Cell Tissue Res DOI 10.1007/s00441-014-2082-7

REVIEW

Neurofilament dynamics and involvement in neurological disorders Benoit J. Gentil & Michael Tibshirani & Heather D. Durham

Received: 9 October 2014 / Accepted: 19 November 2014 # Springer-Verlag Berlin Heidelberg 2015

Abstract Neurons are extremely polarised cells in which the cytoskeleton, composed of microtubules, microfilaments and neurofilaments, plays a crucial role in maintaining structure and function. Neurofilaments, the 10-nm intermediate filaments of neurons, provide structure and mechanoresistance but also provide a scaffolding for the organization of the nucleus and organelles such as mitochondria and ER. Disruption of neurofilament organization and expression or metabolism of neurofilament proteins is characteristic of certain neurological syndromes including Amyotrophic Lateral Sclerosis, Charcot-Marie-Tooth sensorimotor neuropathies and Giant Axonal Neuropathy. Microfluorometric live imaging techniques have been instrumental in revealing the dynamics of neurofilament assembly and transport and their functions in organizing intracellular organelle networks. The insolubility of neurofilament proteins has limited identifying interactors by conventional biochemical techniques but yeast two-hybrid experiments have revealed new roles for oligomeric, nonfilamentous structures including vesicular trafficking. Although having long half-lives, new evidence points to degradation of subunits by the ubiquitin–proteasome system as a mechanism of normal turnover. Although certain E3-ligases ubiquitinating neurofilament proteins have been identified, the overall process of neurofilament degradation is not well understood. We review these mechanisms of neurofilament homeostasis and abnormalities in motor neuron and peripheral nerve disorders. Much remains to discover about the disruption of processes that leads to their pathological aggregation and accumulation and the relevance to pathogenesis. Understanding these mechanisms is crucial for identifying novel therapeutic strategies. B. J. Gentil (*) : M. Tibshirani : H. D. Durham Department of Neurology/Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada e-mail: [email protected]

Keywords Neurofilaments . Charcot-Marie-Tooth disease . Peripheral neuropathies . Amyotrophic Lateral Sclerosis

Introduction This review summarises the biology of neurofilaments (NF)s, with an emphasis on recently discovered aspects of assembly and dynamics and the role of NFs in pathogenesis of neurodegenerative disease, in particular Charcot-Marie-Tooth (CMT) peripheral neuropathies and amyotrophic lateral sclerosis (ALS). Neurons that are affected in CMT and ALS are highly polarized, extending axons long distances into the periphery to form neuromuscular junctions and sensory receptors in muscle and skin. Their cytoskeleton, composed of actin microfilaments, microtubules and NFs, provides the tracks along which transport may occur as well as a frame for determining cellular morphology . Microtubules are long tracks made of tubulin and distributed throughout mature neurons; they are responsible for long-distance bidirectional transport (see, for review, Conde and Caceres 2009). Importantly, mature neurons contain a large population of stable microtubules, resistant to the microtubule depolymerizing agent nocodazole, with half-lives exceeding several hours compared to several minutes (Baas and Black 1990; Baas et al. 1993; Brown et al. 1993). The role of actin in axonal growth cones, developing dendrites and dendritic spines is particularly well documented (Kapitein and Hoogenraad 2011). However, actin is present as short, branched filaments of 1.5–2 μm oriented more or less perpendicular to the plasma membrane, whereas a subset of actin is aligned to microtubules in dendrites and axons, which suggest that actin plays a role in short random and local movement (Fifkova and Delay 1982; Markham and Fifkova 1986; Bearer and Reese 1999; Bearer et al. 1999; Ligon and Steward 2000a, b; Mironov 2006; Mironov and Symonchuk 2006). Finally, IFs form non-polar

Cell Tissue Res

structrues, are characterised by their poor solubility in detergents and are tissue-specific where they serve several functions. Neurofilaments are the 10-nm intermediate filaments specific to neurons Neurofilament (NF) proteins are referred to as type IV intermediate filament (IF) proteins and are exclusively expressed in neurons in both the central (CNS) and peripheral (PNS) nervous systems. NFs in mature neurons are composed of the NF triplet proteins: the light neurofilament protein (NFL, 68 kDa) and two other subunits referred to as NFM (mid-sized 160 kDa) and NFH (high molecular weight 205 kDa). They can also include α-internexin (66 kDa), as described in spinal cord and optic nerve (Yuan, et al. 2006b), and peripherin (58 kDa), particularly in small-diameter sensory neurons of the dorsal root ganglia (Escurat et al. 1990). Expression of these NF proteins is developmentally regulated and reflects neuronal maturation. The following sequence of IF expression is observed in neurons as a result of maturation: NFL and NFM proteins are expressed after vimentin in murine central and peripheral neurons at days 9–10 post-gestation (Cochard and Paulin 1984), whereas NFH appears later as a result of axonal maturation concomitant with myelination (Haynes et al. 2005). α-internexin is expressed early in embryonic neuronal differentiation and then declines in the PNS but is retained in certain regions of the CNS particularly in neurons of the optic nerve (Kaplan et al. 1990). Following α-internexin, peripherin is expressed, being most abundant in late embryonic development and early postnatal brain. Peripherin expression declines in mature neurons being prominent post-natally only in smaller diameter dorsal root ganglia sensory neurons (Escurat et al. 1990). Following neuronal injury, developmentally regulated IF can be re-expressed (e.g., peripherin in motor neurons) (Troy et al. 1990; Beaulieu et al. 2002). NF proteins have the classical IF structure of an amino terminal ‘head’ domain, a central α-helical 'rod' domain required for assembly and a variable length carboxy terminal ‘tail’ domain. The central domains of the NF proteins retain a high degree of homology and mediate polymer assembly. The core human NF proteins α-internexin, peripherin and NFL can form homopolymers or co-assemble into filamentous structures but NFM and NFH cannot; they form heteropolymers with a core NF protein. The head and tail domains are variable, although the latter is generally rich in glutamine repeats and in NFM and NFH contains multiple KSP repeat domains that can be highly phosphorylated, particularly in axons. Neurofilament assembly The head and tail domains of the NF triplet proteins have multiple phosphorylation sites that are involved in regulation

of NF assembly, interaction with other cytoskeletal elements and organelles and transport of NF oligomers (Sihag and Nixon 1989, 1990, 1991; Giasson et al. 1996; Nakamura et al. 2000). The C-termini of NFM and NFH protrude from the filament core and form crossbridges to other NFs (Beck et al. 2010), microtubules and organelles (Rao et al. 2002a). The phosphorylation state of this domain is believed to participate in spacing cytoskeletal elements and to contribute to radial growth and diameter of axons (Dale and Garcia 2012). The α-helical rod domain of the NF subunits contains long stretches of heptad repeats that align in a head-to-tail fashion to form coiled-coil dimers. The dimers undergo antiparallel association to form tetramers, which then assemble into ropelike long filaments of 10 nm diameter (Herrmann and Aebi 2000). These filaments are insoluble in non-ionic detergents such as Triton X-100. To determine the composition of NF and other IF, buffers containing urea have been used to solubilise them into intermediate oligomeric forms (Carden and Eagles 1986). Dialysis of concentrated guanidine or urea solutions containing solubilised subunits results in limited reassembly of IFs in vitro (Liem and Hutchison 1982; Zackroff et al. 1982; Cohlberg et al. 1987, 1995; Hatzfeld and Weber 1990; Steinert 1990), to what are considered physiologically relevant assembly intermediates (Athlan and Mushynski 1997). NFs have long been considered as obligate heteropolymers of NFL, NFM and NFH in a ratio of 4:2:1 (Scott et al. 1985). However, NFs can also exist as heteropolymers of NFL/α-internexin/ NFM and NFH in a ratio of 4:2:2:1 in the optic nerve and mature CNS (Yuan et al. 2006b) and in heteropolymers of NFL/NFM/peripherin and NFH in a ratio of 4:2:1:1 (Yuan et al. 2012). Interestingly, while NFM and NFH require a core subunit such as NFL to assemble (Leung and Liem 1996), a contingent of NFL homodimers was detected in samples of mouse spinal cord and axons of cultured murine dorsal root ganglia (DRG) sensory neurons using a modified version of blue native-polyacrylamide gel electrophoresis (BN-PAGE) (Athlan and Mushynski 1997). IFs differ from microfilaments or microtubules in not being polarized. IFs lengthen by ‘end-to-end annealing’ of existing oligopolymers, rather than individual subunit proteins (Herrmann et al. 1999; Wickert et al. 2005; Uchida et al. 2013). Moreover, subunit exchange can occur along the entire length of the filament (intercalary subunit exchange) (Colakoglu and Brown 2009). NFs as long as 183 μm have been measured in cultured mouse cortical neurons (Brown 1997; Uchida et al. 2013) and in bullfrog olfactory nerve, average NF length was 118 μm (Burton and Wentz 1992). Phosphorylation of serine residues of the head domain of NFL (serine 2, serine 51 and serine 55) and NFM (serine 23) by PKA and/or PKC plays a major role in regulating NF disassembly/assembly (Sihag et al. 1988, 1999; Sihag and Nixon 1991; Doroudchi and Durham 1996; Giasson and Mushynski 1996, 1998). Polymerisation of rat NFL protein in vitro is also regulated by

