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.
F-box and WD repeat domain-containing 7 (Fbxw7/hAgo/hCdc4/Fbw7) is a p53-dependent tumor suppressor and leads to ubiquitination-mediated suppression of several oncoproteins including c-Myc, cyclin E, Notch, c-Jun and others. Our previous study has indicated that low expression of Fbxw7 was negatively correlated with c-Myc, cyclin E and mutant-p53 in hepatocellular carcinoma (HCC) tissues. But the role and mechanisms of Fbxw7 in HCC are still unknown. Here, we investigated the function of Fbxw7 in HCC cell lines and the anti-tumor activity of recombinant human adenovirus-p53 injection (rAd-p53, Gendicine) administration in vitro and in vivo. Fbxw7-specific siRNA enhanced expression of c-Myc and cyclin E proteins and increased proliferation in cell culture. rAd-p53 inhibited tumor cell growth with Fbxw7 upregulation and c-Myc and cyclin E downregulation in vitro and a murine HCC model. This effect could be partially reverted using Fbxw7-specific siRNA. Here, we suggest that the activation of Fbxw7 by adenoviral delivery of p53 leads to increased proteasomal degradation of c-Myc and cyclin E enabling growth arrest and apoptosis. Addressing this pathway, we identified that rAd-p53 could be a potential therapeutic agent for HCC.  相似文献   

2.
Growth factor-dependent accumulation of the cyclin D1 proto-oncogene is balanced by its rapid phosphorylation-dependent proteolysis. Degradation is triggered by threonine 286 phosphorylation, which promotes its ubiquitination by an unknown E3 ligase. We demonstrate that Thr286-phosphorylated cyclin D1 is recognized by a Skp1-Cul1-F box (SCF) ubiquitin ligase where FBX4 and alphaB crystallin govern substrate specificity. Overexpression of FBX4 and alphaB crystallin triggered cyclin D1 ubiquitination and increased cyclin D1 turnover. Impairment of SCF(FBX4-alphaB crystallin) function attenuated cyclin D1 ubiquitination, promoting cyclin D1 overexpression and accelerated cell-cycle progression. Purified SCF(FBX4-alphaB crystallin) catalyzed polyubiquitination of cyclin D1 in vitro. Consistent with a putative role for a cyclin D1 E3 ligase in tumorigenesis, FBX4 and alphaB crystallin expression was reduced in tumor-derived cell lines and a subset of primary human cancers that overexpress cyclin D1. We conclude that SCF(FBX4-alphaB crystallin) is an E3 ubiquitin ligase that promotes ubiquitin-dependent degradation of Thr286-phosphorylated cyclin D1.  相似文献   

3.
4.
Microtubule depolymerases of the kinesin‐13 family play important roles in various cellular processes and are frequently overexpressed in different cancer types. Despite the importance of their correct abundance, remarkably little is known about how their levels are regulated in cells. Using comprehensive screening on protein microarrays, we identified 161 candidate substrates of the multi‐subunit ubiquitin E3 ligase SCFFbxw5, including the kinesin‐13 member Kif2c/MCAK. In vitro reconstitution assays demonstrate that MCAK and its closely related orthologs Kif2a and Kif2b become efficiently polyubiquitylated by neddylated SCFFbxw5 and Cdc34, without requiring preceding modifications. In cells, SCFFbxw5 targets MCAK for proteasomal degradation predominantly during G2. While this seems largely dispensable for mitotic progression, loss of Fbxw5 leads to increased MCAK levels at basal bodies and impairs ciliogenesis in the following G1/G0, which can be rescued by concomitant knockdown of MCAK, Kif2a or Kif2b. We thus propose a novel regulatory event of ciliogenesis that begins already within the G2 phase of the preceding cell cycle.  相似文献   

