persons worldwide, or Parkinson's disease (PD), with an incidence
E-mail address: ldupuis@unistra.fr (L. Dupuis). between 16 and 20 per 100,000. A number of NDDs are much less

0163-7258/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.pharmthera.2011.03.004
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 349

frequent with incidences ranging from 2 to 3 per 100,000 for body. This mechanism is involved in cell to cell communication in the
amyotrophic lateral sclerosis (ALS) to a few cases worldwide for the nervous system and its dysfunction might account for non cell
rarest NDDs. NDDs can be either inherited or appear sporadically in autonomy of neuronal death in NDDs. Cytoplasmic dynein is thus at a
non-previously affected families. Indeed, a number of NDDs are nexus of various pathophysiological mechanisms in NDDs. Here, we
monogenic diseases (HD for instance) with dominant or recessive will first review the structure of the molecular motor and the
transmission patterns. However, for the most frequent NDDs (AD, PD, potential consequences of its dysfunction. We will then critically
and ALS), there are only rare familial cases, but a majority of sporadic review the evidence that involve directly or indirectly dynein in
cases, with currently unknown causes. The socio-economic burden of neurodegenerative diseases, such as motor neuron diseases (includ-
NDDs is strongly increasing in western countries, mostly because a ing ALS), basal ganglia degeneration (mostly HD and PD) and
number of the most frequent NDDs are associated with aging, the dementia (including AD).
current increase in lifespan thus mechanically leading to increased
frequencies of NDDs. 2. Cytoplasmic dynein: structure and function
The common point to all NDDs is the occurrence of neuronal
degeneration. This degenerative phenotype covers several distinct 2.1. Cytoplasmic dynein, a retrograde motor
events that include disappearance of cell bodies but also shrinkage of
axonal or dendritic compartments. Indeed, it appears that the symp- Dyneins are a large group of molecular motors that include
toms of a number of NDDs are caused by loss of synaptic connections multiple axonemal motors (axonemal dyneins) and two complexes
rather than loss of neuronal cell bodies themselves (Dupuis & Loeffler, termed “cytoplasmic dynein”. Dynein motors are very different in
2009; Palop & Mucke, 2010; van Spronsen & Hoogenraad, 2010). The their structures, localizations and functions (Pfister et al., 2005, 2006),
very different symptoms elicited by NDDs are caused by the nature of and we will here focus on cytoplasmic dynein 1. Indeed, despite there
the affected neurons: the degeneration might be widespread like in are two molecular complexes called “cytoplasmic dynein” (cytoplas-
prion diseases for instance, or affect only a small neuronal population, mic dyneins 1 and 2), cytoplasmic dynein 2 appears mostly present in
with consequences limited to functions controlled by this neuronal ciliated cells and is involved in intraflagellar transport, at least in
subset. This is for instance the case in PD, in which the primarily lower eukaryotes (Porter et al., 1999). The expression pattern of
affected cells are dopaminergic neurons from the substantia nigra. cytoplasmic dynein 2 is consistent with this view (Mikami et al.,
These neurons are involved in the fine tuning of movement initiation 2002). Despite the recent discovery of human mutations in the heavy
and their degeneration leads to tremor and bradykinesia. This initial chain of cytoplasmic dynein 2 (Dagoneau et al., 2009), little is known
cell selectivity remains however relative. Indeed, it is increasingly about cytoplasmic dynein 2 function in mammals.
recognized that despite this apparent starting point, degeneration Cytoplasmic dynein 1 (later referred as dynein in this review) is
involves a number of other cell types (Boillee et al., 2006; Dupuis & expressed in most tissues, including brain, testis, lung, liver and
Loeffler, 2009; Dupuis & Echaniz-Laguna, 2010). kidney (Yoshida et al., 1992) and is involved in the transport of
There are a number of common themes between NDDs. The most cargoes on microtubules, from the periphery to the cell center in a
salient pathological feature of NDDs is the occurrence of protein process called “retrograde transport”. Dynein, initially named MAP1C,
aggregates (Cuervo et al., 2010; Krainc, 2010). Among the different was discovered in 1987 (Paschal et al., 1987) and was initially identi-
NDDs, these aggregates might be either intracellular, such as AD- fied as a component of microtubule preparations. Dynein was shown
associated neurofibrillary tangles or extracellular such as amyloid to possess ATP-hydrolyzing activity and microtubule translocation
plaques in AD. The biochemical composition of protein aggregates are properties (Paschal et al., 1987) and to be able to produce force in a
also differing between NDDs, and are most often revealing underlying direction opposite to that observed for kinesin-1, suggesting that it
pathogenic pathways (Neumann et al., 2006; Kabashi et al., 2008; constituted a retrograde motor (Paschal & Vallee, 1987). Later work
Sreedharan et al., 2008; Crews & Masliah, 2010). A second theme has shown that dynein is able to move cargoes using variable step
common to almost all NDDs is that neuronal degeneration is not cell sizes, but also lateral steps across the microtubule surface, and
autonomous, meaning that neuronal degeneration is due to events processive runs toward both the minus- and plus-end of the micro-
both intrinsic and extrinsic to neurons (Ilieva et al., 2009). A third tubule (Mallik et al., 2004; Ross et al., 2006). ATP-binding activity is
mechanism appears common to most NDDs: axonal transport localized on the C-terminal two-thirds of the dynein heavy chain
impairment. Exchange of cargoes between the neuronal cell body protein (Gee et al., 1997) and is due to the presence of six tandemly
and the axonal tip is called axonal transport. Long distance transport is AAA (ATPase associated with cellular activities) modules necessary
carried out on microtubules, while short distance appears mediated for ATP hydrolysis (Takahashi et al., 2004). The dimerization of two
by the actin cytoskeleton. Microtubules constitute the railroad tracks dynein heavy chains is necessary for motor activity. The two globular
on which molecular motors are able to carry cargoes from the cell heads contain in their C-terminus the motor domain, where ATP is
body to the synapse (anterograde axonal transport) or from the hydrolyzed to produce the strength necessary for dynein displace-
synapse to the cell body (retrograde axonal transport). Anterograde ment along the microtubules, and the microtubule-binding site which
axonal transport is carried out by kinesin like motors, while is localized between the AAA4 and AAA5. The tail domain, located at
retrograde axonal transport is performed mostly by a single motor, the N-terminus of dynein heavy chain, is involved in the dimerization
cytoplasmic dynein. A large body of evidence has demonstrated that of the two heavy chains (Hirokawa et al., 2010).
axonal transport machinery is impaired during neurodegeneration,
and likely contributes to it (Chevalier-Larsen & Holzbaur, 2006; De 2.2. Cytoplasmic dynein structure
Vos et al., 2008; Morfini et al., 2009).
In this review, we will focus on one major actor involved in these Apart from the two heavy chains of 530 kD (Paschal et al., 1987;
different common themes in NDDs. Cytoplasmic dynein is the major Holzbaur & Vallee, 1994), that constitute the motors themselves,
motor of retrograde axonal transport (Levy & Holzbaur, 2006). As dynein is a multi complex protein composed of a number of non-
such, the impairment of its function appears able to lead to axonal catalytic subunits, notably two intermediate chains of 74 kD, and four
transport dysfunction. Cytoplasmic dynein is also the molecular light intermediate chains of 55 kD and a number of less characterized
motor responsible for transport of misfolded proteins for their light chains such as LC8, Tctex1 or Rp3 (King et al., 1998; Hirokawa
degradation, and is thus crucially involved in the appearance and et al., 2010).
clearance of protein aggregates. Last, dynein is the molecular motor The tail domain of dynein heavy chain interacts with intermediate
responsible for retrograde signalling from the synapse to the cell light and light chains to form the cargo-binding complex. The accessory
350 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

proteins of the dynein complex are probably crucial in the cargo selec- 2.4. A motor with multiple functions
tivity of the motor (Kardon & Vale, 2009) (Fig. 1).
Dynein has a large variety of functions and is involved in a number
of different cell processes. The most well documented function is the
2.3. Regulation of dynein activity minus end-directed transport of membranous organelles (Schroer
et al., 1989). In neurons, dynein is responsible for retrograde axonal
Dynein activity requires a number of other proteins. Indeed, transport (Schnapp & Reese, 1989). Dynein is able to transport a large
dynein is associated with dynactin (Karki & Holzbaur, 1995; Vaughan variety of cargoes such as mitochondria, proteins (neurofilaments,
& Vallee, 1995), another multi-protein complex with ten subunits trophic factors like brain-derived neurotrophic factor, RNA particles
including P150Glued, p135Glued, p62, p50 (dynamitin) and Arp1 (see for review (Chevalier-Larsen & Holzbaur, 2006)). Dynein also
(Schroer, 2004) (Fig. 1). Dynactin interacts with dynein through participates in endoplasmic reticulum membrane tubules organisation
dynein intermediate chain and p150Glued (Vaughan, 2005). Dynactin (Terasaki, 1990), is required in vitro for the formation of endoplasmic
was first shown to be required for cargo binding (Karki & Holzbaur, reticulum networks (Allan, 1995), and for the centrosomal localization
1995; Vaughan & Vallee, 1995; Schroer, 2004). In particular, the Arp1 of the Golgi complex (Corthesy-Theulaz et al., 1992). Dynein maintains
subunit of dynactin binds to β-III spectrin a filamentous protein that is the microtubule networks (Ross et al., 2008a) and is involved in the
found on the cytosolic side of a number of intracellular vesicles lysomal trafficking (Lin & Collins, 1992), and in the vesicular transport
(Holleran et al., 2001) (Fig. 1). A second function of dynactin is to from early to late endosomes (Aniento et al., 1993). Dynein is also
increase dynein processivity (King & Schroer, 2000). Despite these involved in the clearance of protein aggregates since mutation in
multiple roles of dynactin, how these different dynactin functions are dynein impairs their autophagic clearance (Ravikumar et al., 2005).
integrated remains largely unknown. Dynein is required for mitosis since it was found on kinetochores (Pfarr
Dynein activity is modulated by a number of ubiquitous cofactors et al., 1990) and drives them to the spindle poles (Hoogenraad et al.,
such as UNC-83 (Fridolfsson et al., 2010), LIS1-Ndel1-Nde1 and 2001; Matanis et al., 2002). Dynein is also able to drive lipid droplets
Bicaudal-D (see for review (Dienstbier & Li, 2009) that have been (Gross et al., 2000), participates indirectly in their formation (Bostrom
reviewed elsewhere (Kardon & Vale, 2009). More recently, multiple et al., 2005; Andersson et al., 2006; Bostrom et al., 2007) and break-
evidence point to a regulation of motor activities, and in particular of down (Eschbach et al., 2011).
dynein, by signalling pathways, notably by kinases. First, dynein has
been shown to interact with huntingtin and huntingtin-associated 3. Potential consequences of dynein
protein 1 (Caviston et al., 2007) and Akt mediated phosphorylation of dysfunction and their links to neurodegeneration
huntingtin is able to promote anterograde transport through kinesin-
1 recruitment, while dephosphorylation of huntingtin stimulates Given the very large requirement for dynein in many basic cell
dynein dependent retrograde transport (Colin et al., 2008). Thus, processes, a number of dynein functions have been, directly or
huntingtin and HAP-1 appear to integrate the different directions of indirectly, linked with NDDs. First, dysfunction in dynein transport
transport. Other kinases are likely involved in the regulation of dynein was long associated with decreased retrograde axonal transport (Levy
activity. Indeed, two cyclin dependent kinases, CDK-5 and PCT-1 and & Holzbaur, 2006). This decreased transport has potentially two
the cyclin CCY-1 have been shown to negatively regulate dynein in the consequences: first, it is expected to decrease the transport of
nematode (Ou et al., 2010) and the JNK kinase pathway might also be organelles back to the cell body. This has been especially studied for
involved in the regulation of dynein (Morfini et al., 2006). axonal transport of mitochondria and dynein and kinesin-1 constitute

Fig. 1. Schematic representation of dynein–dynactin complex. Dynein–dynactin complex drive cargoes along microtubules from theirs minus ends toward theirs plus ends. Dynein is
a large, multimeric protein complex composed of two heavy chains, three intermediate chains, four light intermediate chains (not represented) and light chains. Dynactin, which
increases dynein processivity, is itself a large protein complex, with numerous subunits, including P150Glued, p135Glued, p62, p50 (dynamitin), Arp1, p27, p24 and actin capping
protein. Attachment of cargoes to dynein–dynactin complex requires the interaction with β3-Spectrin, ankyrin (not represented) and Arp1 (a dynactin subunit).
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 351

the major motors for fast axonal transport of mitochondria (Hollen- In the specific context of NDDs, dynein dysfunction is generally
beck & Saxton, 2005; Pilling et al., 2006; Frederick & Shaw, 2007) considered as a pathogenic event in post-mitotic neurons. However,
(Fig. 2). However, the exact consequences of decreased retrograde dynein is also crucial for mitosis and through its function in mitotic
transport on mitochondrial bioenergetic functions remain unex- spindle orientation, dynein appears critically involved in neurogen-
plored. Second, decreased retrograde axonal transport should also esis (Zhang et al., 2009; Godin et al., 2010). Impairment of dynein
lead to decreased retrograde signalling through decreased transport function at adult age might thus also decrease neurogenesis and have
of signalling endosomes (Fig. 2). Indeed, axonal injury activates a an impact on cognition.
number of signalling events that lead to transport signal to the Dynein is thus involved in multiple pathways relevant to NDDs
nucleus through dynein dependent processes (Delcroix et al., 2003; pathogenesis (Fig. 2). It should be noted that a number of these dynein
Heerssen et al., 2004; Yudin et al., 2008; Rishal & Fainzilber, 2010). dependent processes have been studied in the context of develop-
More generally, synaptogenesis and synaptic maintenance are largely ment, but not necessarily in the context of neurodegeneration.
driven by retrograde messengers (Regehr et al., 2009). This is espe-
cially true for neurotrophin retrograde signalling that is completely
dependent upon the formation of signalling endosomes carried to the 4. Cytoplasmic dynein in motor neuron diseases
cell body by dynein (Heerssen et al., 2004; Mallik & Gross, 2004;
Zweifel et al., 2005; Cosker et al., 2008). 4.1. Motor neuron diseases
Besides axonal transport, dynein dysfunction might impact
general neuronal physiology. First, dynein is critically involved in Motor neuron diseases are a group of heterogenous pathologies
intracellular membranes trafficking. For instance, dynein is required that all involve degeneration of the lower motor neurons, and a
for endosomal and lysosomal transport (Hirokawa, 1998; Mallik & relative sparing of sensory innervation. Among MNDs, ALS, spinal
Gross, 2004) (Fig. 2). Indeed, both dynein and endosomal transport muscular atrophy (SMA) and spino-bulbar muscular atrophy (SBMA)
are required for dendritic morphogenesis (Satoh et al., 2008; Zheng are the most frequent (Brooks et al., 2000).
et al., 2008; Braunstein et al., 2010). This role of dynein in dendritic Childhood spinal muscular atrophy (hereafter called spinal
morphogenesis is thought to be linked to dynein role in retrograde muscular atrophy, SMA) is a child-onset motor neuron disease
movement of neurofilaments (Wagner et al., 2004). Second, disrup- involving mutations in the survival of motor neuron gene (smn1)
tion of endosomal trafficking might lead to abnormal autophagic (Lefebvre et al., 1995). SMA patients are divided in three clinical
vesicle trafficking (Ravikumar et al., 2005; Jahreiss et al., 2008) groups according to the severity of their disease, with type I SMA
(Fig. 2). Third, signalling events might also be affected by abnormal being the most severe form leading to death before two years of age
trafficking of endo-lysosomal membranes. For instance, cholesterol and type III SMA the less severe form with muscle weakness starting
sensing in cells requires endosomal trafficking and is dynein after 18 months of age. SMA is caused by mutations in the SMN1 gene
dependent (Johansson et al., 2007; Rocha et al., 2009). Dynein and the severity of the disease is linked to the potential compensation
dysfunction might thus also affect transduction pathways in response by SMN2 protein products.
to environmental cues. Last, dynein interacts with HDAC6 and this is Spinal and bulbar muscular atrophy (SBMA), or Kennedy's disease,
required for the formation of aggresome (Kawaguchi et al., 2003; is a monogenic motor neuron disease caused by expansion of a CAG
Zhou et al., 2009) a subcellular structure located at the microtubule repeat in the first exon of the androgen receptor (AR) gene (La Spada
organising center and required for misfolded protein clearance et al., 1991). This leads to an AR protein with an expanded polygluta-
(Fig. 2). mine (polyQ) tract only in individuals bearing an expansion of the

