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.
During cell division, chromosome segregation is governed by the interaction of spindle microtubules with the kinetochore. A dramatic remodeling of interpolar microtubules into an organized central spindle between the separating chromatids is required for the initiation and execution of cytokinesis. Central spindle organization requires mitotic kinesins, microtubule-bundling protein PRC1, and Aurora B kinase complex. However, the precise role of PRC1 in central spindle organization has remained elusive. Here we show that PRC1 recruits CLASP1 to the central spindle at early anaphase onset. CLASP1 belongs to a conserved microtubule-binding protein family that mediates the stabilization of overlapping microtubules of the central spindle. PRC1 physically interacts with CLASP1 and specifies its localization to the central spindle. Repression of CLASP1 leads to sister-chromatid bridges and depolymerization of spindle midzone microtubules. Disruption of PRC1-CLASP1 interaction by a membrane-permeable peptide abrogates accurate chromosome segregation, resulting in sister chromatid bridges. These findings reveal a key role for the PRC1-CLASP1 interaction in achieving a stable anti-parallel microtubule organization essential for faithful chromosome segregation. We propose that PRC1 forms a link between stabilization of CLASP1 association with central spindle microtubules and anti-parallel microtubule elongation.To ensure that each daughter cell receives the full complement of the genome in each cell division, chromosomes move poleward, and non-kinetochore fibers become bundled at the onset of anaphase, initiating assembly of the central spindle, a set of anti-parallel microtubules that serves to concentrate key regulators of cytokinesis (13). Chromosomal passengers are a group of evolutionarily conserved proteins that orchestrates chromosome segregation and central spindle plasticity (4, 5). This protein complex containing Aurora B, Survivin, INCENP, and Borealin is relocated from the kinetochore to the central spindle upon anaphase onset (59). Perturbation of their function results in defects in metaphase chromosome alignment, chromosome segregation, and cytokinesis (10).Among the central spindle maintenance components, only two have been reported to mediate the microtubule bundling in the central spindle. One is centralspindlin, a heterotetramer containing CeMKLP1/ZEN-4 and RhoGAP/CYK-4 (11), and the other one is an evolutionarily conserved protein, PRC1 (also named Feo in fruit fry, Ase1 in yeast, and MAP65 in plant cells). PRC1 is a non-motor microtubule-binding and -bundling protein in human cells originally identified as a Cdc2 substrate essential for cytokinesis (12, 13). Similar microtubule regulatory activities have been reported in yeast, fruit fly, and plant cells. It is well known that overexpression of wild type PRC1 in HeLa cells can result in thick microtubule bundles in cells at interphase (13). Bundling activity of PRC1, as well as centralspindlin, is required for the organization of the central spindle as well as for the successful progression of cytokinesis. PRC1 molecules accumulate on the midline of a central spindle with the cell cycle progression to anaphase. As a non-motor microtubule-binding protein, transportation of PRC1 to the midline is promoted by its association to kinesin, KIF4A, and timing of this progression is controlled by the dephosphorylation of Thr-481 on PRC1 when the cell exits metaphase by phosphatase Cdc14 (14). Our recent study shows that prevention of the phosphorylation of PRC1 at Thr-470 causes an inhibition in PRC1 oligomerization in vitro and an aberrant organization of central spindle in vivo, suggesting that this phosphorylation-dependent PRC1 oligomerization ensures that central spindle assembly occurs at the appropriate time in the cell cycle (15).Spatiotemporal regulation of microtubule organization and dynamics is responsible for the mitotic apparatus such as the central spindle. However, it has remained elusive as to how the central spindle microtubule organization and dynamics are regulated. There are large varieties of microtubule-associated proteins responsible for regulation of the dynamic behavior of microtubules and microtubule-mediated transport. Among these, proteins that associate with the tips of microtubules are called +TIPs, for “plus-end tracking proteins.” These proteins have been shown to be important in different organisms and cellular systems (16). Using yeast two-hybrid assay, CLASPs were identified as interacting partners of the CLIPs and characterized as new +TIP proteins (17).The microtubule-binding protein CLASP is emerging as an important microtubule regulator in the formation of the mitotic apparatus (1822). CLASP is required for promoting plus-end growth of spindle microtubules in prometaphase (23). Although the molecular mechanisms underlying its regulation of microtubule dynamics remain elusive, it is generally believed that CLASP orchestrates microtubule dynamics via its physical interacting with EB1, CLIP170, and microtubules (17, 24).To delineate the molecular function of PRC1 in central spindle organization and spatiotemporal regulation, we carried out a new search for PRC1-interacting proteins. Our studies show that PRC1 physically interacts with CLASP1, and the two proteins cooperate in the organization of the central spindle. Our studies provide a novel regulatory mechanism in which the PRC1 complex operates central spindle organization in mitosis.  相似文献   

2.
Stathmin is an important regulator of microtubule polymerization and dynamics. When unphosphorylated it destabilizes microtubules in two ways, by reducing the microtubule polymer mass through sequestration of soluble tubulin into an assembly-incompetent T2S complex (two α:β tubulin dimers per molecule of stathmin), and by increasing the switching frequency (catastrophe frequency) from growth to shortening at plus and minus ends by binding directly to the microtubules. Phosphorylation of stathmin on one or more of its four serine residues (Ser16, Ser25, Ser38, and Ser63) reduces its microtubule-destabilizing activity. However, the effects of phosphorylation of the individual serine residues of stathmin on microtubule dynamic instability have not been investigated systematically. Here we analyzed the effects of stathmin singly phosphorylated at Ser16 or Ser63, and doubly phosphorylated at Ser25 and Ser38, on its ability to modulate microtubule dynamic instability at steady-state in vitro. Phosphorylation at either Ser16 or Ser63 strongly reduced or abolished the ability of stathmin to bind to and sequester soluble tubulin and its ability to act as a catastrophe factor by directly binding to the microtubules. In contrast, double phosphorylation of Ser25 and Ser38 did not affect the binding of stathmin to tubulin or microtubules or its catastrophe-promoting activity. Our results indicate that the effects of stathmin on dynamic instability are strongly but differently attenuated by phosphorylation at Ser16 and Ser63 and support the hypothesis that selective targeting by Ser16-specific or Ser63-specific kinases provides complimentary mechanisms for regulating microtubule function.Stathmin is an 18-kDa ubiquitously expressed microtubule-destabilizing phosphoprotein whose activity is modulated by phosphorylation of its four serine residues, Ser16, Ser25, Ser38, and Ser63 (17). Several classes of kinases have been identified that phosphorylate stathmin, including kinases associated with cell growth and differentiation such as members of the mitogen-activated protein kinase (MAPK)2 family, cAMP-dependent protein kinase (15, 811), and kinases associated with cell cycle regulation such as cyclin-dependent kinase 1 (3, 1214). Phosphorylation of stathmin is required for cell cycle progression through mitosis and for proper assembly/function of the mitotic spindle (3, 1316). Inhibition of stathmin phosphorylation produces strong mitotic phenotypes characterized by disassembly and disorganization of mitotic spindles and abnormal chromosome distributions (3, 1314).Stathmin is known to destabilize microtubules in two ways. One is by binding to soluble tubulin and forming a stable complex that cannot polymerize into microtubules, consisting of one molecule of stathmin and two molecules of tubulin (T2S complex) (1724). Addition of stathmin to microtubules in equilibrium with soluble tubulin results in sequestration of the tubulin and a reduction in the level of microtubule polymer (1718, 22, 2528). In addition to reducing the amount of assembled polymer, tubulin sequestration by stathmin has been shown to increase the switching frequency at microtubule plus ends from growth to shortening (called the catastrophe frequency) as the microtubules relax to a new steady state (17, 29). The second way is by binding directly to microtubules (2730). The direct binding of stathmin to microtubules increases the catastrophe frequency at both ends of the microtubules and considerably more strongly at minus ends than at plus ends (27). Consistent with its strong catastrophe-promoting activity at minus ends, stathmin increases the treadmilling rate of steady-state microtubules in vitro (27). These results have led to the suggestion that stathmin might be an important cellular regulator of minus-end microtubule dynamics (27).Phosphorylation of stathmin diminishes its ability to regulate microtubule polymerization (3, 14, 2526). Phosphorylation of Ser16 or Ser63 appears to be more critical than phosphorylation of Ser25 and Ser38 for the ability of stathmin to bind to soluble tubulin and to inhibit microtubule assembly in vitro (3, 25). Inhibition of stathmin phosphorylation induces defects in spindle assembly and organization (3, 14) suggesting that not only soluble tubulin-microtubule levels are regulated by phosphorylation of stathmin, but the dynamics of microtubules could also be regulated in a phosphorylation-dependent manner.It is not known how phosphorylation at any of the four serine residues of stathmin affects its ability to regulate microtubule dynamics and, specifically, its ability to increase the catastrophe frequency at plus and minus ends due to its direct interaction with microtubules. Thus, we determined the effects of stathmin individually phosphorylated at either Ser16 or Ser63 and doubly phosphorylated at both Ser25 and Ser38 on dynamic instability at plus and minus ends in vitro at microtubule polymer steady state and physiological pH (pH 7.2). We find that phosphorylation of Ser16 strongly reduces the direct catastrophe-promoting activity of stathmin at plus ends and abolishes it at minus ends, whereas phosphorylation of Ser63 abolishes the activity at both ends. The effects of phosphorylation of individual serines correlated well with stathmin''s reduced abilities to form stable T2S complexes, to inhibit microtubule polymerization, and to bind to microtubules. In contrast, double phosphorylation of Ser25 and Ser38 did not alter the ability of stathmin to modulate dynamic instability at the microtubule ends, its ability to form a stable T2S complex, or its ability to bind to microtubules. The data further support the hypotheses that phosphorylation of stathmin on either Ser16 or Ser63 plays a critical role in regulating microtubule polymerization and dynamics in cells.  相似文献   

