S17

Journal of Alzheimer’s Disease 40 (2014) S17–S22 DOI 10.3233/JAD-132315 IOS Press

Review

Crosstalk between Axonal Classical Microtubule-Associated Proteins and End Binding Proteins during Axon Extension: Possible Implications in Neurodegeneration a,b ´ C.L. Sayasa,b,1,∗ and Jes´us Avila a Centro

de Biolog´ıa Molecular “Severo Ochoa” (CSIC-UAM), Universidad Aut´onoma de Madrid, Campus de Cantoblanco, Madrid, Spain b Centro de Investigaci´ on Biom´edica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain

Accepted 26 December 2013

Abstract. During neuronal development, spherical neuroblasts differentiate into mature neurons through the extension of a long axon and several shorter dendrites. Morphological changes that underlie neuronal differentiation are mostly driven by the microtubular cytoskeleton. Regulation of microtubule dynamics and stability during axon and dendrite extension relies on the action of different families of microtubular proteins, such as classical microtubule-associated proteins (MAPs) and microtubule plus-end tracking proteins (+TIPs). This review article addresses recent research on the crosstalk between the main axonal MAPs, tau and MAP1B, and end binding proteins (EBs), the core +TIPs, during axon outgrowth in developing neuronal cells. Furthermore, we discuss the potential implications of the dysregulation of the interplay between tau and EBs in neurodegenerative disorders such as Alzheimer’s disease. Keywords: Alzheimer’s disease, axon outgrowth, MAPs, MAP1B, microtubule dynamics, neurodegeneration, neuronal development, tau, +TIPs

INTRODUCTION Neuronal development takes place following a complex program of morphological changes that gives rise to highly polarized neurons. Mature neurons possess two morphologically and functionally different types 1 Current address: Centro de Investigaciones Biom´ edicas de Canarias (CIBICAN), Universidad de La Laguna, Tenerife, Spain. ∗ Correspondence to: C.L. Sayas, Centro de Investigaciones Biom´edicas de Canarias (CIBICAN), Instituto de Tecnolog´ias Biom´edicas (ITB), Departamento de Anatom´ia, Facultad de Medicina, Campus de Ciencias de la Salud, Universidad de La Laguna, Tenerife, Spain. Tel.: +349229336; E-mails: [email protected]; [email protected].

of cytoplasmic extensions: a single axon, long and thin, which transmits signals, and multiple shorter dendrites, which receive signals. Deciphering the mechanisms that govern neuronal polarization and development is essential to understand synaptic transmission, plasticity, and neurodegeneration. Microtubule (MT) cytoskeleton is crucial for neuronal polarization, neurite and axon extension during neuronal differentiation. In mature neurons, MTs play an important role in the maintenance of cell shape and axonal transport. Hence, MT dynamics and stability have to be tightly regulated processes in both differentiating and adult neurons. Different types of interacting

ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

S18

´ C.L. Sayas and J. Avila / Crosstalk between Axonal Classical Microtubule-Associated Proteins

proteins participate in the regulation of MT dynamics/stability in neurons [1]. Among them are classical structural MT-associated proteins (MAPs), which bind along the MT lattice and MT plus-end tracking proteins (+TIPs), which specifically accumulate at plus-ends of growing MTs. Recent reports indicate that these two types of microtubular proteins might act coordinately to regulate axon extension during neuronal development ([1], Sayas et al, unpublished data). We will discuss the possible consequences of the dysregulation of the functional interplay between the classical MAP tau and end binding proteins (EBs) in neurodegenerative disorders such as Alzheimer’s disease (AD). MT ORGANIZATION IN NEURONS In neurons, MTs form dense parallel fascicles (bundles) that are required for the growth and maintenance of axons and dendrites. These MTs are nucleated at the centrosome [2], released by the action of katanin, a MT-severing protein [3–5], and then transported as short polymers into neurites, axons, and dendrites by motor proteins [6, 7]. Besides MT transport, MT polymerization occurs in every neuronal compartment [8]. Organization of MTs differs between axons and dendrites in two major aspects: 1) axonal MTs present a uniform orientation, with their plus-ends facing the axon tip, whereas MT orientation in dendrites is mixed, with their plus-ends facing either the cell body or the dendritic tip; 2) the expression of MAPs is different, with MAP2 mostly found in dendrites and MAP1B and tau mainly present in axons [9]. Hence, it is possible to identify the type of cytoplasmatic extension based on the specific organization of the MTs present in each one. ROLES OF CLASSICAL MAPS IN NEURITE/AXON EXTENSION Classical or structural MAPs bind along MTs and stabilize them [10]. Tau and microtubule-associated protein 1B (MAP1B) are classical MAPs, mostly present in neurons [10]. MAP1B is the first MAP expressed, with highest expression levels during axon extension [11, 12]. Downregulation of MAP1B levels in the neonatal brain coincides with the completion of axonal extension and the initiation of active synaptogenesis [13]. This reduction in MAP1B protein levels is the consequence of a gradual decrease of MAP1B mRNA levels by the end of axonogenesis [14]. This process is regulated by the fragile X mental retarda-

