首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Mislocalization and aggregation of Aβ and Tau combined with loss of synapses and microtubules (MTs) are hallmarks of Alzheimer disease. We exposed mature primary neurons to Aβ oligomers and analysed changes in the Tau/MT system. MT breakdown occurs in dendrites invaded by Tau (Tau missorting) and is mediated by spastin, an MT‐severing enzyme. Spastin is recruited by MT polyglutamylation, induced by Tau missorting triggered translocalization of TTLL6 (Tubulin‐Tyrosine‐Ligase‐Like‐6) into dendrites. Consequences are spine loss and mitochondria and neurofilament mislocalization. Missorted Tau is not axonally derived, as shown by axonal retention of photoconvertible Dendra2‐Tau, but newly synthesized. Recovery from Aβ insult occurs after Aβ oligomers lose their toxicity and requires the kinase MARK (Microtubule‐Affinity‐Regulating‐Kinase). In neurons derived from Tau‐knockout mice, MTs and synapses are resistant to Aβ toxicity because TTLL6 mislocalization and MT polyglutamylation are prevented; hence no spastin recruitment and no MT breakdown occur, enabling faster recovery. Reintroduction of Tau re‐establishes Aβ‐induced toxicity in TauKO neurons, which requires phosphorylation of Tau's KXGS motifs. Transgenic mice overexpressing Tau show TTLL6 translocalization into dendrites and decreased MT stability. The results provide a rationale for MT stabilization as a therapeutic approach.  相似文献   