3.
Infection with cagA-positive Helicobacter pylori is the strongest risk factor for the development of gastric carcinoma. The cagA gene product CagA, which is delivered into gastric epithelial cells, specifically binds to and aberrantly activates SHP-2 oncoprotein. CagA also interacts with and inhibits partitioning-defective 1 (PAR1)/MARK kinase, which phosphorylates microtubule-associated proteins to destabilize microtubules and thereby causes epithelial polarity defects. In light of the notion that microtubules are not only required for polarity regulation but also essential for the formation of mitotic spindles, we hypothesized that CagA-mediated PAR1 inhibition also influences mitosis. Here, we investigated the effect of CagA on the progression of mitosis. In the presence of CagA, cells displayed a delay in the transition from prophase to metaphase. Furthermore, a fraction of the CagA-expressing cells showed spindle misorientation at the onset of anaphase, followed by chromosomal segregation with abnormal division axis. The effect of CagA on mitosis was abolished by elevated PAR1 expression. Conversely, inhibition of PAR1 kinase elicited mitotic delay similar to that induced by CagA. Thus, CagA-mediated inhibition of PAR1, which perturbs microtubule stability and thereby causes microtubule-based spindle dysfunction, is involved in the prophase/metaphase delay and subsequent spindle misorientation. Consequently, chronic exposure of cells to CagA induces chromosomal instability. Our findings reveal a bifunctional role of CagA as an oncoprotein: CagA elicits uncontrolled cell proliferation by aberrantly activating SHP-2 and at the same time induces chromosomal instability by perturbing the microtubule-based mitotic spindle. The dual function of CagA may cooperatively contribute to the progression of multistep gastric carcinogenesis.Helicobacter pylori is a spiral-shaped bacterium first described in 1984 by Marshall and Warren (1). H. pylori inhabits at least half of the world''s human population. Clinically isolated H. pylori strains can be divided into two major subtypes based on their ability to produce a 120- to 145-kDa protein called cytotoxin-associated gene A antigen (CagA)2 (25). More than 90–95% of H. pylori strains isolated in East Asian countries such as Japan, Korea, and China are cagA-positive, whereas 40–50% of those isolated in Western countries are cagA-negative. Infection with a cagA-positive H. pylori strain is associated with severe atrophic gastritis, peptic ulcerations, and gastric adenocarcinoma (612).H. pylori cagA-positive strains deliver the CagA protein into host cells via the cag pathogenicity island-encoded type IV secretion system (4, 5, 13, 14). Translocated CagA then localizes to the inner surface of the plasma membrane, where it undergoes tyrosine phosphorylation by Src family kinases or Abl kinase at the Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs present in the C-terminal region of CagA (1517). Tyrosine-phosphorylated CagA then binds specifically to SHP-2 tyrosine phosphatase and deregulates its phosphatase activity (1821). Recent studies have revealed that gain-of-function mutations of SHP-2 are associated with a variety of human malignancies, indicating that SHP-2 is a bona fide human oncoprotein. Furthermore, transgenic expression of CagA in mice induces gastrointestinal and hematological malignancies in a manner that is dependent on CagA tyrosine phosphorylation (22). These findings suggest a critical role of CagA-SHP-2 interaction in the oncogenic potential of CagA.A polarized epithelial monolayer is characterized by the presence of well developed cell-cell interaction apparatuses such as tight junctions and adherens junctions. The tight junctions act as a paracellular barrier in polarized epithelial cells and play an essential role in the establishment and maintenance of epithelial cell polarity by delimiting the apical and basolateral membrane domains. CagA disrupts the tight junctions and causes loss of epithelial apical-basal polarity (23, 24). The disruption of tight junctions by CagA is mediated by the specific interaction of CagA with partitioning-defective 1 (PAR1) (25, 26). PAR1 is a serine/threonine kinase originally isolated in Caenorhabditis elegans and highly conserved from yeast to humans (27, 28). In mammals, there are four PAR1 isoforms, which may have redundant roles in polarity regulation. PAR1 acts as a master regulator for the regulation of cell polarity in various cell systems. During epithelial polarization, PAR1 specifically localizes to the basolateral membrane, whereas atypical PKC complexed with PAR3 and PAR6 (aPKC complex) specifically localizes to the apical membrane as well as the tight junctions (2931). This asymmetric distribution of the two kinases, PAR1 and aPKC complex, ensures formation and maintenance of epithelial apical-basal polarity. Notably, mammalian PAR1 kinases were originally identified as microtubule affinity-regulating kinases (MARKs), which phosphorylate microtubule-associated proteins (MAPs) such as Tau, MAP2, and MAP4 on their tubulin-binding repeats. The PAR1/MARK-dependent phosphorylation causes MAPs to detach from and thereby destabilize microtubules (32, 33). Importantly, microtubules form a mitotic spindle, which plays an indispensable role in chromosomal alignment and separation during mitosis, raising the possibility that PAR1 regulates mitosis through controlling stability of the mitotic spindle. Indeed, during mitosis, MAPs undergo a severalfold higher level of phosphorylation (34, 35), and microtubule dynamics increase ∼20-fold (36). This in turn raises the intriguing possibility that CagA influences chromosomal stability by subverting MAP phosphorylation through systemic inhibition of PAR1.In this study, the effects of CagA on microtubule-dependent cellular events, especially dynamics of the mitotic spindle and chromosomal segregation during mitosis, were examined. The results of this work provide evidence that CagA perturbs mitotic spindle checkpoint and thereby causes chromosomal instability. Given the role of chromosomal instability in cell transformation, the newly identified CagA activity may play a crucial role in the development of gastric carcinoma.  相似文献   

4.
Formins assemble non-branched actin filaments and modulate microtubule dynamics during cell migration and cell division. At the end of mitosis formins contribute to the generation of actin filaments that form the contractile ring. Rho small GTP-binding proteins activate mammalian diaphanous-related (mDia) formins by directly binding and disrupting an intramolecular autoinhibitory mechanism. Although the Rho-regulated activation mechanism is well characterized, little is known about how formins are switched off. Here we reveal a novel mechanism of formin regulation during cytokinesis based on the following observations; 1) mDia2 is degraded at the end of mitosis, 2) mDia2 is targeted for disposal by post-translational ubiquitin modification, 3) forced expression of activated mDia2 yields binucleate cells due to failed cytokinesis, and 4) the cytokinesis block is dependent upon mDia2-mediated actin assembly as versions of mDia2 incapable of nucleating actin but that still stabilize microtubules have no effect on cytokinesis. We propose that the tight control of mDia2 expression and ubiquitin-mediated degradation is essential for the completion of cell division. Because of the many roles for formins in cell morphology, we discuss the relevance of mDia protein turnover in other processes where ubiquitin-mediated proteolysis is an essential component.Formin proteins play a role in diverse processes such as cell migration (1, 2), vesicle trafficking (3, 4), tumor suppression (5, 6), and microtubule stabilization (7, 8). Formins also play an essential and conserved role in cytokinesis (911). Proper cell division is essential in all animals to maintain the integrity of their genome. Failure to complete cytokinesis can result in genomic instability and ultimately lead to disease such as cancer (12).The members of the mDia2 family of formins are autoregulated Rho effectors that remodel the cytoskeleton by nucleating and elongating non-branched actin filaments (13). The amino terminus of mDia contains a GTPase binding domain (GBD) that directs interaction with specific Rho small GTP-binding proteins. The adjacent Dia inhibitory domain (DID) mediates mDia autoregulation through its interaction with the carboxyl-terminal diaphanous autoregulatory domain (DAD) (14, 15). Between the DID and DAD domains lie the conserved formin homology 1 (FH1) and FH2 domains. The FH1 domain is a proline-rich region that mediates binding to other proteins such as profilin, Src, and Dia-interacting protein (1619). In contrast, the FH2 domain binds monomeric actin to generate filamentous actin (F-actin) and can also bind microtubules directly to induce their stabilization (8, 20).Although the mechanism of mDia activation is well characterized, little is known about its inactivation. Previous reports have suggested that formins can cycle between active, partially active, and inactive states (21, 22) due to GTP hydrolysis upon Rho binding to GTPase-activating proteins. Another formin inactivation mechanism is through mDia interactions with Dia-interacting protein (23). In the context of cortical actin assembly, Dia-interacting protein negatively regulates mDia2 actin polymerization but has no effect on mDia1 actin polymerization despite its ability to interact with both proteins directly (17). Because of the fundamental role for formins in cell division, we sought to identify how mDia2 is inactivated in mitosis.During cell division, the expression level and activity of many proteins (e.g. cyclins and Aurora and Polo kinases) are tightly regulated (24). A unifying regulatory mechanism among these proteins is ubiquitin-mediated proteolysis. In this study we find that mDia2 protein levels are constant from S phase into mitosis and dramatically decrease at the end of mitosis due to ubiquitin-mediated degradation. Failure to inhibit mDia2 actin assembly results in multinucleation, which supports an essential role for the tight regulation of mDia2 during cell division.  相似文献   