Cell Tissue Res

protein kinase N (Manser et al. 2008). Interestingly, the chaperone protein HSP27 plays a role in maintaining NF proteins in the detergent soluble fraction and is believed to be important for chaperoning oligomeric structures (Ackerley et al. 2006). Uchida et al. followed NF assembly and disassembly in cultured cortical neurons by expressing NF proteins fused to proteins that convert from green to red fluorescence upon photoactivation. Following activation of a region of a neuron, redistribution of “red” segments of NF into those in the nonactivated region (green) was observed, reflecting fragmentation and reassembly. The time course of the dynamic formation of chimeric NFs was estimated at 270 min (Uchida et al. 2013). Given detergent insolubility and dynamics of NF polymers, it is likely that NF severing occurs through an active process but the mechanism is unknown. Neurofilament transport NFs move along microtubule tracks driven by the motor protein dynein for retrograde transport and kinesin 1A for anterograde transport (Uchida et al. 2009), the latter controlled by Cdk5/p35 (Holmgren et al. 2013). Early pulse chase studies showed coordinate slow anterograde axonal transport of radiolabelled NF proteins at a rate of 0.1–1 mm/day (Hoffman and Lasek 1975; Lasek et al. 1992; Millecamps and Julien 2004). These results implied assembly of NFs in perikarya and transport as filaments to nerve endings at this rate. However, the use of time–lapse live imaging of fluorescently tagged NF proteins expressed in cultured neurons demonstrated that NF transport is more complex. NFs alternate between two distinct kinetic states: a mobile state where they rapidly move short distances at the rate of fast transport with interruptions of transport and a more stationary phase (Brown et al. 2005; Trivedi et al. 2007). Interestingly, the average length of moving NFs is relatively short ranging from 4.1 to 8.6 μm (Trivedi et al. 2007). Longer NFs appear stationary. It is not fully understood how pausing of NFs is regulated but certain evidence supports phosphorylation of C-terminal domains being one of the signals. Immunoreactivity of the C-terminal phosphoepitope recognized by the antibody RT97 coincided with decreased association of NFH and kinesin, implying reduced transport (Yabe et al. 1999), whereas inhibition of Cdk5/p35 activity, known to phosphorylate KSP subdomains, accelerated NFH transport (Ackerley et al. 2003). Mutations of Cdk5/p35 sites in KSP subdomains of NFH, in order to mimic permanent phosphorylation, decreased NFH transport in cultured neurons. On the other hand, the rate of NF transport was normal in genetically modified (knockin) mice expressing NFH or NFM with C-terminal truncations, or NFH or NFM with all KSP repeats deleted. However, in these mice, only one of NFM or NFH was modified; phosphorylation of the remaining subunit was enhanced, leaving the possibility that the KSP domains of either subunit might subserve the

transport function (Rao et al. 2002b, 2003; Garcia et al. 2003; Yuan et al. 2003, 2006a). Finally, it is difficult to study transport and identify the regulating signals independent of NF assembly because of the relationship between transport rate and NF length (Gibb et al. 1998; Ackerley et al. 2003; Trivedi et al. 2007; Yates et al. 2009; Uchida et al. 2013). Neurofilament metabolism Studies of NF protein turnover show a particularly long halflife. Using a Tet off system in order to stop expression of a human NFL transgene in mice, half-life was estimated at 3 weeks (Millecamps et al. 2007). Similar NF protein halflives were found in retinal ganglion cell axons following injection of radiolabelled proline into optic nerve (Nixon and Logvinenko 1986). Interestingly, approximately 10 % of radiolabelled NF proteins remained for several months suggesting existence of a very stable pool (Pant 1988). Little is known about mechanisms regulating NF turnover. Proteases with affinity for dephosphorylated NF proteins, including calcium-activated proteases like calpain, are present in axons and can degrade NFs (Pant 1988). However, this proteolytic system is more likely activated following injury rather than playing a major role in normal NF turnover. Evidence for a role of the ubiquitin proteasome system in NF degradation is accumulating. Although the mechanisms responsible for regulating ubiquitination of NF proteins have yet to be determined in detail, the E3 ligase tripartite motif protein 2 (TRIM2) can mediate ubiquitination of NFL (Balastik et al. 2008), whereas NFM is a target of the E3 ligase CHIP (Wang et al. 2011). The E3 adaptor, gigaxonin, also targets NF proteins, peripherin and other IF proteins to the proteasome for degradation and loss of function mutation leads to IF accumulation in the disorder Giant Axonal Neuropathy (GAN) (Mahammad et al. 2013). NF accumulation is also a key feature of in vivo knockout of Ubiquitin Specific Protease 14 expression (USP14), a component of the 19S proteasome subunit regulating de-ubiquitinylation of proteins entering the proteasome (Crimmins et al. 2006; Chen et al. 2009; Mialki et al. 2013; Peth et al. 2013). What is uncertain is how IF proteins, which naturally form oligomers, are converted to monomeric form to undergo proteasome degradation. Neurofilament functions Neurons are polarised cells with highly specialised compartments (perikaryon, dendrites, axon) in which NF proteins have a role in neuronal organisation. The NF network’s mechanical properties provide protection from repetitive mechanical stress that occurs with movement. The viscoelastic properties of NFs are influenced by subunit composition, with NFH and NFM playing a key role in increasing stretching properties by lowering resistance (Brown et al. 1998). NFs

Cell Tissue Res

in vitro could be stretched and remain stretched at more than 2-fold their original length. This stretching induced a drastic reduction in filament diameter and a lateral aggregation of stretched polypeptides consistent with an α-helix to β-sheet transition of the coiled-coil domains as described for the myosin coil domain (Kreplak et al. 2005). Much of what we have learned about NF function has been a result of genetic manipulation of the expression of NF subunits in mice and cultured cells. NFs are essential for radial growth and maintenance of axonal calibre and for myelination, which are major determinants of the speed of action potential conduction (Zhu et al. 1997; Elder et al. 1998a, b; Jacomy et al. 1999). The formation of NFs and more importantly the extension of NFH and NFM tail domains from the filament core is crucial for spacing of cytoskeletal components and regulating axonal calibre (Rao et al. 2003). The NFM tail domain is particularly important in myelin-dependent radial growth of the axon as mice genetically modified to express a C-terminal truncated form show reduced axon caliber and slower nerve conduction speed (Garcia et al. 2003); however, phosphorylation of KSP repeats is not essential in this process (Garcia et al. 2009). Expansion of KSP repeats in the NFM tail domain has shown to increase axonal calibre in mice (Barry et al. 2012). It is interesting to note that the number of KSP repeats within NFM increases with head–body length in mammals suggesting that this subdomain could contribute to an increase in axonal calibre and nerve conduction velocity as larger mammals evolved (Barry et al. 2012). While there is no doubt about the role of NF proteins in myelination, little is known about the signalling mechanism to Schwann cells. Consistent with those functions, NFL knockout mice are viable and functional but exhibit an absence of NF, smaller axonal calibre and reduced myelination (Zhu et al. 1997; Ishihara et al. 2001). This lack of NFs in NFL−/− mice demonstrates the key role of NFL in the assembly of the other subunits. Peripherin gene knockout in mice has limited effects on axonal calibre and myelination, consistent with expression in small calibre dorsal root ganglia sensory neurons but does disrupt development of this subset of neurons (Lariviere et al. 2002). Finally, α-internexin knockout mice do not show any abnormal developmental phenotype related to axonal growth or changes in axonal calibre but do show an absence of filaments, consistent with persistent expression in the optic system (Kaplan et al. 1990; Levavasseur et al. 1999). In some of these mouse models, loss of NFs was accompanied by increase in tubulin expression and microtubule content that could compensate in part for depletion of NFs (Liu et al. 2013). NFs, as other IFs, are networked with other cytoskeletal elements to organize the cellular environment and position organelles. For example, NFs have a role in positioning of the