Fig. 2. Dynein functions relevant to neurodegenerative diseases. Dynein is involved in basic cellular functions (general, right of the figure) and axonal transport related functions (left
of the figure). Dynein is responsible for retrograde axonal transport of organelles, including mitochondria (1), and for transport of signaling endosomes (2) that convey signals of
neurotrophins. Dynein is responsible for multiple transport of organelles in the cell body, for instance autophagosomes (3) and ER-to-Golgi transport (4). In neurons, dynein is
required for dendrite morphogenesis through still incompletely understood mechanisms (5). Last, dynein is responsible for the transport of misfolded proteins in the aggresome (6).
352 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

polyQ tract over 36 glutamines. SBMA is an X-linked, gender specific evidence also point to dynein as key to MNDs pathogenesis. First,
disease since only male carriers of a pathogenic mutation are affected. mutations in the dynactin P150glued have been documented in familial
Amyotrophic lateral sclerosis (ALS) is the most frequent motor cases of ALS and distal SBMA, a rare MND (Puls et al., 2003; Munch
neuron disease of adult onset. In ALS, not only lower motor neurons et al., 2004; Puls et al., 2005). One of these mutant proteins bearing
degenerate, but also upper motor neurons (corticospinal motor the G59S mutation, impairs dynein/dynactin function (Levy et al.,
neurons). The prognosis of ALS is very poor with death generally 2006) and, when overexpressed in transgenic mice, the G59S
occurring two to five years after the diagnosis. A subset of ALS cases mutation in p150Glued leads to enlarged lysosomes, accumulation of
(20%) are inherited (familial ALS), but the vast majority of ALS cases vesicles and distal degeneration of neuromuscular junctions (Lai et al.,
remain of unknown origin. Several genes including angiogenin 2007; Chevalier-Larsen et al., 2008; Laird et al., 2008). However, only
(Greenway et al., 2006), vapb (Nishimura et al., 2004), and more one of these three mouse strains displayed frank paralysis and
recently tardbp (encoding TDP43) (Kabashi et al., 2008; Kuhnlein shortened survival (Laird et al., 2008), while the other two mouse
et al., 2008; Rutherford et al., 2008; Sreedharan et al., 2008; Yokoseki strains did not progress to paralysis. Furthermore, it appears that
et al., 2008), fus (Kwiatkowski et al., 2009; Ticozzi et al., 2009) and fig4 G59S overexpression is not sufficient to impair retrograde axonal
(Chow et al., 2009) have been genetically linked to familial forms of transport, which suggests that other functions of the dynein/dynactin
ALS, but how these specific mutations lead to ALS is currently complex, including clearance of protein aggregates are instrumental
unknown (Lagier-Tourenne & Cleveland, 2009). On the contrary, the in the mild pathology of these mice (Levy et al., 2006).
mechanisms underlying ALS linked to mutations in the sod1 gene, the
first gene linked to familial ALS in 1993, have been extensively studied 4.3.2. Other dynein-related genes linked to MNDs
(Boillee et al., 2006; Gonzalez de Aguilar et al., 2007; Dupuis & Other genetic mutations linked to ALS are likely to alter dynein
Loeffler, 2009). function. First, mutations in the GTP exchange factor alsin were
observed in atypical juvenile forms of motor neuron diseases mostly
4.2. Indirect evidence of dynein involvement in motor neuron diseases affecting the upper motor neuron (Hadano et al., 2001; Yang et al.,
2001). These mutations were called ALS2 and later found also in
A number of cellular functions involving dynein appear impaired families with typical hereditary spastic paraplegia (HSP) (Eymard-
in MNDs. First, a large body of literature documented abnormalities in Pierre et al., 2002). Alsin functions as a promoter of Rab5 activity and
axonal transport. The possibility that axonal transport defects regulates endolysosomal trafficking (Otomo et al., 2003; Topp et al.,
participate in motor neuron disease pathology stems from observa- 2004; Devon et al., 2006; Hadano et al., 2006). Whether ALS2
tions of axonal swellings in pathological images of ALS motor neurons. mutations directly or indirectly affect dynein activity is still to be
For instance, Binet & Meininger (1988) observed abnormalities in studied (Fig. 3).
cytoskeletal proteins such as tubulin in otherwise pathologically Mutations in a component of the endosomal sorting complex
normal axons and Breuer et al. (1987) observed increased antero- (ESCRT) CHMP2B have been found in ALS and fronto-temporal
grade and decreased retrograde axonal transport in nerves of ALS dementia (FTD) (Skibinski et al., 2005; Parkinson et al., 2006; Cox
patients. Sasaki & Iwata (1996) observed a marked increase in et al., 2010). The ESCRT complex is required for the transport of
mitochondria and lysosomes in the proximal axon of ALS patients, membrane proteins to multivesicular bodies. Furthermore, FTD-
suggesting that these organelles were not properly targeted in the related mutations in CHMP2B perturb endosomal trafficking (van
axon possibly due to fast axonal transport impairment. Analysis in der Zee et al., 2008; Urwin et al., 2010). The mutations in CHMP2B lead
animal models have shown impairments of both fast and slow axonal to protein aggregates through compromised autophagy and impair
transport, very early in the disease process and in both directions clearance of mutant TDP43 (Filimonenko et al., 2007). The involve-
(Zhang et al., 1997; Williamson & Cleveland, 1999; Sasaki et al., 2005; ment of dynein in these events has not been investigated but is likely
De Vos et al., 2007; Perlson et al., 2009; Shi et al., 2010). Similar since transport to MVBs is dynein dependent (Lee et al., 2007; Saksena
evidence have been obtained in animal models of SBMA (Katsuno et al., 2007; Hurley, 2008; Raiborg & Stenmark, 2009; Stuffers et al.,
et al., 2006), thus suggesting that impairment in axonal transport is a 2009) (Fig. 3).
general feature of MNDs. In all, available evidence strongly indicate Mutations in the gene encoding for the VAPB protein have been
that axonal transport is compromised in both directions in motor linked with familial forms of ALS and have been termed als8/VAPB
neuron disease. (Nishimura et al., 2004). A mutation (P56S) in the VAPB gene has been
Other pathological hallmarks of MNDs indirectly indicate an reported in a Brazilian pedigree of Portuguese origin, but also in two
involvement of dynein in MNDs. First, MNs from ALS patients display others unrelated families (Funke et al., 2010; Millecamps et al., 2010).
a very pronounced fragmentation of the Golgi apparatus and the Another mutation (T46I) in a British family has been recently
endoplasmic reticulum (ER) (Gonatas et al., 2006). Analogous pictures described (Chen et al., 2010). There exists a phenotypic variability
are observed in vitro and in vivo after disruption of dynein either by between als8 patients ranging from pure typical ALS (both upper
knock-out, siRNA, dynamitin overexpression or mutant p150glued motor neuron and lower motor neuron involvement) to late onset
overexpression (Burkhardt et al., 1997; Harada et al., 1998; Caviston SMA (lower motor neuron only involvement). In some instances,
et al., 2007; Chevalier-Larsen et al., 2008; Laird et al., 2008). Second, autonomic dysfunction has been observed (Nishimura et al., 2004;
MNs from ALS patients display a number of protein aggregates that Marques et al., 2006). However, in all patients reported to date, there
are mostly ubiquitin positive, and include TDP43 as the major protein is an involvement of lower motor neuron, progressing, or not, to both
aggregated (Deng et al., 2010). Mutant proteins linked to ALS, such as lower motor neuron and upper motor neuron involvement. Thus,
mutant forms of SOD1 or TDP43, misfold, aggregate and impair the als8-linked pathologies are either pure ALS or pathologies close to
activity of the proteasome. Since dynein is in general required to ALS. VAPB is one of the members of the VAP family of proteins which
achieve clearance of misfolded proteins, dynein activity is a potential are binding partners of VAMP/synaptobrevin. VAPB is an integral
modulator of protein aggregation in ALS. membrane protein localized to the endoplasmic reticulum and Golgi
intermediates. VAPB has been shown to modulate dynein dependent
4.3. Genetic evidence of dynein involvement in MNDs transport through its interactions with lipid-binding proteins carrying
a FFAT motif (Peretti et al., 2008; Raychaudhuri & Prinz). Among FFAT
4.3.1. Dynactin mutations in MNDs motif containing proteins and VAPB interaction partners, the
Mutations in the dynein heavy chain do not appear to cause oxysterol binding protein family (OSBPs) directly bind to cholesterol
MNDs (Ahmad-Annuar et al., 2003), but a number of indirect genetic and its oxidized derivatives (Raychaudhuri & Prinz). One OSBP, ORP1L
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 353

Fig. 3. Dynein involvement in motor neuron diseases. P150Glued, a component of the dynactin complex is mutated in some familial MND cases and overexpression of dynamitin in
mouse leads to motor neuron disease. Mutant androgen receptor (linked to SBMA) and variants of Elp3, associated with ALS, impact dynein activity indirectly. Last, mutations in
genes that modulate dynein activity in endosomal trafficking, such as VAPB, CHMP2B or ALS2 were found in familial ALS cases. Gene mutants in MNDs or associated with MNDs are
indicated by dark grey circles.

is found in late endosomes and has recently been found to be sensor of expanded AR, is directly responsible for the modulation of dynein
intracellular cholesterol levels (Rocha et al., 2009). ORP1L binds to activity remains unclear.
VAPB in conditions of low cholesterol, and this leads to removal of
dynein and dynactin from late endosomes, inhibiting their perinuclear
4.5. Conclusion
transport and the subsequent cellular responses to cholesterol (Rocha
et al., 2009). Thus, VAPB is a master regulator of endosomal transport
In all, despite a striking amount of knowledge on the involvement
and dynein function (Fig. 3).
of axonal transport and endosomal trafficking, most definitive
Last, variants in Elongator protein 3(Elp3), a component of the
evidence linking dynein to MNDs are indirect. Genetic mutations
Elongator protein complex, have been associated in one study with
leading to MNDs affect modulators of dynein function, while most
sporadic ALS (Simpson et al., 2009). Elongator, among other functions,
experimental evidence used overexpression of interaction partners.
promotes tubulin acetylation through the acetyl transferase activity of
For instance, disrupting dynein function through the overexpression
Elp3 (Creppe et al., 2009). Tubulin acetylation promotes anchoring of
of dynamitin or of Bicaudal D2, leads not only to dynein/dynactin
dynein to microtubules and facilitates dynein mediated transport. It
interaction disruption but also to mild motor neuron degeneration
was proposed that the dysfunction in tubulin acetylation triggered by
(LaMonte et al., 2002; Teuling et al., 2008). Furthermore, over-
Elp3 variants would decrease dynein mediated transport, leading to
expression of mutant p150Glued disrupted endosomal trafficking, led
neurodegeneration (Nguyen et al., 2010) (Fig. 3). Further work is
protein aggregation and cell death (Levy et al., 2006; Lai et al., 2007;
clearly needed to ascertain this working hypothesis.
Chevalier-Larsen et al., 2008; Laird et al., 2008) (Fig. 3). Nevertheless,
Thus, dynein involvement in MNDs is indirectly suggested by the
the use of such interaction partners to study dynein function is
occurrence of several mutations in modulators of some dynein-
uncertain due to their probable or demonstrated dynein independent
dependent transport events, including dynactin, CHMP2B, VAPB, ELP3
functions. Two mutations in the dynein heavy chain gene have been
and ALS2. However there are currently no direct evidence linking
identified in mutant mouse strains. These mutations appear to
genetically dynein and MNDs.
decrease the processivity of the mutant, which might underlie the
relatively benign phenotype of heterozygous mice (Ori-McKenney
et al., 2010). Dynein mutant mice have been initially thought to lead
4.4. Potential mechanisms underlying dynein involvement in MNDs
to motor neuron disease (Hafezparast et al., 2003) on the basis of
abnormal reflexes and potential motor neuron loss. However, three
How pathogenic mechanisms leading to MNDs might lead to
laboratories failed to reproduce the observation of motor neuron loss
dynein dysfunction remains unclear. A couple of studies have
and on the contrary showed that dynein mutant mice displayed
suggested that mutant forms of SOD1 directly bind to dynein and
proprioceptive sensory neuropathy (Chen et al., 2007; Ilieva et al.,
that this interaction was responsible for SOD1 aggregation (Zhang
2008; Dupuis et al., 2009). Thus, up to now, there currently lacks
et al., 2007; Strom et al., 2008). Furthermore, mutant SOD1 over-
direct evidence of dynein involvement in MNDs.
expression altered dynein and dynactin localization, suggesting that
mutant SOD1 led to dynein dysfunction (Ligon et al., 2005). Recent
work however failed to reproduce the direct interaction between 5. Cytoplasmic dynein in basal ganglia degeneration
mutant SOD1 and dynein (El-Kadi et al., 2010), and if existing, this
direct interaction might be non specific due to the transport of 5.1. Basal ganglia diseases
misfolded proteins by dynein to degradation. To our knowledge, there
is currently no study linking other causes of ALS such as TDP43 or FUS Basal ganglia are a group of subcortical structures that includes the
mutations to dynein. striatum, the globus pallidus, substantia nigra and subthalamic nuclei.
The picture appears clearer for SBMA. Indeed, Brady and Although heterogeneous, these structures are all involved in senso-
colleagues have demonstrated that the expanded polyQ AR was able rimotor functions and in particular in the selection, planning and
to inhibit both anterograde and retrograde axonal transport in initiation of voluntary movements. Basal ganglia are highly inter-
isolated squid axoplasm (Szebenyi et al., 2003). This effect was later connected with other brain structures, notably the thalamus and the
shown to be dependent upon the JNK pathway (Morfini et al., 2006), cortex, and are involved in neural circuits called basal ganglia loops
although whether increased JNK activity, as elicited by the polyQ (cortico-basal ganglia-thalamo-cortical loops) whose dysfunction
354 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