5.
Cyclin E is required for S phase entry. The subsequent ubiquitin-dependent degradation of cyclin E contributes to an orderly progression of the S phase. It has been shown that phosphorylation of threonine 380 (Thr380) in cyclin E provides a signal for its ubiquitin-dependent proteolysis. We report that SKP2, an F-box protein and a substrate-targeting component of the SCF(SKP2) ubiquitin E3 ligase complex, mediates cyclin E degradation. In vitro, SKP2 specifically interacted with the cyclin E peptide containing the phosphorylated-Thr380 but not with a cognate nonphosphorylated peptide. In vivo, expression of SKP2 induced cyclin E polyubiquitination and degradation. Conversion of Thr380 into nonphosphorylatable amino acids caused significant resistance of cyclin E to SKP2. The presence of the CDK inhibitor p27(Kip1) also prevented the SKP2-dependent degradation of cyclin E. Our findings suggest that SKP2 regulates cyclin E stability, thus contributing to the control of S phase progression.  相似文献   

6.
Maintaining accurate progression through the cell cycle requires the proper temporal expression and regulation of cyclins. The mammalian D-type cyclins promote G1-S transition. D1 cyclin protein stability is regulated through its ubiquitylation and resulting proteolysis catalyzed by the SCF E3 ubiquitin ligase complex containing the F-box protein, Fbx4. SCF E3-ligase-dependent ubiquitylation of D1 is trigged by an increase in the phosphorylation status of the cyclin. As inhibition of ubiquitin-dependent D1 degradation is seen in many human cancers, we set out to uncover how D-type cyclin phosphorylation is regulated. Here we show that in S. cerevisiae, a heterotrimeric protein phosphatase 2A (PP2ACdc55) containing the mammalian PPP2R2/PR55 B subunit ortholog Cdc55 regulates the stability of the G1 cyclin Cln2 by directly regulating its phosphorylation state. Cells lacking Cdc55 contain drastically reduced Cln2 levels caused by degradation due to cdk-dependent hyperphosphorylation, as a Cln2 mutant unable to be phosphorylated by the yeast cdk Cdc28 is highly stable in cdc55-null cells. Moreover, cdc55-null cells become inviable when the SCFGrr1 activity known to regulate Cln2 levels is eliminated or when Cln2 is overexpressed, indicating a critical relationship between SCF and PP2A functions in regulating cell cycle progression through modulation of G1-S cyclin degradation/stability. In sum, our results indicate that PP2A is absolutely required to maintain G1-S cyclin levels through modulating their phosphorylation status, an event necessary to properly transit through the cell cycle.  相似文献   

7.
Aneuploidy is a common feature of human solid tumors and is often associated with poor prognosis. There is growing evidence that oncogenic signaling pathways, which are universally dysregulated in cancer, contribute to the promotion of aneuploidy. However, the mechanisms connecting signaling pathways to the execution of mitosis and cytokinesis are not well understood. Here, we show that hyperactivation of the ERK1/2 MAP kinase pathway in epithelial cells impairs cytokinesis, leading to polyploidization and aneuploidy. Mechanistically, deregulated ERK1/2 signaling specifically downregulates expression of the F-box protein Fbxw7β, a substrate-binding subunit of the SCFFbxw7 ubiquitin ligase, resulting in the accumulation of the mitotic kinase Aurora A. Reduction of Aurora A levels by RNA interference or pharmacological inhibition of MEK1/2 reverts the defect in cytokinesis and decreases the frequency of abnormal cell divisions induced by oncogenic H-RasV12. Reciprocally, overexpression of Aurora A or silencing of Fbxw7β phenocopies the effect of H-RasV12 on cell division. In vivo, conditional activation of MEK2 in the mouse intestine lowers Fbxw7β expression, resulting in the accumulation of cells with enlarged nuclei. We propose that the ERK1/2/ Fbxw7β/Aurora A axis identified in this study contributes to genomic instability and tumor progression.  相似文献   