nucleus through protein complexes called ‘linkers of the nucleoskeleton and cytoskeleton’ (LINC) (Mellad et al. 2011). The LINC is a multiprotein complex that anchors nuclear lamins and IFs. An important component of the LINC is nesprin3 (Morgan et al. 2011), which interacts with intermediate filaments on the nuclear outer membrane and organizes and/or stabilizes the perinuclear cytoskeleton. NFs form a network around the nucleus (Fig. 1) suggesting a possible interaction between NFs and nesprin3. Interestingly, mutations in nesprin cause a complex disease, called autosomal recessive arthrogryposis, which has a phenotype resembling Charcot-Marie-Tooth caused by mutations in NEFL (Attali et al. 2009). NFs have a role in organising the cellular architecture. NFL has a role in positioning of axonal mitochondria and endoplasmic reticulum (Rao et al. 2002a, 2011) and is an important regulator of mitochondrial morphology, dynamics and motility (Gentil et al. 2011). NFL may also bind to other cytoskeletal components though linkers such as plectins, which mediate interactions with the actin microfilament network (Foisner et al. 1988). NFs also anchor at the plasmalemma, possibly through β-spectrins (Macioce et al. 1999), plectins (Wiche and Winter 2011), plakophilin 4 or Neural plakophilin-related armadillo protein (Koutras and Levesque 2011). Functions associated with NF proteins are believed to be through the formation of filamentous structures; however, whether NF proteins have functions independent of formation of filaments has been difficult to identify because of their insolubility in buffers amenable to classical methods of studying protein– protein interactions. Interacting partners have so far been found by immunoprecipitation of Triton X-100 soluble small oligomers, representing only 10 % of the total amount of NFs and may not reflect the full range of NF functions. NF oligomers have been identified that could subserve nonstructural functions as well as serve as mediators of subunit exchange within NFs. As a function associated with non-structural functions, NFL oligomers have been found to interact with NMDA receptors (Ehlers et al. 1998). Related to this, a role for fragments of α-internexin in NMDA receptor signal transduction also has been reported. After activation of NMDA receptors, α-internexin protected the retrograde signalling of ERK1/2 to the nucleus by hindering desphosphorylation of ERK1/2 (Karpova et al. 2013). Functions in signalling pathways have also been identified through screening a yeast two-hybrid library using NFL as bait (Table 1). We identified the U1 small nuclear ribonucleoprotein 70 kDa polypeptideA, Snw1 and Smarcd3 as NFL binding partners, proteins that are involved in RNA splicing and epigenetic control of gene transcription. Other NFL partners (i.e., necdin and amyloid precursor like protein 1) are involved in neurite outgrowth and plasticity and might be linked to the normal role of NFL in dendritic arborization (Zhang

Cell Tissue Res

Fig. 1 Confocal imaging of NFH (a) labelling by indirect immunocytochemistry in cultured motor neurons showing the NF network surrounding the nucleus and extending into the processes. Scale bar 10 μm. Confocal image centered on the motor neuron’s nucleus showing co-immunolabelling of NFH (b) and Nesprin3 (c) and staining of nucleus with Hoechst (d). The merged image (e) shows

Nesprin3 (red) labelling as punctate throughout the neuronal cell body but is concentrated around the nucleus . Scale bar 15 μm. The area delineated by the white square is magnified in e’ and magnification of the perinuclear region shows the relationship of Nesprin3 (red), NFH (green) and nucleus (blue), where Nesprin3 and filamentous NFH are adjacent (arrow) (blue) Scale bar (e’) 1 μm

et al. 2002). Our group also identified new interacting partners for peripherin involved in vesicle exocytosis (Gentil et al. 2014). Altogether, these observations suggest that NF proteins may have functions beyond those associated with being components of neurofilaments. Such roles have long been suspected but the evidence is now emerging.

Neurofilament disorders and disease mechanisms NFs abnormalities may cause a wide spectrum of disorders but are particularly associated with diseases affecting neurons projecting on long distance and with thick axons. For this review, we will focus particularly on the effect of neurofilament mutations and abnormalities in Charcot-Marie-Tooth

Cell Tissue Res Table 1

NFL-interacting proteins identified by screening a murine brain library using the yeast two-hybrid technique

Interacting protein

Bait

Number of clones

Functions

OMIM

U1snRNP70 Smarcd3 Snw1 HSP8A (HSC70)

NFL NFL NFL NFL

1 3 1 2

Splicing Chromatin remodelling Splicing Chaperonne

180740 601737 603055 600816

Necdin P116RIP APLP1

NFL NFL NFL

1 2 1

Neuritogenesis Neurites outgrowth Neuronal plasticity

612935 602117 104775

Yeast transformations and two-hybrid screens were carried out using L40 yeast as previously described (Deloulme et al. 2003; Gentil et al. 2014). Briefly, pLex9-NFL was used as bait and was transformed singly into L40 yeast. Cells were grown on DOB-UW then transformed with a mouse brain cDNA library (Clonetech) using the YEAST-1 kit according to the manufacturer’s instructions (Sigma-Aldrich). Primary transformants were analyzed on DOBUWLH medium plates to select for interaction and sequencing was performed on plasmid isolated using Yeast RPM® Kit (Q-BIOgen)

disease, Giant Axonal Neuropathy, diabetic neuropathy and Amyotrophic Lateral Sclerosis, which have common axonal dysfunction in the PNS.

Peripheral neuropathies: CMT, giant axonal neuropathy and diabetic neuropathy Charcot-Marie-Tooth disease More than 30 genes involved in Schwann cells function (CMT1) or neuronal homeostasis (CMT2) are affected in the inherited peripheral neuropathy Charcot-Marie-Tooth disease (CMT) causing distal muscle weakness. A direct role of NFs in peripheral neuropathies is demonstrated by mutations in the NEFL gene causing CMT. NEFL is mutated in two subtypes of CMT: CMT1F, a demyelinating form and CMT2E, an axonal form. Several mutations in the N-terminal head domain (aa 1–93) of NFL (P8R, P8Q, P8L, P22S and E89K) and in the central ‘rod’ domain (aa 94–400) (N97S, A148V, E210X, I213M, Q333P and E397K) have been identified (Mersiyanova et al. 2000; Yoshihara et al. 2002; Jordanova et al. 2003; Fabrizi et al. 2004, 2007; Zuchner et al. 2004; Kabzinska et al. 2006; Miltenberger-Miltenyi et al. 2007; Yum et al. 2009). NFL mutations disrupt NF assembly and transport (Perez-Olle et al. 2002, 2004; Zhai et al. 2007; Tradewell et al. 2009; Yates et al. 2009) but we have demonstrated, using a cellular model of CMT2E/1 F, that they also affect mitochondrial morphology (Tradewell et al. 2009) by interfering with mitochondrial dynamics even before disruption of NF assembly and organization (Gentil et al. 2011). CMT-causing mutations in different functional domains appear to affect NF function differently (Fabrizi et al. 2007). For example, NFs made of NFLP8R form more stable filaments, whereas NFs made of NFLQ333P undergo an anarchic assembly by failure to make making proper coil-coiled dimers, as analysed by seminative BN-PAGE (Gentil et al. 2013).

Interestingly, overexpression of chaperones (HSPA1/ HSP70, HSPB1/HSP27) can maintain function of mutant NFL but different chaperones protect different NFL mutants (Gentil et al. 2013). Mutations in HSPB1 also cause a form of CMT that is characterised by NF abnormalities (Evgrafov et al. 2004). These HSPB1 mutants have higher chaperone activity compared to the wild type protein and induce the collapse of the NF network and perikaryal accumulation, consistent with a role of this chaperone in NF assembly and turnover (Evgrafov et al. 2004; Ackerley et al. 2006; AlmeidaSouza et al. 2010). The HSPB1 mutants impair axonal transport, in part by sequestering p150, a linker between cargo proteins and the retrograde transport motor, dynein, in detergent-insoluble perikaryal aggregates (Ackerley et al. 2006). Expression of HSPB1 mutants also impacts anterograde transport and induces NFs aggregation by affecting the binding of NFs to kinesin, a mechanism that is mediated by Cdk5 phosphorylation of NFs (Holmgren et al. 2013). The importance of NFs in HSPB1 pathogenesis is enlightened by the absence of HSPB1 toxicity in motor neurons from Nefl knockout mice (Zhai et al. 2007). Giant axonal neuropathy and other rare genetic peripheral neuropathies GAN, due to mutations in the gene encoding gigaxonin, is a rare pediatric autosomal recessive neurodegenerative disease affecting both the PNS and CNS. This peripheral neuropathy is characterized by intermittent axonal swellings filled with NF, thus its name but also by collapse of IF networks in multiple cell types (Bruno et al. 2004; Mahammad et al. 2013). Mutant forms of gigaxonin are unable to target intermediate filament proteins for proteasomal degradation (Mahammad et al. 2013). An early onset neuropathy caused by mutations in TRIM2 also shows NF accumulations suggesting that regulation of NF homeostasis is crucial and is a key contributor of