2.
EMBO J 32: 2920–2937 10.1038/emboj.2013.207; published online September242013Microtubule loss from axons and dendrites is a key contributor to nervous system degeneration during Alzheimer disease. Previous evidence suggested a simple pathway by which tau dissociation from microtubules in the axon allows excess severing of microtubules by katanin. Now, new evidence has emerged for a more complex pathway by which abnormal tau invasion into dendrites, triggered by Aβ oligomers, results in excess severing of microtubules by spastin.Alzheimer disease (AD) is a member of a category of neurodegenerative disorders called tauopathies (Wang and Liu, 2008). These are diseases of the nervous system in which tau becomes abnormally phosphorylated, and thereby detaches from microtubules. As the microtubules lose tau, they diminish in number and density, and this loss of microtubule mass negatively impacts the capacity of the neuron to maintain axonal transport and synaptic connections. Terms such as disintegrate or ‘fall apart'' are often used to describe the effect on the microtubules as they lose tau, but to date there has been very little information on how this happens. There is no mechanistic evidence to support the view that the microtubules become less stable and simply disassemble by their normal dynamic properties.One possibility is that tau normally protects microtubules from being destroyed by various proteins in the axon that would otherwise cut them into pieces or in some other way break them down. This presumably reflects a physiological mechanism wherein the regulation of tau dissociation from the microtubule via signalling pathways controls when and where microtubule breakage normally occurs. When a pathological condition causes tau to detach from microtubules, they become extremely sensitive to such factors. In addition, there is strong evidence that the abnormal tau, whether soluble or filamentous, can elicit toxic gain-of-function effects on the axon (Wang and Liu, 2008).To make matters even more complex, AD is not a pure tauopathy. Beta amyloid (Aβ) accumulates abnormally in the brain during AD, and this prompts tau to become hyperphosphorylated and lose association with microtubules. However, the Aβ can also elicit microtubule loss, independent of tau dissociation from the microtubules. In AD, there is also a loss of microtubules from dendrites, and this introduces an additional degree of complexity. Tau is normally less enriched in dendrites than axons. In AD, tau invades dendrites abnormally through deregulation of its normal sorting mechanism, and this somehow leads to microtubule loss from dendrites (Zempel et al, 2010).Microtubule loss is a common end point of multiple pathways, some involving loss of tau function, others involving gain-of-function effects of abnormal tau, and still others working through tau-dependent Aβ toxicity. All of this is not to say that the effects on microtubules are the only reason or even the principal explanation for axonal degeneration in AD, but the loss of microtubules is an important contributor to nervous system degeneration. Preventing or reversing the effects on microtubules could help stave off degeneration and hence provide patients with additional years of cognitive health and better quality of life.Microtubule assembly and disassembly occur from the ends of a microtubule, mainly (and often exclusively) at the plus end of the microtubule in living cells. Proteins that regulate microtubule stability affect the rate of these dynamics at microtubule ends. In recent years, a great deal of attention has focused on a category of proteins, termed microtubule-severing proteins. These proteins are enzymes that yank at the microtubule anywhere along its length to pull out a tubulin subunit, and thereby ‘cut'' the microtubule by causing it to break into pieces (Roll-Mecak and Vale, 2008). If the microtubule is sufficiently stable in the region of the break, the parent microtubule is cut into two shorter microtubules that persist, with minimal disassembly of either of the two pieces. If a microtubule is severed in its more labile region, the breakage could cause a great deal of disassembly. If the tubulin being yanked is situated at one of the ends of a microtubule, the result would be a shortening of the microtubule from that end; that is, disassembly. Thus, microtubule severing in the axon can certainly lead to microtubule loss, either by cutting the polymer all the way to subunits, inducing disassembly directly from microtubule ends or promoting disassembly as a secondary effect to the cutting.To date, the AAA enzymes katanin and spastin are the best studied of the microtubule-severing proteins (Yu et al, 2008). Spastin was originally identified as the product of the gene whose mutations are the chief cause of hereditary spastic paraplegia. Curiously, neurons express levels of spastin and katanin that are theoretically high enough to completely sever all of the microtubules in the neuron to subunits (Solowska et al, 2008), and yet this does not happen. Various regulatory mechanisms presumably keep the activities of the severing proteins in check. One of these mechanisms, in the case of katanin, is microtubule-bound tau, which protects the microtubule from being accessed by katanin (Qiang et al, 2006).Could microtubule loss in AD be due, at least in part, to excess microtubule severing due to deregulation of microtubule-severing proteins? We have posited that heightened severing of the microtubules by katanin, as the microtubules lose association with tau, is a contributing factor to the degradation of microtubules in the axons of AD patients (Sudo and Baas, 2011). A role for spastin in this pathway is questionable, because tau does not appear to protect microtubules against spastin as effectively as it does against katanin (Qiang et al, 2006). However, we now know that spastin is far from irrelevant to AD, as an exciting new article from the Mandelkow and Dawson laboratories implicates spastin in an entirely different pathway for microtubule loss in AD (Zempel et al, 2013). Whereas the katanin pathway is more relevant to axons, this new spastin pathway is more relevant to dendrites.In this new work, Zempel et al (2013) exposed mature primary neurons to oligomers of Aβ and observed microtubule breakdown in dendrites that had been invaded by tau. They found that the missorting of tau leads to an elevation of TTLL6 (Tubulin-Tyrosine-Ligase-Like-6) in dendrites, and this results in a marked increase in the polyglutamylation status of the microtubules. Because spastin has a strong preference for polyglutamylated microtubules, the microtubules become more sensitive to spastin-induced severing. Exactly why katanin is not a factor remains unclear, as polyglutamylation renders microtubules more sensitive to both of the severing proteins, not just spastin (Lacroix et al, 2010). Perhaps some of the tau that invades the dendrite is able to bind to microtubules and protect them from katanin, or perhaps katanin is less potent in dendrites because their microtubules are poorly acetylated, as katanin prefers acetylated microtubules to unacetylated ones (Sudo and Baas, 2011). Whatever the case, these new studies suggest that spastin, a protein whose mutations cause an entirely different neurodegenerative disease, is also a major factor in AD.What are the implications of these findings for AD treatment? In recent years, there have been encouraging results on rodent models for AD, in which behavioural improvement and enhanced neuronal vitality were observed when the animals were treated with drugs that stabilize microtubules against disassembly (Zhang et al, 2012). Such drugs are currently in clinical trials for AD (Barten et al, 2012). This strategy is based on the presumption that the microtubule loss that occurs in AD is due to destabilization of the microtubules. However, the results discussed here suggest that the primary cause of the microtubule loss could be something quite different, namely excess severing of microtubules. In this regard, it is relevant that both katanin and spastin seem to have a preference for severing stable microtubules (Lacroix et al, 2010; Sudo and Baas, 2011). Therefore, while treatment with a microtubule-stabilizing drug would mitigate disassembly that occurs as an aftereffect of microtubule severing, the severing events themselves would likely be increased (Figure 1). Heightened microtubule severing in axons and dendrites, even if the total levels of microtubule mass are preserved, could result in a gradual shift from a normal distribution of long and short microtubules to a predominance of microtubules too short to support sustained excursions of organelle transport. Long microtubules are also necessary as compression-bearing struts that prevent axons and dendrites from collapsing on themselves. We suspect that appropriate treatment regimes can be devised to prevent such dire consequences from happening, but we would advocate for the development of new drugs that inhibit microtubule-severing proteins. Such drugs may prove to be a better approach (on their own or in combination with a stabilizing drug) for preserving the fidelity of axonal and dendritic microtubules in AD patients. Given that the structure of the severing proteins is known, it may be straightforward to develop inhibitors, especially to their ATPase domains.Open in a separate windowFigure 1Microtubules in axons and dendrites consist of a stable region towards the minus end of the microtubule and a labile region towards the plus end, as well as a pool of free tubulin subunits. Microtubule severing is a normal event in the neuron, when tightly regulated. Abnormal (deregulated) microtubule severing is posited to account for microtubule loss in AD. Severing in the stable region of the microtubule would create two new microtubules, with fairly minimal disassembly of either one. Severing in the labile region of the microtubule would result in notably more disassembly. Severing at the end of the microtubule would result in disassembly. Because known microtubule-severing proteins favour the stable region of the microtubule, treatment of AD with a microtubule-stabilizing drug may mitigate disassembly that occurs as an aftereffect of the severing, but the severing events themselves would likely increase.  相似文献   