8.
Mitotic progression is regulated by ubiquitin E3 ligase complexes to carefully orchestrate eukaryotic cell division. Here, we show that a relatively new E3 ligase component belonging to the SCF (Skip-Cullin1-F-box protein) E3 ligase family, SCFFBXL2, impairs cell proliferation by mediating cyclin D3 polyubiquitination and degradation. Both cyclin D3 and FBXL2 colocalize within the centrosome. FBXL2 overexpression led to G2/M-phase arrest in transformed epithelia, resulting in the appearance of supernumerary centrosomes, tetraploidy and nuclei where condensed chromosomes are arranged on circular monopolar spindles typical of mitotic arrest. RNAi-mediated knockdown of cyclin D3 recapitulated effects of SCFFBXL2 expression. SCFFBXL2 impaired the ability of cyclin D3 to associate with centrosomal assembly proteins [Aurora A, polo-like kinase 4 (Plk4), CDK11]. Thus, these results suggest a role for SCFFBXL2 in regulating the fidelity of cellular division.  相似文献   

9.
Mitotic progression is regulated by ubiquitin E3 ligase complexes to carefully orchestrate eukaryotic cell division. Here, we show that a relatively new E3 ligase component belonging to the SCF (Skip-Cullin1-F-box protein) E3 ligase family, SCFFBXL2, impairs cell proliferation by mediating cyclin D3 polyubiquitination and degradation. Both cyclin D3 and FBXL2 colocalize within the centrosome. FBXL2 overexpression led to G2/M-phase arrest in transformed epithelia, resulting in the appearance of supernumerary centrosomes, tetraploidy and nuclei where condensed chromosomes are arranged on circular monopolar spindles typical of mitotic arrest. RNAi-mediated knockdown of cyclin D3 recapitulated effects of SCFFBXL2 expression. SCFFBXL2 impaired the ability of cyclin D3 to associate with centrosomal assembly proteins [Aurora A, polo-like kinase 4 (Plk4), CDK11]. Thus, these results suggest a role for SCFFBXL2 in regulating the fidelity of cellular division.Key words: F-box protein, centrosome, mitosis, cyclin D3, Aurora A  相似文献   

10.
Fbw7 and Cdh1 are substrate-recognition subunits of the SCF- and APC-type E3 ubiquitin ligases, respectively. There is emerging evidence suggesting that both Fbw7 and Cdh1 function as tumor suppressors by targeting oncoproteins for destruction. Loss of Fbw7, but not Cdh1, is frequently observed in various human tumors. However, it remains largely unknown how Fbw7 mechanistically functions as a tumor suppressor and whether there is a signaling crosstalk between Fbw7 and Cdh1. Here, we report that Fbw7-deficient cells not only display elevated expression levels of SCFFbw7 substrates, including cyclin E, but also have increased expression of various APCCdh1 substrates. We further defined cyclin E as the critical signaling link by which Fbw7 governs APCCdh1 activity, as depletion of cyclin E in Fbw7-deficient cells results in decreased expression of APCCdh1 substrates to levels comparable to those in wild-type (WT) cells. Conversely, ectopic expression of cyclin E recapitulates the aberrant APCCdh1 substrate expression observed in Fbw7-deficient cells. More importantly, 4A-Cdh1 that is resistant to Cdk2/cyclin E-mediated phosphorylation, but not WT-Cdh1, reversed the elevated expression of various APCCdh1 substrates in Fbw7-deficient cells. Overexpression of 4A-Cdh1 also resulted in retarded cell growth and decreased anchorage-independent colony formation. Altogether, we have identified a novel regulatory mechanism by which Fbw7 governs Cdh1 activity in a cyclin E-dependent manner. As a result, loss of Fbw7 can lead to aberrant increase in the expression of both SCFFbw7 and APCCdh1 substrates. Our study provides a better understanding of the tumor suppressor function of Fbw7, and suggests that Cdk2/cyclin E inhibitors could serve as effective therapeutic agents for treating Fbw7-deficient tumors.  相似文献   