5.
Microtubule plus-end tracking proteins (+TIPs) control microtubule dynamics in fundamental processes such as cell cycle, intracellular transport, and cell motility, but how +TIPs are regulated during mitosis remains largely unclear. Here we show that the endogenous end-binding protein family EB3 is stable during mitosis, facilitates cell cycle progression at prometaphase, and then is down-regulated during the transition to G1 phase. The ubiquitin-protein isopeptide ligase SIAH-1 facilitates EB3 polyubiquitination and subsequent proteasome-mediated degradation, whereas SIAH-1 knockdown increases EB3 stability and steady-state levels. Two mitotic kinases, Aurora-A and Aurora-B, phosphorylate endogenous EB3 at Ser-176, and the phosphorylation triggers disruption of the EB3-SIAH-1 complex, resulting in EB3 stabilization during mitosis. Our results provide new insight into a regulatory mechanism of +TIPs in cell cycle transition.Microtubule dynamics are essential in many cellular processes, including cell motility, intracellular transport, accurate mitosis, and cytokinesis in all eukaryotes. The regulatory factors for microtubule dynamics can be classified into two main types as follows: microtubule-destabilizing proteins, such as stathmin/Op18 (1) and the Kinesin-13 family (also known as MCAK/KIF2 family) (2), and microtubule-stabilizing proteins, the classic superfamily of microtubule-associated proteins (3). Additionally, the plus-end tracking proteins (+TIPs)3 have recently been identified; this family specifically accumulates at the ends of growing microtubules and regulates the microtubule plus-end targeting to the cell cortex or mitotic kinetochores (4, 5).The EB1 family is a member of the +TIPs family and consists of three homologs in mammals: EB1, EB2/RP1 (henceforth, EB2), and EB3 (6). As EB1 was originally identified as a protein that interacts with the well characterized tumor suppressor adenomatous polyposis coli (APC) protein (7), the function of EB1 has been investigated extensively. EB1 interacts with other +TIPs, including APC, p150glued, CLIPs, and CLASP1/2, and the interaction network controls microtubule orientation and microtubule-cortex interaction during cell migration (5, 8, 9). EB1 functions not only in the regulation of interphase microtubule dynamics but also in mitotic spindle regulation. For accurate chromosomal segregation, sister chromatids become aligned to the metaphase plate during metaphase, and the alignment requires spindle-kinetochore attachment. Two models have been proposed; in the first, termed the “search-and-capture” model, EB1 localized at the growing microtubule plus-ends searches for binding partners located on kinetochores (10, 11). In the second model proposed recently, EB1 makes kinetochore fibers and centrosomal microtubules connect, and it is essential for the formation of a functional bipolar spindle (12). Thus, EB1 is thought to be a master controller of microtubule plus-ends; however, little is known about other EB1 family members. Given that EB3 is localized on the microtubule network and binds to APC and CLIPs identically to EB1, it is possible that EB3 acts as an EB1 analog in cells (1315).Cell division is precisely regulated by several post-translational modifications of proteins, mainly reversible phosphorylation and ubiquitination, which is followed by degradation. Accurate mitotic phase progression requires the appropriate phosphorylation of various proteins by mitotic kinases (16, 17). One of the key mitotic kinases is the Aurora family that has been highly conserved from yeast to humans. There are three homologs (Aurora-A, -B, and -C) in human and mouse (18). Although their homology at the protein level is more than 84%, their functions and subcellular localizations are distinct. Aurora-A is located in the centrosomes and spindle and is required for mitotic entry, centrosome maturation/separation, and spindle assembly (19). Aurora-B is a chromosomal passenger protein that localizes on the inner centromere of the chromosomes until metaphase to regulate the spindle-kinetochore attachment, and from anaphase, it translocates to the central spindle and then accumulates in the midbody for cytokinesis (20, 21). The numerous substrates of the Aurora family include regulatory factors for microtubule dynamics, such as the microtubule-destabilizing proteins MCAK and stathmin, which help to establish the bipolar attachment and spindle assembly, respectively (2224). It is possible that the Aurora family regulates the EB1 family by phosphorylation.In this study, we performed yeast two-hybrid screening and obtained the EB1 yeast homolog Bim1 as a protein that interacts with Ipl1, a yeast counterpart of Aurora. Here we demonstrate the novel regulatory mechanisms of EB3 by two cell cycle-dependent post-translational modifications, phosphorylation and ubiquitin-proteasome-mediated degradation.  相似文献   

6.
In Xenopus egg extracts, spindles assembled around sperm nuclei contain a centrosome at each pole, while those assembled around chromatin beads do not. Poles can also form in the absence of chromatin, after addition of a microtubule stabilizing agent to extracts. Using this system, we have asked (a) how are spindle poles formed, and (b) how does the nucleation and organization of microtubules by centrosomes influence spindle assembly? We have found that poles are morphologically similar regardless of their origin. In all cases, microtubule organization into poles requires minus end–directed translocation of microtubules by cytoplasmic dynein, which tethers centrosomes to spindle poles. However, in the absence of pole formation, microtubules are still sorted into an antiparallel array around mitotic chromatin. Therefore, other activities in addition to dynein must contribute to the polarized orientation of microtubules in spindles. When centrosomes are present, they provide dominant sites for pole formation. Thus, in Xenopus egg extracts, centrosomes are not necessarily required for spindle assembly but can regulate the organization of microtubules into a bipolar array.During cell division, the correct organization of microtubules in bipolar spindles is necessary to distribute chromosomes to the daughter cells. The slow growing or minus ends of the microtubules are focused at each pole, while the plus ends interact with the chromosomes in the center of the spindle (Telzer and Haimo, 1981; McIntosh and Euteneuer, 1984). Current concepts of spindle assembly are based primarily on mitotic spindles of animal cells, which contain centrosomes. Centrosomes are thought to be instrumental for organization of the spindle poles and for determining both microtubule polarity and the spindle axis. In the prevailing model, termed “Search and Capture,” dynamic microtubules growing from two focal points, the centrosomes, are captured and stabilized by chromosomes, generating a bipolar array (Kirschner and Mitchison, 1986). However, while centrosomes are required for spindle assembly in some systems (Sluder and Rieder, 1985; Rieder and Alexander, 1990; Zhang and Nicklas, 1995a ,b), in other systems they appear to be dispensable (Steffen et al., 1986; Heald et al., 1996). Furthermore, centrosomes are not present in higher plant cells and in female meiosis of most animal species (Bajer and Mole, 1982; Gard, 1992; Theurkauf and Hawley, 1992; Albertson and Thomson, 1993; Lambert and Lloyd, 1994). In the absence of centrosomes, bipolar spindle assembly seems to occur through the self-organization of microtubules around mitotic chromatin (McKim and Hawley, 1995; Heald et al., 1996; Waters and Salmon, 1997). The observation of apparently different spindle assembly pathways raises several questions: Do different types of spindles share common mechanisms of organization? How do centrosomes influence spindle assembly? In the absence of centrosomes, what aspects of microtubule self-organization promote spindle bipolarity?To begin to address these questions, we have used Xenopus egg extracts, which can be used to reconstitute different types of spindle assembly. Spindle assembly around Xenopus sperm nuclei is directed by centrosomes (Sawin and Mitchison, 1991). Like other meiotic systems (Bastmeyer et al., 1986; Steffen et al., 1986), Xenopus extracts also support spindle assembly around chromatin in the absence of centrosomes through the movement and sorting of randomly nucleated microtubules into a bipolar structure (Heald et al., 1996). In this process, the microtubule-based motor cytoplasmic dynein forms spindle poles by cross-linking and sliding microtubule minus ends together. Increasing evidence suggests that the function of dynein in spindle assembly depends on its interaction with other proteins, including dynactin, a dynein-binding complex, and NuMA1 (nuclear protein that associates with the mitotic apparatus) (Merdes et al., 1996; Echeverri et al., 1996; Gaglio et al., 1996). In this paper, we demonstrate that both in the presence and absence of centrosomes, spindle pole assembly occurs by a common dynein-dependent mechanism. We show that when centrosomes are present, they are tethered to spindle poles by dynein. In the absence of dynein function, microtubules are still sorted into an antiparallel array, indicating that other aspects of microtubule self-assembly independent of pole formation promote spindle bipolarity around mitotic chromatin. Since centrosomes are dispensable for pole formation in this system, what is their function? We show here that if only one centrosome is present, it acts as a dominant site for microtubule nucleation and focal organization, resulting in a monopolar spindle. Therefore, although centrosomes are not required in this system, they can influence spindle pole formation and bipolarity.  相似文献   

