CELL STRUCTURE AND FUNCTION 35: 15–22 (2010) © 2010 by Japan Society for Cell Biology Review

Unique Post-Translational Modifications in Specialized Architecture

Koji Ikegami∗ and Mitsutoshi Setou Department of Molecular Anatomy, Molecular Imaging Advanced Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan

ABSTACT. (MTs) play specialized roles in a wide variety of cellular events, e.g. molecular transport, cell motility, and cell division. Specialized MT architectures, such as bundles, axonemes, and centrioles, underlie the function. The specialized function and highly organized structure depend on interactions with MT-binding proteins. MT-associated proteins (e.g. MAP1, MAP2, and tau), molecular motors (kinesin and dynein), plus-end tracking proteins (e.g. CLIP-170), and MT-severing proteins (e.g. katanin) interact with MTs. How can the MT-binding proteins know temporospatial information to associate with MTs and to properly play their roles? Post-translational modifications (PTMs) including , , and polyglycylation can provide molecular landmarks for the proteins. Recent efforts to identify modification- regulating enzymes (TTL, carboxypeptidase, polyglutamylase, polyglycylase) and to generate genetically manipulated animals enable us to understand the roles of the modifications. In this review, we present recent advances in understanding regulation of MT function, structure, and stability by PTMs.

Key words: microtubule/post-translational modification/detyrosination/glutamylation/glycylation

Introduction between MT and a variety of its binding partners. The mole- cules contain various proteins with a wide range of mass, The microtubule (MT) is one of three major cytoskeletons structure, and function. Molecular motor proteins, kinesins highly conserved in eukaryotes. As the name indicates, the and dyneins, use MT as a molecular railway and walk on it. MT has a tubular structure with a diameter of 25 nm (Fig. MT-associated proteins (MAPs) starch up the MTs, and 1). The tubule wall is composed of thirteen protofilaments, thus stabilize them. Plus-end tracking proteins (+TIPs) which is further constructed by heterodimers of two small accumulate on the distal tip of the MT. Some other molec- globular proteins, α- and β- (Mohri, 1968) (Fig. 1). ular species can sever and thus destabilize MT. Those MT- MTs play specific roles in a variety of cellular events, which interacting proteins associate with MTs in designated sub- range from directional molecular transport, ciliary or flagellar cellular positions to play their roles properly. motility, and chromosome segregation, to cytokinesis. To What leads the MT-interacting proteins to their work play these specialized roles, the MT constructs higher-order place and how are they regulated, as different kinesin architecture in related subcellular compartments, such as motors deliver their cargos to each specific destination in a bundles (in neuronal processes), axonemes (in flagella and neuronal cell (Setou et al., 2000; Setou et al., 2002; Setou et cilia), and centrioles (in basal bodies or spindle bodies) al., 2004)? Two mechanisms can be assumed. One is a well- (Fig. 1). known regulatory system, the post-translational modifica- How does the simple tubular MT form the highly orga- tions (PTMs) of MT-interacting proteins. The most popular nized architecture? The mechanism relies on interactions example is the of MAPs. Evidence has accumulated that the phosphorylation of MAPs affects *To whom correspondence should be addressed: Koji Ikegami, Depart- their function. The phosphorylation of tau inhibits the func- ment of Molecular Anatomy, Molecular Imaging Advanced Research tion of tau to promote MT assembly (Nishida et al., 1982; Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan. Yamamoto et al., 1983; Lindwall and Cole, 1984; Hoshi et Tel: +81–53–435–2085, Fax: +81–53–435–2292 al., 1987; Wada et al., 1998). The phosphorylation of MAP2 E-mail: [email protected] counteracts MAP2-mediated microtubule polymerization Abbreviations: mAb, monoclonal antibody; MAP, microtubule-associated protein; MT, microtubule; PTM, post-translational modification; TTL, (Jameson et al., 1980; Nishida et al., 1981; Yamamoto et ligase. al., 1983; Nishida et al., 1987; Hoshi et al., 1988). Hence,

15 K. Ikegami and M. Setou

Fig. 1. Microtubule in cell. Microtubule-enriched subcellular compartments or structures are depicted. Microtubule is shown in red. Microtubule- constructed higher architectures, such as bundle, axoneme, and centriole, are highlighted in boxes. Individual microtubule with diameter of 25 nm is composed of 13 protofilaments, which is further constructed from multiple heterodimers of α- and β-tubulins. PTMs currently known are listed below. phosphorylation of MAPs negatively regulates MTs dynam- review the currently available evidence for the roles of the ics and stability (Drewes et al., 1997). CRMP2 phosphory- PTMs in mammals with emphasis on recent progress. We lation inactivates the activity of CRMP2 to promote micro- also present some evidence obtained from genetically modi- tubule assembly and controls neuronal polarity (Yoshimura fied non-mammalian organisms. Readers are referred to et al., 2005). Moreover, MAPs phosphorylation indirectly some review articles for other general modifications (acety- modulates kinesin motor trafficking through affecting lation, phosphorylation, and palmitoylation) (Rosenbaum, MAPs-MT binding affinity (Sato-Harada et al., 1996). 2000; Westermann and Weber, 2003; Verhey and Gaertig, The multi-microtubule hypothesis can support another 2007; Hammond et al., 2008). mechanism. The heterogeneities of tubulin molecules underlie the model (Kobayashi and Mohri, 1977). The heterogeneities are made by two independent mechanisms: Detyrosination/Tyrosination genome-encoded multi-tubulin genes and PTMs. Mamma- lian genome encodes 6 to 8 different tubulin molecules for The cycle of detyrosination and tyrosination is a PTM of each α- and β-tubulins. Those different tubulins show dif- tubulin that has been investigated for the longest time. This ferent tissue-specific expression patterns. PTMs can more PTM occurs in most α-tubulins. In this cycle, a tyrosine that drastically contribute to the heterogeneities. is encoded as the C-terminal by genome is Tubulin is subjected to the detyrosination/tyrosination removed (Argaraña et al., 1978), and a free tyrosine is re- cycle, the removal of penultimate glutamate, polyglutamy- added to the C-terminal of detyrosinated α-tubulin (Barra et lation, polyglycylation, , phosphorylation, and al., 1974). Long-living, i.e. stable MTs are enriched by palmitoylation (Fig. 1). These modifications, except for detyrosinated tubulins while highly dynamic MTs contain acetylation, occur in the C-terminal region, which surfaces abundantly tyrosinated tubulins (Wehland and Weber, on MT lattice where many MT-binding proteins interact. In 1987). A recent work demonstrates that tubulin detyrosina- this review, we focus on unique PTMs (detyrosination/ tion inhibits MT disassembly through suppressing interac- tyrosination, polyglutamylation, and polyglycylation), and tions of MT-depolymerizing motor, MCAK or KIF2 with

