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.
Fbxl7, a component of the Skp1·Cul1·F-box protein type ubiquitin E3 ligase, regulates mitotic cell cycle progression. Here we demonstrate that overexpression of Fbxl7 in lung epithelia decreases the protein abundance of survivin, a member of the inhibitor of apoptosis family. Fbxl7 mediates polyubiquitylation and proteasomal degradation of survivin by interacting with Glu-126 within its carboxyl-terminal α helix. Furthermore, both Lys-90 and Lys-91 within survivin serve as ubiquitin acceptor sites. Ectopically expressed Fbxl7 impairs mitochondrial function, whereas depletion of Fbxl7 protects mitochondria from actions of carbonyl cyanide m-chlorophenylhydrazone, an inhibitor of oxidative phosphorylation. Compared with wild-type survivin, cellular expression of a survivin mutant protein deficient in its ability to interact with Fbxl7 (E126A) and a ubiquitylation-resistant double point mutant (KK90RR/KK91RR) rescued mitochondria to a larger extent from damage induced by overexpression of Fbxl7. Therefore, these data suggest that the Skp1·Cul1·F-box protein complex subunit Fbxl7 modulates mitochondrial function by controlling the cellular abundance of survivin. The results raise opportunities for F-box protein targeting to preserve mitochondrial function.  相似文献   

2.
SCFhFBH1 can act as helicase and E3 ubiquitin ligase   总被引:3,自引:1,他引:2  
Kim JH  Kim J  Kim DH  Ryu GH  Bae SH  Seo YS 《Nucleic acids research》2004,32(8):2287-2297
In our previous study, we found that a human F-box DNA helicase, named hFBH1, interacted with SKP1 to form an SCF (SKP1–Cul1–F-box protein) complex together with CUL1 and ROC1 in an F-box-dependent manner. The complex immunoprecipitated from crude cell extracts catalyzed polyubiquitin formation in the presence of the ubiquitin-activating and ubiquitin-conjugating enzymes, E1 and E2, respectively. In this report, we characterized the enzymatic properties of the recombinant SCFhFBH1 complex purified from insect cells expressing hFBH1, SKP1, CUL1 and ROC1. The SCFhFBH1 complex was isolated as a single tight complex that retained DNA helicase, DNA-dependent ATPase and E3 ubiquitin ligase activities. The helicase and ATPase activities residing in the SCFhFBH1 complex were indistinguishable from those of the hFBH1 protein alone. Moreover, the ubiquitin ligase activity of the SCFhFBH1 complex was hardly affected by single-stranded or double-stranded DNA. The multiple activities present in this complex act independently of each other, suggesting that the SCFhFBH1 complex can catalyze a ubiquitination reaction while acting as a DNA helicase or translocating along DNA. The potential roles of the SCFhFBH1 complex in DNA metabolism based upon the enzymatic activities associated with this complex are discussed.  相似文献   

3.
The ubiquitin ligases, SCF complexes, consist of Cul1, Skp1, Rbx1 and the substrate recognition components F-box proteins. Previous studies have reported that one of these F-box proteins, Fbl12, which is produced by Fbxl12 gene, regulates both cell cycle and differentiation. In this paper, we show that the intronic region of Fbxl12 gene acts as an alternative promoter and induces expression of a short form of Fbl12 that lacks F-box domain (Fbl12ΔF). We also found that UV irradiation increases Fbl12ΔF mRNA in cells. Finally, Fbl12ΔF may promote the subcellular localization of Fbl12 from nucleus to cytoplasm through their binding. Our data provide the possibility that Fbl12ΔF induced by alternative promoter controls the SCFFbl12 activity in response to UV stimulation.  相似文献   