11.
Mitogenic induction of cyclin D1, the allosteric regulator of CDK4/6, is a key regulatory event contributing to G1 phase progression. Following the G1/S transition, cyclin D1 activation is antagonized by GSK3β-dependent threonine-286 (Thr-286) phosphorylation, triggering nuclear export and subsequent cytoplasmic degradation mediated by the SCFFbx4-αBcrystallin E3 ubiquitin ligase. Although cyclin D1 overexpression occurs in numerous malignancies, overexpression of cyclin D1 alone is insufficient to drive transformation. In contrast, cyclin D1 mutants refractory to phosphorylation-dependent nuclear export and degradation are acutely transforming. This raises the question of whether overexpression of cyclin D1 is a significant contributor to tumorigenesis or an effect of neoplastic transformation. Significantly, recent work strongly supports a model wherein nuclear accumulation of cyclin D1-dependent kinase during S-phase is a critical event with regard to transformation. The identification of mutations within SCFFbx4-αBcrystallin ligase in primary tumors provides mechanistic insight into cyclin D1 accumulation in human cancer. Furthermore, analysis of mouse models expressing cyclin D1 mutants refractory to degradation indicate that nuclear cyclin D1/CDK4 kinase triggers DNA re-replication and genomic instability. Collectively, these new findings provide a mechanism whereby aberrations in post-translational regulation of cyclin D1 establish a cellular environment conducive to mutations that favor neoplastic growth.  相似文献   

12.
Tazarotene-induced gene 1 (TIG1) is considered to be a tumor suppressor gene that is highly expressed in normal or well-differentiated colon tissues, while downregulation of TIG1 expression occurs in poorly differentiated colorectal cancer (CRC) tissues. However, it is still unclear how TIG1 regulates the tumorigenesis of CRC. Polo-like kinases (Plks) are believed to play an important role in regulating the cell cycle. The performance of PLK2 in CRC is negatively correlated with the differentiation status of CRC tissues. Here, we found that PLK2 can induce the growth of CRC cells and that TIG1 can prevent PLK2 from promoting the proliferation of CRC cells. We also found that the expression of PLK2 in CRC cells was associated with low levels of Fbxw7 protein and increased expression of cyclin E1. When TIG1 was coexpressed with PLK2, the changes in Fbxw7/cyclin E1 levels induced by PLK2 were reversed. In contrast, silencing TIG1 promoted the proliferation of CRC, and when PLK2 was also silenced, the proliferation of CRC cells induced by TIG1 silencing was significantly inhibited. The above research results suggest that TIG1 can regulate the tumorigenesis of CRC by regulating the activity of PLK2.  相似文献   