16 Unique Post-Translational Modifications in Specialized Microtubule Architecture

Fig. 2. Enzyme for tubulin PTMs. Identified enzymes for modifications are shown in red boldface. Carboxypeptidase for generating detyrosinated tubulin is still only a candidate (yellow). Unidentified enzymes are shown in light gray.

MTs (Peris et al., 2009), although detyrosination has been al., 2006). Indeed, CLIP-170 preferentially binds in vitro to thought not to modify MT stability for a long time (Webster tyrosinated α-tubulin (Peris et al., 2006) and to the –EEY/F et al., 1990). Detyrosinated tubulin is further subjected to motif seen in the α-tubulin C-terminal (Honnappa et al., next deglutamylation, i.e. removal of penultimate glutamate, 2006; Mishima et al., 2007). Similar results are observed in producing Δ2-tubulin (Paturle-Lafanechère et al., 1991) a most primitive eukaryote, the budding yeast. Genetical (Fig. 1). In the current model, Δ2-tubulin is thought to be deletion of phenylalanine from the C-terminal of α-tubulin excluded from the detyrosination/tyrosination cycle results in mislocalization of Bik1p, a yeast homolog of (Westermann and Weber, 2003; Rüdiger et al., 1994). CLIP-170 (Badin-Larçon et al., 2004). An enzyme that accounts for the detyrosination/tyrosina- Another line of evidence indicates that the modification tion cycle is the oldest enzyme identified as a tubulin- affects the interaction between molecular motors and MTs. modifying enzyme. The enzyme for re-addition of tyrosine Kinesin-1 shows more strong binding affinity to MT enriched to detyrosinated α-tubulin, termed tubulin tyrosine ligase by detyrosinated tubulin in vitro (Liao and Gundersen, (TTL) (Raybin and Flavin, 1977) (Fig. 2), is purified from 1998). In vivo, kinesin-1 preferentially binds to detyrosi- porcine brain (Murofushi, 1980), and the polypeptide is nated tubulin-rich MTs (Dunn et al., 2008). Kinesin-1 selec- completely identified (Ersfeld et al., 1993). In contrast, the tively accumulates in a future axon in early stage of neu- enzyme removing the terminal tyrosine is not fully identi- ronal growth (Jacobson et al., 2006). The axonal shaft is fied, while recent researches propose a cytosolic carboxy more enriched by detyrosinated tubulins than dendrites and peptidase, CCP1/Nna1 as a strong candidate (Kalinina et cell body during early neuronal differentiation (Arregui et al., 2007; Rodriguez de la Vega et al., 2007) (Fig. 2). al., 1991). In light of the knowledge that has accumulated, Enzymes performing the removal of penultimate glutamate our group has recently provided clear evidence that tubulin from detyrosinated tubulin remain to be identified. Enzymes detyrosination is a spatial cue for neuronal polarity determi- re-adding glutamate to Δ2-tubulin might be present, though nation through preferential interaction between KIF5-loop8 currently nobody suggests this idea. and detyrosinated tubulin-rich MTs (Konishi and Setou, Recent progress for understanding the physiological roles 2009). The reason why neurons cultured from the TTL-null of this modification cycle is achieved by a generation of a animal form multiple axons (Erck et al., 2005) can be TTL-null mouse line. The TTL-null animal provides evi- explained by this model, whereby kinesin-1 might fail to dence for a vital role of tyrosinated tubulin. The mouse find the future axon since MTs are uniformly detyrosinated. shows malformation of layer in cerebral cortex, disruption Such a model allows us to speculate that the neuronal of cortico-thalamic loop, and perinatal lethality along with degeneration observed in Purkinje cell degeneration (pcd) prominent loss of tyrosinated tubulins (Erck et al., 2005). mouse (Mullen et al., 1976; Landis and Mullen, 1978), a Neurons cultured from the animal display abnormal neurite spontaneous mutant of CCP1/Nna1 (Fernandez-Gonzalez et growth along with forming multiple axons (Erck et al., al., 2002), might result from inefficient intracellular trans- 2005). In the neurons, the cytoplasmic linker protein ports caused by lowered binding affinity between kinesin-1 (CLIP)-170, a +TIP containing -rich cytoskeleton- and MTs that lack detyrosinated tubulins. associated protein (CAP-Gly) domains, is dispersed from the growth cone (Erck et al., 2005). Fibroblasts cultured from the TTL-null animal display mislocalization of CLIP- 170 and abnormal orientations of spindle bodies (Peris et

17 K. Ikegami and M. Setou

Fig. 3. Polyglutamylation and polyglycylation. (A) Schematic view of polyglutamylation and polyglycylation. Poly-glutamate (EEE···) or -glycine (GGG···) chain is attached to a glutamate residue (E) in a protein backbone. Note that the polyglutamate chain has negative charges. (B) More detailed diagram of polyglutamylation. The first glutamate is bound to a γ-carboxyl group of a glutamate residue. Subsequent addition of glutamates is achieved by bonds between α-carboxyl group and amino group. Note that the polyglutamate chain provides extra carboxyl groups that result in increases in total negative charges.