tion protein (FMRP), which interacts with MAP1B mRNA and regulates its translation [14]. Contrary to MAP1B, tau expression levels and molecular complexity reach a peak upon synaptic formation and neuronal maturation [15–21]. Curiously, tau is a RNA binding protein but nothing is known about the regulation of tau mRNA translation in relation with its axonal expression. Both MAP1B and tau are direct MT regulators that promote MT nucleation, polymerization, and stabilization, in vitro and in vivo, but tau is a more potent MT stabilizer than MAP1B and induces MT bundling [22–25]. Both MAPs regulate also MT dynamics [26–29]. In cells, MAP1B is present both in cytosol and bound to dynamic MTs whereas tau mostly localizes along MTs, stabilizing them [22, 27]. In non-neuronal cells, ectopic expression of MAP1B enhances the population of dynamic MTs while tau overexpression induces extension of neuritelike protrusions and MT stabilization through the formation of bundles [16, 27]. In developing neurons, MAP1B and tau mainly localize in growing axons, particularly at their distal region [30, 31]. In line with their localization, both MAP1B and tau play important roles during neurite and axon extension. Downregulation of MAP1B or tau in neuron-like cells or primary neurons, using antisense oligos or by siRNA-mediated knockdown, leads to a delay in neurite/axon extension [32–36]. Hippocampal neurons from Map1b hypomorphous or TAU knockout mice present shorter axons than wildtype neurons [33, 37]. However, while tau-deficient mice are viable and do not present noticeable alterations in brain structure or behavioral deficits until they age [37–40], MAP1B-deficient mice show more dramatic phenotypes due to abnormal brain development that range from mild to severe (lyssencephaly-like), depending on the mice genetic background [41–44]. Mice deficient in both MAP1B and tau present more severe phenotypes related to brain development than tau-deficient mice [45].

ROLES OF EBS IN NEURITE/AXON EXTENSION MT plus-end tracking proteins (+TIPs) specifically accumulate at the plus-ends of growing MTs [46, 47]. +TIPs act as local regulators of MT dynamics and influence different dynamic instability parameters [46, 47]. +TIPs also provide a link between MT distal ends and actin, cell cortex, or organelles [46, 47].

´ C.L. Sayas and J. Avila / Crosstalk between Axonal Classical Microtubule-Associated Proteins

+TIPs are ubiquitous proteins, highly conserved among eukaryotes. +TIPs comprise a group of structurally unrelated proteins that range from motor proteins to MT polymerases/depolymerases, adaptor or regulatory proteins [46, 47]. Diverse +TIPs have been shown to play important roles during different stages of neuronal development such as migration, axon extension and guidance, dendritic spine formation, and maintenance of proper communication between neurons [48]. We will focus on the known roles of the family of EB proteins during neurite and axon extension. The EB protein family consists of three members (EB1-3) and is considered as the ‘core’ +TIP family [47], since EB1/3 track MT ends autonomously and mark all growing MTs [49–54]. Every known+TIP interacts with EB1/3 and many of them require EB1like proteins for plus-end tracking. Furthermore, many +TIPs interact with each other at MT plus-ends [47]. During neuronal development, EB1/3 are present in all neuronal compartments, indicating the existence of local MT polymerization throughout the neuron [8]. In differentiating neuroblastoma cells, EB1 regulates MT growth rate, growth distance, and duration and EB1 downregulation leads to reduction in neurite length [55]. Of the three family members, EB3 is predominantly expressed in brain, in particular in neurons [56]. EB3 is localized at the plus-ends of MTs that enter filopodia in advancing growth cones where it interacts with the F-actin-binding protein drebrin [57]. This interaction is crucial for the proper formation of growth cones and neurite extension. Of note, EB3 has also been found at growing ends of dynamic MTs that enter dendritic spines [58]. In spines, EB3 modulates actin dynamics through its interaction with p140Cap, a protein that binds the F-actin binding protein cortactin [58]. EB3 might act therefore as a linker between MTs and F-actin in neurons. In adult neurons, both EB1 and EB3 have been shown to be concentrated and stabilized in the axon initial segment (AIS), where they interact directly with the scaffold protein ankyrin G (ankG) [59]. EBs coordinate a molecular and functional interplay between MTs and ankG in the AIS [59]. Furthermore, EB1 is crucial for axonal targeting of potassium channel Kv1 (also enriched in the AIS), which regulate action potential propagation, an evolutionarily conserved function important for the control of motor behavior [60]. Therefore, EBs (EB1/3) function as local regulators of MT dynamics during neurite and axon outgrowth and play central roles in the maintenance of neuronal polarity through their actions on the AIS.