13.
Millman SE  Pagano M 《EMBO reports》2011,12(5):384-385
Evidence for the destruction of the anti-apoptotic protein MCL1 during prolonged mitotic arrest comes from three papers, one in The EMBO Journal and two in Nature, thus shedding light on the mechanism of apoptosis induction under these conditions.EMBO Rep (2011) advance online publication. doi:10.1038/nature09732EMBO Rep (2011) advance online publication. doi:10.1038/nature09779EMBO Rep (2011) advance online publication. doi:10.1038/emboj.2010.112During mitosis, eukaryotic cells have to properly align their chromosomes. Only after the kinetochore of each chromosome is attached to a polar microtubule can a cell satisfy the ‘spindle assembly checkpoint'', which prevents the mis-segregation of chromosomes. Failure to correctly segregate chromosomes before cell division might contribute to chromosome instability and tumorigenesis. To counteract chromosome aberrations, the cell initiates the apoptotic programme. It has become clear through the use of microtubule-poisoning, chemotherapeutic agents—such as paclitaxel and vincristine—that prolonged activation of the spindle checkpoint can induce mitotic arrest and, subsequently, programmed cell death. The molecular mechanisms responsible for initiating apoptosis during mitotic arrest have remained poorly defined. Two recent papers in Nature (Inuzuka et al, 2011; Wertz et al, 2011) and a report published by the Clarke group last year in The EMBO Journal (Harley et al, 2010) highlight the destruction of MCL1 during prolonged mitotic arrest and shed light on the mechanisms of apoptosis induction.Myeloid cell leukaemia 1 (MCL1) is an anti-apoptotic member of the B-cell lymphoma 2 (BCL2) family of proteins. MCL1, like BCL2 and BCLxL, prevents the downstream activation of BAX and BAK, which are responsible for mitochondrial outer-membrane permeabilization, initiation of the caspase cascade and induction of apoptosis (Youle & Strasser, 2008). Ubiquitination and proteolysis of MCL1 have been reported, but a mechanism for MCL1 degradation following spindle checkpoint activation remains unknown. Now, the studies referenced above suggest that degradation of MCL1 during prolonged mitotic arrest is essential for the induction of apoptosis. Given its prominent role in driving the cell cycle, as well as in safeguarding the fidelity of this process, it is not surprising that the ubiquitin-proteasome system (UPS) has a key role in dictating the activation of the intrinsic apoptotic pathway in cells arrested in mitosis. However, it is surprising that two E3 ubiquitin ligase complexes simultaneously facilitate this degradation event.Harley and colleagues describe the regulation of MCL1 by APC/CCdc20(anaphase-promoting complex/cyclosome and its activator Cdc20). This multi-subunit RING E3 ubiquitin ligase is active in mitosis, and ubiquitinates substrates such as securin and cyclin B, thereby allowing progression into anaphase. In their report, Harley and co-workers (2010) demonstrate a Cdk1/cyclin-B-mediated, site-specific phosphorylation (Thr 92 in humans) of MCL1 upon mitotic arrest, followed by its proteolytic destruction by APC/CCdc20. Thus, like the sand of an hourglass flipped at each entry into mitosis, the level of MCL1 steadily decreases. If time ‘runs out'' due to a prolonged mitotic arrest (that is, if MCL1 is completely destroyed), then apoptosis is initiated (Fig 1A). Both phosphorylation at Thr 92 and the presence of a conserved destruction or ‘D''-box motif (a characteristic of APC/C substrates) are required for MCL1 proteolysis, although the precise role of phosphorylation in promoting degradation remains unclear.Open in a separate windowFigure 1Two models for proteolytic destruction of MCL1 during mitotic arrest. (A) Schematic illustration of the effects of APC/CCdc20 and Cdk1/cyclin B on the degradation of MCL1 during prolonged arrest in mitosis. (B) Schematic illustration of the effects of SCFFbw7, JNK/p38/CKII and Cdk1/cyclin B on MCL1 degradation during mitotic arrest. PP2A is a protein phosphatase that is reported to associate with MCL1. Dashed lines represent inactive processes. Question marks denote unknown mechanisms. APC/CCdc20, anaphase-promoting complex/cyclosome and its activator Cdc20; SAC, spindle assembly checkpoint.Interestingly, the stability of MCL1 in asynchronous cells seems to be unaffected when the ability of APC/CCdc20 to target MCL1 is compromised by knockdown of Cdc20, or when phosphorylation at Thr 92 is ablated. Although the spindle assembly checkpoint is believed to inhibit APC/CCdc20 activity, the degradation of some targets, such as the CDK-inhibitor p21 and cyclin A, is not affected. Consequently, it is possible that MCL1 can be destroyed through Cdc20 during mitotic arrest.More recently, in two reports in Nature (Inuzuka et al, 2011; Wertz et al, 2011), it is shown that MCL1 interacts with another E3 ubiquitin ligase, SCFFbxw7. Similarly to the APC/C, the SCF (Skp1/Cul1/F-box protein) is a multi-subunit, RING E3 ubiquitin ligase. The F-box protein provides the specificity for target recognition, often by using specific interaction domains to bind to substrates. In the case of Fbxw7 (also known as Fbw7 and hCdc4), a series of WD40 domains form a pocket that dictates the binding of several substrates. For all known substrates, one or two phosphorylated degradation motifs (phospho-degrons) are recognized by Fbxw7 (Welcker & Clurman, 2008), and MCL1 seems to follow this trend. Briefly, two Fbxw7 degrons—Ser 121/Glu 125 and Ser 159/Thr 163—with different binding affinities were identified in MCL1. Inuzuka and colleagues report that these sites are phosphorylated in a GSK3-dependent manner, supporting a previous report that demonstrated a role for GSK3 in controlling MCL1 degradation (Maurer et al, 2006). They also demonstrate