leads to motor disorders. (See for reviews: (Alexander et al., 1990; proteinopathy (Farrer et al., 2009). Perry syndrome mutations are
Middleton & Strick, 2000)). distinct from the G59S mutation found in motor neuron diseases cases
For instance, Parkinson's disease (PD), the most frequent disease of (G71R, G71E, G71A, T72P and Q74P) but are nevertheless located in
basal ganglia, is due to the selective loss of dopamine neurons in the the same protein domain (Farrer et al., 2009) (Fig. 5). Interestingly,
substantia nigra. These neurons are involved in the nigro-striatal the biochemical properties of Perry syndrome associated mutant
pathway, and their degeneration leads to dopaminergic denervation of P150Glued were similar to those of MND associated G59S mutant. The
the striatum and globus pallidus and, as a consequence, to bradyki- functional consequences of Perry mutations on dynein function
nesia, rigidity, and resting tremors (Lees et al., 2009). PD is considered remain however unknown. A more direct evidence of dynein
as a mostly sporadic disease despite only a handful of environmental involvement in basal ganglia degeneration was recently provided by
causes have been identified. Among these, mitochondrial toxins, such our phenotypic analysis of mice with a point mutation in the dynein
as MPTP and rotenone are able to lead to experimental parkinsonism heavy chain. Mice bearing the Cramping allele, previously thought to
under defined experimental conditions (Bove et al., 2005). 10% of PD develop motor neuron degeneration, displayed motor and behav-
are familial cases linked to mutations within PARK genes. A number of ioural abnormalities including hindlimb clasping, early muscle
these genes have been identified, and include α-Synuclein (PARK1), weakness, incoordination and hyperactivity reminiscent of mild HD
Parkin (PARK2), PINK1 (PARK6) and DJ1 (PARK7) (Gupta et al., 2008). animal models. In addition to these phenotypic similarities, these
Another disease affecting basal ganglia is Huntington's disease dynein mutant mice exhibit a striatal atrophy and ventricle
(HD). HD is a monogenic neurodegenerative disease leading to dege- enlargement without associated degeneration of striatal neurons or
neration of striatal neurons and clinically characterized by loss of of substantia nigra dopaminergic neurons. In vitro, dynein mutant
cognitive functions associated with major motor disturbance typified striatal neurons displayed strongly impaired neuritic morphology
by hyperkinesia (Berardelli et al., 1999). HD is caused by expansion in (Braunstein et al., 2010). Most interestingly, the analogy between
the CAG repeats of the huntingtin gene, leading to expanded polyglu- dynein mutant mice and HD animal models was also noted in the
tamine tract in the huntingtin protein (Jou & Myers, 1995; Walker, periphery, with dynein mutant mice displaying increased adiposity
2007). The length of polyglutamine expansion is correlated with the and compromised brown adipose tissue thermogenesis in a manner
severity of the disease (Andrew et al., 1993; Rubinsztein et al., 1993). similar to HD models (Weydt et al., 2006; Eschbach et al., 2011). Thus,
a point mutation in the dynein heavy chain is able to mimic, a mild
5.2. Indirect evidence of dynein involvement in basal ganglia diseases HD-like phenotype. To our knowledge, these data provide the first
direct proof that modifying dynein itself promotes a phenotype mild
A number of indirect evidence suggest that dynein function might but similar to HD.
be altered in PD and HD. First, axonal dystrophy, suggestive of axonal
transport impairment has been observed in PD brains (Lach et al.,
1992). Second, MPTP, a PD relevant injury, leads to defective axonal 5.3.2. Other dynein-related genes in basal ganglia diseases
transport, including increased dynein dependent transport (Morfini Mutations in other genes linked to PD further indirectly support
et al., 2007), while axonal transport defects have been reported in the involvement of dynein in PD pathology. First, Saha et al. (2004)
various animal or cellular models of HD (Gunawardena et al., 2003; had suggested that α-synuclein was associated with motor proteins
Szebenyi et al., 2003; Lee et al., 2004; Trushina et al., 2004; Sinadinos and vesicles. Alpha-synuclein mutations have been reported to
et al., 2009). prevent the transport of the protein as well as its binding to brain
Dynein is known to be involved in Golgi apparatus morphology, vesicles (Jensen et al., 1998; Saha et al., 2004). From a functional point
and dynein disruption results in the fragmentation of this organelle. of view, alpha synuclein overexpression impairs microtubule depen-
Interestingly, PD brains as well as cellular and animal models also dent trafficking (Lee et al., 2006) and leads to Golgi fragmentation
display important fragmentation of the Golgi apparatus (Fujita et al., (Gosavi et al., 2002). More precisely, alpha-synuclein overexpression
2006; Lin et al., 2009; Winslow et al., 2010) and the situation appears is able to block ER-to Golgi trafficking in yeast and mammalian cells,
similar in HD at least in cellular models (Caviston et al., 2007; Pardo through inhibition of the small G-protein Rab1 and other ER/Golgi
et al., 2010). SNARES (Cooper et al., 2006; Gitler et al., 2008; Thayanidhi et al.,
Dynein is crucially required for autophagy. Interestingly, both 2010). The involvement of dynein in these functions of alpha-
mutant alpha-synuclein and mutant huntingtin are degraded by synuclein has not been investigated up to now.
autophagy in a dynein dependent manner (Ravikumar et al., 2005) More direct evidence link recessive familial PD cases to dynein
and the autophagic process itself appears largely abnormal in both HD dysfunction. Autophagy of mitochondria, a dynein dependent process,
and PD (reviewed elsewhere (Banerjee et al., 2010; Wong & Cuervo, appears as a genetic target in some familial PD cases. Mitochondria
2010)). For instance, it has been most recently shown using various that are unable to restore their membrane potential after a fission
mouse and cellular models of HD that HD is associated with a primary event are committed to mitophagy (Twig et al., 2008; Wikstrom et al.,
defect in cargo recognition of autophagic vacuoles (Martinez-Vicente 2009). Interestingly, such dysfunctional mitochondria are bound by
et al., 2010). In a similar manner, alpha-synuclein overexpression, a parkin, a protein encoded by a gene mutant in PARK2 (Narendra et al.,
situation occurring in some familial PD cases due to duplication of 2008; Vives-Bauza et al., 2009). Parkin binding to impaired mito-
alpha-synuclein gene locus (Ross et al., 2008b), inhibits macro- chondria is thought to mediate their transport to perinuclear areas
autophagy (Winslow et al., 2010). Thus, both PD and HD appear to where these damaged organelles might be degraded through
involve abnormal autophagic response, which suggests potential autophagy (Narendra et al., 2008; Vives-Bauza et al., 2009).
involvement of dynein. Importantly, Parkin recruitment to dysfunctional mitochondria is
dependent upon stabilization of PINK1, mutant in PARK6, in
5.3. Genetic evidence of dynein involvement in basal ganglia diseases mitochondria (Matsuda et al., 2010; Narendra et al., 2010; Vives-
Bauza et al., 2010). Parkin-bound mitochondria are then docked to
5.3.1. Dynein and dynactin mutations in basal ganglia diseases dynein through the tubulin desacetylase called HDAC6 and this
Two lines of evidence show involvement of dynactin and dynein in docking triggers their transport to the aggresome through dynein
degeneration of basal ganglia. First, Farrer and collaborators described dependent transport (Hubbert et al., 2002; Kawaguchi et al., 2003;
five mutations in p150Glued linked to an atypical PD resistant to L- Olzmann et al., 2007; Jiang et al., 2008). Interestingly, parkin and
DOPA, called Perry syndrome and characterized by early-onset HDAC6 are also involved in the transport of misfolded DJ-1, the
parkinsonism, depression, weight loss, hypoventilation, and TDP-43 product of PARK7, to the aggresome (Olzmann et al., 2007). Thus,
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 355

dynein dependent transport of mitochondria and misfolded proteins (Miller & Sheetz, 2004). Consistently, inhibition of complex I of the
appears to be crucially involved in PD. electron transport chain with MPP + increased cytoplasmic dynein-
Besides PD, a number of evidence link dynein to huntingtin. First, dependent retrograde fast axonal transport (FAT) and reduced
both huntingtin and its associated protein HAP-1 interact with kinesin-1-mediated anterograde FAT through different mechanisms
microtubules (DiFiglia et al., 1995), dynein (Caviston et al., 2007) and that involve caspase 3 activation (Serulle et al., 2007) and protein
p150Glued (Engelender et al., 1997; Gauthier et al., 2004). Huntingtin kinase C (Morfini et al., 2007).
appears to facilitate dynein mediated transport (Caviston et al., 2007) In HD, recent work by Saudou, Humbert and colleagues showed
but is also required for anterograde axonal transport (Gauthier et al., that a specific serine residue in huntingtin, S421, act as a molecular
2004). Functionally, mutant huntingtin impairs retrograde axonal switch between anterograde and retrograde transport (Colin et al.,
transport (Szebenyi et al., 2003; Trushina et al., 2004) while wild type 2008; Zala et al., 2008). This serine residue can be phosphorylated by
overexpression increases retrograde axonal transport, presumably Akt and these authors had previously shown dysregulated Akt in HD
dynein dependent (Her & Goldstein, 2008). Furthermore, mutant htt (Colin et al., 2005). Independently, Morfini, Brady and colleagues have
striatal neurons exhibit dysmorphic dendrites (Laforet et al., 2001), in a shown that mutant huntingtin impairs axonal transport through JNK
manner similar to that of dynein mutant striatal neurons. In all, available activation, and directly provided evidence of JNK mediated phosphor-
evidence show that huntingtin and HAP-1 act as a docking platform to ylation of kinesin. Whether JNK phosphorylates dynein is currently
modulate vesicular cargo attachment to molecular motors, including elusive and how this relates to other signalling pathways, including
dynein (Caviston et al., 2007) (Fig. 4). Akt and huntingtin phosphorylation deserve further investigations.
Interestingly, HDAC6 has also been involved in the pathophysiology
of HD (Dompierre et al., 2007; Pandey et al., 2007) through its tubulin 5.5. Conclusion
deacetylation activity. Microtubule acetylation levels promote kinesin-1
binding and transport (Reed et al., 2006) and the recruitment of dynein In all, these observations point to dynein involvement to fin basal
to microtubules (Dompierre et al., 2007). In HD brain, tubulin ganglia neurodegeneration. Defects in axonal transport, Golgi ER
acetylation is reduced which in turn, could be responsible for decrease trafficking as well autophagy occurring in HD and PD indicate that
vesicular transport in HD brains (Fig. 4). This mechanism that directly dynein could be a central point in the pathology, an hypothesis
involves dynein, could be the key to explain the decrease of BDNF strengthened by striatal atrophy in dynein mutant mice. However,
transport in striatal neurons (Dompierre et al., 2007). dynein mutant mice do not display any neuronal loss. Interestingly, a
Thus, dynein involvement in PD and HD is indirectly suggested by large number of gene mutants in PD, as well as huntingtin, in basal
the occurrence of several mutations in modulators of some dynein- ganglia diseases, are known to modulate directly or indirectly at least
dependent transport events, including dynactin, huntingtin, parkin, a subset of dynein functions. Nevertheless, dynein independent
PINK1, and potentially alpha-synuclein. In mice, a mutation in dynein effects of these proteins in these diseases also occur, as exemplified
itself is able to lead to a disorder of basal ganglia, without associated by the transcriptional role of huntingtin (Zuccato et al., 2001; Cui et
neurodegeneration. There are currently no known mutations in dynein al., 2006). Further studies with cell targeted loss of function of dynein
in such disorders. will be necessary to provide evidence of the direct involvement of
dynein in PD and HD.
5.4. Potential mechanisms underlying
dynein involvement in basal ganglia diseases 6. Cytoplasmic dynein in Alzheimer's disease

Few studies investigated the causes of dynein dysfunction in PD 6.1. Alzheimer's disease
and HD. In PD, dynein dysfunction might be caused by mitochondrial
dysfunction. Indeed, mitochondrial toxins differentially modulate Alzheimer's disease (AD) is the most common form of dementia,
mitochondrial transport, in particular dynein mediated transport with about 25 million persons affected worldwide. Its prevalence is

Fig. 4. Dynein involvement in basal ganglia diseases. P150Glued, a component of the dynactin complex is mutated in Perry syndrome and the Cramping mutation in dynein heavy
chain leads to striatal atrophy in mouse. Alpha-synuclein, mutant in familial PD is a cargo of dynein and regulates ER-to-Golgi trafficking. Huntingtin (Htt) and its associated protein
HAP-1 are binding partners of dynein/dynactin. HDAC6, a direct dynein binding partner regulates mitophagy through its association with Parkin and PINK1, two gene mutants in PD
cases. When misfolded, DJ-1, also linked to PD, is transported to the aggresome through dynein dependent transport. Gene mutants in PD and HD are indicated by dark grey circles.
356 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