S19

FUNCTIONAL INTERPLAY BETWEEN AXONAL MAPS AND EBS DURING AXON OUTGROWTH MTs in neurites and axons are bundled into fascicles by the cross-linking action of MAPs, and on entering the growth cone they splay out with their plus-ends oriented distally [9]. EB1/3 accumulate at MT plusends in a comet-like pattern, in contrast with classical MAPs that bind along the MT lattice [47] (Fig. 1). The number of binding sites for EBs is higher at the very tip of MT distal ends and decays at a constant rate, leading to the typical comet-shape [47]. EBs undergo a fast exchange on/off MT growing ends [61]. Therefore, a pool of EB proteins exists in the cytosol and its interaction with MTs is susceptible to regulation. Our recent findings indicate that classical MAPs regulate EB1/3 localization and function during neurite and axon extension [1]. We showed that MAP1B overexpression displaces EBs from MT plus-ends whereas MAP1B downregulation enhances EB1/3 interaction with MTs [1]. In MAP1B-deficient neurons, the behavior of EB proteins is altered, leading to changes in MT dynamics [1]. This gives rise to MT overstabilization and looping in growth cones and contributes to growth cone remodeling and a delay in axon outgrowth in Map1b-/- neurons [1]. The effect of MAP1B on EB proteins is direct; MAP1B interacts with EB1/3 and sequesters them in the cytosol of elongating neurites/axons and growth cones of developing neuronal cells (Fig. 1a, b). In this way, MAP1B reduces the effective pool of cytosolic EBs that is available to interact with MT plus-ends and keeps MT highly dynamic during neurite/axon outgrowth. Of note, while EB1 levels remains constant, expression levels of tau and EB3 increase as neurons become mature and establish synaptic contacts [17, 58]. Our data indicate that tau exerts a different type of crosstalk with EB proteins in extending neurites/axons of differentiating neuronal cells (Sayas et al, unpublished data; Fig. 1a). In line with this, MAP2, another classical neuronal (dendritic) MAP that shares with tau conserved MT-binding repeats and an amino-terminal projection domain [62], has been shown to modulate the localization of EB3 [63]. MAP2 induces EB3 redistribution to MT bundles in a prolonged response to NMDA in dendrites of mature neurons [63]. From these studies, EBs emerge as key effectors of the actions of classical MAPs during axon extension. Thus, MAP1B and tau may act as local regulators of MT dynamics by modulating the localization and

S20

´ C.L. Sayas and J. Avila / Crosstalk between Axonal Classical Microtubule-Associated Proteins

play between tau and EBs may still occur in adult neurons. In this context, it would be interesting to study whether hyperphosphorylated tau regulates the binding of EBs to MTs. If this is the case, the abnormal localization of tau in AD or tauopathies might lead to mislocalization of EB1/3 proteins in affected brains. Given that EBs are the main known regulators of MT dynamics [65], dysregulation of EBs distribution and function could further alter MT organization under pathological situations. As a consequence, many known EBs-interacting partners might also be mislocalized in AD or tauopathies. Misdistribution of the whole set of +TIPs, which normally form protein hubs at MT plus-ends, might have extra deleterious effects on MT architecture, and subsequently, on MT-dependent axonal transport. Research should be done to confirm whether a disruption of the crosstalk between tau and EBs occurs upon neurodegeneration and how it contributes to the pathology. CONCLUSIONS

Fig. 1. Interplay between axonal MAPs and +TIPs during neuronal development. In the axon shaft (a) and growth cone (b), MAP1B localizes along MTs and in the cytosol. MAP1B interacts directly with EB1/3 and sequesters them in the cytosol of extending axons and growth cones (a and b). Moreover, microtubular MAP1B displaces EB1/3 from the MT lattice. In this way, MAP1B contributes to keep MTs highly dynamic during neuronal differentiation. On the other hand, tau, which binds MTs and induces their bundling and stabilization (detail in a), crosstalks with EB1/3, regulating MT dynamics and stability during neuronal development.

function of EB1/3. Notably, MAP1B and tau regulate EBs in different manners indicating that their roles during axon extension are not overlapping, contrary to the general view.