Polyglutamylation deficient mouse shows impaired synaptic transmission and a decrease in the amount of synaptic vesicles in synaptic Tubulin polyglutamylation was first reported in pig brain terminals associated with a gross loss of polyglutamylation MT (Eddé et al., 1990). This modification is highly intrigu- on α-tubulin in brain homogenate (Ikegami et al., 2007). ing from two points. First, the form of modification is Trafficking of KIF1A (kinesin-3 family), but neither KIF3A unique. A long chain of polypeptides constructed from mul- (kinesin-2 family) nor KIF5A (kinesin-1 family), is impaired tiple glutamates are covalently linked on a γ-carboxy group in the neurons cultured from the mouse (Ikegami et al., of glutamate residue in tubulin C-terminal region (Fig. 3). 2007). In vitro, binding of KIF1A to MT is most affected by Second, the long polyglutamate chain increases remarkably the loss of polyglutamylated α-tubulin (Ikegami et al., negative charges in the tubulin C-terminal (Fig. 3), which 2007). This KIF1-selective impairment can be explained by readily suggests the important roles of the charges in the the finding that KIF1 has a longer K-loop than KIF3 or interaction between MT and its binding partners. Studies KIF5 (Okada and Hirokawa, 1999). using a specific monoclonal antibody (mAb) GT335 against Evidence has accumulated for the spatial and temporal glutamylated tubulin reveal that polyglutamylated tubulins distribution of polyglutamylated tubulins. In neuronal cells, are enriched in neuronal cells (Wolff et al., 1992), and that polyglutamylated β-tubulin concentrates in the somato- axonemal and centriolar MTs are also highly modified dendritic region, whereas polyglutamylated α-tubulin accu- (Fouquet et al., 1994; Bobinnec et al., 1998b). mulates in the neuronal processes (Ikegami et al., 2006; A recent significant breakthrough in the area of poly- Ikegami et al., 2007) (Fig. 4). In mouse brain, polyglutamy- glutamylation research is the success in identifying lated β-tubulin emerges after birth, whereas modified α- modification-performing enzymes, polyglutamylases (or tubulin is already prominent at birth (Audebert et al., 1994). glutamate ligases) (Fig. 2). Interestingly, the enzymes are In culture, polyglutamylated β-tubulin emerges and increases members of a protein family homologous to TTL. The first in parallel with neuronal growth, while modified α-tubulin identified TTLL1 is a subunit of the polyglutamylase com- is detected immediately after starting the culture (Audebert plex purified from mouse brain (Janke et al., 2005). Further et al., 1994). The late increase in level of polyglutamylated studies demonstrate that TTLL4, 5, 6, 7, 9, 11, and 13 have β-tubulin may be involved in postnatal growth and matura- tubulin polyglutamylase activity (Ikegami et al., 2006; van tion of neuronal processes, especially dendrites. Indeed, Dijk et al., 2007; Wloga et al., 2008; Mukai et al., 2009). knockdown of TTLL7 in PC12 cells demonstrates that Currently, the glutamate-removing enzyme, deglutamylase TTLL7-mediated β-tubulin polyglutamylation is required for or glutamatase remains to be identified. CCP 2 to 6, which neurite growth (Ikegami et al., 2006). What involves poly- are members of cytosolic carboxypeptidases with similarity glutamylated β-tubulin in neurite growth currently remains to CCP1/Nna1 (Kalinina et al., 2007; Rodriguez de la Vega to be elucidated. However, MAP1A is a plausible candidate. et al., 2007), might harbor the enzymes. This hypothesis can be supported by three independent lines The identification of enzymes enables us to better under- of evidence: i) strong dependency of MAP1A-MT interac- stand the physiological roles of tubulin polyglutamylation tion on level of tubulin polyglutamylation (Bonnet et al., in detail. A genetically manipulated mouse line, which lacks 2001; Ikegami et al., 2007); ii) postnatal increase in level of PGs1 (Campbell et al., 2002), a component of the TTLL1- MAP1A in developing mouse brain (Schoenfeld et al., 1989); containing polyglutamylase complex (Regnard et al., 2003; iii) the important role of MAP1A in activity-dependent Janke et al., 2005), provides strong evidence. The PGs1- dendritic growth and branching (Szebenyi et al., 2005).

18 Unique Post-Translational Modifications in Specialized Microtubule Architecture

Fig. 4. Distribution of polyglutamylated and polyglycylated tubulins. Note that the subunits of tubulin that are well polyglutamylated are different between somato-dendrites and axons. Currently, no evidence is available that polyglycylated tubulin is detected in centrioles.