4.
5.
The SCF complex, which consists of the invariable components Skp1, Cul1, and Rbx1 as well as a variable F-box protein, functions as an E3 ubiquitin ligase. The mechanism by which the activity of this complex is regulated, however, has been unclear. The application of tandem affinity purification has now resulted in the identification of a novel Cul1-binding protein: TATA-binding protein-interacting protein 120A (TIP120A, also called CAND1). Immunoprecipitation, immunoblot, and immunofluorescence analyses with mammalian cells revealed that TIP120A physically associates with Cul1 in the nucleus and that this interaction is mediated by a central region of Cul1 distinct from its binding sites for Skp1 and Rbx1. Furthermore, TIP120A was shown to interact selectively with Cul1 that is not modified by NEDD8. The Cul1-TIP120A complex does not include Skp1, raising the possibility that TIP120A competes with Skp1 for binding to Cul1. These observations thus suggest that TIP120A may function as a negative regulator of the SCF complex by binding to nonneddylated Cul1 and thereby preventing assembly of this ubiquitin ligase.  相似文献   

6.
The SCF ubiquitin ligase complex consists of four components, Skp1, Cul1, ROC1/Rbx1, and a variable subunit F-box protein, which serves as a receptor for target proteins. The F-box proteins consist of an N-terminal ∼40 amino acid F-box domain that binds to Skp1 and the C-terminal substrate-binding domain. We have reported previously that Fbs1 and Fbs2 are N-linked glycoprotein-specific F-box proteins. In addition, other three F-box proteins, Fbg3, Fbg4, and Fbg5, show high homology to Fbs1 and Fbs2, but their functions remain largely unknown. Here we report that Skp1 assists in correct folding of exogenously expressed F-box proteins. Fbs2 as well as Fbg3, Fbg4, and Fbg5 proteins formed SCF complexes but did not bind to N-glycoproteins when exogenously expressed alone. However, co-expression of Fbs2 and Fbg5 with Skp1 facilitated their binding to glycoproteins that reacted with ConA. Furthermore, Skp1 increased the cellular concentrations of F-box proteins by preventing aggregate formation. These observations suggest that Skp1 plays an important role in stabilizing the conformation of these F-box proteins, which increases their expression levels and substrate-binding.  相似文献   

7.
The ubiquitin protein ligase SCF(Skp2) is composed of Skp1, Cul1, Roc1/Rbx1 and the F-box protein Skp2, the substrate-recognition subunit. Levels of Skp2 decrease as cells exit the cell cycle and increase as cells re-enter the cycle. Ectopic expression of Skp2 in quiescent fibroblasts causes mitogen-independent S-phase entry. Hence, mechanisms must exist for limiting Skp2 protein expression during the G(0)/G(1) phases. Here we show that Skp2 is degraded by the proteasome in G(0)/G(1) and is stabilized when cells re-enter the cell cycle. Rapid degradation of Skp2 in quiescent cells depends on Skp2 sequences that contribute to Cul1 binding and interference with endogenous Cul1 function in serum-deprived cells induces Skp2 expression. Furthermore, recombinant Cul1-Roc1/Rbx1-Skp1 complexes can catalyse Skp2 ubiquitylation in vitro. These results suggest that degradation of Skp2 in G(0)/G(1) is mediated, at least in part, by an autocatalytic mechanism involving a Skp2-bound Cul1-based core ubiquitin ligase and imply a role for this mechanism in the suppression of SCF(Skp2) ubiquitin protein ligase function during the G(0)/G(1) phases of the cell cycle.  相似文献   

8.
The accurate replication of genetic information is critical to maintaining chromosomal integrity. Cdc6 functions in the assembly of pre-replicative complexes and is specifically required to load the Mcm2-7 replicative helicase complex at replication origins. Cdc6 is targeted for protein degradation by multiple mechanisms in Saccharomyces cerevisiae, although only a single pathway and E3 ubiquitin ligase for Cdc6 has been identified, the SCFCdc4 (Skp1/Cdc53/F-box protein) complex. Notably, Cdc6 is unstable during the G1 phase of the cell cycle, but the ubiquitination pathway has not been previously identified. Using a genetic approach, we identified two additional E3 ubiquitin ligase components required for Cdc6 degradation, the F-box protein Dia2 and the Hect domain E3 Tom1. Both Dia2 and Tom1 control Cdc6 turnover during G1 phase of the cell cycle and act separately from SCFCdc4. Ubiquitination of Cdc6 is significantly reduced in dia2Δ and tom1Δ cells. Tom1 and Dia2 each independently immunoprecipitate Cdc6, binding to a C-terminal region of the protein. Tom1 and Dia2 cannot compensate for each other in Cdc6 degradation. Cdc6 and Mcm4 chromatin association is aberrant in tom1Δ and dia2Δ cells in G1 phase. Together, these results present evidence for a novel degradation pathway that controls Cdc6 turnover in G1 that may regulate pre-replicative complex assembly.  相似文献   