7.
8.
Mutations in adenomatous polyposis coli (APC) protein is a major contributor to tumor initiation and progression in several tumor types. These mutations affect APC function in the Wnt-β-catenin signaling and influence mitotic spindle anchoring to the cell cortex and orientation. Here we report that the mitotic anchoring and orientation function of APC is regulated by cyclin A/cdk2. Knockdown of cyclin A and inhibition of cdk2 resulted in cells arrested in mitosis with activation of the spindle assembly checkpoint. The mitotic spindle was unable to form stable attachments to the cell cortex, and this resulted in the spindles failing to locate to the central position in the cells and undergo dramatic rotation. We have demonstrated that cyclin A/cdk2 specifically associates with APC in late G2 phase and phosphorylates it at Ser-1360, located in the mutation cluster region of APC. Mutation of APC Ser-1360 to Ala results in identical off-centered mitotic spindles. Thus, this cyclin A/cdk2-dependent phosphorylation of APC affects astral microtubule attachment to the cortical surface in mitosis.Adenomatous polyposis coli (APC)5 was initially identified as a tumor suppressor in familial colon cancers. It is a regulator of Wnt-β-catenin signaling and thereby regulates progression into the cell cycle, but also has Wnt-independent mitotic roles in spindle anchoring and kinetochore function (13). These latter functions of APC are mediated through its ability to bind microtubules and the end-binding protein, EB1 (2). Loss or mutation of APC has been demonstrated to increase chromosomal instability, although whether this is through its Wnt-dependent or independent functions is unclear (3). The mitotic defects caused by APC mutation and depletion are characterized by an inability to locate the center of the cell and failure of chromosomal alignment (4). It was also associated with a loss of normal spindle orientation in small intestinal crypts of APCMin/+ mice (5), suggesting that disruption of the normal mitotic functions of APC are likely to be major contributors to the chromosomal instability observed.APC interaction with EB1 is regulated by phosphorylation of its C-terminal domain by cyclin B/cdk1 in mitosis (6, 7). The majority of APC mutations occurs in a region from codons 1,000 to 1,500 called the mutation cluster region (MCR) and result in truncations of the C-terminal half of the protein, which includes the β-catenin, microtubule, and EB1 binding sites of APC (1, 2). Depletion of either APC or EB1 produce almost identical mitotic defects, indicating their interaction is critical to normal spindle formation (4, 8). However, expression of various truncation mutants across the MCR revealed interesting differences the spindle defects observed, suggesting that this role of APC in spindle function is not solely due to interaction with EB1 (4). Progression into mitosis is regulated by cyclin B/cdk1, but the timing of its activation is regulated by cyclin A/cdk2 (912), which in turn is regulated by the dual specificity phosphatase cdc25B in G2 phase (13). Cyclin A is destroyed at prometaphase (14) suggesting that its activity is required for not only entry into mitosis but during the early part of mitosis itself. The majority of substrates identified for cyclin A/cdk2 are nuclear, where the majority of cyclin A/cdk2 is localized in G2, but reports also suggest that cyclin A is capable of localizing to both the cytoplasm and centrosomes (9, 14, 15), thus there are likely to be additional substrates for this complex in the cytoplasmic compartment. In vitro studies using Xenopus extracts have demonstrated that cyclin A/cdk is capable of increasing microtubule nucleation at the centrosomes (16). Thus it is likely that cyclin A in association with its cdk partner has roles in not only promoting entry into mitosis but also in establishing mitosis, possibly by influencing the mitotic machinery.We have used siRNA to knockdown cyclin A2, the major cyclin A isoform in somatic cells, and cdk2 inhibitors to examine the role of the G2 phase cyclin A/cdk2 complex in cell cycle progression. We demonstrate that knockdown of cyclin A delayed progression through mitosis and activation of the spindle assembly checkpoint. Spindle anchoring was also defective, a phenotype identical to APC-truncating mutants. We demonstrate that cyclin A/cdk2 binds to APC in late G2 phase/early mitosis and phosphorylates Ser-1360, and that the lack of this phosphorylation of APC results in identical mitotic defects to the absence of cyclin A/cdk2.  相似文献   

9.
During mitosis, establishment of structurally and functionally sound bipolar spindles is necessary for maintaining the fidelity of chromosome segregation. Tumor-associated microtubule-associated protein (TMAP), also known as cytoskeleton-associated protein 2 (CKAP2), is a mitotic spindle-associated protein whose level is frequently up-regulated in various malignancies. Previous reports have suggested that TMAP is a potential regulator of mitotic spindle assembly and dynamics and that it is required for chromosome segregation to occur properly. So far, there have been no reports on how its mitosis-related functions are regulated. Here, we report that TMAP is hyper-phosphorylated at the C terminus specifically during mitosis. At least four different residues (Thr-578, Thr-596, Thr-622, and Ser-627) were responsible for the mitosis-specific phosphorylation of TMAP. Among these, Thr-622 was specifically phosphorylated by Cdk1-cyclin B1 both in vitro and in vivo. Interestingly, compared with the wild type, a phosphorylation-deficient mutant form of TMAP, in which Thr-622 had been replaced with an alanine (T622A), induced a significant increase in the frequency of metaphase cells with abnormal bipolar spindles, which often displayed disorganized, asymmetrical, or narrow and elongated morphologies. Formation of these abnormal bipolar spindles subsequently resulted in misalignment of metaphase chromosomes and ultimately caused a delay in the entry into anaphase. Moreover, such defects resulting from the T622A mutation were associated with a decrease in the rate of protein turnover at spindle microtubules. These findings suggest that Cdk1-cyclin B1-mediated phosphorylation of TMAP is important for and contributes to proper regulation of microtubule dynamics and establishment of functional bipolar spindles during mitosis.Tumor-associated microtubule-associated protein (TMAP),3 also known as cytoskeleton-associated protein 2 (CKAP2), LB-1, and se20-10, is frequently up-regulated in various malignancies, including gastric adenocarcinoma, diffuse B-cell lymphoma, and cutaneous T-cell lymphoma (13), and detected in various cancer cell lines (1, 4). Knockdown of TMAP significantly reduces the rate of cell growth (5, 6), indicating that it is essential for normal cell growth. However, the cellular functions of TMAP remain largely unknown. Recent findings indicate that TMAP plays an essential role in mitosis. Expression of TMAP changes in a cell cycle-dependent manner; its expression is relatively low during G1, starts to incline during G1/S transition, and peaks at G2/M phases of the cell cycle (5, 7). TMAP primarily localizes at mitotic spindle and spindle poles during mitosis (1, 4, 8, 9). During late stages of mitosis, however, TMAP localizes near the chromatin region and to the midbody microtubules (8). TMAP has microtubule-stabilizing properties (4, 8, 9), and its overexpression induces mitotic spindle defects, including monopolar spindle formation, and arrests cells at mitosis as a result (8). Similar to other mitotic regulators, TMAP is a substrate of the anaphase-promoting complex (8). TMAP is degraded during mitotic exit by the anaphase-promoting complex-Cdh1 in a KEN box-dependent manner. Results of the experiments using a nondegradable mutant of TMAP suggested that proper regulation of the TMAP protein level is functionally important for establishment of bipolar spindles and completion of cytokinesis. Recently, we also have shown that siRNA-mediated depletion of TMAP in mammalian cells results in chromosome missegregation, characterized by chromatin bridge formation and malformation of interphase nuclei, and such phenotype was associated with a reduction in the spindle assembly checkpoint activity (6). These findings suggest that TMAP is a potential regulator of mitotic spindle function and dynamics and that proper regulation of its protein level and functions is necessary for establishment of bipolar spindles as well as for maintaining the fidelity of the chromosome segregation process.At the onset of mitosis, the microtubule network undergoes extensive rearrangements to form a unique bipolar structure, called the mitotic spindle. Multiple factors have been shown to associate with the mitotic spindle and regulate its function by influencing its assembly and dynamics (10, 11). Establishment of a functional bipolar mitotic spindle is critical for faithful segregation of sister chromatids and maintenance of genomic stability. In support of this notion, disruption or depletion of factors involved in regulation of the spindle microtubule dynamics or establishment of spindle bipolarity have been shown to induce spindle dysfunction and ultimately chromosome missegregation (1214).The cyclin-dependent kinase 1 (Cdk1) in complex with cyclin B1 (Cdk1-cyclin B1) is one of the key mitotic kinases. The kinase activity of Cdk1-cyclin B1 governs the entry into mitosis from G2 phase of the cell cycle (15, 16). Through mediating phosphorylation of a variety of substrates, Cdk1-cyclin B1 also plays an important role in multiple processes during mitosis, including chromosome condensation, nuclear envelope breakdown, centrosome separation, regulation of spindle microtubule dynamics, and metaphase to anaphase transition (1720). In particular, a number of regulators of microtubules are among Cdk1-cyclin B1 substrates (21). For instance, phosphorylation of a kinesin-related motor protein, Eg5, by Cdk1-cyclin B1 is necessary for its centrosomal localization and ultimately for the centrosome separation process to occur properly (18). Also, Cdk1-cyclin B1-mediated phosphorylation of some of the effectors of microtubule dynamics has been shown to regulate their microtubule-stabilizing or -destabilizing activities during mitosis (22, 23). These suggest that the assembly and maintenance of bipolar spindles during mitosis are under regulation of Cdk1-cyclin B1.We have recently reported that TMAP is phosphorylated specifically during mitosis (24), which led us to hypothesize that the mitotic functions of TMAP are regulated by timely phosphorylation. In the present study, we identified multiple, mitosis-specific phosphorylation sites on TMAP, one of which is phosphorylated by Cdk1-cyclin B1, and investigated the functional importance of Cdk1-cyclin B1-mediated phosphorylation of TMAP during mitosis.  相似文献   