The roles of tubulin polyglutamylation in flagella or cilia Polyglycylation are still subject to controversy. The PGs1-deficient mouse shows malformation of sperm flagella, but not cilia in Polyglycylated tubulin is abundantly detected in axonemal trachea, nasal duct, or oviduct (Campbell et al., 2002), MTs (Redeker et al., 1994) (Fig. 4). In polyglycylation, a suggesting that tubulin polyglutamylation is required for poly-glycine chain is bound to a glutamate residue in tubu- axoneme construction and that the modification has distinct lin C-terminal region. In contrast to polyglutamylation, roles in flagella and cilia. Studies where modification- polyglycylation does not alter the net charge of the tubulin specific antibodies are used provide supportive evidence. C-terminal (Fig. 3). This simple addition of a mere poly- Ciliary motility of human respiratory epithelium is signifi- peptide that has no outstanding characteristic had made it cantly inhibited by injection of mAb GT335 (Million et al., difficult for researchers to anticipate the function of the 1999). A similar result is observed in a work where sea modification. However, the roles of tubulin polyglycylation urchin sperm is used. The flagellar motility was affected by are now beginning to be unveiled, as TTLLs 3, 8, and 10 injection of another mAb B3 into the sperm (Gagnon et al., are identified as polyglycylation-performing enzymes, the 1996). Although the numerous studies employing anti- polyglycylases (or glycine ligases) (Ikegami et al., 2008; bodies indicate the importance of tubulin polyglutamylation Ikegami and Setou, 2009; Rogowski et al., 2009; Wloga et in ciliary or flagellar motility, Tetrahymena, a ciliated pro- al., 2009b). tozoan swims normally even if it lacks both TTLL1 and While understanding the physiological roles of the tubu- TTLL9 (Wloga et al., 2008). The role of tubulin poly- lin polyglycylation in mammals has been slow to emerge, glutamylation may differ in organism species. To resolve elegant works using Tetrahymena have provided interesting these questions, we anticipate the development of mamma- evidence for the roles of tubulin polyglycylation. Mutations lian models, the TTLLs of which are genetically modified. in polyglycylation sites of β-tubulin, but not of α-tubulin, A recent work investigating zebrafish provides evidence have been reported to impair cellular motility and cytokine- for broad function of tubulin polyglutamylation in the cilia sis (Xia et al., 2000). Ultrastructurally, the axonemes of the of the whole body. A knockdown of Fleer, a newly identi- mutant lose the central-pair MTs and B-subfibers (Thazhath fied regulator of tubulin polyglutamylation, or TTLL6 et al., 2002), suggesting that tubulin polyglycylation has results in structural and functional abnormalities throughout a pivotal role in constructing ciliary axonemes. More the whole body (Pathak et al., 2007). However, the function recently, it has been revealed that tubulin polyglycylation by of the modification in centrioles is blurrier. The only evi- TTLL3 is essential for proper cilia assembly in Tetrahymena dence available is that mAb GT335 destabilizes the centri- and zebrafish (Wloga et al., 2009b). Interestingly, TTLL3- oles when injected into living cells (Bobinnec et al., 1998a). deficient Tetrahymena shows a counter-increase in poly- To achieve a more direct and accurate understanding glutamylation along with a decrease in tubulin polyglycyla- requires more robust investigations, e.g. those examining tion (Wloga et al., 2009b), which is consistent with the polyglutamylation-performing TTLL knockout animals. concept of cross-talk of PTMs in α- and β-tubulin C-termini (Redeker et al., 2005). The increase in polyglutamylation