that Fbxw7 affects MCL1 stability during the DNA damage response. Wertz and co-workers provide evidence that, during mitotic arrest, the degrons in MCL1 are instead phosphorylated by JNK, p38 and CKII. Interestingly, when Wertz and colleagues investigated the degradation of MCL1 during mitotic arrest, they discovered a dependence on Fbxw7 similar to that reported for Cdc20 (Fig 1B). Furthermore, a functional Fbxw7–MCL1 interaction was required for the induction of apoptosis in ovarian cancer and T-ALL cell lines treated with microtubule-targeting chemotherapies. This observation presents a dilemma. Which ubiquitin ligase complex—APC/CCdc20 or SCFFbxw7—targets MCL1 for destruction during mitotic arrest? Do they compete or cooperate?There are several approaches that could be taken to investigate these questions. Perhaps the most promising direction is through understanding the role of various MCL1 phosphorylation events, particularly phosphorylation of Thr 92. The reports collectively demonstrate that Thr 92 and the Fbxw7 degrons are phosphorylated in mitotic cells. It is interesting that Thr 92 phosphorylation is specifically induced at mitosis, and Wertz and colleagues suggest that this event might drive the dissociation of a phosphatase to allow Fbxw7 degron phosphorylation (Fig 1B). However, the results so far are preliminary, and a more complete understanding of the mechanism by which Cdk1/cyclin B phosphorylation of MCL1 promotes proteolysis, and whether this is through Cdc20 and/or Fbxw7, is essential. Although MCL1 degradation after mitotic arrest is unlikely to be associated with the activity of GSK3, is there an induction of GSK3-dependent phosphorylation of MCL1 under other conditions? This important question has been studied previously, but it requires further investigation. Perhaps additional ‘priming'' kinases are involved, as is suspected to be the case for cyclin E and c-Myc, two other substrates of Fbxw7.The concept of a protein being targeted by two ubiquitin ligases is not new. For example, similarly to MCL1, p21 and MLL are targeted by both APC/CCdc20 and an SCF complex (SCFSkp2). Several APC/CCdh1 substrates (for example, Cdc25A and claspin) are also degraded via SCFβTrCP (Frescas & Pagano, 2008). However, in these instances, APC/C and SCF target the substrates at different phases of the cell cycle. The case of MCL1 is less clear. Wertz and colleagues show that mitotic arrest specifically induces binding of MCL1 to Fbxw7. Conversely, Inuzuka and colleagues provide data suggesting that Fbxw7 loss affects the non-mitotic stability of MCL1. Additionally, in an earlier paper, the Fbxw7 degron was reported to be phosphorylated by GSK3 during cytokine withdrawal (Maurer et al, 2006). Thus, we are left with a picture in which Fbxw7 targets MCL1 during mitotic arrest, but it might also target MCL1 at other points during the cell cycle or in response to external stimuli. With regard to Cdc20-mediated degradation of MCL1, mutation of the D-box seems to stabilize MCL1 only during mitotic arrest, although Cdc20 remains bound to MCL1 in non-mitotic cells. Thus, there might be differences in the conditions for recognition by either Fbxw7 or Cdc20 that merit further investigation. It is also worth mentioning that deubiquitinating enzymes (DUBs) might counteract the activity of Fbxw7, Cdc20, or both. In fact, the DUB USP9X was found to associate with MCL1 (Schwickart et al, 2010).Assuming that both E3 ligases target the same pool of MCL1 at the same time during mitotic arrest, why are there two modes of regulation? It could be that the ligases cooperate to lower MCL1 levels. It is possible that Fbxw7 and Cdc20 together deplete MCL1 to a point at which apoptosis can be initiated; if either ligase is compromised, apoptotic induction is inefficient. Alternatively, there might be a particularly relevant growth condition or cell-type specificity that favours the activity of one complex over the other. For example, it is possible that in tissues that give rise to human cancers harbouring Fbxw7 mutations (for example, T-ALLs or ovarian carcinomas), SCFFbxw7 acts as the predominant ligase. Finally, there could be redundancy or competition between the different E3 ligases. Perhaps untransformed cells maintain both systems, to protect against apoptosis evasion in the face of spindle dysfunction. Alternatively, one or both of these systems might be compromised in the cell-culture models. Notably, the situation is further complicated by reports indicating that other ligases seem to affect MCL1 stability: Mule/Huwe1 (Zhong et al, 2005) and SCFβTrCP (Ding et al, 2007). Silencing of Mule stabilizes MCL1, although Wertz and colleagues did not observe dramatic changes in MCL1 stability after Mule depletion during mitotic arrest. Instead, three groups did not observe stabilization of MCL1 after βTrCP silencing (Wertz et al, 2011; Inuzuka et al, 2011; Dehan et al, 2009). Moreover, the interaction between MCL1 and βTrCP seems to be mediated by BimEL (a βTrCP substrate), as indicated by increased binding under conditions when BimEL is degraded (rather than under conditions when MCL1 is degraded) and by the fact that some BimEL mutants lose their ability to bind to βTrCP, regardless of their binding to MCL1 (Dehan et al, 2009).Although the details of MCL1 regulation at mitotic arrest have only begun to unfold, it is clear that this pathway holds promise for furthering our understanding of the regulation of apoptosis. Microtubule-poisoning agents have historically been reliable chemotherapeutics, so, identifying cellular components that regulate MCL1 degradation during mitotic arrest is not only a way to stratify patients for a positive response to such drugs, but might also lead to the identification of novel targets for pharmacological intervention.  相似文献   