10.
CLIP-170, the founding member of microtubule “plus ends tracking” proteins, is involved in many critical microtubule-related functions, including recruitment of dynactin to the microtubule plus ends and formation of kinetochore-microtubule attachments during metaphase. Although it has been reported that CLIP-170 is a phosphoprotein, neither have individual phosphorylation sites been identified nor have the associated kinases been extensively studied. Herein, we identify Cdc2 as a kinase that phosphorylates CLIP-170. We show that Cdc2 interacts with CLIP-170 mediating its phosphorylation on Thr287 in vivo. Significantly, expression of CLIP-170 with a threonine 287 to alanine substitution (T287A) results in its mislocalization, accumulation of Plk1 and cyclin B, and block of the G2/M transition. Finally, we found that depletion of CLIP-170 leads to centrosome reduplication and that Cdc2 phosphorylation of CLIP-170 is required for the process. These results demonstrate that Cdc2-mediated phosphorylation of CLIP-170 is essential for the normal function of this protein during cell cycle progression.Microtubule dynamics consist of alternating phases of growth and shortening, a pattern of behavior known as dynamic instability (1). This process is tightly regulated by a group of proteins that bind specifically to the plus ends of the growing microtubules (2). Cytoplasmic linker protein (CLIP)3 -170, the founding member of the microtubule plus end family (3), is composed of three separate regions: N terminus, central coiled-coil region, and C terminus. In addition to two conserved cytoskeleton-associated protein glycine-rich (CAP-Gly) domains, the N terminus has three serine-rich regions. The N-terminal domain plays an essential role in microtubule targeting (4), the long central coiled-coil domain is responsible for dimerization of the protein, and the C-terminal region, which contains two zinc-finger domains interferes with microtubule binding by interacting with the N terminus (5). Experiments in a variety of organisms have demonstrated that CLIP-170 plays an important role in microtubule dynamics (6, 7). In addition to its positive role in regulating microtubule growth in both yeast and humans (8, 9), CLIP-170 is involved in recruitment of dynactin to the microtubule plus ends and in linking microtubules to the cortex through Cdc42 and IQGAP (10, 11). The role of CLIP-170 during mitosis was recently examined by loss-of-function approaches. It was shown that CLIP-170 localizes to unattached kinetochores in prometaphase and that such localization is essential for the formation of kinetochore-microtubule attachments (12, 13).It was previously reported that CLIP-170 is a phosphoprotein and that overall phosphorylation of CLIP-170 affects its microtubule binding ability (14). More recently, metabolic labeling experiments indicated that CLIP-170 is phosphorylated at multiple sites (15). However, individual phosphorylation sites have not been identified. Moreover, the FKBP12-rapamycin-associated protein (FRAP) is the only kinase identified to date for CLIP-170 (15). Therefore, to fully understand the regulation of CLIP-170, it is important to identify individual phosphorylation sites and the responsible kinases. In this communication, we describe a novel kinase/substrate partnership between Cdc2 and CLIP-170. We provide evidence that Cdc2 phosphorylates CLIP-170 at Thr287, and the Cdc2-mediated phosphorylation of CLIP-170 is essential for its localization at microtubule plus ends in the G2 phase and the G2/M transition.  相似文献   

11.
Mitotic centromere-associated kinesin (MCAK) plays an essential role in spindle formation and in correction of improper microtubule-kinetochore attachments. The localization and activity of MCAK at the centromere/kinetochore are controlled by Aurora B kinase. However, MCAK is also abundant in the cytosol and at centrosomes during mitosis, and its regulatory mechanism at these sites is unknown. We show here that cyclin-dependent kinase 1 (Cdk1) phosphorylates T537 in the core domain of MCAK and attenuates its microtubule-destabilizing activity in vitro and in vivo. Phosphorylation of MCAK by Cdk1 promotes the release of MCAK from centrosomes and is required for proper spindle formation. Interfering with the regulation of MCAK by Cdk1 causes dramatic defects in spindle formation and in chromosome positioning. This is the first study demonstrating that Cdk1 regulates the localization and activity of MCAK in mitosis by directly phosphorylating the catalytic core domain of MCAK.Chromosomes are properly attached to the mitotic spindles, and chromosome movement is tightly linked to the structure and dynamics of spindle microtubules during mitosis. Important regulators of microtubule dynamics are the kinesin-13 proteins (37). This kinesin superfamily is defined by the localization of the conserved kinesin core motor domain in the middle of the polypeptide (19). Kinesin-13 proteins induce microtubule depolymerization by disassembling tubulin subunits from the polymer end (6). Among them, mitotic centromere-associated kinesin (MCAK) is the best-characterized member of the family. It depolymerizes microtubules in vitro and in vivo, regulates microtubule dynamics, and has been implicated in correcting misaligned chromosomes (12, 14, 16, 24). In agreement with these observations, both overexpression and inhibition of MCAK result in a disruption of microtubule dynamics, leading further to improper spindle assembly and errors in chromosome alignment and segregation (7, 11, 15, 22, 33). The importance of MCAK in ensuring the faithful segregation of chromosomes is consistent with the observation that MCAK is highly expressed in several types of cancer and thus is likely to be involved in causing aneuploidy (25, 32).While MCAK is found both in the cytoplasm and at the centromeres throughout the cell cycle, it is highly enriched on centrosomes, the centromeres/kinetochores, and the spindle midzone during mitosis (18, 21, 36, 38). In accordance with its localizations, MCAK affects many aspects throughout mitosis, from spindle assembly and maintenance (3, 10, 36) to chromosome positioning and segregation (14, 21, 35). Thus, the precise control of the localization and activity of MCAK is crucial for maintaining genetic integrity during mitosis. Regulation of MCAK on the centromeres/kinetochores by Aurora B kinase in mitosis has been intensively investigated (1, 28, 29, 43). The data reveal that MCAK is phosphorylated on several serine/threonine residues by Aurora B, which inhibits the microtubule-destabilizing activity of MCAK and regulates its localization on chromosome arms/centromeres/kinetochores during mitosis (1, 18, 28). Moreover, in concert with Aurora B, ICIS (inner centromere KinI stimulator), a protein targeting the inner centromeres in an MCAK-dependent manner, may regulate MCAK at the inner centromeres and prevent kinetochore-microtubule attachment errors in mitosis by stimulating the activity of MCAK (27). Interestingly, hSgo2, a recently discovered inner centromere protein essential for centromere cohesion, has been reported to be important in localizing MCAK to the centromere and in spatially regulating its mitotic activity (13). These data highlight that the activity and localization of MCAK on the centromeres/kinetochores during mitosis are tightly controlled by Aurora B and its cofactors. Remarkably, MCAK concentrates at spindle poles from prophase to telophase during mitosis (18); however, only a few studies have been done to deal with that issue. Aurora A-depleted prometaphase cells delocalize MCAK from spindle poles but accumulate the microtubule-stabilizing protein ch-TOG at poles (5), implying that Aurora A might influence the centrosomal localization of MCAK in mitosis. Aurora A is also found to be important for focusing microtubules at aster centers and for facilitating the transition from asters to bipolar spindles in Xenopus egg extracts (42). In addition, it has been revealed that Ca2+/calmodulin-dependent protein kinase II gamma (CaMKII gamma) suppresses MCAK''s activity, which is essential for bipolar spindle formation in mitosis (11). More work is required to gain insight into the regulatory mechanisms of MCAK at spindle poles during mitosis.Deregulated cyclin-dependent kinases (Cdks) are very often linked to genomic and chromosomal instability (20). Cyclin B1, the regulatory subunit of Cdk1, is localized to unattached kinetochores and contributes to efficient microtubule attachment and proper chromosome alignment (2, 4). We observed that knockdown of cyclin B1 induces defects in chromosome alignment and mitotic spindle formation (N.-N. Kreis, M. Sanhaji, A. Krämer, K. Sommor, F. Rödel, K. Strebhardt, and J. Yuan, submitted for publication). Yet, how Cdk1/cyclin B1 carries out these functions is not very well understood. In this context, it is extremely interesting to investigate the relationship between the essential mitotic kinase Cdk1 and the microtubule depolymerase MCAK in human cells.  相似文献   