19 K. Ikegami and M. Setou

Table I. ENZYMES AND FUNCTIONS OF UNIQUE TUBULIN PTMS

PTMs Enzymes Functions Detyrosination/tyrosination (CCP1/Nna1)a)/TTL · Regulating CLIP-170 binding and localization · Modulating kinesin-1 (KIF5) binding and trafficking · Regulating MT stability Polyglutamylation TTLL1, 4, 5, 6, 7, 9, 11, 13 · Modulating kinesin-3 (KIF1) binding and trafficking · Modulating MAP1-MTs interaction · Essential for sperm flagella formation · Important for neurite extension Polyglycylation TTLL3, 8, 10 · Acting as a suppressor of polyglutamylation · Essential for cilia formation a) The detyrosination-performing enzyme is still only a candidate. could explain the cause of abnormal cilia formation, as tier. It is amply clear that we are standing on the threshold over-polyglutamylation, manipulated by the overexpression of a vast field of studies on tubulin PTMs. of the polyglutamylase TTLL6, destabilizes axonemal microtubules (Wloga et al., 2009a). These findings can pro- Acknowledgements. This review was written as part of the Young Scien- vide a cue for understanding the molecular machinery tist Award for Best Presentation of the Japan Society for Cell Biology st whereby tubulin polyglycylation performs its function. The awarded to Koji Ikegami at the 61 JSCB Annual Meeting. The authors would like to express their heartfelt thanks to Prof. Kozo Kaibuchi, the MT-severing enzyme katanin might be one of the machiner- chair of the 61st JSCB Annual Meeting, and the organizing committee of ies since the phenotypes of the mutant MT are copied in the 61st JSCB Annual Meeting for giving us this opportunity to write this katanin-deficient organism (Sharma et al., 2007), though it review. We are also grateful to the present and former members of the remains unclear whether or not katanin recognizes polygly- Setou laboratory of the Hamamatsu University School of Medicine and the cylation or polyglutamylation. Given these intriguing clues, Mitsubishi Kagaku Institute of Life Sciences for their technical assistance, the development of genetically manipulated mammalian valuable discussions, and encouragement. models are now eagerly awaited. References Argaraña, C.E., Barra, H.S., and Caputto, R. 1978. Release of [14C] tyrosine from tubulinyl-[14C] tyrosine by brain extract. Separation of a Conclusions carboxypeptidase from tubulin-tyrosine ligase. Mol. Cell. Biochem., 19: 17–21. Our understanding of the unique tubulin PTMs has pro- Arregui, C., Busciglio, J., Caceres, A., and Barra, H.S. 1991. Tyrosinated ceeded dramatically during the past few years. The recent and detyrosinated microtubules in axonal processes of cerebellar macro- advances are summarized in Table I. The progress achieved neurons grown in culture. J. Neurosci. Res., 28: 171–181. is due in large part to the identification of modification- Audebert, S., Koulakoff, A., Berwald-Netter, Y., Gros, F., Denoulet, P., performing enzymes and the development of genetically and Eddé, B. 1994. Developmental regulation of polyglutamylated manipulated organisms. Research on the roles of tyrosination/ alpha- and beta-tubulin in mouse brain neurons. J. Cell Sci., 107: 2313– 2322. detyrosination cycle are becoming ever more sophisticated. Badin-Larçon, A.C., Boscheron, C., Soleilhac, J.M., Piel, M., Mann, C., The identification of polyglutamylases and polyglycylases Denarier, E., Fourest-Lieuvin, A., Lafanechère, L., Bornens, M., and has enabled research studies in the field of polyglutamylation Job, D. 2004. Suppression of nuclear oscillations in Saccharomyces and polyglycylation to enter the next stage. Function in cerevisiae expressing Glu tubulin. Proc. Natl. Acad. Sci. USA, 101: mammals can be inferred from studies employing more 5577–5582. primitive organisms, like small ciliates, which are easily Barra, H.S., Arce, C.A., Rodríguez, J.A., and Caputto, R. 1974. Some manipulated genetically. Findings obtained from gene- common properties of the protein that incorporates tyrosine as a single unit and the microtubule proteins. Biochem. Biophys. Res. Commun., 60: knockdown in zebrafish have provided valuable information 1384–1390. to anticipate the functions of PTMs in mammals. Nonethe- Bobinnec, Y., Khodjakov, A., Mir, L.M., Rieder, C.L., Eddé, B., and less, more direct evidence is expected to be obtained from Bornens, M. 1998a. Centriole disassembly in vivo and its effect on genetically manipulated mice. Those animals will enable us centrosome structure and function in vertebrate cells. J. Cell Biol., 143: to better understand the physiological function of the modi- 1575–1589. fications more directly. The additional and multiplicative Bobinnec, Y., Moudjou, M., Fouquet, J.P., Desbruyères, E., Eddé, B., and Bornens, M. 1998b. Glutamylation of centriole and cytoplasmic tubulin effects of other PTMs will be a major theme in future in proliferating non-neuronal cells. Cell Motil. Cytoskeleton, 39: 223– works. In certain cases, the coordinated or competitive 232. regulation of PTMs will be a problem. Spatio-temporal Bonnet, C., Boucher, D., Lazereg, S., Pedrotti, B., Islam, K., Denoulet, P., dynamic regulation of multiple PTMs will be the next fron- and Larcher, J.C. 2001. Differential binding regulation of microtubule-