Cell Tissue Res

axonopathies (Ylikallio et al. 2013). Disruption of TRIM2 and UPS14 expression in mice results in ataxia and motor neuron dysfunction characterised by NF accumulation in axons (Balastik et al. 2008). Interestingly, in Autosomal Spastic Ataxia of Charlevoix-Saguenay (ARSACS), caused by mutations in SACS, the phenotype of ataxia and peripheral neuropathy is characterized at the cellular level by accumulation of NF bundles in affected neurons, particularly evident in dendrites (Bouchard et al. 1979; Engert et al. 2000; Lariviere et al. 2014). Considering the long half-life of NF proteins it is likely that these accumulations impact multiple neuronal functions in which they participate. Diabetic neuropathies NF proteins are also implicated in metabolic disorder. A small percentage of individuals with autoimmune type 1 diabetes have autoantibodies against peripherin, as well as small sensory fibre and autonomic neuropathy (Boitard et al. 1992; Pleau et al. 1993; Gelber et al. 1994). In addition to the PNS, peripherin is also embryonically expressed in β-islet cells before being down-regulated in the adult upon remodelling of the pancreas (Durant et al. 2003; Puertas et al. 2007). It is not known whether peripherin autoantibodies are damaging to the β-islet cells and therefore play a causative role in the diabetes, or are simply an early marker of their denervation. However, the association of the presence of these antibodies in individuals with peripheral neuropathy affecting peripherinexpressing neurons suggests a possible role in this neurological complication. Do NFs play any role in diabetic neuropathy in individuals without peripherin autoantibodies? Altered expression and phosphorylation of NF proteins has been reported in rat models of diabetic neuropathy. NF protein levels were reduced in distal axons (Yagihashi et al. 1990; Schmidt et al. 1997a, b). Perikaryal NFs were more highly phosphorylated in DRG neurons (Fernyhough et al. 1999). These findings could simply reflect the consequences of injury. Amyotrophic lateral sclerosis (ALS) ALS is a very complex syndrome affecting both upper and motor neurons with multiple causes, the majority of which remain unresolved. Characteristic neuropathological findings in ALS implicating NFs in pathogenesis are abnormal accumulation of bundled NFs in axonal spheroids, hyperphosphorylation of NFM and NFH tail domains and the presence of NF proteins in perikaryal inclusions (Carpenter 1968; Sobue et al. 1990; Itoh et al. 1992). Also, rare mutations in the peripherin gene and in the KSP repeats motifs of NFH have been found in patients with sporadic ALS (Figlewicz et al. 1994; Al-Chalabi et al. 1999; Gros-Louis et al. 2004).

The cause of abnormal accumulation and bundling of NFs in ALS is unknown but evidence points to abnormal NF assembly and transport as a result of disrupted stoichiometry and an altered phosphorylation state of subunits. The generation of transgenic mice carrying a mutant NFL transgene, in which Ser-55 was mutated to Asp to mimic permanent phosphorylation associated with NF disassembly (Gibb et al. 1996), resulted in pathological accumulation of NFs in neuronal cell bodies including motor neurons that resembled findings in ALS (Gibb et al. 1998). In addition, elevated levels of NFL and phosphorylated NFH (NFH-P) have been detected in cerebral spinal fluid, blood and plasma samples from ALS patients compared with healthy controls, possibly due to decreased structural integrity of motor axons and dendrites (Boylan et al. 2013; Gaiottino et al. 2013). NFH-P levels in serum correlated with a faster decline in the ALSFRS-R score, a measure of clinical progression, suggesting that NFH-P could serve as a possible prognostic biomarker (Boylan et al. 2013). Motor neurons in autopsy specimens from sporadic ALS patients are depleted of NFL mRNA compared with controls, associated with altered stoichiometry of the NF triplet proteins that is important for NF assembly (Bergeron et al. 1994). Several proteins implicated in ALS by mutation or abnormal metabolism (mutant SOD1, TDP43, and RGNEF) destabilize NEFL mRNA by interacting with its 3′-UTR (Hadano et al. 2001; Kwiatkowski et al. 2009; Vance et al. 2009; Volkening et al. 2009; Leblond et al. 2014). NEFL mRNA stability is also regulated by miRNAs, several of which have altered expression in ALS spinal cord (Ishtiaq et al. 2014). In addition, increase in NFH levels is predicted from the downregulation of miR9 in ALS, a regulator of NEFH mRNA stability (Haramati et al. 2010). Toxic forms of peripherin generated by alternative splicing are also observed in ALS and affect motor neuron survival and NF assembly (Robertson et al. 2003; Xiao et al. 2008). Collectively, these abnormalities predict altered stoichiometry of NFL:NFH although the relative contribution to ALS pathogenesis is not known. Certainly, the high content of NFs in motor neurons and the characteristic NFs that course through their dendrites are vulnerability factors (Wuerker and Palay 1969). Decreasing the NFL: NFH ratio by manipulating expression in transgenic mice caused dendritic retraction in spinal motor neurons (Kong et al. 1998; Zhang et al. 2002), a common pathological finding in ALS (Nakano and Hirano 1987; Karpati et al. 1988; Takeda et al. 2014). Expression of SOD1 with ALS-associated mutations has been shown to cause NF bundling in cultured motor neurons (Durham et al. 1997) as well as dendritic atrophy (Tibshirani and Durham, personal communication) and in differentiated iPSC cells derived from individuals with SOD1 mutation (Chen et al. 2014). Retraction of dendrites would be expected to compromise central connections important for motor control.

Cell Tissue Res

Conclusion Almost a century after their first identification by Remak as neurofibrils and the development of silver impregnation techniques to stain them by Max Bielschowsky, there is still mystery surrounding NF function and dynamics. Recent advances in genetics and live cell imaging are shedding light on the intricacies of NF metabolism and their multiple roles in neuronal homeostasis and disease. Acknowledgments We thank Sandra Minotti for her precious help in preparing spinal cord and dorsal root ganglia cultures and Patrick Bouchard for confocal microscopy. This work is supported by a grant from the ARSACS Foundation.

References Ackerley S, Thornhill P, Grierson AJ, Brownlees J, Anderton BH, Leigh PN, Shaw CE, Miller CC (2003) Neurofilament heavy chain side arm phosphorylation regulates axonal transport of neurofilaments. J Cell Biol 161:489–495 Ackerley S, James PA, Kalli A, French S, Davies KE, Talbot K (2006) A mutation in the small heat-shock protein HSPB1 leading to distal hereditary motor neuronopathy disrupts neurofilament assembly and the axonal transport of specific cellular cargoes. Hum Mol Genet 15: 347–354 Al-Chalabi A, Andersen PM, Nilsson P, Chioza B, Andersson JL, Russ C, Shaw CE, Powell JF, Leigh PN (1999) Deletions of the heavy neurofilament subunit tail in amyotrophic lateral sclerosis. Hum Mol Genet 8:157–164 Almeida-Souza L, Goethals S, de Winter V, Dierick I, Gallardo R, Van Durme J, Irobi J, Gettemans J, Rousseau F, Schymkowitz J, Timmerman V, Janssens S (2010) Increased monomerization of mutant HSPB1 leads to protein hyperactivity in Charcot-MarieTooth neuropathy. J Biol Chem 285:12778–12786 Athlan ES, Mushynski WE (1997) Heterodimeric associations between neuronal intermediate filament proteins. J Biol Chem 272:31073– 31078 Attali R, Warwar N, Israel A, Gurt I, McNally E, Puckelwartz M, Glick B, Nevo Y, Ben-Neriah Z, Melki J (2009) Mutation of SYNE-1, encoding an essential component of the nuclear lamina, is responsible for autosomal recessive arthrogryposis. Hum Mol Genet 18: 3462–3469 Baas PW, Black MM (1990) Individual microtubules in the axon consist of domains that differ in both composition and stability. J Cell Biol 111:495–509 Baas PW, Ahmad FJ, Pienkowski TP, Brown A, Black MM (1993) Sites of microtubule stabilization for the axon. J Neurosci 13:2177–2185 Balastik M, Ferraguti F, Pires-da Silva A, Lee TH, Alvarez-Bolado G, Lu KP, Gruss P (2008) Deficiency in ubiquitin ligase TRIM2 causes accumulation of neurofilament light chain and neurodegeneration. Proc Natl Acad Sci U S A 105:12016–12021 Barry DM, Stevenson W, Bober BG, Wiese PJ, Dale JM, Barry GS, Byers NS, Strope JD, Chang R, Schulz DJ, Shah S, Calcutt NA, Gebremichael Y, Garcia ML (2012) Expansion of neurofilament medium C terminus increases axonal diameter independent of increases in conduction velocity or myelin thickness. J Neurosci 32: 6209–6219 Bearer EL, Reese TS (1999) Association of actin filaments with axonal microtubule tracts. J Neurocytol 28:85–98