12.
The ordered arrangement of cortical microtubules in growing plant cells is essential for anisotropic cell expansion and, hence, for plant morphogenesis. These arrays are dismantled when the microtubule cytoskeleton is rearranged during mitosis and reassembled following completion of cytokinesis. The reassembly of the cortical array has often been considered as initiating from a state of randomness, from which order arises at least partly through self-organizing mechanisms. However, some studies have shown evidence for ordering at early stages of array assembly. To investigate how cortical arrays are initiated in higher plant cells, we performed live-cell imaging studies of cortical array assembly in tobacco (Nicotiana tabacum) Bright Yellow-2 cells after cytokinesis and drug-induced disassembly. We found that cortical arrays in both cases did not initiate randomly but with a significant overrepresentation of microtubules at diagonal angles with respect to the cell axis, which coincides with the predominant orientation of the microtubules before their disappearance from the cell cortex in preprophase. In Arabidopsis (Arabidopsis thaliana) root cells, recovery from drug-induced disassembly was also nonrandom and correlated with the organization of the previous array, although no diagonal bias was observed in these cells. Surprisingly, during initiation, only about one-half of the new microtubules were nucleated from locations marked by green fluorescent protein-γ-tubulin complex protein2-tagged γ-nucleation complexes (γ-tubulin ring complex), therefore indicating that a large proportion of early polymers was initiated by a noncanonical mechanism not involving γ-tubulin ring complex. Simulation studies indicate that the high rate of noncanonical initiation of new microtubules has the potential to accelerate the rate of array repopulation.Higher plant cells feature ordered arrays of microtubules at the cell cortex (Ledbetter and Porter, 1963) that are essential for cell and tissue morphogenesis, as revealed by disruption of cortical arrays by drugs that cause microtubule depolymerization (Green, 1962) or stabilization (Weerdenburg and Seagull, 1988) and by loss-of-function mutations in a wide variety of microtubule-associated proteins (Baskin, 2001; Whittington et al., 2001; Buschmann and Lloyd, 2008; Lucas et al., 2011). The structure of these arrays is thought to control the pattern of cell growth primarily by its role in the deposition of cellulose microfibrils, the load-bearing component of the cell wall (Somerville, 2006). Functional relations between cortical microtubules and cellulose microfibrils have been proposed since the early sixties, even before cortical microtubules had been visualized (Green, 1962). Recent live-cell imaging studies have confirmed that cortical microtubules indeed guide the movement of cellulose synthase complexes that produce cellulose microfibrils (Paredez et al., 2006) and have shown further that microtubules position the insertion of most cellulose synthase complexes into the plasma membrane (Gutierrez et al., 2009). These activities of ordered cortical microtubules are proposed to facilitate the organization of cell wall structure, creating material properties that underlie cell growth anisotropy.While organization of the interphase cortical array appears to be essential for cell morphogenesis, this organization is disrupted during the cell cycle as microtubules are rearranged to create the preprophase band, spindle, and phragmoplast during mitosis and cytokinesis (for review, see Wasteneys, 2002). Upon completion of cytokinesis, an organized interphase cortical array is regenerated, but the pathway for this reassembly is not well understood.The plant interphase microtubule array is organized and maintained without centrosomes as organizing centers (for review, see Wasteneys, 2002; Bartolini and Gundersen, 2006; Ehrhardt and Shaw, 2006), and microtubule self-organization is proposed to play an important role in cortical microtubule array ordering (Dixit and Cyr, 2004). In electron micrographs, microtubules have been observed to be closely associated with the plasma membrane (Hardham and Gunning, 1978), and live-cell imaging provides evidence for attachment of microtubules to the cell cortex (Shaw et al., 2003; Vos et al., 2004). The close association to the plasma membrane restricts the cortical microtubules to a quasi two-dimensional plane where they interact through polymerization-driven collisions (Shaw et al., 2003; Dixit and Cyr, 2004). Microtubule encounters at shallow angles (<40°) have a high probability of leading to bundling, while microtubule encounters at steeper angles most likely result in induced catastrophes or microtubule crossovers (Dixit and Cyr, 2004). Several computational modeling studies have since shown that these types of interactions between surface-bound dynamical microtubules can indeed explain spontaneous coalignment of microtubules (Allard et al., 2010; Eren et al., 2010; Hawkins et al., 2010; Tindemans et al., 2010).The question of how the orientation of the cortical array is established with respect to the cell axis is less well understood. One possibility is that microtubules are selectively destabilized with respect to cellular coordinates (Ehrhardt and Shaw, 2006). Indeed, recent results from biological observations and modeling suggest that catastrophic collisions induced at the edges between cell faces or heighted catastrophe rates in cell caps could be sufficient to selectively favor microtubules in certain orientation and hence determine the final orientation of the array (Allard et al., 2010; Eren et al., 2010; Ambrose et al., 2011; Dhonukshe et al., 2012).To date, all models of cortical array assembly assume random initial conditions. However, experimental work by Wasteneys and Williamson (1989a, 1989b) in Nitella tasmanica showed that, during array reassembly after drug-induced disruption, microtubules were initially transverse. This was followed by a less ordered phase and later by the acquisition of the final transverse order. A nonrandom initial ordering was also observed in tobacco (Nicotiana tabacum) Bright Yellow-2 (BY-2) cells by Kumagai et al. (2001), who concluded that the process of transverse array establishment starts with longitudinal order but did not provide quantitative data for the process of array assembly. The initial conditions for the cortical microtubule array formation are important to consider, as they may strongly influence the speed at which order is established and could even prevent it from being established over a biologically relevant time scale.In this study, we used live-cell imaging to follow and record the whole transition from the cortical microtubule-free state to the final transverse array and used digital tracking algorithms to quantify the microtubule order. Nucleation stands out as a central process to characterize during array initiation. Lacking a central body to organize microtubule nucleations, the higher plant cell has dispersed nucleation complexes (Wasteneys and Williamson, 1989a, 1989b; Chan et al., 2003; Shaw et al., 2003; Murata et al., 2005; Pastuglia et al., 2006; Nakamura et al., 2010). Therefore, we performed high time resolution observations to quantify nucleation complex recruitment, nucleation rates, and microtubule nucleation angles. We found evidence for a highly nonrandom initial ordering state that features diagonal microtubule orientation and an atypical microtubule initiation mechanism. Simulation analysis indicates that these atypical nucleations have the potential to accelerate the recovery of cortical array density.  相似文献   