20 Unique Post-Translational Modifications in Specialized Microtubule Architecture

associated proteins MAP1A, MAP1B, and MAP2 by tubulin poly- nucleosome assembly protein 1. FEBS Lett., 582: 1129–1134. glutamylation. J. Biol. Chem., 276: 12839–12848. Ikegami, K. and Setou, M. 2009. TTLL10 can perform tubulin glycylation Campbell, P.K., Waymire, K.G., Heier, R.L., Sharer, C., Day, D.E., when co-expressed with TTLL8. FEBS Lett., 583: 1957–1963. Reimann, H., Jaje, J.M., Friedrich, G.A., Burmeister, M., Bartness, T.J., Jacobson, C., Schnapp, B., and Banker, G.A. 2006. A change in the selec- Russell, L.D., Young, L.J., Zimmer, M., Jenne, D.E., and MacGregor, tive translocation of the Kinesin-1 motor domain marks the initial speci- G.R. 2002. Mutation of a novel gene results in abnormal development fication of the axon. Neuron, 49: 797–804. of spermatid flagella, loss of intermale aggression and reduced body fat Jameson, L., Frey, T., Zeeberg, B., Dalldorf, F., and Caplow, M. 1980. in mice. Genetics, 162: 307–320. Inhibition of microtubule assembly by phosphorylation of microtubule- Drewes, G., Ebneth, A., Preuss, U., Mandelkow, E.M., and Mandelkow, E. associated proteins. Biochemistry, 19: 2472–2479. 1997. MARK, a novel family of protein kinases that phosphorylate Janke, C., Rogowski, K., Wloga, D., Regnard, C., Kajava, A.V., Strub, microtubule-associated proteins and trigger microtubule disruption. Cell, J.M., Temurak, N., van Dijk, J., Boucher, D., van Dorsselaer, A., 89: 297–308. Suryavanshi, S., Gaertig, J., and Eddé, B. 2005. Tubulin polyglut- Dunn, S., Morrison, E.E., Liverpool, T.B., Molina-París, C., Cross, R.A., amylase enzymes are members of the TTL domain protein family. Alonso, M.C., and Peckham, M. 2008. Differential trafficking of Kif5c Science, 308: 1758–1762. on tyrosinated and detyrosinated microtubules in live cells. J. Cell Sci., Kalinina, E., Biswas, R., Berezniuk, I., Hermoso, A., Aviles, F.X., and 121: 1085–1095. Fricker, L.D. 2007. A novel subfamily of mouse cytosolic carboxy- Eddé, B., Rossier, J., Le Caer, J.P., Desbruyères, E., Gros, F., and Denoulet, peptidases. FASEB J., 21: 836–850. P. 1990. Posttranslational glutamylation of alpha-tubulin. Science, 247: Kobayashi, Y. and Mohri, H. 1977. Microheterogeneity of alpha and beta 83–85. subunit of tubulin from microtubules of starfish (Asterias amurensis) Erck, C., Peris, L., Andrieux, A., Meissirel, C., Gruber, A.D., Vernet, M., sperm flagella. J. Mol. Biol., 116: 613–617. Schweitzer, A., Saoudi, Y., Pointu, H., Bosc, C., Salin, P.A., Job, D., and Konishi, Y. and Setou, M. 2009. Tubulin tyrosination navigates the Wehland, J. 2005. A vital role of tubulin-tyrosine-ligase for neuronal kinesin-1 motor domain to axons. Nat. Neurosci., 12: 559–567. organization. Proc. Natl. Acad. Sci. USA, 102: 7853–7858. Landis, S.C. and Mullen, R.J. 1978. The development and degeneration of Ersfeld, K., Wehland, J., Plessmann, U., Dodemont, H., Gerke, V., and Purkinje cells in pcd mutant mice. J. Comp. Neurol., 177: 125–143. Weber, K. 1993. Characterization of the tubulin-tyrosine ligase. J. Cell Liao, G. and Gundersen, G.G. 1998. Kinesin is a candidate for cross- Biol., 120: 725–732. bridging microtubules and intermediate filaments. Selective binding of Fernandez-Gonzalez, A., La Spada, A.R., Treadaway, J., Higdon, J.C., kinesin to detyrosinated tubulin and vimentin. J. Biol. Chem., 273: Harris, B.S., Sidman, R.L., Morgan, J.I., and Zuo, J. 2002. Purkinje cell 9797–9803. degeneration (pcd) phenotypes caused by mutations in the axotomy- Lindwall, G. and Cole, R.D. 1984. Phosphorylation affects the ability of induced gene, Nna1. Science, 295: 1904–1906. tau protein to promote microtubule assembly. J. Biol. Chem., 259: 5301– Fouquet, J.P., Eddé, B., Kann, M.L., Wolff, A., Desbruyeres, E., and 5305. Denoulet, P. 1994. Differential