estimated to be about 25 to 33% in people aged of more than 85 years. endosomal trafficking of APP and accelerate amyloid plaque deposi-
From a clinical point of view, patients slowly lose episodic memory. tion. Further work is needed to resolve this discrepancy.
Disease progression includes aphasia, apraxia and agnosia and begins The second typical lesions of AD are neurofibrillary tangles (NFT),
with an early clinical stage called mild cognitive impairment (MCI). mainly composed of accumulations of hyperphosphorylated tau.
AD is mostly a sporadic disease, with a few familial cases linked to Interestingly, Tau is a microtubule associated protein, that was
mutations in APP or in presenilins. Familial cases display earlier onset recently shown to differentially regulate kinesin and dynein motor
and more severe disease course. Concerning sporadic cases, a major activity (Dixit et al., 2008). Tau is also a binding partner for dynactin,
risk factor is the apoE4 allele, which increases the risk of AD of three and enhances dynactin binding to microtubules (Magnani et al.,
times in heterozygotes and fifteen times in homozygotes. Pathology of 2007). Last, very recently, Tau reduction was shown to prevent Aβ
AD includes two typical lesions, amyloid plaques, that are extracel- induced defect in axonal transport, both in anterograde and retro-
lular aggregates mainly composed of amyloid β peptides (Aβ), and grade directions (Vossel et al., 2010). Thus, this crucial player in AD
neurofibrillary tangles largely composed of intracellular hyperphos- pathogenesis appears also a potential modulator of dynein-dependent
phorylated Tau aggregates. The clinics, genetics and mechanisms of transports.
AD have been largely reviewed elsewhere (Blennow et al., 2006;
Bettens et al., 2010; Crews & Masliah, 2010).
6.3. Conclusions
6.2. Evidence for dynein involvement in AD
Despite little doubt that axonal transport machinery and intracel-
Evidence involving dynein in AD are largely indirect and dynein lular trafficking events are involved in AD pathogenesis, the direct
might be involved in both amyloid plaques and neurofibrillary tangle evidence linking dynein to AD remain scarce, and the links provided
neurotoxicity. Indeed, as would be expected if AD is associated with here between AD and dynein remain largely speculative. Further work
decreased dynein function, AD is associated with a number of axonal should be focused on more direct studies.
defects and swellings, and these defects are present in human AD
brains, but also in animal models long before cognitive deficits are
obvious (Stokin et al., 2005; Stokin & Goldstein, 2006). However, 7. Cytoplasmic dynein in other neurodegenerative diseases
observations of axonal defects and swellings should not be taken as
proofs of axonal transport defects or of dynein involvement. In the 7.1. Prion diseases
case of AD, such evidence are still lacking.
APP, the precursor of Aβ peptide, is transported by fast axonal Prion diseases are also known as transmissible spongiform en-
anterograde transport, is the subject to complex trafficking events cephalopathies (TSEs). TSEs are uniformly lethal neurodegenerative
(Tang, 2009) and accumulates in pathological enlarged endosomes diseases, that are caused by an original infectious agent called prion.
(Cataldo et al., 2000; Kimura et al., 2009). A number of evidence Prions are abnormally folded isoforms of the cellular isoform of the
suggest that Aβ is transported to the synapse where it is secreted prion protein (PrPC). PrPC is necessary for prion diseases, and the
(Sisodia et al., 1993; Lazarov et al., 2002, 2005) and a number of pathology requires the conversion of PrPC to the abnormal confor-
laboratories have focused their efforts in determining whether the mation PrPSc. For review see Aguzzi and Heikenwalder (2006); Aguzzi
amyloid cascade might be related with axonal transport impairment. et al. (2008).
APP has been found to be interacting with kinesin-1 (Kamal et al., Dynein might be involved at different steps of prion mediated
2000), but this has since been refuted (Lazarov et al., 2005). More neurodegeneration. First, dynein was thought to be key in prion
recently, Aβ peptide was shown to disrupt axonal transport in neuroinvasion. Prions are formed in the periphery and then “trans-
cultured neurons (Rui et al., 2006; Pigino et al., 2009; Decker et al., ported” in the CNS through unclear mechanisms in a process called
2010). Whatever the underlying mechanism, most evidence linking neuroinvasion. Several mechanisms might account for this phenom-
axonal transport machinery to amyloid plaques suggest an involve- enon. First, PrPC might be converted to PrPSc locally, this conversion
ment of rather anterograde transport than retrograde transport. For then spreading from place to place (“Domino” theory). Alternatively,
instance, the reduction in kinesin gene dosage enhanced axonal PrPSc might be transported through microtubule dependent transport
pathology in AD animal models (Stokin et al., 2005), while no similar (“Streetcar hypothesis”) (Aguzzi, 2003; Heikenwalder et al., 2007). In
evidence have been published concerning dynein. The function of APP the latter scenario, dynein could be the crucial motor for prion
itself in axonal transport is also unclear since APP deletions did not transport. This hypothesis is substantiated by reports of PrPC transport
modify axonal transport, while APP overexpression leads to axono- by fast axonal transport in both anterograde (Borchelt et al., 1994) and
pathy in an Aβ-independent manner (Stokin et al., 2008). The links retrograde directions (Moya et al., 2004) and by numerous evidence of
between APP and axonal transport in general, and dynein in intracellular transport of both PrPC and PrPSc between the cell surface,
particular, thus remain largely unclear. the ER, the Golgi and endosomes (Campana et al., 2005). However,
Recent studies showed that APP accumulates in enlarged endo- impairment of fast axonal transport through four-repeat tau over-
somes during normal aging and AD. Endosomal pathology in AD is a expression does not modify prion neuroinvasion (Kunzi et al., 2002)
very early event that precedes Aβ deposition (Cataldo et al., 2000). and dynein mutant mice do not display delayed scrapie incubation
Interestingly, increasing endocytosis through Rab5 overexpression times (Hafezparast et al., 2005). These results suggest that a lack of
largely mimics AD-related endosomal pathology, increases Aβ dynein involvement prion neuroinvasion, but do not provide strong
deposition and promotes neuronal apoptosis (Grbovic et al., 2003; arguments for this.
Cataldo et al., 2004; Laifenfeld et al., 2007). Dynein function is Dynein dysfunction might also be the consequence of prion
required for Rab5-mediated increase in endocytosis (Driskell et al., mediated neurotoxicity. Indeed, the onset of prion disease in mice is
2007), suggesting that dynein function might increase Aβ deposition associated with important defects in retrograde axonal transport
through facilitation of Rab5 mediated endocytosis. However, knock- (Ermolayev et al., 2009a, 2009b). From a mechanistic point of view,
ing down dynein in neurons mimicked aging-induced endosomal this impairment in retrograde transport might be due to abnormal
pathology, led to accumulation of APP in endosomes and increased localization of the small G-protein Rab7 which mediates dynein/
beta-secretase cleavage of APP, leading to increased production of Aβ dynactin attachment to late endosomes (Ermolayev et al., 2009a)
(Kimura & Yanagisawa, 2007; Kimura et al., 2009). Thus, dynein might (Fig. 5). In all, these results suggest that dynein dysfunction con-
be required for Aβ production, but its loss might also interfere with tributes rather to the later events of prion diseases.
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 357

Fig. 5. Dynein involvement in peripheral neuropathies. Dynein cargoes, including NF‐L or mitochondrial mitofusin, are mutant in CMT families. Gigaxonin deletion impairs dynein
motor activity through increased expression of microtubule associated proteins MAP8 and MAP1C. Rab7 mutation in CMT constitutively activate Rab7 and alter dynein dépendant
endosomal trafficking. Genr mutants in peripheral neurophathie are indicated by dark grey circles.

7.2. Peripheral neuropathies and was demonstrated to be essential for axonal retrograde transport
(Deinhardt et al., 2006). It is thus probable that CMT2B mutations
A vast array of NDDs are peripheral neuropathies, that affect either affect, in a still incompletely understood manner, dynein dependent
sensory neurons or motor neurons or both. For instance, the most transport (Fig. 5).
common causes of peripheral neuropathies in western countries are
diabetes and alcoholism, while HIV and leprosy are the most common 7.3. Spinocerebellar ataxia
infectious causes. These pathologies are extremely diverse, with
different etiologies (genetic, toxic, infectious…) and very diverse Spinocerebellar ataxias (SCAs) are a heterogenous group of genetic
clinical pictures and disease course. Peripheral neuropathies can thus disorders, characterized by progressive cerebellar ataxia leading to
be acute or chronic, symmetrical or not, be axonal or involving postural abnormalities, and progressive motor incoordination. SCAs
demyelination and involve only parts of the neuron or its whole. For are also associated with a number of other non-motor signs, including
review see Fricker et al. (2008). A number of peripheral neuropathies cognitive deficits (Durr, 2010). There are currently 18 different SCA
are inherited, and we would like here to focus on a few examples that loci, and most of these are associated with polyglutamine expansions.
link these diseases to dynein function. Interestingly, ataxin 3, mutated in SCA3, is involved in aggresome
A number of inherited neuropathies are associated with docu- formation (Burnett & Pittman, 2005). Ataxin 3 possesses a deubiqui-
mented defects in axonal transport (reviewed in (De Vos et al., tinilating enzyme activity, is colocalized with HDAC6 and dynein and
2008)). In Charcot–Marie–Tooth disease (CMT) type 2E, caused by appears indispensable for aggresome formation. The involvement of
mutations in the neurofilament light gene (NF-L), NF-L mutants dynein in SCA3 pathogenesis appears thus plausible (Fig. 6).
aggregates perturb the cytoskeletal network, in particular microtu- Mutations in β-III spectrin trigger another SCA, SCA5 (Ikeda et al.,
bules, and disrupt the normal localization of mitochondria (Perez-Olle 2006). β-III spectrin interacts with Arp1, a subunit of dynactin, and is
et al., 2004). The overexpression of CMT2E-linked NF-L leads to found in a complex with dynactin (Holleran et al., 2001). It is
impaired axonal transport in vitro (Brownlees et al., 2002) and in vivo noteworthy that β-III spectrin is required for dynein-mediated
(Dequen et al., 2010). Furthermore, in a mouse model of CMT2A translocation of late endosomes to microtubule minus ends and that
caused by mutations in mitofusin, axonal transport of mitochondria this requires the activities of Rab7-RILP and ORP1L (Johansson et al.,
was compromised (Baloh et al., 2007; Misko et al., 2010), and in giant 2007). Thus β-III spectrin is hypothesized to facilitate protein
axonal neuropathy, caused by mutations in gigaxonin, deletion of trafficking by linking dynein/dynactin to its cargoes. β-III spectrin
gigaxonin in mouse led to impaired retrograde axonal transport null mice display a SCA5-like phenotype, with Purkinje cell degen-
(Allen et al., 2005; Ding et al., 2006). This impairment of axonal eration and progressive motor impairment, that could be due to loss
transport appears not only true for axonal but also for demyelinating of neuronal and astrocytic transporter of glutamate, in particular of
hereditary neuropathies. Indeed, in an animal model of X-linked EAAT4 (Perkins et al., 2010; Stankewich et al., 2010). Interestingly,
demyelinating/type I Charcot–Marie–Tooth neuropathy (CMT1X), an SCA5-associated mutations in β-III spectrin disrupt binding to Arp1,
inherited peripheral neuropathy caused by mutations in the gap and impair intracellular trafficking of EAAT4 (Clarkson et al., 2010).
junction protein connexin32, Vavlitou et al., 2010 observed reduced Consistently, SCA5 mutations impair axonal transport in drosophila
axonal transport and axonal pathology before demyelination occurs. (Lorenzo et al., 2010) and their toxicity is enhanced by dynein or
Dynein dysfunction appears directly involved in at least a subset of dynactin loss of functions. In all, it appears that SCA5 associated
these diseases. First, the deletion in gigaxonin in mouse leads to mutations in β-III spectrin impair dynein function through defective
increased MAP8 and MAP1B LC levels. This, in turn, traps dynein and binding to dynactin, leading to abnormalities in glutamate transporter
impairs axonal retrograde transport (Allen et al., 2005; Ding et al., intracellular trafficking and axonal transport impairment (Fig. 6).
2006). More directly, mutations in the G-protein Rab7 trigger CMT2B
(Verhoeven et al., 2003) leading to constitutive activity of mutant 8. Conclusions
proteins (De Luca et al., 2008; Spinosa et al., 2008; McCray et al.,
2010). Rab7 controls dynein/dynactin recruitment to lysosomes and Since the discovery of dynein, almost 25 years ago, research has
late endosomes through its effector protein RILP (Jordens et al., 2001) largely demonstrated that this molecular motor might play a key role
358 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

Fig. 6. Dynein involvement in ataxias. β3-spectrin that is required for anchoring dynein/dynactin to vesicular cargoes is mutated in SCA5, while ataxin3, mutant in SCA3, is
colocalized with dynein and required for the formation of the aggresome. Gene mutants in SCAs are indicated by dark grey symbols.

in neurodegenerative diseases. Indeed, a number of cellular processes pour la recherche et le developpement de moyens de lutte contre les
that are almost entirely dynein-dependent are crucially involved in maladies neurodégénératives (AREMANE).
NDDS, notably retrograde axonal transport, endosomal and lysosomal
trafficking and protein degradation. Moreover, a number of dynein References
interactors or regulators are mutant in various neurodegenerative
diseases, clearly showing that the motor activity of dynein is certainly Aguzzi, A. (2003). Prions and the immune system: a journey through gut, spleen, and
critical for neuronal survival. nerves. Adv Immunol 81, 123−171.
Aguzzi, A., & Heikenwalder, M. (2006). Pathogenesis of prion diseases: current status
Despite this wealth of knowledge, it has to be pointed out that to
and future outlook. Nat Rev Microbiol 4, 765−775.
date, most of the available evidence of involvement of dynein itself is Aguzzi, A., Sigurdson, C., & Heikenwaelder, M. (2008). Molecular mechanisms of prion
indirect, and mostly based on dynamitin overexpression or on over- pathogenesis. Annu Rev Pathol 3, 11−40.
expression of mutant P150Glued or other mutant interactors of dynein. Ahmad-Annuar, A., Shah, P., Hafezparast, M., Hummerich, H., Witherden, A. S.,
Morrison, K. E., et al. (2003). No association with common Caucasian genotypes
In this context, a number of these proteins have been recently shown in exons 8, 13 and 14 of the human cytoplasmic dynein heavy chain gene
to possess dynein independent function. This includes notably dynac- (DNCHC1) and familial motor neuron disorders. Amyotroph Lateral Scler Other Mot
tin and bicaudal D which regulate not only the function of dynein but Neuron Disord 4, 150−157.
Alexander, G. E., Crutcher, M. D., & DeLong, M. R. (1990). Basal ganglia-thalamocortical
also that of kinesins (Berezuk & Schroer, 2007; Splinter et al., 2010). circuits: parallel substrates for motor, oculomotor, “prefrontal” and “limbic”
Similarly, huntingtin appears to modulate not only dynein but also functions. Prog Brain Res 85, 119−146.
kinesin and is documented to have transcriptional effects on BDNF Allan, V. (1995). Protein phosphatase 1 regulates the cytoplasmic dynein-driven
formation of endoplasmic reticulum networks in vitro. J Cell Biol 128, 879−891.
expression and PGC1α co-activator function (Zuccato et al., 2001; Cui Allen, E., Ding, J., Wang, W., Pramanik, S., Chou, J., Yau, V., et al. (2005). Gigaxonin-
et al., 2006). Moreover, besides a regulation of intracellular transport, controlled degradation of MAP1B light chain is critical to neuronal survival. Nature
p150Glued protein has been shown to act as a docking protein and has 438, 224−228.
Andersson, L., Bostrom, P., Ericson, J., Rutberg, M., Magnusson, B., Marchesan, D., et al.
been recently suggested to affect gene transcription through direct
(2006). PLD1 and ERK2 regulate cytosolic lipid droplet formation. J Cell Sci 119,
modulation of transcription factors (Lee et al., 2009; Shrum et al., 2246−2257.
2009). Thus direct proofs of involvement of dynein motor itself in Andrew, S. E., Goldberg, Y. P., Kremer, B., Telenius, H., Theilmann, J., Adam, S., et al.
(1993). The relationship between trinucleotide (CAG) repeat length and clinical
NDDs are largely lacking. This lack of evidence might well be due to
features of Huntington's disease. Nat Genet 4, 398−403.
the obligate need of dynein for embryonic development. Aniento, F., Emans, N., Griffiths, G., & Gruenberg, J. (1993). Cytoplasmic dynein-
Last, it remains unknown whether the motor activity of dynein is dependent vesicular transport from early to late endosomes. J Cell Biol 123,
rate-limiting in neuronal survival or whether it is rather the quality of 1373−1387.
Baloh, R. H., Schmidt, R. E., Pestronk, A., & Milbrandt, J. (2007). Altered axonal
the material transported that bears any importance. In this respect, it mitochondrial transport in the pathogenesis of Charcot–Marie–Tooth disease from
is striking to note that dynein heterozygous mice appeared grossly mitofusin 2 mutations. J Neurosci 27, 422−430.
normal while a point mutation on a single dynein heavy chain allele is Banerjee, R., Beal, M. F., & Thomas, B. (2010). Autophagy in neurodegenerative
disorders: pathogenic roles and therapeutic implications. Trends Neurosci 33(12),
sufficient to lead to a number of neurologic and peripheral pheno- 541−549.
types (Harada et al., 1998; Hafezparast et al., 2003; Dupuis et al., 2009; Berardelli, A., Noth, J., Thompson, P. D., Bollen, E. L., Curra, A., Deuschl, G., et al. (1999).
Braunstein et al., 2010) suggesting that the mutant allele possess a Pathophysiology of chorea and bradykinesia in Huntington's disease. Mov Disord
14, 398−403.
toxic gain of function over at least one crucial function for dynein in Berezuk, M. A., & Schroer, T. A. (2007). Dynactin enhances the processivity of kinesin-2.
neuronal physiology. Here again, genetic deletion of dynein in Traffic 8, 124−129.
neurons of interest might show the dose dependency of neuronal Bettens, K., Sleegers, K., & Van Broeckhoven, C. (2010). Current status on Alzheimer
disease molecular genetics: from past, to present, to future. Hum Mol Genet 19,
survival to dynein motor activity.
R4−R11.
Binet, S., & Meininger, V. (1988). Modifications of microtubule proteins in ALS nerve
Acknowledgments precede detectable histologic and ultrastructural changes. Neurology 38,
1596−1600.
Blennow, K., de Leon, M. J., & Zetterberg, H. (2006). Alzheimer's disease. Lancet 368,
Work in our laboratory is supported by grants from Fondation 387−403.
pour la Recherche Médicale, Association pour la Recherche sur la Boillee, S., Vande Velde, C., & Cleveland, D. W. (2006). ALS: a disease of motor neurons
Sclérose Latérale Amyotrophique (ARSla), Amyotrophic Lateral and their nonneuronal neighbors. Neuron 52, 39−59.
Borchelt, D. R., Koliatsos, V. E., Guarnieri, M., Pardo, C. A., Sisodia, S. S., & Price, D. L.
Sclerosis Association (grant 1698), Fondation Thierry Latran, Associ- (1994). Rapid anterograde axonal transport of the cellular prion glycoprotein in the
ation française de lutte contre les myopathies (AFM) and association peripheral and central nervous systems. J Biol Chem 269, 14711−14714.
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 359