13.
Helicobacter pylori CagA plays a key role in gastric carcinogenesis. Upon delivery into gastric epithelial cells, CagA binds and deregulates SHP-2 phosphatase, a bona fide oncoprotein, thereby causing sustained ERK activation and impaired focal adhesions. CagA also binds and inhibits PAR1b/MARK2, one of the four members of the PAR1 family of kinases, to elicit epithelial polarity defect. In nonpolarized gastric epithelial cells, CagA induces the hummingbird phenotype, an extremely elongated cell shape characterized by a rear retraction defect. This morphological change is dependent on CagA-deregulated SHP-2 and is thus thought to reflect the oncogenic potential of CagA. In this study, we investigated the role of the PAR1 family of kinases in the hummingbird phenotype. We found that CagA binds not only PAR1b but also other PAR1 isoforms, with order of strength as follows: PAR1b > PAR1d ≥ PAR1a > PAR1c. Binding of CagA with PAR1 isoforms inhibits the kinase activity. This abolishes the ability of PAR1 to destabilize microtubules and thereby promotes disassembly of focal adhesions, which contributes to the hummingbird phenotype. Consistently, PAR1 knockdown potentiates induction of the hummingbird phenotype by CagA. The morphogenetic activity of CagA was also found to be augmented through inhibition of non-muscle myosin II. Because myosin II is functionally associated with PAR1, perturbation of PAR1-regulated myosin II by CagA may underlie the defect of rear retraction in the hummingbird phenotype. Our findings reveal that CagA systemically inhibits PAR1 family kinases and indicate that malfunctioning of microtubules and myosin II by CagA-mediated PAR1 inhibition cooperates with deregulated SHP-2 in the morphogenetic activity of CagA.Infection with Helicobacter pylori strains bearing cagA (cytotoxin-associated gene A)-positive strains is the strongest risk factor for the development of gastric carcinoma, the second leading cause of cancer-related death worldwide (13). The cagA gene is located within a 40-kb DNA fragment, termed the cag pathogenicity island, which is specifically present in the genome of cagA-positive H. pylori strains (46). In addition to cagA, there are ∼30 genes in the cag pathogenicity island, many of which encode a bacterial type IV secretion system that delivers the cagA-encoded CagA protein into gastric epithelial cells (710). Upon delivery into gastric epithelial cells, CagA is localized to the plasma membrane, where it undergoes tyrosine phosphorylation at the C-terminal Glu-Pro-Ile-Tyr-Ala motifs by Src family kinases or c-Abl kinase (1114). The C-terminal Glu-Pro-Ile-Tyr-Ala-containing region of CagA is noted for the structural diversity among distinct H. pylori isolates. Oncogenic potential of CagA has recently been confirmed by a study showing that systemic expression of CagA in mice induces gastrointestinal and hematological malignancies (15).When expressed in gastric epithelial cells, CagA induces morphological transformation termed the hummingbird phenotype, which is characterized by the development of one or two long and thin protrusions resembling the beak of the hummingbird. It has been thought that the hummingbird phenotype is related to the oncogenic action of CagA (7, 1619). Pathophysiological relevance for the hummingbird phenotype in gastric carcinogenesis has recently been provided by the observation that infection with H. pylori carrying CagA with greater ability to induce the hummingbird phenotype is more closely associated with gastric carcinoma (2023). Elevated motility of hummingbird cells (cells showing the hummingbird phenotype) may also contribute to invasion and metastasis of gastric carcinoma.In host cells, CagA interacts with the SHP-2 phosphatase, C-terminal Src kinase, and Crk adaptor in a tyrosine phosphorylation-dependent manner (16, 24, 25) and also associates with Grb2 adaptor and c-Met in a phosphorylation-independent manner (26, 27). Among these CagA targets, much attention has been focused on SHP-2 because the phosphatase has been recognized as a bona fide oncoprotein, gain-of-function mutations of which are found in various human malignancies (17, 18, 28). Stable interaction of CagA with SHP-2 requires CagA dimerization, which is mediated by a 16-amino acid CagA-multimerization (CM)2 sequence present in the C-terminal region of CagA (29). Upon complex formation, CagA aberrantly activates SHP-2 and thereby elicits sustained ERK MAP kinase activation that promotes mitogenesis (30). Also, CagA-activated SHP-2 dephosphorylates and inhibits focal adhesion kinase (FAK), causing impaired focal adhesions. It has been shown previously that both aberrant ERK activation and FAK inhibition by CagA-deregulated SHP-2 are involved in induction of the hummingbird phenotype (31).Partitioning-defective 1 (PAR1)/microtubule affinity-regulating kinase (MARK) is an evolutionally conserved serine/threonine kinase originally isolated in C. elegans (3234). Mammalian cells possess four structurally related PAR1 isoforms, PAR1a/MARK3, PAR1b/MARK2, PAR1c/MARK1, and PAR1d/MARK4 (3537). Among these, PAR1a, PAR1b, and PAR1c are expressed in a variety of cells, whereas PAR1d is predominantly expressed in neural cells (35, 37). These PAR1 isoforms phosphorylate microtubule-associated proteins (MAPs) and thereby destabilize microtubules (35, 38), allowing asymmetric distribution of molecules that are involved in the establishment and maintenance of cell polarity.In polarized epithelial cells, CagA disrupts the tight junctions and causes loss of apical-basolateral polarity (39, 40). This CagA activity involves the interaction of CagA with PAR1b/MARK2 (19, 41). CagA directly binds to the kinase domain of PAR1b in a tyrosine phosphorylation-independent manner and inhibits the kinase activity. Notably, CagA binds to PAR1b via the CM sequence (19). Because PAR1b is present as a dimer in cells (42), CagA may passively homodimerize upon complex formation with the PAR1 dimer via the CM sequence, and this PAR1-directed CagA dimer would form a stable complex with SHP-2 through its two SH2 domains.Because of the critical role of CagA in gastric carcinogenesis (7, 1619), it is important to elucidate the molecular basis underlying the morphogenetic activity of CagA. In this study, we investigated the role of PAR1 isoforms in induction of the hummingbird phenotype by CagA, and we obtained evidence that CagA-mediated inhibition of PAR1 kinases contributes to the development of the morphological change by perturbing microtubules and non-muscle myosin II.  相似文献   

14.
15.
Degradation of the M phase cyclins triggers the exit from M phase. Cdc14 is the major phosphatase required for the exit from the M phase. One of the functions of Cdc14 is to dephosphorylate and activate the Cdh1/APC/C complex, resulting in the degradation of the M phase cyclins. However, other crucial targets of Cdc14 for mitosis and cytokinesis remain to be elucidated. Here we systematically analyzed the positions of dephosphorylation sites for Cdc14 in the budding yeast Saccharomyces cerevisiae. Quantitative mass spectrometry identified a total of 835 dephosphorylation sites on 455 potential Cdc14 substrates in vivo. We validated two events, and through functional studies we discovered that Cdc14-mediated dephosphorylation of Smc4 and Bud3 is essential for proper mitosis and cytokinesis, respectively. These results provide insight into the Cdc14-mediated pathways for exiting the M phase.All cells proliferate following a fixed, highly coordinated cycle. Mitosis especially requires elaborate coordination for proper chromosome segregation, mitotic spindle disassembly, and cytokinesis. Much of this activity is facilitated by numerous, diverse phosphorylation and dephosphorylation signals that orchestrate the precise progression of M phase.Prior to mitosis, sister chromatids resulting from DNA replication during S phase are held together by the cohesion complex. Then, during prophase, chromosomes are condensed by the condensin (Smc2/4) complex (1) and microtubules are remodeled to form the mitotic spindle (2). Subsequently, in metaphase, the microtubules of the spindle apparatus attach to the chromosome kinetochores (3) and dissolution of the sister chromatids is triggered by the separase-mediated cleavage of cohesin (4, 5). Finally, Cdc14, Cdh1, and APC/C work together in telophase to degrade the M phase cyclins (6), promote decondensation of chromosomes (7), and finish cytokinesis (8, 9).Cdc14, a dual-specificity phosphatase that removes the phosphate group on both phosphotyrosine and phosphoserine/threonine residues (10), is required for mitosis (11, 12). Specifically, Cdc14 function is essential in late M phase: cells carrying a defective mutation arrest in telophase (13), whereas overexpression of Cdc14 results in G1 arrest (12). Cdc14 triggers mitotic cyclin-dependent kinase (CDK)1 inactivation, enabling cells to exit mitosis through dephosphorylation and activation of the inhibitors of CDKs. At interphase, Cdc14 is a subunit of the mitotic exit network (1417), which usually localizes to the nucleolus. However, the Cdc14 early anaphase release network initiates the release of Cdc14 from its inhibitor, Net1/Cfi1 (18), and the mitotic exit network promotes further release of Cdc14 from its inhibitor, allowing it to spread into the nucleus and cytoplasm, where it dephosphorylates its major targets (8, 9), leading to exit from mitosis. In addition to this essential role in late M phrase, Cdc14 substrates have also been identified in other stages of the cell cycle (19).Cdc14 putatively regulates 27 proteins (1922). Some studies have documented the substrates of Cdc14 via in vitro phosphatase assay, whereas others have provided in vivo evidence. However, dephosphorylation sites have been identified for only five of the target proteins (17, 2225), suggesting that spurious relationships cannot be ruled out. Also, experiments have not been carried out to demonstrate whether these modifications entail direct or indirect regulation. Therefore, our understanding of Cdc14 function and regulation during mitosis in metazoans is incomplete. Conceivably, Cdc14 may regulate many more substrates involved in aspects of chromosome condensation and cytokinesis. To examine this possibility we performed a systematic phosphoproteomic screen to identify new in vivo pathways regulated by Cdc14. Using this approach, we identified both known and potentially novel substrates of Cdc14, as well as their dephosphorylation sites. Many potentially novel substrates are physically associated with Cdc14 in public databases. We also provide biochemical evidence for direct dephosphorylation of the substrates, characterize the specificity of dephosphorylation in two substrates, Smc4 and Bud3, and further study their regulation and critical role in mitosis and cytokinesis.  相似文献   

16.
17.
The role of the herpes simplex virus type 1 tegument protein VP22 during infection is as yet undefined. We have previously shown that VP22 has the unusual property of efficient intercellular transport, such that the protein spreads from single expressing cells into large numbers of surrounding cells. We also noted that in cells expressing VP22 by transient transfection, the protein localizes in a distinctive cytoplasmic filamentous pattern. Here we show that this pattern represents a colocalization between VP22 and cellular microtubules. Moreover, we show that VP22 reorganizes microtubules into thick bundles which are easily distinguishable from nonbundled microtubules. These bundles are highly resistant to microtubule-depolymerizing agents such as nocodazole and incubation at 4°C, suggesting that VP22 has the capacity to stabilize the microtubule network. In addition, we show that the microtubules contained in these bundles are modified by acetylation, a marker for microtubule stability. Analysis of infected cells by both immunofluorescence and measurement of microtubule acetylation further showed that colocalization between VP22 and microtubules, and induction of microtubule acetylation, also occurs during infection. Taken together, these results suggest that VP22 exhibits the properties of a classical microtubule-associated protein (MAP) during both transfection and infection. This is the first demonstration of a MAP encoded by an animal virus.