distribution of glutamylated tubulin Million, K., Larcher, J., Laoukili, J., Bourguignon, D., Marano, F., and during spermatogenesis in mammalian testis. Cell Motil. Cytoskeleton, Tournier, F. 1999. Polyglutamylation and polyglycylation of alpha- and 27: 49–58. beta-tubulins during in vitro ciliated cell differentiation of human respi- Gagnon, C., White, D., Cosson, J., Huitorel, P., Eddé, B., Desbruyères, E., ratory epithelial cells. J. Cell Sci., 112: 4357–4366. Paturle-Lafanechère, L., Multigner, L., Job, D., and Cibert, C. 1996. Mishima, M., Maesaki, R., Kasa, M., Watanabe, T., Fukata, M., Kaibuchi, The polyglutamylated lateral chain of alpha-tubulin plays a key role in K., and Hakoshima, T. 2007. Structural basis for tubulin recognition by flagellar motility. J. Cell Sci., 109: 1545–1553. cytoplasmic linker protein 170 and its autoinhibition. Proc. Natl. Acad. Hammond, J.W., Cai, D., and Verhey, K.J. 2008. Tubulin modifications Sci. USA, 104: 10346–10351. and their cellular functions. Curr. Opin. Cell Biol., 20: 71–76. Mohri, H. 1968. Amino-acid composition of “Tubulin” constituting micro- Honnappa, S., Okhrimenko, O., Jaussi, R., Jawhari, H., Jelesarov, I., tubules of sperm flagella. Nature, 217: 1053–1054. Winkler, F.K., and Steinmetz, M.O. 2006. Key interaction modes of Mukai, M., Ikegami, K., Sugiura, Y., Takeshita, K., Nakagawa, A., and dynamic +TIP networks. Mol. Cell, 23: 663–671. Setou, M. 2009. Recombinant mammalian tubulin polyglutamylase Hoshi, M., Nishida, E., Miyata, Y., Sakai, H., Miyoshi, T., Ogawara, H., TTLL7 performs both initiation and elongation of polyglutamylation on and Akiyama, T. 1987. Protein kinase C phosphorylates tau and induces beta-tubulin through a random sequential pathway. Biochemistry, 48: its functional alterations. FEBS Lett., 217: 237–241. 1084–1093. Hoshi, M., Akiyama, T., Shinohara, Y., Miyata, Y., Ogawara, H., Nishida, Mullen, R.J., Eicher, E.M., and Sidman, R.L. 1976. Purkinje cell degener- E., and Sakai, H. 1988. Protein-kinase-C-catalyzed phosphorylation of ation, a new neurological mutation in the mouse. Proc. Natl. Acad. Sci. the microtubule-binding domain of microtubule-associated protein 2 USA, 73: 208–212. inhibits its ability to induce tubulin polymerization. Eur. J. Biochem., Murofushi, H. 1980. Purification and characterization of tubulin-tyrosine 174: 225–230. ligase from porcine brain. J. Biochem., 87: 979–984. Ikegami, K., Mukai, M., Tsuchida, J., Heier, R.L., MacGregor, G.R., and Nishida, E., Kuwaki, T., and Sakai, H. 1981. Phosphorylation of Setou, M. 2006. TTLL7 is a mammalian beta-tubulin polyglutamylase microtubule-associated proteins (MAPs) and pH of the medium control required for growth of MAP2-positive neurites. J. Biol. Chem., 281: interaction between MAPs and actin filaments. J. Biochem., 90: 575– 30707–30716. 578. Ikegami, K., Heier, R.L., Taruishi, M., Takagi, H., Mukai, M., Shimma, S., Nishida, E., Kotani, S., Kuwaki, T., and Sakai, H. 1982. In Biological Taira, S., Hatanaka, K., Morone, N., Yao, I., Campbell, P.K., Yuasa, S., Functions of Microtubules and Related Structures (H. Sakai, H. Mohri, Janke, C., Macgregor, G.R., and Setou, M. 2007. Loss of alpha-tubulin and G. Borisy, eds.). Academic Press, New York, pp285–295. polyglutamylation in ROSA22 mice is associated with abnormal target- Nishida, E., Hoshi, M., Miyata, Y., Sakai, H., Kadowaki, T., Kasuga, M., ing of KIF1A and modulated synaptic function. Proc. Natl. Acad. Sci. Saijo, S., Ogawara, H., and Akiyama, T. 1987. Tyrosine phosphoryla- USA, 104: 3213–3218. tion by the epidermal growth factor receptor kinase induces functional Ikegami, K., Horigome, D., Mukai, M., Livnat, I., MacGregor, G.R., and alterations in microtubule-associated protein 2. J. Biol. Chem., 262: Setou, M. 2008. TTLL10 is a protein polyglycylase that can modify 16200–16204.