3.
Two related enzymes, katanin and spastin, use the energy from ATP hydrolysis to sever microtubules. Two new studies (one in this issue; see McNally et al., p. 881) show that microtubule severing by katanin provides a means for increasing microtubule density in meiotic spindles. Interestingly, loss of spastin leads to a sparser microtubule array in axons and synaptic boutons. Together, these studies hint at a wider role for microtubule-severing enzymes in the formation and organization of noncentrosomal microtubule arrays by generating new seeds for microtubule growth.  相似文献   

4.
Tubulin polyglutamylation is a post‐translational modification of the microtubule cytoskeleton, which is generated by a variety of enzymes with different specificities. The “tubulin code” hypothesis predicts that modifications generated by specific enzymes selectively control microtubule functions. Our recent finding that excessive accumulation of polyglutamylation in neurons causes their degeneration and perturbs axonal transport provides an opportunity for testing this hypothesis. By developing novel mouse models and a new glutamylation‐specific antibody, we demonstrate here that the glutamylases TTLL1 and TTLL7 generate unique and distinct glutamylation patterns on neuronal microtubules. We find that under physiological conditions, TTLL1 polyglutamylates α‐tubulin, while TTLL7 modifies β‐tubulin. TTLL1, but not TTLL7, catalyses the excessive hyperglutamylation found in mice lacking the deglutamylase CCP1. Consequently, deletion of TTLL1, but not of TTLL7, prevents degeneration of Purkinje cells and of myelinated axons in peripheral nerves in these mice. Moreover, loss of TTLL1 leads to increased mitochondria motility in neurons, while loss of TTLL7 has no such effect. By revealing how specific patterns of tubulin glutamylation, generated by distinct enzymes, translate into specific physiological and pathological readouts, we demonstrate the relevance of the tubulin code for homeostasis.  相似文献   