9.
A novel class of ubiquitin ligases, termed the SCF complex, consists of invariable components, Skp1 and Cullin, and variable components called F-box proteins, which have a primary role in determining substrate specificity. We have isolated a cDNA encoding the mouse F-box protein Fwd2 (also known as MD6) as a possible constituent of an SCF-type ubiquitin ligase. Fwd2 cDNA contains 1890 bp with a 1362-bp open reading frame and encodes an approximately 51.5-kDa protein. Fwd2 is expressed predominantly in liver and, to a lesser extent, in the testis, lung, heart, and skeletal muscle. Immunofluorescence staining for Fwd2 protein shows a pattern with the cytoplasm. A coimmunoprecipitation assay has revealed the in vivo interaction between Skp1 and Fwd2 through the F-box domain. Fwd2 also interacts with Cul1 through Skp1, suggesting that Skp1, Cul1, and the F-box protein Fwd2 form an SCF complex (SCF(Fwd2)). We have also isolated and determined the nucleotide sequence and genomic organization of the gene that encodes mouse Fwd2. This gene spans approximately 17 kb and consists of six exons and five introns. Our results suggest that Fwd2 is an F-box protein that constitutes an SCF ubiquitin ligase complex and that it plays a critical role in the ubiquitin-dependent degradation of proteins expressed in the liver.  相似文献   

10.
The Ku heterodimer, composed of Ku70 and Ku80, is the initiating factor of the nonhomologous end joining (NHEJ) double-strand break (DSB) repair pathway. Ku is also thought to impede the homologous recombination (HR) repair pathway via inhibition of DNA end resection. Using the cell-free Xenopus laevis egg extract system, we had previously discovered that Ku80 becomes polyubiquitylated upon binding to DSBs, leading to its removal from DNA and subsequent proteasomal degradation. Here we show that the Skp1-Cul1-F box (SCF) E3 ubiquitin ligase complex is required for Ku80 ubiquitylation and removal from DNA. A screen for DSB-binding F box proteins revealed that the F box protein Fbxl12 was recruited to DNA in a DSB- and Ku-sensitive manner. Immunodepletion of Fbxl12 prevented Cul1 and Skp1 binding to DSBs and Ku80 ubiquitylation, indicating that Fbxl12 is the F box protein responsible for Ku80 substrate recognition. Unlike typical F box proteins, the F box of Fbxl12 was essential for binding to both Skp1 and its substrate Ku80. Besides Fbxl12, six other chromatin-binding F box proteins were identified in our screen of a subset of Xenopus F box proteins: β-TrCP, Fbh1, Fbxl19, Fbxo24, Fbxo28 and Kdm2b. Our study unveils a novel function for the SCF ubiquitin ligase in regulating the dynamic interaction between DNA repair machineries and DSBs.  相似文献   

11.
SCF is a ubiquitin ligase and is composed of Skp1, Cul1, F-box protein, and Roc1. The catalytic site of the SCF is the Cul1/Roc1 complex and RING-finger protein Roc1. It was shown earlier that when Cul1 was co-expressed with Roc1 in Sf-9 cells in a baculovirus protein expression system, Cul1 was highly neddylated in the cell, suggesting that Roc1 may function as a Nedd8-E3 ligase. However, there is no direct evidence that Roc1 is a Nedd8-E3 in an in vitro enzyme system. Here we have shown that Roc1 binds to Ubc12, E2 for Nedd8, but not to Ubc9, E2 for SUMO-1 and Roc1 RING-finger mutant, H77A, did not bind to Ubc12. In in vitro neddylation system using purified Cul1/Roc1 complex expressed in bacteria, Roc1 promotes neddylation of Cul1. These results demonstrate that Roc1 functions as a Nedd8-E3 ligase toward Cul1. Furthermore, Roc1 and Cul1 were ubiquitinylated in a manner dependent on the neddylation of Cul1 in vitro. In addition, Cul1 was degraded through the ubiquitin-proteasome pathway, and a non-neddylated mutant Cul1, K720R, was more stable than wild-type in intact cells. Thus, neddylation of Cul1 might regulate SCF function negatively via degradation of Cul1/Roc1 complex.  相似文献   