14.
Cyclin E, an activator of cyclin-dependent kinase 2 (Cdk2), is targeted for proteasomal degradation by phosphorylation-dependent multiubiquitylation via the ubiquitin ligase SCF(hCdc4). SCF ubiquitin ligases are composed of a core of conserved subunits and one variable subunit (an F box protein) involved in substrate recognition. We show here that multiubiquitylation of cyclin E requires the sequential function of two distinct splice variant isoforms of the F box protein hCdc4 known as alpha and gamma. SCF(hCdc4alpha) binds a complex containing cyclin E, Cdk2, and the prolyl cis/trans isomerase Pin1 and promotes the activity of Pin1 without directly ubiquitylating cyclin E. However, due to the action of this SCF(hCdc4alpha)-Pin1 complex, cyclin E becomes an efficient ubiquitylation substrate of SCF(hCdc4gamma). Furthermore, in the context of Cdc4alpha and cyclin E, mutational data suggest that Pin1 isomerizes a noncanonical proline-proline bond, with the possibility that Cdc4alpha may serve as a cofactor for altering the specificity of Pin1.  相似文献   

15.
The F-box protein Fbw7 (also known as Fbxw7, hCdc4 and Sel-10) functions as a substrate recognition component of a SCF-type E3 ubiquitin ligase. SCF(Fbw7) facilitates polyubiquitination and subsequent degradation of various proteins such as Notch, cyclin E, c-Myc and c-Jun. Fbw7 is highly expressed in the nervous system and controls neural stem cell differentiation and apoptosis via Notch and c-Jun during embryonic development (Hoeck et al., 2010). Fbw7 deletion in the neural lineage is perinatal lethal and thus prohibits studying the role of Fbw7 in the adult nervous system. fbw7 mRNA is highly expressed in the postnatal brain and to gain insights into the function of Fbw7 in postnatal neurogenesis we analysed Fbw7 function in the cerebellum. We generated conditional Fbw7-knockout mice (fbw7?Cb) by inactivating Fbw7 specifically in the cerebellar anlage. This resulted in decreased cerebellar size, reduced Purkinje cell number and defects in axonal arborisation. Moreover, Fbw7-deficient cerebella showed supranumeral fissures and aberrant progenitor cell migration. Protein levels of the Fbw7 substrates Notch1 and N-terminally phosphorylated c-Jun were upregulated in fbw7?Cb mice. Concomitant deletion of c-Jun, and also the junAA knock-in mutation which specifically abrogates c-Jun N-terminal phosphorylation, rescued Purkinje cell numbers and arborisation in the fbw7?Cb background. Taken together these data demonstrate that Fbw7 is essential during cerebellar development, and identify N-terminally phosphorylated c-Jun as an important substrate of SCF(Fbw7) during neurogenesis.  相似文献   