The correct formation and proper function of the nervous system relies on the ability of neurons to polarize and undergo major morphological changes. Rearrangements of MT cytoskeleton underlie these changes in cell shape. MT assembly and dynamics are regulated by a plethora of proteins, such as classical MAPs and +TIPs [10, 46, 47, 64]. Recent findings indicate that axon extension is regulated by the coordinated actions of axonal MAPs (MAP1B and tau) and the core + TIPs (EBs) ([1], Sayas et al., unpublished data). From these studies MAPs have emerged as direct regulators of EBs localization and function during neurite and axon outgrowth. It remains to be elucidated whether the interplay between classical MAPs and EBs, or its disruption, contributes to neurodegeneration in disorders such as AD.

POSSIBLE IMPLICATIONS IN ALZHEIMER’S DISEASE

ACKNOWLEDGMENTS

Neurofibrillary tangles, a histopathological hallmark of AD, are composed of paired helical filaments made of hyperphosphorylated tau [20]. Abnormal tau hyperphosphorylation is also a hallmark of tauopathies, other related neurodegenerative disorders [64]. Since tau modulates the interaction of EBs with MTs during neuronal development (Sayas et al., unpublished data) and their levels remain high in adult neurons [17, 58], it is likely that the inter-

C.L. Sayas was supported by CSIC and CIBERNED (Madrid), and is currently supported by the IMBRAIN project. The authors would like to acknowledge the IMBRAIN project (FP7-REGPOT-2012-CT201231637-IMBRAIN), funded under the 7th Framework Programme (Capacities). This work was supported by grants from the Spanish Research Council (CSIC) (Plan Nacional), CIBERNED and Comunidad de Madrid (Spain).

´ C.L. Sayas and J. Avila / Crosstalk between Axonal Classical Microtubule-Associated Proteins

Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=2080).

[18]

REFERENCES [19] [1]

[2]

[3]

[4]

[5]

[6]

[7] [8]

[9]

[10]

[11]

[12]

[13]

[14]

[15]

[16]

[17]

Tortosa E, Galjart N, Avila J, Sayas CL (2013) MAP1B regulates microtubule dynamics by sequestering EB1/3 in the cytosol of developing neuronal cells. EMBO J 32, 1293-1306. Yu W, Centonze VE, Ahmad FJ, Baas PW (1993) Microtubule nucleation and release from the neuronal centrosome. J Cell Biol 122, 349-359. Ahmad FJ, Yu W, McNally FJ, Baas PW (1999) An essential role for katanin in severing microtubules in the neuron. J Cell Biol 145, 305-315. Karabay A, Yu W, Solowska JM, Baird DH, Baas PW (2004) Axonal growth is sensitive to the levels of katanin, a protein that severs microtubules. J Neurosci 24, 5778-5788. Yu W, Solowska JM, Qiang L, Karabay A, Baird D, Baas PW (2005) Regulation of microtubule severing by katanin subunits during neuronal development. J Neurosci 25, 55735583. Hirokawa N, Noda Y (2008) Intracellular transport and kinesin superfamily proteins, KIFs: Structure, function, and dynamics. Physiol Rev 88, 1089-1118. Baas PW, Buster DW (2004) Slow axonal transport and the genesis of neuronal morphology. J Neurobiol 58, 3-17. Stepanova T, Slemmer J, Hoogenraad CC, Lansbergen G, Dortland B, De Zeeuw CI, Grosveld F, van Cappellen G, Akhmanova A, Galjart N (2003) Visualization of microtubule growth in cultured neurons via the use of EB3-GFP (endbinding protein 3-green fluorescent protein). J Neurosci 23, 2655-2664. Conde C, Caceres A (2009) Microtubule assembly, organization and dynamics in axons and dendrites. Nat Rev 10, 319-332. Avila J, Dominguez J, Diaz-Nido J (1994) Regulation of microtubule dynamics by microtubule-associated protein expression and phosphorylation during neuronal development. Int J Dev Biol 38, 13-25. Tucker RP, Binder LI, Matus AI (1988) Neuronal microtubule-associated proteins in the embryonic avian spinal cord. J Comp Neurol 271, 44-55. Tucker RP, Garner CC, Matus A (1989) In situ localization of microtubule-associated protein mRNA in the developing and adult rat brain. Neuron 2, 1245-1256. Riederer BM, Guadano-Ferraz A, Innocenti GM (1990) Difference in distribution of microtubule-associated proteins 5a and 5b during the development of cerebral cortex and corpus callosum in cats: dependence on phosphorylation. Brain Res Dev Brain Res 56(2), 235-43. Lu R, Wang H, Liang Z, Ku L, O’Donnell WT, Li W, Warren ST, Feng Y (2004) The fragile X protein controls microtubuleassociated protein 1B translation and microtubule stability in brain neuron development. Proc Natl Acad Sci U S A 101, 15201-15206. Kanai Y, Hirokawa N (1995) Sorting mechanisms of tau and MAP2 in neurons: Suppressed axonal transit of MAP2 and locally regulated microtubule binding. Neuron 14, 421-432. Knops J, Kosik KS, Lee G, Pardee JD, Cohen-Gould L, McConlogue L (1991) Overexpression of tau in a nonneuronal cell induces long cellular processes. J Cell Biol 114, 725-733. Mareck A, Fellous A, Francon J, Nunez J (1980) Changes in composition and activity of microtubule-associated