5.
BACKGROUNDTubulins, building blocks of microtubules, are modified substrates of diverse post-translational modifications including phosphorylation, polyglycylation and polyglutamylation. Polyglutamylation of microtubules, catalyzed by enzymes from the tubulin tyrosine ligase-like (TTLL) family, can regulate interactions with molecular motors and other proteins. Due to the diversity and functional importance of microtubule modifications, strict control of the TTLL enzymes has been suggested.AIMTo characterize the interaction between never in mitosis gene A-related kinase 5 (NEK5) and TTLL4 proteins and the effects of TTLL4 phosphorylation.METHODSThe interaction between NEK5 and TTLL4 was identified by yeast two-hybrid screening using the C-terminus of NEK5 (a.a. 260–708) as bait and confirmed by immunoprecipitation. The phosphorylation sites of TTLL4 were identified by mass spectrometry and point mutations were introduced.RESULTSHere, we show that NEK5 interacts with TTLL4 and regulates its polyglutamylation activity. We further show that NEK5 can also interact with TTLL5 and TTLL7. The silencing of NEK5 increases the levels of polyglutamylation of proteins by increasing the activity of TTLL4. The same effects were observed after the expression of the catalytically inactive form of NEK5. This regulation of TTLL4 activity involves its phosphorylation at Y815 and S1136 amino acid residues.CONCLUSIONOur results demonstrate, for the first time, the regulation of TTLL activity through phosphorylation, pointing to NEK5 as a potential effector kinase. We also suggest a general control of tubulin polyglutamylation through NEK family members in human cells.  相似文献   

6.
The formation of interstitial axonal branches involves the severing of microtubules at sites where new branches form. Here we wished to ascertain whether basic fibroblast growth factor (bFGF) enhances axonal branching through alterations in proteins involved in the severing of microtubules. We found that treatment of cultured hippocampal neurons with bFGF heightens expression of both katanin and spastin, which are proteins that sever microtubules in the axon. In addition, treatment with bFGF enhances phosphorylation of tau at sites expected to cause it to dissociate from microtubules. This is important because tau regulates the access of katanin to the microtubule. In live-cell imaging experiments, axons of neurons treated with bFGF displayed greater numbers of dynamic free ends of microtubules, as well as greater numbers of short mobile microtubules. Entirely similar enhancement of axonal branching, short microtubule transport, and frequency of microtubule ends was observed when spastin was overexpressed in the neurons. Depletion of either katanin or spastin with siRNA diminished but did not eliminate the enhancement in branching elicited by bFGF. Collectively, these results indicate that bFGF enhances axonal branch formation by augmenting the severing of microtubules through both a spastin-based mode and a katanin-based mode.  相似文献   

7.
In 1999, mutations in the gene encoding the microtubule severing AAA ATPase spastin were identified as a major cause of a genetic neurodegenerative condition termed hereditary spastic paraplegia (HSP). This finding stimulated intense study of the spastin protein and over the last decade, a combination of cell biological, in vivo, in vitro and structural studies have provided important mechanistic insights into the cellular functions of the protein, as well as elucidating cell biological pathways that might be involved in axonal maintenance and degeneration. Roles for spastin have emerged in shaping the endoplasmic reticulum and the abscission stage of cytokinesis, in which spastin appears to couple membrane modelling to microtubule regulation by severing.  相似文献   

8.
9.
Neurons express two different microtubule-severing proteins, namely P60-katanin and spastin. Here, we performed studies on cultured neurons to ascertain whether these two proteins participate differently in axonal branch formation. P60-katanin is more highly expressed in the neuron, but spastin is more concentrated at sites of branch formation. Overexpression of spastin dramatically enhances the formation of branches, whereas overexpression of P60-katanin does not. The excess spastin results in large numbers of short microtubules, whereas the excess P60-katanin results in short microtubules intermingled with longer microtubules. We hypothesized that these different microtubule-severing patterns may be due to the presence of molecules such as tau on the microtubules that more strongly shield them from being severed by P60-katanin than by spastin. Consistent with this hypothesis, we found that axons depleted of tau show a greater propensity to branch, and that this is true whether or not the axons are also depleted of spastin. We propose that there are two modes by which microtubule severing is orchestrated during axonal branch formation, one based on the local concentration of spastin at branch sites and the other based on local detachment from microtubules of molecules such as tau that regulate the severing properties of P60-katanin.  相似文献   