12.
The multiprotein von Hippel-Lindau (VHL) tumor suppressor and Skp1-Cul1-F-box protein (SCF) complexes belong to families of structurally related E3 ubiquitin ligases. In the VHL ubiquitin ligase, the VHL protein serves as the substrate recognition subunit, which is linked by the adaptor protein Elongin C to a heterodimeric Cul2/Rbx1 module that activates ubiquitylation of target proteins by the E2 ubiquitin-conjugating enzyme Ubc5. In SCF ubiquitin ligases, F-box proteins serve as substrate recognition subunits, which are linked by the Elongin C-like adaptor protein Skp1 to a Cul1/Rbx1 module that activates ubiquitylation of target proteins, in most cases by the E2 Cdc34. In this report, we investigate the functions of the Elongin C and Skp1 proteins in reconstitution of VHL and SCF ubiquitin ligases. We identify Elongin C and Skp1 structural elements responsible for selective interaction with their cognate Cullin/Rbx1 modules. In addition, using altered specificity Elongin C and F-box protein mutants, we investigate models for the mechanism underlying E2 selection by VHL and SCF ubiquitin ligases. Our findings provide evidence that E2 selection by VHL and SCF ubiquitin ligases is determined not solely by the Cullin/Rbx1 module, the target protein, or the integrity of the substrate recognition subunit but by yet to be elucidated features of these macromolecular complexes.  相似文献   

13.
BRCA1 mutations account for a significant proportion of familial breast and ovarian cancers. In addition, reduced BRCA1 protein is associated with sporadic cancer cases in these tissues. At the cellular level, BRCA1 plays a critical role in multiple cellular functions such as DNA repair and cell cycle checkpoint control. Its protein level is regulated in a cell cycle-dependent manner. However, regulation of BRCA1 protein stability is not fully understood. Our earlier study showed that the amino terminus of BRCA1 harbors a degron sequence that is sufficient and necessary for conferring BRCA1 degradation. In the current study, we used mass spectrometry to identify Skp1 that regulates BRCA1 protein stability. Small interfering RNA screening that targets all human F-box proteins uncovered FBXO44 as an important protein that influences BRCA1 protein level. The Skp1-Cul1-F-box-protein44 (SCFFBXO44) complex ubiquitinates full-length BRCA1 in vitro. Furthermore, the N terminus of BRCA1 mediates the interaction between BRCA1 and FBXO44. Overexpression of SCFFBXO44 reduces BRCA1 protein level. Taken together, our work strongly suggests that SCFFBXO44 is an E3 ubiquitin ligase responsible for BRCA1 degradation. In addition, FBXO44 expression pattern in breast carcinomas suggests that SCFFBXO44-mediated BRCA1 degradation might contribute to sporadic breast tumor development.  相似文献   

14.
The concentrations and functions of many cellular proteins are regulated by the ubiquitin pathway. Cullin family proteins bind with the RING-finger protein Roc1 to recruit the ubiquitin-conjugating enzyme (E2) to the ubiquitin ligase complex (E3). Cul1 and Cul7, but not other cullins, bind to an adaptor protein, Skp1. Cul1 associates with one of many F-box proteins through Skp1 to assemble various SCF-Roc1 E3 ligases that each selectively ubiquitinate one or more specific substrates. Here, we show that Cul3, but not other cullins, binds directly to multiple BTB domains through a conserved amino-terminal domain. In vitro, Cul3 promoted ubiquitination of Caenorhabditis elegans MEI-1, a katanin-like protein whose degradation requires the function of both Cul3 and BTB protein MEL-26. We suggest that in vivo there exists a potentially large number of BCR3 (BTB-Cul3-Roc1) E3 ubiquitin ligases.  相似文献   