[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

S21

proteins during brain development. Nature 284, 353355. Schoenfeld TA, Obar RA (1994) Diverse distribution and function of fibrous microtubule-associated proteins in the nervous system. Int Rev Cytol 151, 67-137. Vanier MT, Neuville P, Michalik L, Launay JF (1998) Expression of specific tau exons in normal and tumoral pancreatic acinar cells. J Cell Sci 111(Pt 10), 1419-1432. Avila J, Lucas JJ, Perez M, Hernandez F (2004) Role of tau protein in both physiological and pathological conditions. Physiol Rev 84, 361-384. Francon J, Lennon AM, Fellous A, Mareck A, Pierre M, Nunez J (1982) Heterogeneity of microtubule-associated proteins and brain development. Eur J Biochem 129, 465-471. Takemura R, Okabe S, Umeyama T, Kanai Y, Cowan NJ, Hirokawa N (1992) Increased microtubule stability and alpha tubulin acetylation in cells transfected with microtubuleassociated proteins MAP1B, MAP2 or tau. J Cell Sci 103(Pt 4), 953-964. Vandecandelaere A, Pedrotti B, Utton MA, Calvert RA, Bayley PM (1996) Differences in the regulation of microtubule dynamics by microtubule-associated proteins MAP1B and MAP2. Cell Motil Cytoskeleton 35, 134-146. Pedrotti B, Islam K (1995) Microtubule associated protein 1B (MAP1B) promotes efficient tubulin polymerisation in vitro. FEBS Lett 371, 29-31. Brandt R, Lee G (1993) Functional organization of microtubule-associated protein tau. Identification of regions which affect microtubule growth, nucleation, and bundle formation in vitro. J Biol Chem 268, 3414-3419. Panda D, Samuel JC, Massie M, Feinstein SC, Wilson L (2003) Differential regulation of microtubule dynamics by three- and four-repeat tau: Implications for the onset of neurodegenerative disease. Proc Natl Acad Sci U S A 100, 9548-9553. Goold RG, Owen R, Gordon-Weeks PR (1999) Glycogen synthase kinase 3beta phosphorylation of microtubule-associated protein 1B regulates the stability of microtubules in growth cones. J Cell Sci 112(Pt 19), 3373-3384. Panda D, Goode BL, Feinstein SC, Wilson L (1995) Kinetic stabilization of microtubule dynamics at steady state by tau and microtubule-binding domains of tau. Biochemistry 34, 11117-11127. Bunker JM, Wilson L, Jordan MA, Feinstein SC (2004) Modulation of microtubule dynamics by tau in living cells: Implications for development and neurodegeneration. Mol Biol Cell 15, 2720-2728. Black MM, Slaughter T, Fischer I (1994) Microtubuleassociated protein 1b (MAP1b) is concentrated in the distal region of growing axons. J Neurosci 14, 857-870. Black MM, Slaughter T, Moshiach S, Obrocka M, Fischer I (1996) Tau is enriched on dynamic microtubules in the distal region of growing axons. J Neurosci 16, 3601-3619. Tymanskyj SR, Scales TM, Gordon-Weeks PR (2012) MAP1B enhances microtubule assembly rates and axon extension rates in developing neurons. Mol Cell Neurosci 49, 110-119. Gonzalez-Billault C, Avila J, Caceres A (2001) Evidence for the role of MAP1B in axon formation. Mol Biol Cell 12, 20872098. Caceres A, Kosik KS (1990) Inhibition of neurite polarity by tau antisense oligonucleotides in primary cerebellar neurons. Nature 343, 461-463. DiTella MC, Feiguin F, Carri N, Kosik KS, Caceres A (1996) MAP-1B/TAU functional redundancy during