Bearer EL, Schlief ML, Breakefield XO, Schuback DE, Reese TS, LaVail JH (1999) Squid axoplasm supports the retrograde axonal transport of herpes simplex virus. Biol Bull 197:257–258 Beaulieu JM, Kriz J, Julien JP (2002) Induction of peripherin expression in subsets of brain neurons after lesion injury or cerebral ischemia. Brain Res 946:153–161 Beck R, Deek J, Jones JB, Safinya CR (2010) Gel-expanded to gelcondensed transition in neurofilament networks revealed by direct force measurements. Nat Mater 9:40–46 Bergeron C, Muntasser S, Somerville MJ, Weyer L, Percy ME (1994) Copper/zinc superoxide dismutase mRNA levels are increased in sporadic amyotrophic lateral sclerosis motorneurons. Brain Res 659: 272–276 Boitard C, Villa MC, Becourt C, Gia HP, Huc C, Sempe P, Portier MM, Bach JF (1992) Peripherin: an islet antigen that is cross-reactive with nonobese diabetic mouse class II gene products. Proc Natl Acad Sci U S A 89:172–176 Bouchard JP, Barbeau A, Bouchard R, Bouchard RW (1979) Electromyography and nerve conduction studies in Friedreich’s ataxia and autosomal recessive spastic ataxia of CharlevoixSaguenay (ARSACS). Can J Neurol Sci 6:185–189 Boylan KB, Glass JD, Crook JE, Yang C, Thomas CS, Desaro P, Johnston A, Overstreet K, Kelly C, Polak M, Shaw G (2013) Phosphorylated neurofilament heavy subunit (pNF-H) in peripheral blood and CSF as a potential prognostic biomarker in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 84:467–472 Brown A (1997) Visualization of single neurofilaments by immunofluorescence microscopy of splayed axonal cytoskeletons. Cell Motil Cytoskeleton 38:133–145 Brown A, Li Y, Slaughter T, Black MM (1993) Composite microtubules of the axon: quantitative analysis of tyrosinated and acetylated tubulin along individual axonal microtubules. J Cell Sci 104(Pt 2): 339–352 Brown HG, Troncoso JC, Hoh JH (1998) Neurofilament-L homopolymers are less mechanically stable than native neurofilaments. J Microsc 191:229–237 Brown A, Wang L, Jung P (2005) Stochastic simulation of neurofilament transport in axons: the “stop-and-go” hypothesis. Mol Biol Cell 16: 4243–4255 Bruno C, Bertini E, Federico A, Tonoli E, Lispi ML, Cassandrini D, Pedemonte M, Santorelli FM, Filocamo M, Dotti MT, Schenone A, Malandrini A, Minetti C (2004) Clinical and molecular findings in patients with giant axonal neuropathy (GAN). Neurology 62:13–16 Burton PR, Wentz MA (1992) Neurofilaments are prominent in bullfrog olfactory axons but are rarely seen in those of the tiger salamander, Ambystoma tigrinum. J Comp Neurol 317:396–406 Carden MJ, Eagles PAM(1986) Chemical cross-linking analyses of ox neurofilaments. Biochem J 234:587–591 Carpenter S (1968) Proximal axonal enlargement in motor neuron disease. Neurology 18:841–851 Chen PC, Qin LN, Li XM, Walters BJ, Wilson JA, Mei L, Wilson SM (2009) The proteasome-associated deubiquitinating enzyme Usp14 is essential for the maintenance of synaptic ubiquitin levels and the development of neuromuscular junctions. J Neurosci: Off J Soc Neurosci 29:10909–10919 Chen H, Qian K, Du Z, Cao J, Petersen A, Liu H, Blackbourn LW 4th, Huang CL, Errigo A, Yin Y, Lu J, Ayala M, Zhang SC (2014) Modeling ALS with iPSCs reveals that mutant SOD1 misregulates neurofilament balance in motor neurons. Cell Stem Cell 14:796–809 Cochard P, Paulin D (1984) Initial expression of neurofilaments and vimentin in the central and peripheral nervous system of the mouse embryo in vivo. J Neurosci 4:2080–2094 Cohlberg JA, Hajarian H, Sainte-Marie S (1987) Discrete soluble forms of middle and high molecular weight neurofilament proteins in dilute aqueous buffers. J Biol Chem 262:17009–17015

Cell Tissue Res Cohlberg JA, Hajarian H, Tran T, Alipourjeddi P, Noveen A (1995) Neurofilament protein heterotetramers as assembly intermediates. J Biol Chem 270:9334–9339 Colakoglu G, Brown A (2009) Intermediate filaments exchange subunits along their length and elongate by end-to-end annealing. J Cell Biol 185:769–777 Conde C, Caceres A (2009) Microtubule assembly, organization and dynamics in axons and dendrites. Nat Rev Neurosci 10:319–332 Crimmins S, Jin Y, Wheeler C, Huffman AK, Chapman C, Dobrunz LE, Levey A, Roth KA, Wilson JA, Wilson SM (2006) Transgenic rescue of ataxia mice with neuronal-specific expression of ubiquitin-specific protease 14. J Neurosci 26:11423–11431 Dale JM, Garcia ML (2012) Neurofilament phosphorylation during development and disease: which came first, the phosphorylation or the accumulation? J Amino Acids 2012:382107 Deloulme JC, Gentil BJ, Baudier J (2003) Monitoring of S100 homodimerization and heterodimeric interactions by the yeast twohybrid system. Microsc Res Tech 60:560–568 Doroudchi MM, Durham HD (1996) Activation of protein kinase C induces neurofilament fragmentation, hyperphosphorylation of perikaryal neurofilaments and proximal dendritic swellings in cultured motor neurons. J Neuropathol Exp Neurol 55:246–256 Durant S, Geutskens S, Van Blokland SC, Coulaud J, Alves V, Pleau JM, Versnel M, Drexhage HA, Homo-Delarche F (2003) Proapoptosis and antiapoptosis-related molecules during postnatal pancreas development in control and nonobese diabetic mice: relationship with innervation. Lab Investig 83:227–239 Durham HD, Roy J, Dong L, Figlewicz DA (1997) Aggregation of mutant Cu/Zn superoxide dismutase proteins in a culture model of ALS. J Neuropathol Exp Neurol 56:523–530 Ehlers MD, Fung ET, O’Brien RJ, Huganir RL (1998) Splice variantspecific interaction of the NMDA receptor subunit NR1 with neuronal intermediate filaments. J Neurosci 18:720–730 Elder GA, Friedrich VL Jr, Bosco P, Kang C, Gourov A, Tu PH, Lee VM, Lazzarini RA (1998a) Absence of the mid-sized neurofilament subunit decreases axonal calibers, levels of light neurofilament (NF-L), and neurofilament content. J Cell Biol 141:727–739 Elder GA, Friedrich VL Jr, Kang C, Bosco P, Gourov A, Tu PH, Zhang B, Lee VM, Lazzarini RA (1998b) Requirement of heavy neurofilament subunit in the development of axons with large calibers. J Cell Biol 143:195–205 Engert JC, Berube P, Mercier J, Dore C, Lepage P, Ge B, Bouchard JP, Mathieu J, Melancon SB, Schalling M, Lander ES, Morgan K, Hudson TJ, Richter A (2000) ARSACS, a spastic ataxia common in northeastern Quebec, is caused by mutations in a new gene encoding an 11.5-kb ORF. Nat Genet 24:120–125 Escurat M, Djabali K, Gumpel M, Gros F, Portier MM (1990) Differential expression of two neuronal intermediate-filament proteins, peripherin and the low-molecular-mass neurofilament protein (NFL), during the development of the rat. J Neurosci 10:764–784 Evgrafov OV, Mersiyanova I, Irobi J, Van Den Bosch L, Dierick I, Leung CL, Schagina O, Verpoorten N, Van Impe K, Fedotov V, Dadali E, Auer-Grumbach M, Windpassinger C, Wagner K, Mitrovic Z, Hilton-Jones D, Talbot K, Martin JJ, Vasserman N, Tverskaya S, Polyakov A, Liem RK, Gettemans J, Robberecht W, De Jonghe P, Timmerman V (2004) Mutant small heat-shock protein 27 causes axonal Charcot-Marie-Tooth disease and distal hereditary motor neuropathy. Nat Genet 36:602–606 Fabrizi GM, Cavallaro T, Angiari C, Bertolasi L, Cabrini I, Ferrarini M, Rizzuto N (2004) Giant axon and neurofilament accumulation in Charcot-Marie-Tooth disease type 2E. Neurology 62:1429–1431 Fabrizi GM, Cavallaro T, Angiari C, Cabrini I, Taioli F, Malerba G, Bertolasi L, Rizzuto N (2007) Charcot-Marie-Tooth disease type 2E, a disorder of the cytoskeleton. Brain 130:394–403 Fernyhough P, Gallagher A, Averill SA, Priestley JV, Hounsom L, Patel J, Tomlinson DR (1999) Aberrant neurofilament phosphorylation in