15.
The F-box protein is the substrate recognition subunit of SCF (SKP1/CUL1/F-box) E3 ubiquitin ligase complex, a multicomponent RING-type E3 ligase involved in the regulation of numerous cellular processes by targeting critical regulatory proteins for ubiquitination. However, whether and how F-box proteins are regulated is largely unknown. Here we report that FBXO28, a poorly characterized F-box protein, is a novel substrate of SCF E3 ligase. Pharmaceutical or genetic inhibition of neddylation pathway that is required for the activation of SCF stabilizes FBXO28 and prolongs its half-life. Meanwhile, FBXO28 is subjected to ubiquitination and cullin1-based SCF complex promotes FBXO28 degradation. Moreover, deletion of F-box domain stabilizes FBXO28 and knockdown of endogenous FBXO28 strongly upregulates exogenous FBXO28 expression. Taken together, these data reveal that SCFFBXO28 is the E3 ligase responsible for the self-ubiquitination and proteasomal degradation of FBXO28, providing a new clue for the upstream signaling regulation for F-box proteins.  相似文献   

16.
The SCF complex containing Skp1, Cul1, and the F-box protein FWD1 (the mouse homologue of Drosophila Slimb and Xenopus beta-TrCP) functions as the ubiquitin ligase for IkappaBalpha. FWD1 associates with Skp1 through the F-box domain and also recognizes the conserved DSGXXS motif of IkappaBalpha. The structural requirements for the interactions of FWD1 with IkappaBalpha and with Skp1 have now been investigated further. The D31A mutation (but not the G33A mutation) in the DSGXXS motif of IkappaBalpha abolished the binding of IkappaBalpha to FWD1 and its subsequent ubiquitination without affecting the phosphorylation of IkappaBalpha. The IkappaBalpha mutant D31E still exhibited binding to FWD1 and underwent ubiquitination. These results suggest that, in addition to site-specific phosphorylation at Ser(32) and Ser(36), an acidic amino acid at position 31 is required for FWD1-mediated ubiquitination of IkappaBalpha. Deletion analysis of Skp1 revealed that residues 61-143 of this protein are required for binding to FWD1. On the other hand, the highly conserved residues Pro(149), Ile(160), and Leu(164) in the F-box domain of FWD1 were dispensable for binding to Skp1. Together, these data delineate the structural requirements for the interactions among IkappaBalpha, FWD1, and Skp1 that underlie substrate recognition by the SCF ubiquitin ligase complex.  相似文献   