S22

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47] [48]

[49] [50]

´ C.L. Sayas and J. Avila / Crosstalk between Axonal Classical Microtubule-Associated Proteins laminin-enhanced axonal growth. J Cell Sci 109(Pt 2), 467-477. Yu W, Qiang L, Solowska JM, Karabay A, Korulu S, Baas PW (2008) The microtubule-severing proteins spastin and katanin participate differently in the formation of axonal branches. Mol Biol Cell 19, 1485-1498. Dawson HN, Ferreira A, Eyster MV, Ghoshal N, Binder LI, Vitek MP (2001) Inhibition of neuronal maturation in primary hippocampal neurons from tau deficient mice. J Cell Sci 114, 1179-1187. Harada A, Oguchi K, Okabe S, Kuno J, Terada S, Ohshima T, Sato-Yoshitake R, Takei Y, Noda T, Hirokawa N (1994) Altered microtubule organization in small-calibre axons of mice lacking tau protein. Nature 369, 488-491. Fujio J, Hosono H, Ishiguro K, Ikegami S, Fujita SC (2007) Tau phosphorylation in the mouse brain during aversive conditioning. Neurochem Int 51, 200-208. Ikegami S, Harada A, Hirokawa N (2000) Muscle weakness, hyperactivity, and impairment in fear conditioning in tau-deficient mice. Neurosci Lett 279, 129-132. Meixner A, Haverkamp S, Wassle H, Fuhrer S, Thalhammer J, Kropf N, Bittner RE, Lassmann H, Wiche G, Propst F (2000) MAP1B is required for axon guidance and is involved in the development of the central and peripheral nervous system. J Cell Biol 151, 1169-1178. Gonzalez-Billault C, Demandt E, Wandosell F, Torres M, Bonaldo P, Stoykova A, Chowdhury K, Gruss P, Avila J, Sanchez MP (2000) Perinatal lethality of microtubuleassociated protein 1B-deficient mice expressing alternative isoforms of the protein at low levels. Mol Cell Neurosci 16, 408-421. Edelmann W, Zervas M, Costello P, Roback L, Fischer I, Hammarback JA, Cowan N, Davies P, Wainer B, Kucherlapati R (1996) Neuronal abnormalities in microtubule-associated protein 1B mutant mice. Proc Natl Acad Sci U S A 93, 12701275. Takei Y, Kondo S, Harada A, Inomata S, Noda T, Hirokawa N (1997) Delayed development of nervous system in mice homozygous for disrupted microtubule-associated protein 1B (MAP1B) gene. J Cell Biol 137, 1615-1626. Takei Y, Teng J, Harada A, Hirokawa N (2000) Defects in axonal elongation and neuronal migration in mice with disrupted tau and map1b genes. J Cell Biol 150, 989-1000. Gouveia SM, Akhmanova A (2010) Cell and molecular biology of microtubule plus end tracking proteins: End binding proteins and their partners. Int Rev Cell Mol Biol 285, 1-74. Galjart N (2010) Plus-end-tracking proteins and their interactions at microtubule ends. Curr Biol 20, R528-R537. Jaworski J, Hoogenraad CC, Akhmanova A (2008) Microtubule plus-end tracking proteins in differentiated mammalian cells. Int J Biochem Cell Biol 40, 619-637. Lansbergen G, Akhmanova A (2006) Microtubule plus end: A hub of cellular activities. Traffic 7, 499-507. Bieling P, Kandels-Lewis S, Telley IA, van Dijk J, Janke C, Surrey T (2008) CLIP-170 tracks growing microtubule ends by dynamically recognizing composite EB1/tubulin-binding sites. J Cell Biol 183, 1223-1233.