10.
阿尔茨海默病(AD)是非常普遍的神经变性性疾病并且是老年人痴呆的主要原因。AD患者的症状特点包括进行性的认知障碍、记忆丧失和行为障碍,与大脑中的病理变化密切相关。AD现成为全球最严重的健康和社会经济问题。在AD患者脑中神经纤维网或神经营养障碍的过程中存在tau蛋白的异常。tau蛋白丧失其促微管组装的生物学功能,导致细胞骨架的破坏、丝状物形成和神经缠结,轴突运输损害,进而导致突触蛋白失去功能和神经退行性病变。其数量和结构的改变将会影响其功能而且会出现异常聚集。调节Tau蛋白的异常聚集的分子机制主要是一些翻译后修饰使其结构及构象发生变化。因此,异常磷酸化和截断的tau蛋白作为tau蛋白病理过程的关键机制而引起学者关注。本文描述了tau蛋白的结构和功能及其在AD中的主要病理变化,同时在本文中还涉及到磷酸化的tau蛋白是神经元对氧化应激的代偿反应这一观点。对tau蛋白进行更加全面的解读。  相似文献   

11.
Posttranslational glutamylation of tubulin is present on selected subsets of microtubules in cells. Although the modification is expected to contribute to the spatial and temporal organization of the cytoskeleton, hardly anything is known about its functional relevance. Here we demonstrate that glutamylation, and in particular the generation of long glutamate side chains, promotes the severing of microtubules. In human cells, the generation of long side chains induces spastin-dependent microtubule disassembly and, consistently, only microtubules modified by long glutamate side chains are efficiently severed by spastin in vitro. Our study reveals a novel control mechanism for microtubule mass and stability, which is of fundamental importance to cellular physiology and might have implications for diseases related to microtubule severing.  相似文献   

12.
There is broad agreement that cells reconfigure their microtubules through rapid bouts of assembly and disassembly, as described by the mechanism known as dynamic instability. However, many cell types have complex patterns of microtubule organization that are not entirely explicable by dynamic instability. There is growing evidence that microtubules can be moved into new patterns of organization by forces generated by molecular motor proteins. Studies on several cell types support a model called 'cut and run' in which long microtubules are stationary, but relatively short microtubules are mobile. In this model, cells mobilize their microtubules by severing them into short pieces, using enzymes such as katanin and spastin that break the lattice of the microtubule polymer. After being reorganized, the short microtubules can once again elongate and lose their mobility. Microtubule severing is also crucial for a variation of 'cut and run' in which the severed microtubules are reorganized by means of treadmilling.  相似文献   

13.
Hereditary spastic paraplegias (HSPs) are neurodegenerative diseases caused by mutations in more than 20 genes, which lead to progressive spasticity and weakness of the lower limbs. The most frequently mutated gene causing autosomal dominant HSP is SPG4, which encodes spastin, a protein that belongs to the family of ATPases associated with various cellular activities (AAAs). A number of studies have suggested that spastin regulates microtubule dynamics. We have studied the ATPase activity of recombinant human spastin and examined the effect of taxol-stabilized microtubules on this activity. We used spastin translated from the second ATG and provide evidence that this is the physiologically relevant form. We showed that microtubules enhance the ATPase activity of the protein, a property also described for katanin, an AAA of the same spastin subgroup. Furthermore, we demonstrated that human spastin has a microtubule-destabilizing activity and can bundle microtubules in vitro, providing new insights into the molecular pathogenesis of HSP.  相似文献   