Bostrom, P., Andersson, L., Rutberg, M., Perman, J., Lidberg, U., Johansson, B. R., et al. De Luca, A., Progida, C., Spinosa, M. R., Alifano, P., & Bucci, C. (2008). Characterization of
(2007). SNARE proteins mediate fusion between cytosolic lipid droplets and are the Rab7K157N mutant protein associated with Charcot–Marie–Tooth type 2B.
implicated in insulin sensitivity. Nat Cell Biol 9, 1286−1293. Biochem Biophys Res Commun 372, 283−287.
Bostrom, P., Rutberg, M., Ericsson, J., Holmdahl, P., Andersson, L., Frohman, M. A., et al. De Vos, K. J., Chapman, A. L., Tennant, M. E., Manser, C., Tudor, E. L., Lau, K. F., et al. (2007).
(2005). Cytosolic lipid droplets increase in size by microtubule-dependent complex Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal
formation. Arterioscler Thromb Vasc Biol 25, 1945−1951. transport to reduce axonal mitochondria content. Hum Mol Genet 16, 2720−2728.
Bove, J., Prou, D., Perier, C., & Przedborski, S. (2005). Toxin-induced models of De Vos, K. J., Grierson, A. J., Ackerley, S., & Miller, C. C. (2008). Role of axonal transport in
Parkinson's disease. NeuroRx 2, 484−494. neurodegenerative diseases. Annu Rev Neurosci 31, 151−173.
Braunstein, K. E., Eschbach, J., Rona-Voros, K., Soylu, R., Mikrouli, E., Larmet, Y., et al. Decker, H., Lo, K. Y., Unger, S. M., Ferreira, S. T., & Silverman, M. A. (2010). Amyloid-beta
(2010). A point mutation in the dynein heavy chain gene leads to striatal atrophy peptide oligomers disrupt axonal transport through an NMDA receptor-dependent
and compromises neurite outgrowth of striatal neurons. Hum Mol Genet. doi: mechanism that is mediated by glycogen synthase kinase 3beta in primary cultured
10.1093/hmg/ddq1361. hippocampal neurons. J Neurosci 30, 9166−9171.
Breuer, A. C., Lynn, M. P., Atkinson, M. B., Chou, S. M., Wilbourn, A. J., Marks, K. E., et al. Deinhardt, K., Salinas, S., Verastegui, C., Watson, R., Worth, D., Hanrahan, S., et al.
(1987). Fast axonal transport in amyotrophic lateral sclerosis: an intra-axonal (2006). Rab5 and Rab7 control endocytic sorting along the axonal retrograde
organelle traffic analysis. Neurology 37, 738−748. transport pathway. Neuron 52, 293−305.
Brooks, B. R., Sanjak, M., Belden, D., Juhasz-Poscine, K., & Waclawik, A. (2000). Natural Delcroix, J. D., Valletta, J. S., Wu, C., Hunt, S. J., Kowal, A. S., & Mobley, W. C. (2003). NGF
history of amyotrophic lateral sclerosis. In R. H. J. Brown, V. Meininger, & M. Swash signaling in sensory neurons: evidence that early endosomes carry NGF retrograde
(Eds.), Amyotrophic Lateral Sclerosis (pp. 31−58). London: Dunitz. signals. Neuron 39, 69−84.
Brownlees, J., Ackerley, S., Grierson, A. J., Jacobsen, N. J., Shea, K., Anderton, B. H., et al. Deng, H. X., Zhai, H., Bigio, E. H., Yan, J., Fecto, F., Ajroud, K., et al. (2010). FUS-
(2002). Charcot–Marie–Tooth disease neurofilament mutations disrupt neurofila- immunoreactive inclusions are a common feature in sporadic and non-SOD1
ment assembly and axonal transport. Hum Mol Genet 11, 2837−2844. familial amyotrophic lateral sclerosis. Ann Neurol 67, 739−748.
Burkhardt, J. K., Echeverri, C. J., Nilsson, T., & Vallee, R. B. (1997). Overexpression Dequen, F., Filali, M., Lariviere, R. C., Perrot, R., Hisanaga, S., & Julien, J. P. (2010).
of the dynamitin (p50) subunit of the dynactin complex disrupts dynein- Reversal of neuropathy phenotypes in conditional mouse model of Charcot–Marie–
dependent maintenance of membrane organelle distribution. J Cell Biol 139, Tooth disease type 2E. Hum Mol Genet 19, 2616−2629.
469−484. Devon, R. S., Orban, P. C., Gerrow, K., Barbieri, M. A., Schwab, C., Cao, L. P., et al. (2006).
Burnett, B. G., & Pittman, R. N. (2005). The polyglutamine neurodegenerative protein Als2-deficient mice exhibit disturbances in endosome trafficking associated with
ataxin 3 regulates aggresome formation. Proc Natl Acad Sci U S A 102, 4330−4335. motor behavioral abnormalities. Proc Natl Acad Sci U S A 103, 9595−9600.
Campana, V., Sarnataro, D., & Zurzolo, C. (2005). The highways and byways of prion Dienstbier, M., & Li, X. (2009). Bicaudal-D and its role in cargo sorting by microtubule-
protein trafficking. Trends Cell Biol 15, 102−111. based motors. Biochem Soc Trans 37, 1066−1071.
Cataldo, A. M., Petanceska, S., Terio, N. B., Peterhoff, C. M., Durham, R., Mercken, M., et al. DiFiglia, M., Sapp, E., Chase, K., Schwarz, C., Meloni, A., Young, C., et al. (1995).
(2004). Abeta localization in abnormal endosomes: association with earliest Abeta Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain
elevations in AD and Down syndrome. Neurobiol Aging 25, 1263−1272. neurons. Neuron 14, 1075−1081.
Cataldo, A. M., Peterhoff, C. M., Troncoso, J. C., Gomez-Isla, T., Hyman, B. T., & Nixon, R. A. Ding, J., Allen, E., Wang, W., Valle, A., Wu, C., Nardine, T., et al. (2006). Gene targeting of
(2000). Endocytic pathway abnormalities precede amyloid beta deposition in GAN in mouse causes a toxic accumulation of microtubule-associated protein 8 and
sporadic Alzheimer's disease and Down syndrome: differential effects of APOE impaired retrograde axonal transport. Hum Mol Genet 15, 1451−1463.
genotype and presenilin mutations. Am J Pathol 157, 277−286. Dixit, R., Ross, J. L., Goldman, Y. E., & Holzbaur, E. L. (2008). Differential regulation of
Caviston, J. P., Ross, J. L., Antony, S. M., Tokito, M., & Holzbaur, E. L. (2007). Huntingtin dynein and kinesin motor proteins by tau. Science 319, 1086−1089.
facilitates dynein/dynactin-mediated vesicle transport. Proc Natl Acad Sci U S A 104, Dompierre, J. P., Godin, J. D., Charrin, B. C., Cordelieres, F. P., King, S. J., Humbert, S., et al.
10045−10050. (2007). Histone deacetylase 6 inhibition compensates for the transport deficit in
Chen, H. J., Anagnostou, G., Chai, A., Withers, J., Morris, A., Adhikaree, J., et al. (2010). Huntington's disease by increasing tubulin acetylation. J Neurosci 27, 3571−3583.
Characterisation of the properties of a novel mutation in VAPB in familial ALS. J Biol Driskell, O. J., Mironov, A., Allan, V. J., & Woodman, P. G. (2007). Dynein is required for
Chem 285(51), 40266−40281. receptor sorting and the morphogenesis of early endosomes. Nat Cell Biol 9,
Chen, X. J., Levedakou, E. N., Millen, K. J., Wollmann, R. L., Soliven, B., & Popko, B. (2007). 113−120.
Proprioceptive sensory neuropathy in mice with a mutation in the cytoplasmic Dupuis, L., & Echaniz-Laguna, A. (2010). Skeletal muscle in motor neuron diseases:
Dynein heavy chain 1 gene. J Neurosci 27, 14515−14524. therapeutic target and delivery route for potential treatments. Curr Drug Targets 11
Chevalier-Larsen, E., & Holzbaur, E. L. (2006). Axonal transport and neurodegenerative (10), 1250−1261.
disease. Biochim Biophys Acta 1762, 1094−1108. Dupuis, L., Fergani, A., Braunstein, K. E., Eschbach, J., Holl, N., Rene, F., et al. (2009). Mice
Chevalier-Larsen, E. S., Wallace, K. E., Pennise, C. R., & Holzbaur, E. L. (2008). Lysosomal with a mutation in the dynein heavy chain 1 gene display sensory neuropathy but
proliferation and distal degeneration in motor neurons expressing the G59S lack motor neuron disease. Exp Neurol 215, 146−152.
mutation in the p150Glued subunit of dynactin. Hum Mol Genet 17, 1946−1955. Dupuis, L., & Loeffler, J. P. (2009). Neuromuscular junction destruction during
Chow, C. Y., Landers, J. E., Bergren, S. K., Sapp, P. C., Grant, A. E., Jones, J. M., et al. (2009). amyotrophic lateral sclerosis: insights from transgenic models. Curr Opin
Deleterious variants of FIG4, a phosphoinositide phosphatase, in patients with ALS. Pharmacol 9, 341−346.
Am J Hum Genet 84, 85−88. Durr, A. (2010). Autosomal dominant cerebellar ataxias: polyglutamine expansions and
Clarkson, Y. L., Gillespie, T., Perkins, E. M., Lyndon, A. R., & Jackson, M. (2010). Beta-III beyond. Lancet Neurol 9, 885−894.
spectrin mutation L253P associated with spinocerebellar ataxia type 5 interferes El-Kadi, A. M., Bros-Facer, V., Deng, W., Philpott, A., Stoddart, E., Banks, G., et al. (2010). The
with binding to Arp1 and protein trafficking from the Golgi. Hum Mol Genet 19, legs at odd angles (Loa) mutation in cytoplasmic dynein ameliorates mitochondrial
3634−3641. function in SOD1G93A mouse model for motor neuron disease. J Biol Chem 285,
Colin, E., Regulier, E., Perrin, V., Durr, A., Brice, A., Aebischer, P., et al. (2005). Akt is 18627−18639.
altered in an animal model of Huntington's disease and in patients. Eur J Neurosci Engelender, S., Sharp, A. H., Colomer, V., Tokito, M. K., Lanahan, A., Worley, P., et al.
21, 1478−1488. (1997). Huntingtin-associated protein 1 (HAP1) interacts with the p150Glued
Colin, E., Zala, D., Liot, G., Rangone, H., Borrell-Pages, M., Li, X. J., et al. (2008). Huntingtin subunit of dynactin. Hum Mol Genet 6, 2205−2212.
phosphorylation acts as a molecular switch for anterograde/retrograde transport in Ermolayev, V., Cathomen, T., Merk, J., Friedrich, M., Hartig, W., Harms, G. S., et al.
neurons. EMBO J 27, 2124−2134. (2009a). Impaired axonal transport in motor neurons correlates with clinical prion
Cooper, A. A., Gitler, A. D., Cashikar, A., Haynes, C. M., Hill, K. J., Bhullar, B., et al. (2006). disease. PLoS Pathog 5, e1000558.
Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's Ermolayev, V., Friedrich, M., Nozadze, R., Cathomen, T., Klein, M. A., Harms, G. S., et al.
models. Science 313, 324−328. (2009b). Ultramicroscopy reveals axonal transport impairments in cortical motor
Corthesy-Theulaz, I., Pauloin, A., & Pfeffer, S. R. (1992). Cytoplasmic dynein participates neurons at prion disease. Biophys J 96, 3390−3398.
in the centrosomal localization of the Golgi complex. J Cell Biol 118, 1333−1345. Eschbach, J., Fergani, A., Oudart, H., Robin, J. P., Rene, F., de Aguilar, J. L., et al. (2011).
Cosker, K. E., Courchesne, S. L., & Segal, R. A. (2008). Action in the axon: generation and Mutations in cytoplasmic dynein lead to a Huntington's disease-like defect in energy
transport of signaling endosomes. Curr Opin Neurobiol 18, 270−275. metabolism of brown and white adipose tissues. Biochim Biophys Acta 1812(1), 59−69.
Cox, L. E., Ferraiuolo, L., Goodall, E. F., Heath, P. R., Higginbottom, A., Mortiboys, H., et al. Eymard-Pierre, E., Lesca, G., Dollet, S., Santorelli, F. M., di Capua, M., Bertini, E., et al.
(2010). Mutations in CHMP2B in lower motor neuron predominant amyotrophic (2002). Infantile-onset ascending hereditary spastic paralysis is associated with
lateral sclerosis (ALS). PLoS ONE 5, e9872. mutations in the alsin gene. Am J Hum Genet 71, 518−527.
Creppe, C., Malinouskaya, L., Volvert, M. L., Gillard, M., Close, P., Malaise, O., et al. (2009). Farrer, M. J., Hulihan, M. M., Kachergus, J. M., Dachsel, J. C., Stoessl, A. J., Grantier, L. L.,
Elongator controls the migration and differentiation of cortical neurons through et al. (2009). DCTN1 mutations in Perry syndrome. Nat Genet 41, 163−165.
acetylation of alpha-tubulin. Cell 136, 551−564. Filimonenko, M., Stuffers, S., Raiborg, C., Yamamoto, A., Malerod, L., Fisher, E. M., et al.
Crews, L., & Masliah, E. (2010). Molecular mechanisms of neurodegeneration in (2007). Functional multivesicular bodies are required for autophagic clearance of
Alzheimer's disease. Hum Mol Genet 19, R12−R20. protein aggregates associated with neurodegenerative disease. J Cell Biol 179,
Cuervo, A. M., Wong, E. S., & Martinez-Vicente, M. (2010). Protein degradation, 485−500.
aggregation, and misfolding. Mov Disord 25(Suppl. 1), S49−S54. Frederick, R. L., & Shaw, J. M. (2007). Moving mitochondria: establishing distribution of
Cui, L., Jeong, H., Borovecki, F., Parkhurst, C. N., Tanese, N., & Krainc, D. (2006). an essential organelle. Traffic 8, 1668−1675.
Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mito- Fricker, B., Muller, A., & Rene, F. (2008). Evaluation tools and animal models of
chondrial dysfunction and neurodegeneration. Cell 127, 59−69. peripheral neuropathies. Neurodegener Dis 5, 72−108.
Dagoneau, N., Goulet, M., Genevieve, D., Sznajer, Y., Martinovic, J., Smithson, S., et al. Fridolfsson, H. N., Ly, N., Meyerzon, M., & Starr, D. A. (2010). UNC-83 coordinates
(2009). DYNC2H1 mutations cause asphyxiating thoracic dystrophy and short rib- kinesin-1 and dynein activities at the nuclear envelope during nuclear migration.
polydactyly syndrome, type III. Am J Hum Genet 84, 706−711. Dev Biol 338, 237−250.
360 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