[51]

[52]

[53]

[54]

[55]

[56]

[57]

[58]

[59]

[60]

[61]

[62] [63]

[64]

[65]

Bieling P, Laan L, Schek H, Munteanu EL, Sandblad L, Dogterom M, Brunner D, Surrey T (2007) Reconstitution of a microtubule plus-end tracking system in vitro. Nature 450, 1100-1105. Zimniak T, Stengl K, Mechtler K, Westermann S (2009) Phosphoregulation of the budding yeast EB1 homologue Bim1p by Aurora/Ipl1p. J Cell Biol 186, 379-391. Dixit R, Barnett B, Lazarus JE, Tokito M, Goldman YE, Holzbaur EL (2009) Microtubule plus-end tracking by CLIP170 requires EB1. Proc Natl Acad Sci U S A 106, 492-497. Komarova Y, De Groot CO, Grigoriev I, Gouveia SM, Munteanu EL, Schober JM, Honnappa S, Buey RM, Hoogenraad CC, Dogterom M, Borisy GG, Steinmetz MO, Akhmanova A (2009) Mammalian end binding proteins control persistent microtubule growth. J Cell Biol 184, 691-706. Stepanova T, Smal I, van Haren J, Akinci U, Liu Z, Miedema M, Limpens R, van Ham M, van der Reijden M, Poot R, Grosveld F, Mommaas M, Meijering E, Galjart N (2010) History-dependent catastrophes regulate axonal microtubule behavior. Curr Biol 20, 1023-1028. Nakagawa H, Koyama K, Murata Y, Morito M, Akiyama T, Nakamura Y (2000) EB3, a novel member of the EB1 family preferentially expressed in the central nervous system, binds to a CNS-specific APC homologue. Oncogene 19, 210-216. Geraldo S, Khanzada UK, Parsons M, Chilton JK, GordonWeeks PR (2008) Targeting of the F-actin-binding protein drebrin by the microtubule plus-tip protein EB3 is required for neuritogenesis. Nat Cell Biol 10, 1181-1189. Jaworski J, Kapitein LC, Gouveia SM, Dortland BR, Wulf PS, Grigoriev I, Camera P, Spangler SA, Di Stefano P, Demmers J, Krugers H, Defilippi P, Akhmanova A, Hoogenraad CC (2009) Dynamic microtubules regulate dendritic spine morphology and synaptic plasticity. Neuron 61, 85-100. Leterrier C, Vacher H, Fache MP, d’Ortoli SA, Castets F, Autillo-Touati A, Dargent B (2011) End-binding proteins EB3 and EB1 link microtubules to ankyrin G in the axon initial segment. Proc Natl Acad Sci U S A 108, 8826-8831. Gu C, Zhou W, Puthenveedu MA, Xu M, Jan YN, Jan LY (2006) The microtubule plus-end tracking protein EB1 is required for Kv1 voltage-gated K+channel axonal targeting. Neuron 52, 803-816. Dragestein KA, van Cappellen WA, van Haren J, Tsibidis GD, Akhmanova A, Knoch TA, Grosveld F, Galjart N (2008) Dynamic behavior of GFP-CLIP-170 reveals fast protein turnover on microtubule plus ends. J Cell Biol 180, 729-737. Dehmelt L, Halpain S (2005) The MAP2/Tau family of microtubule-associated proteins. Genome Biol 6, 204. Kapitein LC, Yau KW, Gouveia SM, van der Zwan WA, Wulf PS, Keijzer N, Demmers J, Jaworski J, Akhmanova A, Hoogenraad CC (2011) NMDA receptor activation suppresses microtubule growth and spine entry. J Neurosci 31, 8194-8209. Iqbal K, Liu F, Gong CX, Grundke-Iqbal I (2010) Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res 7, 656-664. Tirnauer JS, Bierer BE (2000) EB1 proteins regulate microtubule dynamics, cell polarity, and chromosome stability. J Cell Biol 149, 761-766.

Crosstalk between axonal classical microtubule-associated proteins and end binding proteins during axon extension: possible implications in neurodegeneration.

During neuronal development, spherical neuroblasts differentiate into mature neurons through the extension of a long axon and several shorter dendrite...
190KB Sizes 0 Downloads 0 Views