The eukaryotic cytoskeleton, which comprises actin microfilaments, intermediate filaments (IFs), and microtubules (MTs), performs a broad range of complex activities within the cell. These include various aspects of cell motility (2, 3), the determination of cell shape and internal architecture (17, 32), and vesicle trafficking and chromosome movement during mitosis (18, 25, 29). Furthermore, the individual components of the cytoskeleton are interlinked to form a dynamic network accessing every area of the cytoplasm (41) and the plasma membrane (10, 39), providing a framework which coordinates multiple cellular processes. The involvement in so many cellular activities is likely to make the cytoskeleton a primary target for exploitation during virus infection of host cells. Surprisingly, however, there is relatively little detailed information on virus interactions with the host cytoskeleton, and it is only recently that data suggesting that viruses may utilize the positioning and dynamics of the cytoskeletal network to their own advantage have begun to emerge.The majority of virus-induced cytoskeletal alterations documented to date involve the overall disruption of one or more elements of the cytoskeleton. For example, retroviruses and poliovirus encode proteases which induce the cleavage of cytoskeleton-associated proteins, thereby broadly increasing the dynamics of the cytoskeleton, resulting in disruption of the cell structure as infection progresses, and the appearance of well-characterized cytopathic effects (20, 43). A more specific disruption of the cytoskeleton occurs during infection by the rhabdovirus vesicular stomatitis virus, where the direct interaction of the virus matrix protein with tubulin results in the inhibition of MT assembly (33). Human immunodeficiency virus and papillomaviruses, on the other hand, encode activities which induce the collapse of the IF network, a property which may promote virus release from the cell (13, 23).By contrast, examples of virus activities which induce cytoskeletal polymerization and/or stabilization are much rarer. One example is the baculovirus Autographa californica nuclear polyhedrosis virus, which has been shown to induce the appearance of thick actin cables between the plasma membrane and the nucleus at early times after infection (8) and to induce actin filaments in the nucleus at late times (7). These features have been proposed to be involved in virus transport from the cell surface to the nucleus and nucleocapsid morphogenesis, respectively. However, the best-characterized viral exploitation of the host cell cytoskeleton is that of vaccinia virus, which has been shown to induce actin polymerization directly behind its virus particle as a means of propelling the virus through the cell (11, 12). The virus protein(s) responsible for this activity has not yet been identified, but it has been shown that disruption of the actin cytoskeleton in infected cells inhibits virus release, indicating that actin is essential to the virus replicative cycle (35).The herpes simplex virus type 1 (HSV-1) structural protein VP22, a component of the viral tegument, has an as yet undefined role in virus replication. However, we have recently shown that VP22 has the unusual property of intercellular transport when it is expressed during both infection and transient transfection (14). Moreover, we demonstrated that such VP22 transport occurs via a mechanism potentially involving actin microfilaments, suggesting that VP22 exhibits a cytoskeletal interaction. In this report, we demonstrate that VP22 interacts with another component of the cellular cytoskeleton, the MT network. We show that VP22 colocalizes with MTs in both transfected and infected cells and induces the appearance of thick MT bundles. Furthermore, we show that these VP22-induced MT bundles are highly stabilized in comparison to normal MTs and are resistant to both drug and cold treatment. As a consequence of VP22-induced stabilization, MTs are extensively modified by acetylation, a property also demonstrated in infected cells. Taken together, these results suggest that VP22 exhibits the properties of a classical cellular MT-associated protein (MAP) with powerful MT-stabilizing properties and represents the first demonstration of a MAP encoded by an animal virus.  相似文献   

18.
19.
In Alzheimer disease (AD) and frontotemporal dementia the microtubule-associated protein Tau becomes progressively hyperphosphorylated, eventually forming aggregates. However, how Tau dysfunction is associated with functional impairment is only partly understood, especially at early stages when Tau is mislocalized but has not yet formed aggregates. Impaired axonal transport has been proposed as a potential pathomechanism, based on cellular Tau models and Tau transgenic mice. We recently reported K369I mutant Tau transgenic K3 mice with axonal transport defects that suggested a cargo-selective impairment of kinesin-driven anterograde transport by Tau. Here, we show that kinesin motor complex formation is disturbed in the K3 mice. We show that under pathological conditions hyperphosphorylated Tau interacts with c-Jun N-terminal kinase- interacting protein 1 (JIP1), which is associated with the kinesin motor protein complex. As a result, transport of JIP1 into the axon is impaired, causing JIP1 to accumulate in the cell body. Because we found trapping of JIP1 and a pathological Tau/JIP1 interaction also in AD brain, this may have pathomechanistic implications in diseases with a Tau pathology. This is supported by JIP1 sequestration in the cell body of Tau-transfected primary neuronal cultures. The pathological Tau/JIP1 interaction requires phosphorylation of Tau, and Tau competes with the physiological binding of JIP1 to kinesin light chain. Because JIP1 is involved in regulating cargo binding to kinesin motors, our findings may, at least in part, explain how hyperphosphorylated Tau mediates impaired axonal transport in AD and frontotemporal dementia.The microtubule-associated protein Tau is predominantly found in the axonal compartment of neurons, where it binds to microtubules (1). In human brain, six isoforms of Tau are expressed, due to alternative splicing of exons 2, 3 and 10 (2). Tau consists of an amino-terminal projection domain followed by 3 or 4 microtubule binding repeats (3R or 4R), due to splicing of exon 10, and a carboxyl-terminal tail region. In the AD3 and FTD brain, Tau forms filamentous inclusions (3). They are found in nerve cell bodies and apical dendrites as neurofibrillary tangles (NFTs), in distal dendrites as neuropil threads, and in the abnormal neurites that are associated with some amyloid plaques (neuritic plaques) (3). Hyperphosphorylation of Tau is thought to be an initiating step (4), as it detaches Tau from microtubules and makes it prone to form aggregates (1, 5). Whereas in AD no mutations have been identified in the MAPT gene encoding Tau, so far 42 intronic and exonic mutations have been found in familial forms of FTD (6). Their identification assisted in the generation of transgenic mouse models that reproduce NFT formation and memory impairment (7).The models were also instrumental in testing hypotheses that had been brought forward to link Tau pathology to functional impairment (810). In particular, defects in axonal transport have been implicated in neurodegenerative disorders (11, 12). Tau binding to microtubules affects axonal transport (13), and in cell culture overexpression of Tau was shown to lead to impaired transport of mitochondria and vesicles (14, 15). Axonal transport defects have also been reproduced in wild-type Tau transgenic mice (16) and in K369I mutant Tau K3 mice (17), whereas Tau expression failed to inhibit axonal transport in other systems (18, 19). This apparent discrepancy may depend on the type of cargos analyzed and, specifically, the experimental paradigm, e.g. using phosphorylated (16, 17, 20) versus non-phosphorylated Tau (18).To dissect Tau-mediated axonal transport defects at a molecular level, we used K3 mice that overexpress human Tau carrying the pathogenic FTD K369I mutation (17). We observed a pronounced hyperphosphorylation of transgenic Tau in many brain areas. Clinically, the mice present with an early onset motor phenotype that is, at least in part, caused by impairment of axonal transport in neurons of the substantia nigra. Interestingly, only selected aspects of anterograde axonal transport were impaired, in particular those of kinesin-I motor complex-driven vesicles and mitochondria. Our data suggest a selective impairment of axonal transport rather than a generalized, non-selective blockage of microtubules that has been established in cell culture systems, which fail to phosphorylate Tau at the high levels that are found in vivo even under physiological conditions. More importantly, in AD and FTD Tau is even more phosphorylated, i.e. hyperphosphorylated at physiological sites and de novo at pathological sites, preventing it from binding to microtubules (1).Based on our findings of an impaired kinesin-I-driven axonal transport in the K3 mice, we speculated that hyperphosphorylated Tau may impair anterograde transport by interfering directly with components of the kinesin-I motor complex rather than disrupting the binding of the kinesin heavy chain (see below) to microtubules. Axonal transport along microtubules is mediated by members of the kinesin superfamily (KIF) of motor proteins (2123). The KIFs typically consist of an ATPase domain that interacts with microtubules and drives movement and a domain that links to cargos, either directly or indirectly, as in the case of KIF5, by assembling with the kinesin light chain (KLC) to form the kinesin-I (KIF5/KLC) motor complex (24). In addition, increasing evidence suggests that scaffolding proteins mediate and regulate the binding of cargos to KIFs (21, 2527). These include the scaffold protein JNK-interacting protein (JIP) that is involved in the linkage of cargos to the kinesin-I motor complex via KLC (25, 2833).Here, by using the K3 mouse model, we identified a novel interaction of Tau and JIP in neurons that causes a trapping of JNK interacting protein 1 (JIP1) in the cell body of K3 mice, cell culture systems, and human AD brain. We found that the pathological interaction of hyperphosphorylated Tau and JIP1 competes with the physiological binding of JIP1 to KLC.  相似文献   

20.
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