Fujita, Y., Ohama, E., Takatama, M., Al-Sarraj, S., & Okamoto, K. (2006). Fragmentation of Ilieva, H. S., Yamanaka, K., Malkmus, S., Kakinohana, O., Yaksh, T., Marsala, M., et al.
Golgi apparatus of nigral neurons with alpha-synuclein-positive inclusions in (2008). Mutant dynein (Loa) triggers proprioceptive axon loss that extends
patients with Parkinson's disease. Acta Neuropathol 112, 261−265. survival only in the SOD1 ALS model with highest motor neuron death. Proc Natl
Funke, A. D., Esser, M., Kruttgen, A., Weis, J., Mitne-Neto, M., Lazar, M., et al. (2010). The Acad Sci U S A 105, 12599−12604.
p.P56S mutation in the VAPB gene is not due to a single founder: the first European Jahreiss, L., Menzies, F. M., & Rubinsztein, D. C. (2008). The itinerary of autophago-
case. Clin Genet 77, 302−303. somes: from peripheral formation to kiss-and-run fusion with lysosomes. Traffic 9,
Gauthier, L. R., Charrin, B. C., Borrell-Pages, M., Dompierre, J. P., Rangone, H., Cordelieres, 574−587.
F. P., et al. (2004). Huntingtin controls neurotrophic support and survival of neurons Jensen, P. H., Nielsen, M. S., Jakes, R., Dotti, C. G., & Goedert, M. (1998). Binding of
by enhancing BDNF vesicular transport along microtubules. Cell 118, 127−138. alpha-synuclein to brain vesicles is abolished by familial Parkinson's disease
Gee, M. A., Heuser, J. E., & Vallee, R. B. (1997). An extended microtubule-binding mutation. J Biol Chem 273, 26292−26294.
structure within the dynein motor domain. Nature 390, 636−639. Jiang, Q., Ren, Y., & Feng, J. (2008). Direct binding with histone deacetylase 6 mediates the
Gitler, A. D., Bevis, B. J., Shorter, J., Strathearn, K. E., Hamamichi, S., Su, L. J., et al. (2008). reversible recruitment of parkin to the centrosome. J Neurosci 28, 12993−13002.
The Parkinson's disease protein alpha-synuclein disrupts cellular Rab homeostasis. Johansson, M., Rocha, N., Zwart, W., Jordens, I., Janssen, L., Kuijl, C., et al. (2007).
Proc Natl Acad Sci U S A 105, 145−150. Activation of endosomal dynein motors by stepwise assembly of Rab7-RILP-
Godin, J. D., Colombo, K., Molina-Calavita, M., Keryer, G., Zala, D., Charrin, B. C., et al. p150Glued, ORP1L, and the receptor betalll spectrin. J Cell Biol 176, 459−471.
(2010). Huntingtin is required for mitotic spindle orientation and mammalian Jordens, I., Fernandez-Borja, M., Marsman, M., Dusseljee, S., Janssen, L., Calafat, J., et al.
neurogenesis. Neuron 67, 392−406. (2001). The Rab7 effector protein RILP controls lysosomal transport by inducing the
Gonatas, N. K., Stieber, A., & Gonatas, J. O. (2006). Fragmentation of the Golgi apparatus recruitment of dynein–dynactin motors. Curr Biol 11, 1680−1685.
in neurodegenerative diseases and cell death. J Neurol Sci 246, 21−30. Jou, Y. S., & Myers, R. M. (1995). Evidence from antibody studies that the CAG repeat in the
Gonzalez de Aguilar, J. L., Echaniz-Laguna, A., Fergani, A., Rene, F., Meininger, V., Loeffler, J. P., Huntington disease gene is expressed in the protein. Hum Mol Genet 4, 465−469.
et al. (2007). Amyotrophic lateral sclerosis: all roads lead to Rome. J Neurochem 101(5), Kabashi, E., Valdmanis, P. N., Dion, P., Spiegelman, D., McConkey, B. J., Vande Velde, C.,
1153−1160. et al. (2008). TARDBP mutations in individuals with sporadic and familial
Gosavi, N., Lee, H. J., Lee, J. S., Patel, S., & Lee, S. J. (2002). Golgi fragmentation occurs in amyotrophic lateral sclerosis. Nat Genet 40, 572−574.
the cells with prefibrillar alpha-synuclein aggregates and precedes the formation of Kamal, A., Stokin, G. B., Yang, Z., Xia, C. H., & Goldstein, L. S. (2000). Axonal transport of
fibrillar inclusion. J Biol Chem 277, 48984−48992. amyloid precursor protein is mediated by direct binding to the kinesin light chain
Grbovic, O. M., Mathews, P. M., Jiang, Y., Schmidt, S. D., Dinakar, R., Summers-Terio, N. B., subunit of kinesin-I. Neuron 28, 449−459.
et al. (2003). Rab5-stimulated up-regulation of the endocytic pathway increases Kardon, J. R., & Vale, R. D. (2009). Regulators of the cytoplasmic dynein motor. Nat Rev
intracellular beta-cleaved amyloid precursor protein carboxyl-terminal fragment Mol Cell Biol 10, 854−865.
levels and Abeta production. J Biol Chem 278, 31261−31268. Karki, S., & Holzbaur, E. L. (1995). Affinity chromatography demonstrates a direct
Greenway, M. J., Andersen, P. M., Russ, C., Ennis, S., Cashman, S., Donaghy, C., et al. binding between cytoplasmic dynein and the dynactin complex. J Biol Chem 270,
(2006). ANG mutations segregate with familial and ‘sporadic’ amyotrophic lateral 28806−28811.
sclerosis. Nat Genet 38, 411−413. Katsuno, M., Adachi, H., Minamiyama, M., Waza, M., Tokui, K., Banno, H., et al. (2006).
Gross, S. P., Welte, M. A., Block, S. M., & Wieschaus, E. F. (2000). Dynein-mediated cargo Reversible disruption of dynactin 1-mediated retrograde axonal transport in
transport in vivo. A switch controls travel distance. J Cell Biol 148, 945−956. polyglutamine-induced motor neuron degeneration. J Neurosci 26, 12106−12117.
Gunawardena, S., Her, L. S., Brusch, R. G., Laymon, R. A., Niesman, I. R., Gordesky-Gold, B., Kawaguchi, Y., Kovacs, J. J., McLaurin, A., Vance, J. M., Ito, A., & Yao, T. P. (2003). The
et al. (2003). Disruption of axonal transport by loss of huntingtin or expression of deacetylase HDAC6 regulates aggresome formation and cell viability in response to
pathogenic polyQ proteins in Drosophila. Neuron 40, 25−40. misfolded protein stress. Cell 115, 727−738.
Gupta, A., Dawson, V. L., & Dawson, T. M. (2008). What causes cell death in Parkinson's Kimura, N., Inoue, M., Okabayashi, S., Ono, F., & Negishi, T. (2009). Dynein dysfunction
disease? Ann Neurol 64(Suppl. 2), S3−S15. induces endocytic pathology accompanied by an increase in Rab GTPases: a
Hadano, S., Benn, S. C., Kakuta, S., Otomo, A., Sudo, K., Kunita, R., et al. (2006). Mice potential mechanism underlying age-dependent endocytic dysfunction. J Biol Chem
deficient in the Rab5 guanine nucleotide exchange factor ALS2/alsin exhibit age- 284, 31291−31302.
dependent neurological deficits and altered endosome trafficking. Hum Mol Genet Kimura, N., & Yanagisawa, K. (2007). Endosomal accumulation of GM1 ganglioside-
15, 233−250. bound amyloid beta-protein in neurons of aged monkey brains. Neuroreport 18,
Hadano, S., Hand, C. K., Osuga, H., Yanagisawa, Y., Otomo, A., Devon, R. S., et al. (2001). A 1669−1673.
gene encoding a putative GTPase regulator is mutated in familial amyotrophic King, S. J., & Schroer, T. A. (2000). Dynactin increases the processivity of the cytoplasmic
lateral sclerosis 2. Nat Genet 29, 166−173. dynein motor. Nat Cell Biol 2, 20−24.
Hafezparast, M., Brandner, S., Linehan, J., Martin, J. E., Collinge, J., & Fisher, E. M. (2005). King, S. M., Barbarese, E., Dillman, J. F., 3rd, Benashski, S. E., Do, K. T., Patel-King, R. S.,
Prion disease incubation time is not affected in mice heterozygous for a dynein et al. (1998). Cytoplasmic dynein contains a family of differentially expressed light
mutation. Biochem Biophys Res Commun 326, 18−22. chains. Biochemistry 37, 15033−15041.
Hafezparast, M., Klocke, R., Ruhrberg, C., Marquardt, A., Ahmad-Annuar, A., Bowen, S., Krainc, D. (2010). Clearance of mutant proteins as a therapeutic target in neurode-
et al. (2003). Mutations in dynein link motor neuron degeneration to defects in generative diseases. Arch Neurol 67, 388−392.
retrograde transport. Science 300, 808−812. Kuhnlein, P., Sperfeld, A. D., Vanmassenhove, B., Van Deerlin, V., Lee, V. M., Trojanowski,
Harada, A., Takei, Y., Kanai, Y., Tanaka, Y., Nonaka, S., & Hirokawa, N. (1998). Golgi J. Q., et al. (2008). Two German kindreds with familial amyotrophic lateral sclerosis
vesiculation and lysosome dispersion in cells lacking cytoplasmic dynein. J Cell Biol due to TARDBP mutations. Arch Neurol 65, 1185−1189.
141, 51−59. Kunzi, V., Glatzel, M., Nakano, M. Y., Greber, U. F., Van Leuven, F., & Aguzzi, A. (2002).
Heerssen, H. M., Pazyra, M. F., & Segal, R. A. (2004). Dynein motors transport activated Unhampered prion neuroinvasion despite impaired fast axonal transport in
Trks to promote survival of target-dependent neurons. Nat Neurosci 7, 596−604. transgenic mice overexpressing four-repeat tau. J Neurosci 22, 7471−7477.
Heikenwalder, M., Julius, C., & Aguzzi, A. (2007). Prions and peripheral nerves: a deadly Kwiatkowski, T. J., Jr., Bosco, D. A., Leclerc, A. L., Tamrazian, E., Vanderburg, C. R., Russ, C.,
rendezvous. J Neurosci Res 85, 2714−2725. et al. (2009). Mutations in the FUS/TLS gene on chromosome 16 cause familial
Her, L. S., & Goldstein, L. S. (2008). Enhanced sensitivity of striatal neurons to axonal amyotrophic lateral sclerosis. Science 323, 1205−1208.
transport defects induced by mutant huntingtin. J Neurosci 28, 13662−13672. Lach, B., Grimes, D., Benoit, B., & Minkiewicz-Janda, A. (1992). Caudate nucleus
Hirokawa, N. (1998). Kinesin and dynein superfamily proteins and the mechanism of pathology in Parkinson's disease: ultrastructural and biochemical findings in
organelle transport. Science 279, 519−526. biopsy material. Acta Neuropathol 83, 352−360.
Hirokawa, N., Niwa, S., & Tanaka, Y. (2010). Molecular motors in neurons: transport Laforet, G. A., Sapp, E., Chase, K., McIntyre, C., Boyce, F. M., Campbell, M., et al. (2001).
mechanisms and roles in brain function, development, and disease. Neuron 68, Changes in cortical and striatal neurons predict behavioral and electrophysiological
610−638. abnormalities in a transgenic murine model of Huntington's disease. J Neurosci 21,
Hollenbeck, P. J., & Saxton, W. M. (2005). The axonal transport of mitochondria. J Cell Sci 9112−9123.
118, 5411−5419. Lagier-Tourenne, C., & Cleveland, D. W. (2009). Rethinking ALS: the FUS about TDP-43.
Holleran, E. A., Ligon, L. A., Tokito, M., Stankewich, M. C., Morrow, J. S., & Holzbaur, E. L. Cell 136, 1001−1004.
(2001). Beta III spectrin binds to the Arp1 subunit of dynactin. J Biol Chem 276, Lai, C., Lin, X., Chandran, J., Shim, H., Yang, W. J., & Cai, H. (2007). The G59S mutation in
36598−36605. p150(glued) causes dysfunction of dynactin in mice. J Neurosci 27, 13982−13990.
Holzbaur, E. L., & Vallee, R. B. (1994). DYNEINS: molecular structure and cellular Laifenfeld, D., Patzek, L. J., McPhie, D. L., Chen, Y., Levites, Y., Cataldo, A. M., et al. (2007).
function. Annu Rev Cell Biol 10, 339−372. Rab5 mediates an amyloid precursor protein signaling pathway that leads to
Hoogenraad, C. C., Akhmanova, A., Howell, S. A., Dortland, B. R., De Zeeuw, C. I., apoptosis. J Neurosci 27, 7141−7153.
Willemsen, R., et al. (2001). Mammalian Golgi-associated Bicaudal-D2 functions in Laird, F. M., Farah, M. H., Ackerley, S., Hoke, A., Maragakis, N., Rothstein, J. D., et al.
the dynein–dynactin pathway by interacting with these complexes. EMBO J 20, (2008). Motor neuron disease occurring in a mutant dynactin mouse model is
4041−4054. characterized by defects in vesicular trafficking. J Neurosci 28, 1997−2005.
Hubbert, C., Guardiola, A., Shao, R., Kawaguchi, Y., Ito, A., Nixon, A., et al. (2002). HDAC6 LaMonte, B. H., Wallace, K. E., Holloway, B. A., Shelly, S. S., Ascano, J., Tokito, M., et al.
is a microtubule-associated deacetylase. Nature 417, 455−458. (2002). Disruption of dynein/dynactin inhibits axonal transport in motor neurons
Hurley, J. H. (2008). ESCRT complexes and the biogenesis of multivesicular bodies. Curr causing late-onset progressive degeneration. Neuron 34, 715−727.
Opin Cell Biol 20, 4−11. La Spada, A. R., Wilson, E. M., Lubahn, D. B., Harding, A. E., & Fischbeck, K. H. (1991).
Ikeda, Y., Dick, K. A., Weatherspoon, M. R., Gincel, D., Armbrust, K. R., Dalton, J. C., et al. Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy.
(2006). Spectrin mutations cause spinocerebellar ataxia type 5. Nat Genet 38, Nature 352, 77−79.
184−190. Lazarov, O., Lee, M., Peterson, D. A., & Sisodia, S. S. (2002). Evidence that synaptically
Ilieva, H., Polymenidou, M., & Cleveland, D. W. (2009). Non-cell autonomous toxicity in released beta-amyloid accumulates as extracellular deposits in the hippocampus of
neurodegenerative disorders: ALS and beyond. J Cell Biol 187, 761−772. transgenic mice. J Neurosci 22, 9785−9793.
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 361