21 K. Ikegami and M. Setou

Okada, Y. and Hirokawa, N. 1999. A processive single-headed motor: dendritic transport. J. Neurobiol., 58: 201–206. kinesin superfamily protein KIF1A. Science, 283: 1152–1157. Sharma, N., Bryant, J., Wloga, D., Donaldson, R., Davis, R.C., Jerka- Pathak, N., Obara, T., Mangos, S., Liu, Y., and Drummond, I.A. 2007. The Dziadosz, M., and Gaertig, J. 2007. Katanin regulates dynamics of zebrafish fleer gene encodes an essential regulator of cilia tubulin poly- microtubules and biogenesis of motile cilia. J. Cell Biol., 178: 1065– glutamylation. Mol. Biol. Cell, 18: 4353–4364. 1079. Paturle-Lafanechère, L., Eddé, B., Denoulet, P., Van Dorsselaer, A., Szebenyi, G., Bollati, F., Bisbal, M., Sheridan, S., Faas, L., Wray, R., Mazarguil, H., Le Caer, J.P., Wehland, J., and Job, D. 1991. Character- Haferkamp, S., Nguyen, S., Caceres, A., and Brady, S.T. 2005. Activity- ization of a major brain tubulin variant which cannot be tyrosinated. driven dendritic remodeling requires microtubule-associated protein 1A. Biochemistry, 30: 10523–10528. Curr. Biol., 15: 1820–1826. Peris, L., Thery, M., Fauré, J., Saoudi, Y., Lafanechère, L., Chilton, J.K., Thazhath, R., Liu, C., and Gaertig, J. 2002. Polyglycylation domain of Gordon-Weeks, P., Galjart, N., Bornens, M., Wordeman, L., Wehland, beta-tubulin maintains axonemal architecture and affects cytokinesis in J., Andrieux, A., and Job, D. 2006. Tubulin tyrosination is a major Tetrahymena. Nat. Cell Biol., 4: 256–259. factor affecting the recruitment of CAP-Gly proteins at microtubule van Dijk, J., Rogowski, K., Miro, J., Lacroix, B., Eddé, B., and Janke, C. plus ends. J. Cell Biol., 174: 839–849. 2007. A targeted multienzyme mechanism for selective microtubule Peris, L., Wagenbach, M., Lafanechère, L., Brocard, J., Moore, A.T., polyglutamylation. Mol. Cell, 26: 437–448. Kozielski, F., Job, D., Wordeman, L., and Andrieux, A. 2009. Motor- Verhey, K.J. and Gaertig, J. 2007. The tubulin code. Cell Cycle, 6: 2152– dependent microtubule disassembly driven by tubulin tyrosination. J. 2160. Cell Biol., 185: 1159–1166. Wada, Y., Ishiguro, K., Itoh, T.J., Uchida, T., Hotani, H., Saito, Raybin, D. and Flavin, M. 1977. Enzyme which specifically adds tyrosine T., Kishimoto, T., and Hisanaga, S. 1998. Microtubule-stimulated to the alpha chain of tubulin. Biochemistry, 16: 2189–2194. phosphorylation of tau at Ser202 and Thr205 by cdk5 decreases its Redeker, V., Levilliers, N., Schmitter, J.M., Le Caer, J.P., Rossier, J., microtubule nucleation activity. J. Biochem., 124: 738–746. Adoutte, A., and Bré, M.H. 1994. Polyglycylation of tubulin: a post- Webster, D.R., Wehland, J., Weber, K., and Borisy, G.G. 1990. Detyrosi- translational modification in axonemal microtubules. Science, 266: nation of alpha tubulin does not stabilize microtubules in vivo. J. Cell 1688–1691. Biol., 111: 113–122. Redeker, V., Levilliers, N., Vinolo, E., Rossier, J., Jaillard, D., Burnette, Wehland, J. and Weber, K. 1987. Turnover of the carboxy-terminal D., Gaertig, J., and Bré, M.H. 2005. Mutations of tubulin glycylation tyrosine of alpha-tubulin and means of reaching elevated levels of sites reveal cross-talk between the C termini of alpha- and beta-tubulin detyrosination in living cells. J. Cell Sci., 88: 185–203. and affect the ciliary matrix in Tetrahymena. J. Biol. Chem., 280: 596– Westermann, S. and Weber, K. 2003. Post-translational modifications 606. regulate microtubule function. Nat. Rev. Mol. Cell Biol., 4: 938–947. Regnard, C., Fesquet, D., Janke, C., Boucher, D., Desbruyéres, E., Wloga, D., Rogowski, K., Sharma, N., Van Dijk, J., Janke, C., Eddé, B., Koulakoff, A., Insina, C., Travo, P., and Eddé, B. 2003. Characterisa- Bré, M.H., Levilliers, N., Redeker, V., Duan, J., Gorovsky, M.A., tion of PGs1, a subunit of a protein complex co-purifying with tubulin Jerka-Dziadosz, M., and Gaertig, J. 2008. Glutamylation on alpha- polyglutamylase. J. Cell Sci., 116: 4181–4190. tubulin is not essential but affects the assembly and functions of a subset Rodriguez de la Vega, M., Sevilla, R.G., Hermoso, A., Lorenzo, J., Tanco, of microtubules in Tetrahymena thermophila. Eukaryot. Cell, 7: 1362– S., Diez, A., Fricker, L.D., Bautista, J.M., and Avilés, F.X. 2007. Nna1- 1372. like proteins are active metallocarboxypeptidases of a new and diverse Wloga, D., Dave, D., Meagley, J., Rogowski, K., Jerka-Dziadosz, M., and M14 subfamily. FASEB J., 21: 851–865. Gaertig, J. 2009a. Hyperglutamylation of tubulin can either stabilize or Rogowski, K., Juge, F., van Dijk, J., Wloga, D., Strub, J.M., Levilliers, N., destabilize microtubules in the same cell. Eukaryot Cell, doi: 10.1128/ Thomas, D., Bré, M.H., Van Dorsselaer, A., Gaertig, J., and Janke, C. EC.00176-09. 2009. Evolutionary divergence of enzymatic mechanisms for posttrans- Wloga, D., Webster, D.M., Rogowski, K., Bré, M.H., Levilliers, N., lational polyglycylation. Cell, 137: 1076–1087. Jerka-Dziadosz, M., Janke, C., Dougan, S.T., and Gaertig, J. 2009b. Rosenbaum, J. 2000. Cytoskeleton: functions for tubulin modifications at TTLL3 is a tubulin glycine ligase that regulates the assembly of cilia. last. Curr. Biol., 10: R801–803. Dev. Cell, 16: 867–876. Rüdiger, M., Wehland, J., and Weber, K. 1994. The carboxy-terminal pep- Wolff, A., de Néchaud, B., Chillet, D., Mazarguil, H., Desbruyères, E., tide of detyrosinated alpha tubulin provides a minimal system to study Audebert, S., Eddé, B., Gros, F., and Denoulet, P. 1992. Distribution of the substrate specificity of tubulin-tyrosine ligase. Eur. J. Biochem., 220: glutamylated alpha and beta-tubulin in mouse tissues using a specific 309–320. monoclonal antibody, GT335. Eur. J. Cell Biol., 59: 425–432. Sato-Harada, R., Okabe, S., Umeyama, T., Kanai, Y., and Hirokawa, N. Xia, L., Hai, B., Gao, Y., Burnette, D., Thazhath, R., Duan, J., Bré, M.H., 1996. Microtubule-associated proteins regulate microtubule function as Levilliers, N., Gorovsky, M.A., and Gaertig, J. 2000. Polyglycylation of the track for intracellular membrane organelle transports. Cell Struct. tubulin is essential and affects cell motility and division in Tetrahymena Funct., 21: 283–295. thermophila. J. Cell Biol., 149: 1097–1106. Schoenfeld, T.A., McKerracher, L., Obar, R., and Vallee, R.B. 1989. MAP Yamamoto, H., Fukunaga, K., Tanaka, E., and Miyamoto, E. 1983. Ca2+- 1A and MAP 1B are structurally related microtubule associated proteins and calmodulin-dependent phosphorylation of microtubule-associated with distinct developmental patterns in the CNS. J. Neurosci., 9: 1712– protein 2 and tau factor, and inhibition of microtubule assembly. J. 1730. Neurochem., 41: 1119–1125. Setou, M., Nakagawa, T., Seog, D.H., and Hirokawa, N. 2000. Kinesin Yoshimura, T., Kawano, Y., Arimura, N., Kawabata, S., Kikuchi, A., and superfamily motor protein KIF17 and mLin-10 in NMDA receptor- Kaibuchi, K. 2005. GSK-3b regulates phosphorylation of CRMP-2 containing vesicle transport. Science, 288: 1796–1802. and neuronal polarity. Cell, 120: 137–149. Setou, M., Seog, D.H., Tanaka, Y., Kanai, Y., Takei, Y., Kawagishi, M., and Hirokawa, N. 2002. Glutamate-receptor-interacting protein GRIP1 (Received for publication, December 25, 2009, accepted, February 18, 2010 directly steers kinesin to dendrites. Nature, 417: 83–87. and published online, February 27, 2010) Setou, M., Hayasaka, T., and Yao, I. 2004. Axonal transport versus

22