sensory neurons of rats with diabetic neuropathy. Diabetes 48:881– 889 Fifkova E, Delay RJ (1982) Cytoplasmic actin in neuronal processes as a possible mediator of synaptic plasticity. J Cell Biol 95:345– 350 Figlewicz DA, Krizus A, Martinoli MG, Meininger V, Dib M, Rouleau GA, Julien JP (1994) Variants of the heavy neurofilament subunit are associated with the development of amyotrophic lateral sclerosis. Hum Mol Genet 3:1757–1761 Foisner R, Leichtfried FE, Herrmann H, Small JV, Lawson D, Wiche G (1988) Cytoskeleton-associated plectin: in situ localization, in vitro reconstitution, and binding to immobilized intermediate filament proteins. J Cell Biol 106:723–733 Gaiottino J, Norgren N, Dobson R, Topping J, Nissim A, Malaspina A, Bestwick JP, Monsch AU, Regeniter A, Lindberg RL, Kappos L, Leppert D, Petzold A, Giovannoni G, Kuhle J (2013) Increased neurofilament light chain blood levels in neurodegenerative neurological diseases. PLoS ONE 8:e75091 Garcia ML, Lobsiger CS, Shah SB, Deerinck TJ, Crum J, Young D, Ward CM, Crawford TO, Gotow T, Uchiyama Y, Ellisman MH, Calcutt NA, Cleveland DW (2003) NF-M is an essential target for the myelin-directed “outside-in” signaling cascade that mediates radial axonal growth. J Cell Biol 163:1011–1020 Garcia ML, Rao MV, Fujimoto J, Garcia VB, Shah SB, Crum J, Gotow T, Uchiyama Y, Ellisman M, Calcutt NA, Cleveland DW (2009) Phosphorylation of highly conserved neurofilament medium KSP repeats is not required for myelin-dependent radial axonal growth. J Neurosci 29:1277–1284 Gelber C, Paborsky L, Singer S, McAteer D, Tisch R, Jolicoeur C, Buelow R, McDevitt H, Fathman CG (1994) Isolation of nonobese diabetic mouse T-cells that recognize novel autoantigens involved in the early events of diabetes. Diabetes 43:33–39 Gentil BJ, Minotti S, Beange M, Baloh RH, Julien JP, Durham HD (2011) Normal role of the low-molecular-weight neurofilament protein in mitochondrial dynamics and disruption in Charcot-Marie-Tooth disease. FASEB J 26:1194–1203 Gentil BJ, Mushynski WE, Durham HD (2013) Heterogeneity in the properties of NEFL mutants causing Charcot-Marie-Tooth disease results in differential effects on neurofilament assembly and susceptibility to intervention by the chaperone-inducer, celastrol. Int J Biochem Cell Biol 45:1499–1508 Gentil BJ, McLean JR, Xiao S, Zhao B, Durham HD, Robertson J (2014) A two-hybrid screen identifies an unconventional role for the intermediate filament peripherin in regulating the subcellular distribution of the SNAP25 interacting protein, SIP30. J Neurochem 131(5): 588–601 Giasson BI, Mushynski WE (1996) Aberrant stress-induced phosphorylation of perikaryal neurofilaments. J Biol Chem 271:30404–30409 Giasson BI, Mushynski WE (1998) Intermediate filament disassembly in cultured dorsal root ganglion neurons is associated with aminoterminal head domain phosphorylation of specific subunits. J Neurochem 70:1869–1875 Giasson BI, Cromlish JA, Athlan ES, Mushynski WE (1996) Activation of cyclic AMP-dependent protein kinase in okadaic acid-treated neurons potentiates neurofilament fragmentation and stimulates phosphorylation of Ser2 in the low-molecular-mass neurofilament subunit. J Neurochem 66:1207–1213 Gibb BJ, Robertson J, Miller CC (1996) Assembly properties of neurofilament light chain Ser55 mutants in transfected mammalian cells. J Neurochem 66:1306–1311 Gibb BJ, Brion JP, Brownlees J, Anderton BH, Miller CC (1998) Neuropathological abnormalities in transgenic mice harbouring a phosphorylation mutant neurofilament transgene. J Neurochem 70: 492–500 Gros-Louis F, Lariviere R, Gowing G, Laurent S, Camu W, Bouchard JP, Meininger V, Rouleau GA, Julien JP (2004) A frameshift deletion in

Cell Tissue Res peripherin gene associated with amyotrophic lateral sclerosis. J Biol Chem 279:45951–45956 Hadano S, Hand CK, Osuga H, Yanagisawa Y, Otomo A, Devon RS, Miyamoto N, Showguchi-Miyata J, Okada Y, Singaraja R, Figlewicz DA, Kwiatkowski T, Hosler BA, Sagie T, Skaug J, Nasir J, Brown RH Jr, Scherer SW, Rouleau GA, Hayden MR, Ikeda JE (2001) A gene encoding a putative GTPase regulator is mutated in familial amyotrophic lateral sclerosis 2. Nat Genet 29: 166–173 Haramati S, Chapnik E, Sztainberg Y, Eilam R, Zwang R, Gershoni N, McGlinn E, Heiser PW, Wills AM, Wirguin I, Rubin LL, Misawa H, Tabin CJ, Brown R Jr, Chen A, Hornstein E (2010) miRNA malfunction causes spinal motor neuron disease. Proc Natl Acad Sci U S A 107:13111–13116 Hatzfeld M, Weber K (1990) The coiled coil of in vitro assembled keratin filaments is a heterodimer of type I and II keratins: use of sitespecific mutagenesis and recombinant protein expression. J Cell Biol 110:1199–1210 Haynes RL, Borenstein NS, Desilva TM, Folkerth RD, Liu LG, Volpe JJ, Kinney HC (2005) Axonal development in the cerebral white matter of the human fetus and infant. J Comp Neurol 484:156– 167 Herrmann H, Aebi U (2000) Intermediate filaments and their associates: multi-talented structural elements specifying cyto architecture and cytodynamics. Curr Opin Cell Biol 12:79–90 Herrmann H, Haner M, Brettel M, Ku NO, Aebi U (1999) Characterization of distinct early assembly units of different intermediate filament proteins. J Mol Biol 286:1403–1420 Hoffman PN, Lasek RJ (1975) The slow component of axonal transport. Identification of major structural polypeptides of the axon and their generality among mammalian neurons. J Cell Biol 66:351–366 Holmgren A, Bouhy D, De Winter V, Asselbergh B, Timmermans JP, Irobi J, Timmerman V (2013) Charcot-Marie-Tooth causing HSPB1 mutations increase Cdk5-mediated phosphorylation of neurofilaments. Acta Neuropathol 126:93–108 Ishihara T, Higuchi M, Zhang B, Yoshiyama Y, Hong M, Trojanowski JQ, Lee VM (2001) Attenuated neurodegenerative disease phenotype in tau transgenic mouse lacking neurofilaments. J Neurosci 21: 6026–6035 Ishtiaq M, Campos-Melo D, Volkening K, Strong MJ (2014) Analysis of novel NEFL mRNA targeting microRNAs in amyotrophic lateral sclerosis. PLoS ONE 9:e85653 Itoh T, Sobue G, Ken E, Mitsuma T, Takahashi A, Trojanowski JQ (1992) Phosphorylated high molecular weight neurofilament protein in the peripheral motor, sensory and sympathetic neuronal perikarya: system-dependent normal variations and changes in amyotrophic lateral sclerosis and multiple system atrophy. Acta Neuropathol 83: 240–245 Jacomy H, Zhu Q, Couillard-Despres S, Beaulieu JM, Julien JP (1999) Disruption of type IV intermediate filament network in mice lacking the neurofilament medium and heavy subunits. J Neurochem 73: 972–984 Jordanova A, De Jonghe P, Boerkoel CF, Takashima H, De Vriendt E, Ceuterick C, Martin JJ, Butler IJ, Mancias P, Papasozomenos S, Terespolsky D, Potocki L, Brown CW, Shy M, Rita DA, Tournev I, Kremensky I, Lupski JR, Timmerman V (2003) Mutations in the neurofilament light chain gene (NEFL) cause early onset severe Charcot-Marie-Tooth disease. Brain 126:590–597 Kabzinska D, Perez-Olle R, Goryunov D, Drac H, Ryniewicz B, Hausmanowa-Petrusewicz I, Kochanski A, Liem RK (2006) Is a novel I214M substitution in the NEFL gene a cause of CharcotMarie-Tooth disease? functional analysis using cell culture models. J Peripher Nerv Syst 11:225–231 Kapitein LC, Hoogenraad CC (2011) Which way to go? Cytoskeletal organization and polarized transport in neurons. Mol Cell Neurosci 46:9–20