Lazarov, O., Morfini, G. A., Lee, E. B., Farah, M. H., Szodorai, A., DeBoer, S. R., et al. (2005). Neumann, M., Sampathu, D. M., Kwong, L. K., Truax, A. C., Micsenyi, M. C., Chou, T. T.,
Axonal transport, amyloid precursor protein, kinesin-1, and the processing et al. (2006). Ubiquitinated TDP-43 in frontotemporal lobar degeneration and
apparatus: revisited. J Neurosci 25, 2386−2395. amyotrophic lateral sclerosis. Science 314, 130−133.
Lee, H. J., Khoshaghideh, F., Lee, S., & Lee, S. J. (2006). Impairment of microtubule-dependent Nguyen, L., Humbert, S., Saudou, F., & Chariot, A. (2010). Elongator - an emerging role in
trafficking by overexpression of alpha-synuclein. Eur J Neurosci 24, 3153−3162. neurological disorders. Trends Mol Med 16, 1−6.
Lee, J. A., Beigneux, A., Ahmad, S. T., Young, S. G., & Gao, F. B. (2007). ESCRT-III dysfunc- Nishimura, A. L., Mitne-Neto, M., Silva, H. C., Richieri-Costa, A., Middleton, S., Cascio, D.,
tion causes autophagosome accumulation and neurodegeneration. Curr Biol 17, et al. (2004). A mutation in the vesicle-trafficking protein VAPB causes late-onset
1561−1567. spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75,
Lee, S. J., Chae, C., & Wang, M. M. (2009). p150/glued modifies nuclear estrogen receptor 822−831.
function. Mol Endocrinol 23, 620−629. Olzmann, J. A., Li, L., Chudaev, M. V., Chen, J., Perez, F. A., Palmiter, R. D., et al. (2007).
Lee, W. C., Yoshihara, M., & Littleton, J. T. (2004). Cytoplasmic aggregates trap Parkin-mediated K63-linked polyubiquitination targets misfolded DJ-1 to aggre-
polyglutamine-containing proteins and block axonal transport in a Drosophila somes via binding to HDAC6. J Cell Biol 178, 1025−1038.
model of Huntington's disease. Proc Natl Acad Sci U S A 101, 3224−3229. Ori-McKenney, K. M., Xu, J., Gross, S. P., & Vallee, R. B. (2010). A cytoplasmic dynein tail
Lees, A. J., Hardy, J., & Revesz, T. (2009). Parkinson's disease. Lancet 373, 2055−2066. mutation impairs motor processivity. Nat Cell Biol 12, 1228−1234.
Lefebvre, S., Burglen, L., Reboullet, S., Clermont, O., Burlet, P., Viollet, L., et al. (1995). Otomo, A., Hadano, S., Okada, T., Mizumura, H., Kunita, R., Nishijima, H., et al. (2003).
Identification and characterization of a spinal muscular atrophy-determining gene. ALS2, a novel guanine nucleotide exchange factor for the small GTPase Rab5, is
Cell 80, 155−165. implicated in endosomal dynamics. Hum Mol Genet 12, 1671−1687.
Levy, J. R., & Holzbaur, E. L. (2006). Cytoplasmic dynein/dynactin function and Ou, C. Y., Poon, V. Y., Maeder, C. I., Watanabe, S., Lehrman, E. K., Fu, A. K., et al. (2010).
dysfunction in motor neurons. Int J Dev Neurosci 24, 103−111. Two cyclin-dependent kinase pathways are essential for polarized trafficking of
Levy, J. R., Sumner, C. J., Caviston, J. P., Tokito, M. K., Ranganathan, S., Ligon, L. A., et al. presynaptic components. Cell 141, 846−858.
(2006). A motor neuron disease-associated mutation in p150Glued perturbs Palop, J. J., & Mucke, L. (2010). Amyloid-beta-induced neuronal dysfunction in
dynactin function and induces protein aggregation. J Cell Biol 172, 733−745. Alzheimer's disease: from synapses toward neural networks. Nat Neurosci 13,
Ligon, L. A., LaMonte, B. H., Wallace, K. E., Weber, N., Kalb, R. G., & Holzbaur, E. L. (2005). 812−818.
Mutant superoxide dismutase disrupts cytoplasmic dynein in motor neurons. Pandey, U. B., Nie, Z., Batlevi, Y., McCray, B. A., Ritson, G. P., Nedelsky, N. B., et al. (2007).
Neuroreport 16, 533−536. HDAC6 rescues neurodegeneration and provides an essential link between
Lin, S. X., & Collins, C. A. (1992). Immunolocalization of cytoplasmic dynein to autophagy and the UPS. Nature 447, 859−863.
lysosomes in cultured cells. J Cell Sci 101(Pt 1), 125−137. Pardo, R., Molina-Calavita, M., Poizat, G., Keryer, G., Humbert, S., & Saudou, F. (2010).
Lin, X., Parisiadou, L., Gu, X. L., Wang, L., Shim, H., Sun, L., et al. (2009). Leucine-rich pARIS-htt: an optimised expression platform to study huntingtin reveals functional
repeat kinase 2 regulates the progression of neuropathology induced by domains required for vesicular trafficking. Mol Brain 3, 17.
Parkinson's-disease-related mutant alpha-synuclein. Neuron 64, 807−827. Parkinson, N., Ince, P. G., Smith, M. O., Highley, R., Skibinski, G., Andersen, P. M., et al.
Lorenzo, D. N., Li, M. G., Mische, S. E., Armbrust, K. R., Ranum, L. P., & Hays, T. S. (2010). (2006). ALS phenotypes with mutations in CHMP2B (charged multivesicular body
Spectrin mutations that cause spinocerebellar ataxia type 5 impair axonal transport protein 2B). Neurology 67, 1074−1077.
and induce neurodegeneration in Drosophila. J Cell Biol 189, 143−158. Paschal, B. M., Shpetner, H. S., & Vallee, R. B. (1987). MAP 1C is a microtubule-activated ATPase
Magnani, E., Fan, J., Gasparini, L., Golding, M., Williams, M., Schiavo, G., et al. (2007). which translocates microtubules in vitro and has dynein-like properties. J Cell Biol 105,
Interaction of tau protein with the dynactin complex. EMBO J 26, 4546−4554. 1273−1282.
Mallik, R., Carter, B. C., Lex, S. A., King, S. J., & Gross, S. P. (2004). Cytoplasmic dynein Paschal, B. M., & Vallee, R. B. (1987). Retrograde transport by the microtubule-
functions as a gear in response to load. Nature 427, 649−652. associated protein MAP 1 C. Nature 330, 181−183.
Mallik, R., & Gross, S. P. (2004). Molecular motors: strategies to get along. Curr Biol 14, Peretti, D., Dahan, N., Shimoni, E., Hirschberg, K., & Lev, S. (2008). Coordinated lipid
R971−R982. transfer between the endoplasmic reticulum and the Golgi complex requires the
Marques, V. D., Barreira, A. A., Davis, M. B., Abou-Sleiman, P. M., Silva, W. A., Jr., Zago, M. A., VAP proteins and is essential for Golgi-mediated transport. Mol Biol Cell 19,
et al. (2006). Expanding the phenotypes of the Pro56Ser VAPB mutation: 3871−3884.
proximal SMA with dysautonomia. Muscle Nerve 34, 731−739. Perez-Olle, R., Jones, S. T., & Liem, R. K. (2004). Phenotypic analysis of neurofilament
Martinez-Vicente, M., Talloczy, Z., Wong, E., Tang, G., Koga, H., Kaushik, S., et al. (2010). light gene mutations linked to Charcot–Marie–Tooth disease in cell culture models.
Cargo recognition failure is responsible for inefficient autophagy in Huntington's Hum Mol Genet 13, 2207−2220.
disease. Nat Neurosci 13, 567−576. Perkins, E. M., Clarkson, Y. L., Sabatier, N., Longhurst, D. M., Millward, C. P., Jack, J., et al.
Matanis, T., Akhmanova, A., Wulf, P., Del Nery, E., Weide, T., Stepanova, T., et al. (2002). (2010). Loss of beta-III spectrin leads to Purkinje cell dysfunction recapitulating the
Bicaudal-D regulates COPI-independent Golgi-ER transport by recruiting the behavior and neuropathology of spinocerebellar ataxia type 5 in humans. J Neurosci
dynein–dynactin motor complex. Nat Cell Biol 4, 986−992. 30, 4857−4867.
Matsuda, N., Sato, S., Shiba, K., Okatsu, K., Saisho, K., Gautier, C. A., et al. (2010). PINK1 Perlson, E., Jeong, G. B., Ross, J. L., Dixit, R., Wallace, K. E., Kalb, R. G., et al. (2009). A
stabilized by mitochondrial depolarization recruits Parkin to damaged mitochon- switch in retrograde signaling from survival to stress in rapid-onset neurodegen-
dria and activates latent Parkin for mitophagy. J Cell Biol 189, 211−221. eration. J Neurosci 29, 9903−9917.
McCray, B. A., Skordalakes, E., & Taylor, J. P. (2010). Disease mutations in Rab7 result in Pfarr, C. M., Coue, M., Grissom, P. M., Hays, T. S., Porter, M. E., & McIntosh, J. R. (1990).
unregulated nucleotide exchange and inappropriate activation. Hum Mol Genet 19, Cytoplasmic dynein is localized to kinetochores during mitosis. Nature 345,
1033−1047. 263−265.
Middleton, F. A., & Strick, P. L. (2000). Basal ganglia and cerebellar loops: motor and Pfister, K. K., Fisher, E. M., Gibbons, I. R., Hays, T. S., Holzbaur, E. L., McIntosh, J. R., et al.
cognitive circuits. Brain Res Brain Res Rev 31, 236−250. (2005). Cytoplasmic dynein nomenclature. J Cell Biol 171, 411−413.
Mikami, A., Tynan, S. H., Hama, T., Luby-Phelps, K., Saito, T., Crandall, J. E., et al. (2002). Pfister, K. K., Shah, P. R., Hummerich, H., Russ, A., Cotton, J., Annuar, A. A., et al. (2006).
Molecular structure of cytoplasmic dynein 2 and its distribution in neuronal and Genetic analysis of the cytoplasmic dynein subunit families. PLoS Genet 2, e1.
ciliated cells. J Cell Sci 115, 4801−4808. Pigino, G., Morfini, G., Atagi, Y., Deshpande, A., Yu, C., Jungbauer, L., et al. (2009).
Millecamps, S., Salachas, F., Cazeneuve, C., Gordon, P., Bricka, B., Camuzat, A., et al. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal
(2010). SOD1, ANG, VAPB, TARDBP, and FUS mutations in familial amyotrophic amyloid beta. Proc Natl Acad Sci U S A 106, 5907−5912.
lateral sclerosis: genotype-phenotype correlations. J Med Genet 47, 554−560. Pilling, A. D., Horiuchi, D., Lively, C. M., & Saxton, W. M. (2006). Kinesin-1 and Dynein
Miller, K. E., & Sheetz, M. P. (2004). Axonal mitochondrial transport and potential are are the primary motors for fast transport of mitochondria in Drosophila motor
correlated. J Cell Sci 117, 2791−2804. axons. Mol Biol Cell 17, 2057−2068.
Misko, A., Jiang, S., Wegorzewska, I., Milbrandt, J., & Baloh, R. H. (2010). Mitofusin 2 is Porter, M. E., Bower, R., Knott, J. A., Byrd, P., & Dentler, W. (1999). Cytoplasmic dynein
necessary for transport of axonal mitochondria and interacts with the Miro/Milton heavy chain 1b is required for flagellar assembly in Chlamydomonas. Mol Biol Cell
complex. J Neurosci 30, 4232−4240. 10, 693−712.
Morfini, G., Pigino, G., Opalach, K., Serulle, Y., Moreira, J. E., Sugimori, M., et al. (2007). 1- Puls, I., Jonnakuty, C., LaMonte, B. H., Holzbaur, E. L., Tokito, M., Mann, E., et al. (2003).
Methyl-4-phenylpyridinium affects fast axonal transport by activation of caspase Mutant dynactin in motor neuron disease. Nat Genet 33, 455−456.
and protein kinase C. Proc Natl Acad Sci U S A 104, 2442−2447. Puls, I., Oh, S. J., Sumner, C. J., Wallace, K. E., Floeter, M. K., Mann, E. A., et al. (2005).
Morfini, G., Pigino, G., Szebenyi, G., You, Y., Pollema, S., & Brady, S. T. (2006). JNK Distal spinal and bulbar muscular atrophy caused by dynactin mutation. Ann Neurol
mediates pathogenic effects of polyglutamine-expanded androgen receptor on fast 57, 687−694.
axonal transport. Nat Neurosci 9, 907−916. Raiborg, C., & Stenmark, H. (2009). The ESCRT machinery in endosomal sorting of
Morfini, G. A., Burns, M., Binder, L. I., Kanaan, N. M., LaPointe, N., Bosco, D. A., et al. (2009). ubiquitylated membrane proteins. Nature 458, 445−452.
Axonal transport defects in neurodegenerative diseases. J Neurosci 29, 12776−12786. Ravikumar, B., Acevedo-Arozena, A., Imarisio, S., Berger, Z., Vacher, C., O'Kane, C. J., et al.
Moya, K. L., Hassig, R., Creminon, C., Laffont, I., & Di Giamberardino, L. (2004). Enhanced (2005). Dynein mutations impair autophagic clearance of aggregate-prone
detection and retrograde axonal transport of PrPc in peripheral nerve. J Neurochem proteins. Nat Genet 37, 771−776.
88, 155−160. Raychaudhuri, S., & Prinz, W. A. (2010). The Diverse Functions of Oxysterol-Binding
Munch, C., Sedlmeier, R., Meyer, T., Homberg, V., Sperfeld, A. D., Kurt, A., et al. (2004). Proteins. Annu Rev Cell Dev Biol 26, 157−177.
Point mutations of the p150 subunit of dynactin (DCTN1) gene in ALS. Neurology Reed, N. A., Cai, D., Blasius, T. L., Jih, G. T., Meyhofer, E., Gaertig, J., et al. (2006).
63, 724−726. Microtubule acetylation promotes kinesin-1 binding and transport. Curr Biol 16,
Narendra, D., Tanaka, A., Suen, D. F., & Youle, R. J. (2008). Parkin is recruited selectively 2166−2172.
to impaired mitochondria and promotes their autophagy. J Cell Biol 183, 795−803. Regehr, W. G., Carey, M. R., & Best, A. R. (2009). Activity-dependent regulation of
Narendra, D. P., Jin, S. M., Tanaka, A., Suen, D. F., Gautier, C. A., Shen, J., et al. (2010). synapses by retrograde messengers. Neuron 63, 154−170.
PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Rishal, I., & Fainzilber, M. (2010). Retrograde signaling in axonal regeneration. Exp
Biol 8, e1000298. Neurol 223, 5−10.
362 J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363