14.
BACKGROUND: Hereditary Spastic Paraplegia (HSP) is a devastating neurological disease causing spastic weakness of the lower extremities and eventual axonal degeneration. Over 20 genes have been linked to HSP in humans; however, mutations in one gene, spastin (SPG4), are the cause of >40% of all cases. Spastin is a member of the ATPases associated with diverse cellular activities (AAA) protein family, and contains a microtubule interacting and organelle transport (MIT) domain. Previous work in cell culture has proposed a role for Spastin in regulating microtubules. RESULTS: Employing Drosophila transgenic methods for overexpression and RNA interference (RNAi), we have investigated the role of Spastin in vivo. We show that Drosophila Spastin (D-Spastin) is enriched in axons and synaptic connections. At neuromuscular junctions (NMJ), Dspastin RNAi causes morphological undergrowth and reduced synaptic area. Moreover, Dspastin overexpression reduces synaptic strength, whereas Dspastin RNAi elevates synaptic currents. By using antibodies against posttranslationally modified alpha-Tubulin, we find that Dspastin regulates microtubule stability. Functional synaptic defects caused by Dspastin RNAi and overexpression were pharmacologically alleviated by agents that destabilize and stabilize microtubules, respectively. CONCLUSIONS: Loss of Dspastin in Drosophila causes an aberrantly stabilized microtubule cytoskeleton in neurons and defects in synaptic growth and neurotransmission. These in vivo data strongly support previous reports, providing a probable cause for the neuronal dysfunction in spastin-linked HSP disease. The role of Spastin in regulating neuronal microtubule stability suggests therapeutic targets for HSP treatment and may provide insight into neurological disorders linked to microtubule dysfunction.  相似文献   

15.
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is associated with protein misfolding, plaque accumulation, neuronal dysfunction, synaptic loss, and cognitive decline. The pathological cascade of AD includes the intracellular Tau hyperphosphorylation and its subsequent aggregation, extracellular Amyloid-β plaque formation and microglia-mediated neuroinflammation. The extracellular release of aggregated Tau is sensed by surveilling microglia through the involvement of various cell surface receptors. Among all, purinergic P2Y12R signaling is involved in microglial chemotaxis towards the damaged neurons. Microglial migration is highly linked with membrane-associated actin remodeling leading to the phagocytosis of extracellular Tau species. Here, we studied the formation of various actin structures such as podosome, lamellipodia and filopodia, in response to extracellular Tau monomers and aggregates. Microglial podosomes are colocalized with actin nucleator protein WASP, Arp2 and TKS5 adaptor protein during Tau-mediated migration. Moreover, the P2Y12 receptors were associated with F-actin-rich podosome structures, which signify the potential of Tau aggregates in microglial chemotaxis through the involvement of actin remodeling.  相似文献   

16.
Mutations in spastin are the most common cause of hereditary spastic paraplegia (HSP) but the mechanisms by which mutant spastin induces disease are not clear. Spastin functions to regulate microtubule organisation, and because of the essential role of microtubules in axonal transport, this has led to the suggestion that defects in axonal transport may underlie at least part of the disease process in HSP. However, as yet there is no direct evidence to support this notion. Here we analysed axonal transport in a novel mouse model of spastin-induced HSP that involves a pathogenic splice site mutation, which leads to a loss of spastin protein. A mutation located within the same splice site has been previously described in HSP. Spastin mice develop gait abnormalities that correlate with phenotypes seen in HSP patients and also axonal swellings containing cytoskeletal proteins, mitochondria and the amyloid precursor protein (APP). Pathological analyses of human HSP cases caused by spastin mutations revealed the presence of similar axonal swellings. To determine whether mutant spastin influenced axonal transport we quantified transport of two cargoes, mitochondria and APP-containing membrane bound organelles, in neurons from mutant spastin and control mice, using time-lapse microscopy. We found that mutant spastin perturbs anterograde transport of both cargoes. In neurons with axonal swellings we found that the mitochondrial axonal transport defects were exacerbated; distal to axonal swellings both anterograde and retrograde transport were severely reduced. These results strongly support a direct role for defective axonal transport in the pathogenesis of HSP because of spastin mutation.  相似文献   