16.
Truncating or missense mutation of cullin 4B (CUL4B) is one of the most prevalent causes underlying X-linked intellectual disability (XLID). CUL4B-RING E3 ubiquitin ligase promotes ubiquitination and degradation of various proteins. Consistent with previous studies, overexpression of wild-type CUL4B in 293 cells enhanced ubiquitylation and degradation of TSC2 or cyclin E. The present study shows that XLID mutant (R388X), (R572C) or (V745A) CULB failed to promote ubiquitination and degradation of TSC2 or cyclin E. Adenoviruses-mediated expression of wild-type CUL4B decreased protein level of TSC2 or cyclin E in cultured neocortical neurons of frontal lobe. Furthermore, shRNA-mediated CUL4B knockdown caused an upregulation of TSC2 or cyclin E. XLID mutant (R388X), (R572C) or (V745A) CUL4B did not downregulate protein expression of TSC2 or cyclin E in neocortical neurons. By promoting TSC2 degradation, CUL4B could positively regulate mTOR activity in neocortical neurons of frontal cortex. Consistent with this hypothesis, CUL4B knockdown-induced upregulation of TSC2 in neocortical neurons resulted in a decreased protein level of active phospho-mTORSer2448 and a reduced expression of active phospho-p70S6KThr389 and phospho-4E-BP1Thr37/46, two main substrates of mTOR-mediated phosphorylation. Wild-type CUL4B also increased protein level of active phospho-mTORSer2448, phospho-p70S6KThr389 or phospho-4E-BP1Thr37/46. XLID CUL4B mutants did not affect protein level of active phospho-mTORSer2448, phospho-p70S6KThr389 or phospho-4E-BP1Thr37/46. Our results suggest that XLID CUL4B mutants are defective in promoting TSC2 degradation and positively regulating mTOR signaling in neocortical neurons.  相似文献   

17.
18.
Cyclin E1 is expressed at the G₁/S phase transition of the cell cycle to drive the initiation of DNA replication and is degraded during S/G₂M. Deregulation of its periodic degradation is observed in cancer and is associated with increased proliferation and genomic instability. We identify that in cancer cells, unlike normal cells, the closely related protein cyclin E2 is expressed predominantly in S phase, concurrent with DNA replication. This occurs at least in part because the ubiquitin ligase component that is responsible for cyclin E1 downregulation in S phase, Fbw7, fails to effectively target cyclin E2 for proteosomal degradation. The distinct cell cycle expression of the two E-type cyclins in cancer cells has implications for their roles in genomic instability and proliferation and may explain their associations with different signatures of disease.  相似文献   

19.
20.
D-type cyclins play a pivotal role in G(1)-S progression of the cell cycle, and their expression is frequently deregulated in cancer. Cyclin D1 has a half-life of only ~30 min as a result of its ubiquitylation and proteasomal degradation, with various F-box proteins, including Fbxo4, Fbxw8, Skp2, and Fbxo31, having been found to contribute to its ubiquitylation. We have now generated Fbxo4-deficient mice and found no abnormalities in these animals. Cyclin D1 accumulation was thus not observed in Fbxo4(-/-) mouse tissues. The half-life of cyclin D1 in mouse embryonic fibroblasts (MEFs) prepared from Fbxo4(-/-), Fbxw8(-/-), and Fbxo4(-/-); Fbxw8(-/-) mice also did not differ from that in wild-type MEFs. Additional depletion of Skp2 and Fbxo31 in Fbxo4(-/-); Fbxw8(-/-) MEFs by RNA interference did not affect cyclin D1 stability. Although Fbxo31 depletion in MEFs increased cyclin D1 abundance, this effect appeared attributable to upregulation of cyclin D1 mRNA. Furthermore, abrogation of the function of the Skp1-Cul1-F-box protein (SCF) complex or the anaphase-promoting complex/cyclosome (APC/C) complexes did not alter the half-life of cyclin D1, whereas cyclin D1 degradation was dependent largely on proteasome activity. Our genetic analyses thus do not support a role for any of the four F-box proteins examined in cyclin D1 degradation during normal cell cycle progression. They suggest the existence of other ubiquitin ligases that target cyclin D1 for proteolysis.  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