Rocha, N., Kuijl, C., van der Kant, R., Janssen, L., Houben, D., Janssen, H., et al. (2009). Stuffers, S., Brech, A., & Stenmark, H. (2009). ESCRT proteins in physiology and disease.
Cholesterol sensor ORP1L contacts the ER protein VAP to control Rab7-RILP-p150 Exp Cell Res 315, 1619−1626.
Glued and late endosome positioning. J Cell Biol 185, 1209−1225. Szebenyi, G., Morfini, G. A., Babcock, A., Gould, M., Selkoe, K., Stenoien, D. L., et al.
Ross, J. L., Wallace, K., Shuman, H., Goldman, Y. E., & Holzbaur, E. L. (2006). Processive (2003). Neuropathogenic forms of huntingtin and androgen receptor inhibit fast
bidirectional motion of dynein–dynactin complexes in vitro. Nat Cell Biol 8, 562−570. axonal transport. Neuron 40, 41−52.
Ross, J. L., Ali, M. Y., & Warshaw, D. M. (2008a). Cargo transport: molecular motors Takahashi, Y., Edamatsu, M., & Toyoshima, Y. Y. (2004). Multiple ATP-hydrolyzing sites
navigate a complex cytoskeleton. Curr Opin Cell Biol 20, 41−47. that potentially function in cytoplasmic dynein. Proc Natl Acad Sci U S A 101,
Ross, O. A., Braithwaite, A. T., Skipper, L. M., Kachergus, J., Hulihan, M. M., Middleton, F. A., 12865−12869.
et al. (2008b). Genomic investigation of alpha-synuclein multiplication and Tang, B. L. (2009). Neuronal protein trafficking associated with Alzheimer disease: from
parkinsonism. Ann Neurol 63, 743−750. APP and BACE1 to glutamate receptors. Cell Adhes Migr 3, 118−128.
Rubinsztein, D. C., Barton, D. E., Davison, B. C., & Ferguson-Smith, M. A. (1993). Analysis Terasaki, M. (1990). Recent progress on structural interactions of the endoplasmic
of the huntingtin gene reveals a trinucleotide-length polymorphism in the region reticulum. Cell Motil Cytoskeleton 15, 71−75.
of the gene that contains two CCG-rich stretches and a correlation between Teuling, E., van Dis, V., Wulf, P. S., Haasdijk, E. D., Akhmanova, A., Hoogenraad, C. C., et al.
decreased age of onset of Huntington's disease and CAG repeat number. Hum Mol (2008). A novel mouse model with impaired dynein/dynactin function develops
Genet 2, 1713−1715. amyotrophic lateral sclerosis (ALS)-like features in motor neurons and improves
Rui, Y., Tiwari, P., Xie, Z., & Zheng, J. Q. (2006). Acute impairment of mitochondrial lifespan in SOD1-ALS mice. Hum Mol Genet 17, 2849−2862.
trafficking by beta-amyloid peptides in hippocampal neurons. J Neurosci 26, Thayanidhi, N., Helm, J. R., Nycz, D. C., Bentley, M., Liang, Y., & Hay, J. C. (2010). Alpha-
10480−10487. synuclein delays endoplasmic reticulum (ER)-to-Golgi transport in mammalian
Rutherford, N. J., Zhang, Y. J., Baker, M., Gass, J. M., Finch, N. A., Xu, Y. F., et al. (2008). cells by antagonizing ER/Golgi SNAREs. Mol Biol Cell 21, 1850−1863.
Novel mutations in TARDBP (TDP-43) in patients with familial amyotrophic lateral Ticozzi, N., Silani, V., Leclerc, A. L., Keagle, P., Gellera, C., Ratti, A., et al. (2009). Analysis of
sclerosis. PLoS Genet 4, e1000193. FUS gene mutation in familial amyotrophic lateral sclerosis within an Italian cohort.
Saha, A. R., Hill, J., Utton, M. A., Asuni, A. A., Ackerley, S., Grierson, A. J., et al. (2004). Neurology 73(15), 1180−1185.
Parkinson's disease alpha-synuclein mutations exhibit defective axonal transport Topp, J. D., Gray, N. W., Gerard, R. D., & Horazdovsky, B. F. (2004). Alsin is a Rab5 and
in cultured neurons. J Cell Sci 117, 1017−1024. Rac1 guanine nucleotide exchange factor. J Biol Chem 279, 24612−24623.
Saksena, S., Sun, J., Chu, T., & Emr, S. D. (2007). ESCRTing proteins in the endocytic Trushina, E., Dyer, R. B., Badger, J. D., 2nd, Ure, D., Eide, L., Tran, D. D., et al. (2004).
pathway. Trends Biochem Sci 32, 561−573. Mutant huntingtin impairs axonal trafficking in mammalian neurons in vivo and in
Sasaki, S., & Iwata, M. (1996). Impairment of fast axonal transport in the proximal axons vitro. Mol Cell Biol 24, 8195−8209.
of anterior horn neurons in amyotrophic lateral sclerosis. Neurology 47, 535−540. Twig, G., Elorza, A., Molina, A. J., Mohamed, H., Wikstrom, J. D., Walzer, G., et al. (2008).
Sasaki, S., Warita, H., Abe, K., & Iwata, M. (2005). Impairment of axonal transport in the Fission and selective fusion govern mitochondrial segregation and elimination by
axon hillock and the initial segment of anterior horn neurons in transgenic mice autophagy. EMBO J 27, 433−446.
with a G93A mutant SOD1 gene. Acta Neuropathol 110, 48−56. Urwin, H., Authier, A., Nielsen, J. E., Metcalf, D., Powell, C., Froud, K., et al. (2010).
Satoh, D., Sato, D., Tsuyama, T., Saito, M., Ohkura, H., Rolls, M. M., et al. (2008). Spatial Disruption of endocytic trafficking in frontotemporal dementia with CHMP2B
control of branching within dendritic arbors by dynein-dependent transport of mutations. Hum Mol Genet 19, 2228−2238.
Rab5-endosomes. Nat Cell Biol 10, 1164−1171. van der Zee, J., Urwin, H., Engelborghs, S., Bruyland, M., Vandenberghe, R., Dermaut, B.,
Schnapp, B. J., & Reese, T. S. (1989). Dynein is the motor for retrograde axonal transport et al. (2008). CHMP2B C-truncating mutations in frontotemporal lobar degener-
of organelles. Proc Natl Acad Sci U S A 86, 1548−1552. ation are associated with an aberrant endosomal phenotype in vitro. Hum Mol
Schroer, T. A. (2004). Dynactin. Annu Rev Cell Dev Biol 20, 759−779. Genet 17, 313−322.
Schroer, T. A., Steuer, E. R., & Sheetz, M. P. (1989). Cytoplasmic dynein is a minus end- van Spronsen, M., & Hoogenraad, C. C. (2010). Synapse pathology in psychiatric and
directed motor for membranous organelles. Cell 56, 937−946. neurologic disease. Curr Neurol Neurosci Rep 10, 207−214.
Serulle, Y., Morfini, G., Pigino, G., Moreira, J. E., Sugimori, M., Brady, S. T., et al. (2007). 1- Vaughan, K. T. (2005). Microtubule plus ends, motors, and traffic of Golgi membranes.
Methyl-4-phenylpyridinium induces synaptic dysfunction through a pathway Biochim Biophys Acta 1744, 316−324.
involving caspase and PKCdelta enzymatic activities. Proc Natl Acad Sci U S A 104, Vaughan, K. T., & Vallee, R. B. (1995). Cytoplasmic dynein binds dynactin through a
2437−2441. direct interaction between the intermediate chains and p150Glued. J Cell Biol 131,
Shi, P., Strom, A. L., Gal, J., & Zhu, H. (2010). Effects of ALS-related SOD1 mutants on 1507−1516.
dynein- and KIF5-mediated retrograde and anterograde axonal transport. Biochim Vavlitou, N., Sargiannidou, I., Markoullis, K., Kyriacou, K., Scherer, S. S., & Kleopa, K. A.
Biophys Acta 1802, 707−716. (2010). Axonal pathology precedes demyelination in a mouse model of X-linked
Shrum, C. K., Defrancisco, D., & Meffert, M. K. (2009). Stimulated nuclear translocation demyelinating/type I Charcot–Marie–Tooth neuropathy. J Neuropathol Exp Neurol
of NF-kappaB and shuttling differentially depend on dynein and the dynactin 69, 945−958.
complex. Proc Natl Acad Sci U S A 106, 2647−2652. Verhoeven, K., De Jonghe, P., Coen, K., Verpoorten, N., Auer-Grumbach, M., Kwon, J. M.,
Simpson, C. L., Lemmens, R., Miskiewicz, K., Broom, W. J., Hansen, V. K., van Vught, P. W., et al. (2003). Mutations in the small GTP-ase late endosomal protein RAB7 cause
et al. (2009). Variants of the elongator protein 3 (ELP3) gene are associated with Charcot–Marie–Tooth type 2B neuropathy. Am J Hum Genet 72, 722−727.
motor neuron degeneration. Hum Mol Genet 18, 472−481. Vives-Bauza, C., Zhou, C., Huang, Y., Cui, M., de Vries, R. L., Kim, J., et al. (2010). PINK1-
Sinadinos, C., Burbidge-King, T., Soh, D., Thompson, L. M., Marsh, J. L., Wyttenbach, A., dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci U S A
et al. (2009). Live axonal transport disruption by mutant huntingtin fragments in 107(1), 378−383.
Drosophila motor neuron axons. Neurobiol Dis 34, 389−395. Vives-Bauza, C., Zhou, C., Huang, Y., Cui, M., de Vries, R. L., Kim, J., et al. (2010). PINK1-
Sisodia, S. S., Koo, E. H., Hoffman, P. N., Perry, G., & Price, D. L. (1993). Identification and dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci
transport of full-length amyloid precursor proteins in rat peripheral nervous U S A 107, 378−383.
system. J Neurosci 13, 3136−3142. Vossel, K. A., Zhang, K., Brodbeck, J., Daub, A. C., Sharma, P., Finkbeiner, S., et al. (2010).
Skibinski, G., Parkinson, N. J., Brown, J. M., Chakrabarti, L., Lloyd, S. L., Hummerich, H., Tau Reduction Prevents A{beta}-Induced Defects in Axonal Transport. Science 330
et al. (2005). Mutations in the endosomal ESCRTIII-complex subunit CHMP2B in (6001), 198.
frontotemporal dementia. Nat Genet 37, 806−808. Wagner, O. I., Ascano, J., Tokito, M., Leterrier, J. F., Janmey, P. A., & Holzbaur, E. L. (2004).
Spinosa, M. R., Progida, C., De Luca, A., Colucci, A. M., Alifano, P., & Bucci, C. (2008). The interaction of neurofilaments with the microtubule motor cytoplasmic dynein.
Functional characterization of Rab7 mutant proteins associated with Charcot– Mol Biol Cell 15, 5092−5100.
Marie–Tooth type 2B disease. J Neurosci 28, 1640−1648. Walker, F. O. (2007). Huntington's disease. Lancet 369, 218−228.
Splinter, D., Tanenbaum, M. E., Lindqvist, A., Jaarsma, D., Flotho, A., Yu, K. L., et al. (2010). Weydt, P., Pineda, V. V., Torrence, A. E., Libby, R. T., Satterfield, T. F., Lazarowski, E. R.,
Bicaudal D2, dynein, and kinesin-1 associate with nuclear pore complexes and et al. (2006). Thermoregulatory and metabolic defects in Huntington's disease
regulate centrosome and nuclear positioning during mitotic entry. PLoS Biol 8, transgenic mice implicate PGC-1alpha in Huntington's disease neurodegeneration.
e1000350. Cell Metab 4, 349−362.
Sreedharan, J., Blair, I. P., Tripathi, V. B., Hu, X., Vance, C., Rogelj, B., et al. (2008). TDP-43 Wikstrom, J. D., Twig, G., & Shirihai, O. S. (2009). What can mitochondrial heterogeneity
mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319, tell us about mitochondrial dynamics and autophagy? Int J Biochem Cell Biol 41,
1668−1672. 1914−1927.
Stankewich, M. C., Gwynn, B., Ardito, T., Ji, L., Kim, J., Robledo, R. F., et al. (2010). Williamson, T. L., & Cleveland, D. W. (1999). Slowing of axonal transport is a very early event
Targeted deletion of betaIII spectrin impairs synaptogenesis and generates ataxic in the toxicity of ALS-linked SOD1 mutants to motor neurons. Nat Neurosci 2, 50−56.
and seizure phenotypes. Proc Natl Acad Sci U S A 107, 6022−6027. Winslow, A. R., Chen, C. W., Corrochano, S., Acevedo-Arozena, A., Gordon, D. E., Peden, A. A.,
Stokin, G. B., Almenar-Queralt, A., Gunawardena, S., Rodrigues, E. M., Falzone, T., Kim, J., et al. (2010). alpha-Synuclein impairs macroautophagy: implications for Parkinson's
et al. (2008). Amyloid precursor protein-induced axonopathies are independent of disease. J Cell Biol 190, 1023−1037.
amyloid-beta peptides. Hum Mol Genet 17, 3474−3486. Wong, E., & Cuervo, A. M. (2010). Autophagy gone awry in neurodegenerative diseases.
Stokin, G. B., & Goldstein, L. S. (2006). Axonal transport and Alzheimer's disease. Annu Nat Neurosci 13, 805−811.
Rev Biochem 75, 607−627. Yang, Y., Hentati, A., Deng, H. X., Dabbagh, O., Sasaki, T., Hirano, M., et al. (2001). The
Stokin, G. B., Lillo, C., Falzone, T. L., Brusch, R. G., Rockenstein, E., Mount, S. L., et al. gene encoding alsin, a protein with three guanine-nucleotide exchange factor
(2005). Axonopathy and transport deficits early in the pathogenesis of Alzheimer's domains, is mutated in a form of recessive amyotrophic lateral sclerosis. Nat Genet
disease. Science 307, 1282−1288. 29, 160−165.
Strom, A. L., Shi, P., Zhang, F., Gal, J., Kilty, R., Hayward, L. J., et al. (2008). Interaction of Yokoseki, A., Shiga, A., Tan, C. F., Tagawa, A., Kaneko, H., Koyama, A., et al. (2008).
amyotrophic lateral sclerosis (ALS)-related mutant copper-zinc superoxide TDP-43 mutation in familial amyotrophic lateral sclerosis. Ann Neurol 63, 538−542.
dismutase with the dynein–dynactin complex contributes to inclusion formation. Yoshida, T., Takanari, H., & Izutsu, K. (1992). Distribution of cytoplasmic and axonemal
J Biol Chem 283, 22795−22805. dyneins in rat tissues. J Cell Sci 101(Pt 3), 579−587.
J. Eschbach, L. Dupuis / Pharmacology & Therapeutics 130 (2011) 348–363 363

Yudin, D., Hanz, S., Yoo, S., Iavnilovitch, E., Willis, D., Gradus, T., et al. (2008). Localized Zheng, Y., Wildonger, J., Ye, B., Zhang, Y., Kita, A., Younger, S. H., et al. (2008). Dynein is
regulation of axonal RanGTPase controls retrograde injury signaling in peripheral required for polarized dendritic transport and uniform microtubule orientation in
nerve. Neuron 59, 241−252. axons. Nat Cell Biol 10, 1172−1180.
Zala, D., Colin, E., Rangone, H., Liot, G., Humbert, S., & Saudou, F. (2008). Phosphory- Zhou, X., Ikenoue, T., Chen, X., Li, L., Inoki, K., & Guan, K. L. (2009). Rheb controls
lation of mutant huntingtin at S421 restores anterograde and retrograde transport misfolded protein metabolism by inhibiting aggresome formation and autophagy.
in neurons. Hum Mol Genet 17, 3837−3846. Proc Natl Acad Sci U S A 106, 8923−8928.
Zhang, B., Tu, P., Abtahian, F., Trojanowski, J. Q., & Lee, V. M. (1997). Neurofilaments and Zuccato, C., Ciammola, A., Rigamonti, D., Leavitt, B. R., Goffredo, D., Conti, L., et al.
orthograde transport are reduced in ventral root axons of transgenic mice that (2001). Loss of huntingtin-mediated BDNF gene transcription in Huntington's
express human SOD1 with a G93A mutation. J Cell Biol 139, 1307−1315. disease. Science 293, 493−498.
Zhang, F., Strom, A. L., Fukada, K., Lee, S., Hayward, L. J., & Zhu, H. (2007). Interaction Zweifel, L. S., Kuruvilla, R., & Ginty, D. D. (2005). Functions and mechanisms of
between familial amyotrophic lateral sclerosis (ALS)-linked SOD1 mutants and the retrograde neurotrophin signalling. Nat Rev Neurosci 6, 615−625.
dynein complex. J Biol Chem 282, 16691−16699.
Zhang, X., Lei, K., Yuan, X., Wu, X., Zhuang, Y., Xu, T., et al. (2009). SUN1/2 and Syne/
Nesprin-1/2 complexes connect centrosome to the nucleus during neurogenesis
and neuronal migration in mice. Neuron 64, 173−187.

You might also like