17.
18.
Ozdowski EF  Gayle S  Bao H  Zhang B  Sherwood NT 《Genetics》2011,189(1):123-135
Microtubules are dynamic structures that must elongate, disassemble, and be cleaved into smaller pieces for proper neuronal development and function. The AAA ATPase Spastin severs microtubules along their lengths and is thought to regulate the balance between long, stable filaments and shorter fragments that seed extension or are transported. In both Drosophila and humans, loss of Spastin function results in reduction of synaptic connections and disabling motor defects. To gain insight into how spastin is regulated, we screened the Drosophila melanogaster genome for deletions that modify a spastin overexpression phenotype, eye size reduction. One suppressor region deleted p21-activated kinase 3 (pak3), which encodes a member of the Pak family of actin-regulatory enzymes, but whose in vivo function is unknown. We show that pak3 mutants have only mild synaptic defects at the larval neuromuscular junction, but exhibit a potent genetic interaction with spastin mutations. Aberrant bouton morphology, microtubule distribution, and synaptic transmission caused by spastin loss of function are all restored to wild type when pak3 is simultaneously reduced. Neuronal overexpression of pak3 induces actin-rich thin projections, suggesting that it functions in vivo to promote filopodia during presynaptic terminal arborization. pak3 therefore regulates synapse development in vivo, and when mutated, suppresses the synaptic defects that result from spastin loss.  相似文献   

19.
Spastin and katanin are ring-shaped hexameric AAA ATPases that sever microtubules, and thus crucially depend on a physical interaction with microtubules. For the first time, we report here the microtubule binding properties of spastin at the single-molecule level, and compare them to katanin. Microscopic fluorescence assays showed that human spastin bound to microtubules by ionic interactions, and diffused along microtubules with a diffusion coefficient comparable to katanin. The microscopic measurement of landing and dissociation rates demonstrated the ionic character of the interaction, which could be mapped to a patch of three lysine residues outside of the catalytic domain of human spastin. This motif is not conserved in Drosophila spastin or katanin, which also bound by non-catalytic parts of the protein. The binding affinities of spastin and katanin were nucleotide-sensitive, with the lowest affinities under ADP,, the highest under ATP-γS conditions. These changes correlated with the formation of higher oligomeric states, as shown in biochemical experiments and electron microscopic images. Vice versa, the artificial dimerization of human spastin by addition of a coiled coil led to a constitutively active enzyme. These observations suggest that dimer formation is a crucial step in the formation of the active complex, and thus the severing process by spastin.  相似文献   

20.
Polyglutamylation is a post-translational modification that generates lateral acidic side chains on proteins by sequential addition of glutamate amino acids. This modification was first discovered on tubulins, and it is important for several microtubule functions. Besides tubulins, only the nucleosome assembly proteins NAP1 and NAP2 have been shown to be polyglutamylated. Here, using a proteomic approach, we identify a large number of putative substrates for polyglutamylation in HeLa cells. By analyzing a selection of these putative substrates, we show that several of them can serve as in vitro substrates for two of the recently discovered polyglutamylases, TTLL4 and TTLL5. We further show that TTLL4 is the main polyglutamylase enzyme present in HeLa cells and that new substrates of polyglutamylation are indeed modified by TTLL4 in a cellular context. No clear consensus polyglutamylation site could be defined from the primary sequence of the here-identified new substrates of polyglutamylation. However, we demonstrate that glutamate-rich stretches are important for a protein to become polyglutamylated. Most of the newly identified substrates of polyglutamylation are nucleocytoplasmic shuttling proteins, including many chromatin-binding proteins. Our work reveals that polyglutamylation is a much more widespread post-translational modification than initially thought and thus that it might be a regulator of many cellular processes.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号