Kaplan MP, Chin SS, Fliegner KH, Liem RK (1990) Alpha-internexin, a novel neuronal intermediate filament protein, precedes the low molecular weight neurofilament protein (NF-L) in the developing rat brain. J Neurosci 10:2735–2748 Karpati G, Carpenter S, Durham H (1988) A hypothesis for the pathogenesis of amyotrophic lateral sclerosis. Rev Neurol 144:672–675 Karpova A, Mikhaylova M, Bera S, Bar J, Reddy PP, Behnisch T, Rankovic V, Spilker C, Bethge P, Sahin J, Kaushik R, Zuschratter W, Kahne T, Naumann M, Gundelfinger ED, Kreutz MR (2013) Encoding and transducing the synaptic or extrasynaptic origin of NMDA receptor signals to the nucleus. Cell 152:1119–1133 Kong J, Tung VW, Aghajanian J, Xu Z (1998) Antagonistic roles of neurofilament subunits NF-H and NF-M against NF-L in shaping dendritic arborization in spinal motor neurons. J Cell Biol 140: 1167–1176 Koutras C, Levesque G (2011) Identification of novel NPRAP/deltacatenin-interacting proteins and the direct association of NPRAP with dynamin 2. PLoS ONE 6:e25379 Kreplak L, Bar H, Leterrier JF, Herrmann H, Aebi U (2005) Exploring the mechanical behavior of single intermediate filaments. J Mol Biol 354:569–577 Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA, Hosler BA, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA, Yan J, Ticozzi N, Siddique T, McKenna-Yasek D, Sapp PC, Horvitz HR, Landers JE, Brown RH Jr (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323:1205–1208 Lariviere RC, Nguyen MD, Ribeiro-da-Silva A, Julien JP (2002) Reduced number of unmyelinated sensory axons in peripherin null mice. J Neurochem 81:525–532 Lariviere R, Gaudet R, Gentil BJ, Girard M, Conte TC, Minotti S, Leclerc-Desaulniers K, Gehring K, McKinney RA, Shoubridge EA, McPherson PS, Durham HD, Brais B (2014) Sacs knockout mice present pathophysiological defects underlying autosomal recessive spastic ataxia of Charlevoix-Saguenay. Hum Mol Genet Lasek RJ, Paggi P, Katz MJ (1992) Slow axonal transport mechanisms move neurofilaments relentlessly in mouse optic axons. J Cell Biol 117:607–616 Leblond CS, Kaneb HM, Dion PA, Rouleau GA (2014) Dissection of genetic factors associated with amyotrophic lateral sclerosis. Exp Neurol. doi:10.1016/j.expneurol.2014.04.013 Leung CL, Liem RK (1996) Characterization of interactions between the neurofilament triplet proteins by the yeast two-hybrid system. J Biol Chem 271:14041–14044 Levavasseur F, Zhu Q, Julien JP (1999) No requirement of alphainternexin for nervous system development and for radial growth of axons. Brain Res Mol Brain Res 69:104–112 Liem RK, Hutchison SB (1982) Purification of individual components of the neurofilament triplet: filament assembly from the 70 000-dalton subunit. Biochemistry 21:3221–3226 Ligon LA, Steward O (2000a) Movement of mitochondria in the axons and dendrites of cultured hippocampal neurons. J Comp Neurol 427: 340–350 Ligon LA, Steward O (2000b) Role of microtubules and actin filaments in the movement of mitochondria in the axons and dendrites of cultured hippocampal neurons. J Comp Neurol 427:351–361 Liu Y, Staal JA, Canty AJ, Kirkcaldie MT, King AE, Bibari O, Mitew ST, Dickson TC, Vickers JC (2013) Cytoskeletal changes during development and aging in the cortex of neurofilament light protein knockout mice. J Comp Neurol 521:1817–1827 Macioce P, Gandolfi N, Leung CL, Chin SS, Malchiodi-Albedi F, Ceccarini M, Petrucci TC, Liem RK (1999) Characterization of NF-L and betaIISigma1-spectrin interaction in live cells. Exp Cell Res 250:142–154

Cell Tissue Res Mahammad S, Murthy SN, Didonna A, Grin B, Israeli E, Perrot R, Bomont P, Julien JP, Kuczmarski E, Opal P, Goldman RD (2013) Giant axonal neuropathy-associated gigaxonin mutations impair intermediate filament protein degradation. J Clin Invest 123:1964–1975 Manser C, Stevenson A, Banner S, Davies J, Tudor EL, Ono Y, Leigh PN, McLoughlin DM, Shaw CE, Miller CC (2008) Deregulation of PKN1 activity disrupts neurofilament organisation and axonal transport. FEBS Lett 582:2303–2308 Markham JA, Fifkova E (1986) Actin filament organization within dendrites and dendritic spines during development. Brain Res 392:263– 269 Mellad JA, Warren DT, Shanahan CM (2011) Nesprins LINC the nucleus and cytoskeleton. Curr Opin Cell Biol 23:47–54 Mersiyanova IV, Perepelov AV, Polyakov AV, Sitnikov VF, Dadali EL, Oparin RB, Petrin AN, Evgrafov OV (2000) A new variant of Charcot-Marie-Tooth disease type 2 is probably the result of a mutation in the neurofilament-light gene. Am J Hum Genet 67:37– 46 Mialki RK, Zhao J, Wei J, Mallampalli DF, Zhao Y (2013) Overexpression of USP14 protease reduces I-kappaB protein levels and increases cytokine release in lung epithelial cells. J Biol Chem 288:15437–15441 Millecamps S, Julien JP (2004) [35S]Methionine metabolic labeling to study axonal transport of neuronal intermediate filament proteins in vivo. Methods Cell Biol 78:555–571 Millecamps S, Gowing G, Corti O, Mallet J, Julien JP (2007) Conditional NF-L transgene expression in mice for in vivo analysis of turnover and transport rate of neurofilaments. J Neurosci 27:4947–4956 Miltenberger-Miltenyi G, Janecke AR, Wanschitz JV, Timmerman V, Windpassinger C, Auer-Grumbach M, Loscher WN (2007) Clinical and electrophysiological features in Charcot-Marie-Tooth disease with mutations in the NEFL gene. Arch Neurol 64:966–970 Mironov SL (2006) Spontaneous and evoked neuronal activities regulate movements of single neuronal mitochondria. Synapse 59:403–411 Mironov SL, Symonchuk N (2006) ER vesicles and mitochondria move and communicate at synapses. J Cell Sci 119:4926–4934 Morgan JT, Pfeiffer ER, Thirkill TL, Kumar P, Peng G, Fridolfsson HN, Douglas GC, Starr DA, Barakat AI (2011) Nesprin-3 regulates endothelial cell morphology, perinuclear cytoskeletal architecture, and flow-induced polarization. Mol Biol Cell 22:4324–4334 Nakamura Y, Hashimoto R, Kashiwagi Y, Aimoto S, Fukusho E, Matsumoto N, Kudo T, Takeda M (2000) Major phosphorylation site (Ser55) of neurofilament L by cyclic AMP-dependent protein kinase in rat primary neuronal culture. J Neurochem 74:949–959 Nakano I, Hirano A (1987) Atrophic cell processes of large motor neurons in the anterior horn in amyotrophic lateral sclerosis: observation with silver impregnation method. J Neuropathol Exp Neurol 46:40–49 Nixon RA, Logvinenko KB (1986) Multiple fates of newly synthesized neurofilament proteins: evidence for a stationary neurofilament network distributed nonuniformly along axons of retinal ganglion cell neurons. J Cell Biol 102:647–659 Pant HC (1988) Dephosphorylation of neurofilament proteins enhances their susceptibility to degradation by calpain. Biochem J 256:665–668 Perez-Olle R, Leung CL, Liem RK (2002) Effects of Charcot-MarieTooth-linked mutations of the neurofilament light subunit on intermediate filament formation. J Cell Sci 115:4937–4946 Perez-Olle R, Jones ST, Liem RK (2004) Phenotypic analysis of neurofilament light gene mutations linked to Charcot-Marie-Tooth disease in cell culture models. Hum Mol Genet 13:2207–2220 Peth A, Kukushkin N, Bosse M, Goldberg AL (2013) Ubiquitinated proteins activate the proteasomal ATPases by binding to Usp14 or Uch37 homologs. J Biolo Chem 288:7781–7790 Pleau JM, Marche PN, Serrano MP, Boitard C, Bach JF (1993) Evidence for antigen driven selection in two monoclonal auto-antibodies derived from nonobese diabetic mice. Mol Immunol 30:1257–1264

Puertas MC, Carrillo J, Pastor X, Ampudia RM, Planas R, Alba A, Bruno R, Pujol-Borrell R, Estanyol JM, Vives-Pi M, Verdaguer J (2007) Peripherin is a relevant neuroendocrine autoantigen recognized by islet-infiltrating B lymphocytes. J Immunol 178:6533–6539 Rao MV, Engle LJ, Mohan PS, Yuan A, Qiu D, Cataldo A, Hassinger L, Jacobsen S, Lee VM, Andreadis A, Julien JP, Bridgman PC, Nixon RA (2002a) Myosin Va binding to neurofilaments is essential for correct myosin Va distribution and transport and neurofilament density. J Cell Biol 159:279–290 Rao MV, Garcia ML, Miyazaki Y, Gotow T, Yuan A, Mattina S, Ward CM, Calcutt NA, Uchiyama Y, Nixon RA, Cleveland DW (2002b) Gene replacement in mice reveals that the heavily phosphorylated tail of neurofilament heavy subunit does not affect axonal caliber or the transit of cargoes in slow axonal transport. J Cell Biol 158:681– 693 Rao MV, Campbell J, Yuan A, Kumar A, Gotow T, Uchiyama Y, Nixon RA (2003) The neurofilament middle molecular mass subunit carboxyl-terminal tail domains is essential for the radial growth and cytoskeletal architecture of axons but not for regulating neurofilament transport rate. J Cell Biol 163:1021–1031 Rao MV, Mohan PS, Kumar A, Yuan A, Montagna L, Campbell J, Veeranna EEM, Julien JP, Nixon RA (2011) The myosin Va head domain binds to the neurofilament-L rod and modulates endoplasmic reticulum (ER) content and distribution within axons. PLoS ONE 6:e17087 Robertson J, Doroudchi MM, Nguyen MD, Durham HD, Strong MJ, Shaw G, Julien JP, Mushynski WE (2003) A neurotoxic peripherin splice variant in a mouse model of ALS. J Cell Biol 160:939–949 Schmidt RE, Beaudet LN, Plurad SB, Dorsey DA (1997a) Axonal cytoskeletal pathology in aged and diabetic human sympathetic autonomic ganglia. Brain Res 769:375–383 Schmidt RE, Dorsey D, Parvin CA, Beaudet LN, Plurad SB, Roth KA (1997b) Dystrophic axonal swellings develop as a function of age and diabetes in human dorsal root ganglia. J Neuropathol Exp Neurol 56:1028–1043 Scott D, Smith KE, O’Brien BJ, Angelides KJ (1985) Characterization of mammalian neurofilament triplet proteins. Subunit stoichiometry and morphology of native and reconstituted filaments. J Biol Chem 260:10736–10747 Sihag RK, Nixon RA (1989) In vivo phosphorylation of distinct domains of the 70-kilodalton neurofilament subunit involves different protein kinases. J Biol Chem 264:457–464 Sihag RK, Nixon RA (1990) Phosphorylation of the amino-terminal head domain of the middle molecular mass 145-kDa subunit of neurofilaments. Evidence for regulation by second messengerdependent protein kinases. J Biol Chem 265:4166–4171 Sihag RK, Nixon RA (1991) Identification of Ser-55 as a major protein kinase A phosphorylation site on the 70-kDa subunit of neurofilaments. Early turnover during axonal transport. J Biol Chem 266:18861–18867 Sihag RK, Jeng AY, Nixon RA (1988) Phosphorylation of neurofilament proteins by protein kinase C. FEBS Lett 233:181–185 Sihag RK, Jaffe H, Nixon RA, Rong X (1999) Serine-23 is a major protein kinase A phosphorylation site on the amino-terminal head domain of the middle molecular mass subunit of neurofilament proteins. J Neurochem 72:491–499 Sobue G, Hashizume Y, Yasuda T, Mukai E, Kumagai T, Mitsuma T, Trojanowski JQ (1990) Phosphorylated high molecular weight neurofilament protein in lower motor neurons in amyotrophic lateral sclerosis and other neurodegenerative diseases involving ventral horn cells. Acta Neuropathol 79:402–408 Steinert PM (1990) The two-chain coiled-coil molecule of native epidermal keratin intermediate filaments is a type I-type II heterodimer. J Biol Chem 265:8766–8774 Takeda T, Uchihara T, Nakayama Y, Nakamura A, Sasaki S, Kakei S, Uchiyama S, Duyckaerts C, Yoshida M (2014) Dendritic retraction,

Cell Tissue Res but not atrophy, is consistent in amyotrophic lateral sclerosiscomparison between Onuf’s neurons and other sacral motor neurons. Acta Neuropathol Commun 2:11 Tradewell ML, Durham HD, Mushynski WE, Gentil BJ (2009) Mitochondrial and axonal abnormalities precede disruption of the neurofilament network in a model of Charcot-Marie-Tooth disease type 2E and are prevented by heat shock proteins in a mutantspecific fashion. J Neuropathol Exp Neurol 68:642–652 Trivedi N, Jung P, Brown A (2007) Neurofilaments switch between distinct mobile and stationary states during their transport along axons. J Neurosci 27:507–516 Troy CM, Muma NA, Greene LA, Price DL, Shelanski ML (1990) Regulation of peripherin and neurofilament expression in regenerating rat motor neurons. Brain Res 529:232–238 Uchida A, Alami NH, Brown A (2009) Tight functional coupling of kinesin-1A and dynein motors in the bidirectional transport of neurofilaments. Mol Biol Cell 20:4997–5006 Uchida A, Colakoglu G, Wang L, Monsma PC, Brown A (2013) Severing and end-to-end annealing of neurofilaments in neurons. Proc Natl Acad Sci U S A 110:E2696–2705 Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, Sreedharan J, Hu X, Smith B, Ruddy D, Wright P, Ganesalingam J, Williams KL, Tripathi V, Al-Saraj S, Al-Chalabi A, Leigh PN, Blair IP, Nicholson G, de Belleroche J, Gallo JM, Miller CC, Shaw CE (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323:1208–1211 Volkening K, Leystra-Lantz C, Yang W, Jaffee H, Strong MJ (2009) Tar DNA binding protein of 43 kDa (TDP-43), 14-3-3 proteins and copper/zinc superoxide dismutase (SOD1) interact to modulate NFL mRNA stability. Implications for altered RNA processing in amyotrophic lateral sclerosis (ALS). Brain Res 1305:168–182 Wang Q, Song F, Zhang C, Zhao X, Zhu Z, Yu S, Xie K (2011) Carboxylterminus of Hsc70 interacting protein mediates 2,5-hexanedioneinduced neurofilament medium chain degradation. Biochem Pharmacol 81:793–799 Wiche G, Winter L (2011) Plectin isoforms as organizers of intermediate filament cytoarchitecture. Biogeosciences 1:14–20 Wickert U, Mucke N, Wedig T, Muller SA, Aebi U, Herrmann H (2005) Characterization of the in vitro co-assembly process of the intermediate filament proteins vimentin and desmin: mixed polymers at all stages of assembly. Eur J Cell Biol 84:379–391 Wuerker RB, Palay SL (1969) Neurofilaments and microtubules in anterior horn cells of the rat. Tissue Cell 1:387–402 Xiao S, Tjostheim S, Sanelli T, McLean JR, Horne P, Fan Y, Ravits J, Strong MJ, Robertson J (2008) An aggregate-inducing peripherin isoform generated through intron retention is upregulated in amyotrophic lateral sclerosis and associated with disease pathology. J Neurosci 28:1833–1840 Yabe JT, Pimenta A, Shea TB (1999) Kinesin-mediated transport of neurofilament protein oligomers in growing axons. J Cell Sci 112(Pt 21):3799–3814

Yagihashi S, Kamijo M, Watanabe K (1990) Reduced myelinated fiber size correlates with loss of axonal neurofilaments in peripheral nerve of chronically streptozotocin diabetic rats. Am J Pathol 136:1365–1373 Yates DM, Manser C, De Vos KJ, Shaw CE, McLoughlin DM, Miller CC (2009) Neurofilament subunit (NFL) head domain phosphorylation regulates axonal transport of neurofilaments. Eur J Cell Biol 88: 193–202 Ylikallio E, Poyhonen R, Zimon M, De Vriendt E, Hilander T, Paetau A, Jordanova A, Lonnqvist T, Tyynismaa H (2013) Deficiency of the E3 ubiquitin ligase TRIM2 in early-onset axonal neuropathy. Hum Mol Genet 22:2975–2983 Yoshihara T, Yamamoto M, Hattori N, Misu K, Mori K, Koike H, Sobue G (2002) Identification of novel sequence variants in the neurofilament-light gene in a Japanese population: analysis of Charcot-Marie-Tooth disease patients and normal individuals. J Peripher Nerv Syst 7:221–224 Yuan A, Rao MV, Kumar A, Julien JP, Nixon RA (2003) Neurofilament transport in vivo minimally requires hetero-oligomer formation. J Neurosci 23:9452–9458 Yuan A, Nixon RA, Rao MV (2006a) Deleting the phosphorylated tail domain of the neurofilament heavy subunit does not alter neurofilament transport rate in vivo. Neurosci Lett 393:264–268 Yuan A, Rao MV, Sasaki T, Chen Y, Kumar A, Veeranna LRK, Eyer J, Peterson AC, Julien JP, Nixon RA (2006b) Alpha-internexin is structurally and functionally associated with the neurofilament triplet proteins in the mature CNS. J Neurosci 26:10006–10019 Yuan A, Sasaki T, Kumar A, Peterhoff CM, Rao MV, Liem RK, Julien JP, Nixon RA (2012) Peripherin is a subunit of peripheral nerve neurofilaments: implications for differential vulnerability of CNS and peripheral nervous system axons. J Neurosci 32:8501–8508 Yum SW, Zhang J, Mo K, Li J, Scherer SS (2009) A novel recessive Nefl mutation causes a severe, early-onset axonal neuropathy. Ann Neurol 66:759–770 Zackroff RV, Idler WW, Steinert PM, Goldman RD (1982) In vitro reconstitution of intermediate filaments form mammalian neurofilament triplet polypeptides. Proc Natl Acad Sci U S A 79:754–757 Zhai J, Lin H, Julien JP, Schlaepfer WW (2007) Disruption of neurofilament network with aggregation of light neurofilament protein: a common pathway leading to motor neuron degeneration due to Charcot-Marie-Tooth disease-linked mutations in NFL and HSPB1. Hum Mol Genet 16:3103–3116 Zhang Z, Casey DM, Julien JP, Xu Z (2002) Normal dendritic arborization in spinal motoneurons requires neurofilament subunit L. J Comp Neurol 450:144–152 Zhu Q, Couillard-Despres S, Julien JP (1997) Delayed maturation of regenerating myelinated axons in mice lacking neurofilaments. Exp Neurol 148:299–316 Zuchner S, Vorgerd M, Sindern E, Schroder JM (2004) The novel neurofilament light (NEFL) mutation Glu397Lys is associated with a clinically and morphologically heterogeneous type of CharcotMarie-Tooth neuropathy. Neuromuscul Disord 14:147–157

Neurofilament dynamics and involvement in neurological disorders.

Neurons are extremely polarised cells in which the cytoskeleton, composed of microtubules, microfilaments and neurofilaments, plays a crucial role in ...
4MB Sizes 0 Downloads 9 Views