17.
Cand1 (Cullin-associated and neddylation-dissociated protein 1) has long been known as a regulator of SCF ubiquitin ligases, but details remained puzzling due to conflicting results from in vitro and in vivo experiments. Three recent reports, one in Cell and two in Nature Communications, propose Cand1 as a protein exchange factor with interesting mechanism that reconciles Cand1 genetics and biochemistry.Most eukaryotic proteins are modified by the small protein ubiquitin at some point during their life. Ubiquitin tags can mark them for degradation in the proteasome, or control other protein properties such as localization, activity, and interactions. Ubiquitin ligases (E3 enzymes) play a particularly important role in the E1-E2-E3 ubiquitylation cascade as they directly select substrates for ubiquitin attachment. E3s define a large protein family with over 600 members in human cells that control ubiquitin transfer onto thousands of substrate proteins1. The complexity of this system comes with conceptual challenges that are particularly apparent for the largest group of E3s, the multisubunit Cullin-RING ubiquitin ligases (CRLs). The archetypal CRLs, the Skp1/Cul1/F-box protein (SCF) complexes, assemble on the Cul1 scaffold, with the small RING protein Rbx1 and E2 bound to the C-terminus, and the adapter protein Skp1 associated with the N-terminal region. Skp1 binds to one of many F-box proteins (FboxP), which confer specificity by selectively recruiting substrate proteins for ubiquitin transfer2 (Figure 1, right). Up to 69 FboxPs in humans, and possibly 700 in plants, compete for the Cul1 core. How cells adjust abundance of the different SCF ligases in response to cell cycle and environmental cues to dynamically match substrate demand is one of the major questions in the field. Since identification of Cand1 over 10 years ago, its involvement in SCF complex formation has been evident3,4. However, its true function was somewhat of a mystery. Cand1 acted as a potent SCF inhibitor in vitro by displacing the FboxP-Skp1 pair from Cul1, but genetic experiments classified Cand1 as a positive regulator of SCF and other CRLs in vivo5. An additional layer of complexity is added by covalent modification of cullins with the ubiquitin-like protein Nedd8. Neddylation (modification with Nedd8) induces a conformational rearrangement of Cul1 that stimulates ubiquitin transfer by the SCF-bound E2 and also obscures the Cand1 binding site on Cul16. Nedd8 deconjugation is catalyzed by the COP9 signalosome (CSN). Strikingly, the paradox observed for Cand1 is also evident for CSN, because CSN clearly functions as a negative regulator of SCF in vitro, yet genetic data suggest a positive role for SCF activity in vivo5. A prevailing model has been that SCF and other CRLs must undergo neddylation cycles whereby deneddylated cullins are sequestered by Cand1, allowing substrate receptor exchange followed by reactivation of the assembled CRL by neddylation. However, mechanistic insight was scarce.Open in a separate windowFigure 1Cand1-driven substrate receptor exchange model (based on Pierce et al.7). Substrate availability protects the stable substrate ubiquitylation state (right). Depletion of substrates enhances CSN-mediated deneddylation shifting the SCF complex into a transition state that either finds new substrates and becomes reactivated by Nedd8 (N8) conjugation, or forms a transient complex with Cand1. The transient complex is highly unstable because of steric interference between F-box protein and Cand1 causing cycles of Cand1 and FboxP-Skp1 eviction. The exchange state allows the repertoire of formed SCF complexes to sample for substrates and, upon engagement, transit into the stable substrate ubiquitylation state.In a recent study published in Cell7, Deshaies and colleagues provide a biochemical framework that not only explains the CSN and Cand1 paradoxes, but also suggests a model for how SCF composition adjusts to varying substrate demand. They used in vitro real-time fluorescence resonance energy transfer (FRET) assays to monitor binding dynamics between FboxP-Skp1 and Cul1-Rbx1 complexes. Fbxw7-Skp1 formed an astonishingly tight complex with Cul1-Rbx1 (KD = 200 fM) that could not be replaced by other FboxP-Skp1 complexes. However, addition of Cand1 accelerated spontaneous dissociation of SCFFbxw7 over one-million-fold. Kinetic measurements demonstrated that Cand1 acts neither as a competitive nor allosteric inhibitor of Fbxw7-Skp1 binding to Cul1-Rbx1. Instead, Cand1 specifically increases the dissociation rate of the FboxP-Skp1 complex while having little effect on association rates. The authors point out that such a kinetic effect is reminiscent of guanine nucleotide exchange factors (GEFs). Accordingly, they suggest the term substrate receptor exchange factor (SREF) for Cand1 and functionally similar factors.Cand1''s SREF activity was beautifully illustrated in vitro using the two different F-box proteins Fbxw7 and β-TrCP. When SCFβ-TrCP was combined with purified Fbxw7-Skp1 in an in vitro ubiquitylation reaction, no ubiquitylation of cyclin E (Fbxw7 substrate) was observed. This was not surprising because the tight binding of β-TrCP-Skp1 to Cul1 was expected to prevent assembly of SCFFbxw7. Remarkably, addition of Cand1 dramatically stimulated cyclin E ubiquitylation, likely through dissociation of β-TrCP-Skp1, thus establishing a new equilibrium of SCFβ-TrCP and SCFFbxw7 complexes. This assay design exposed Cand1 as an activator of SCF in vitro, which is consistent with its positive regulator role revealed by genetic experiments. The important findings that the FboxP-Skp1 complex can remove tightly bound Cand1 from Cul1, and indication of a transient complex of Cand1 with fully assembled SCF led to proposal of a model for SCF dynamics driven by substrate demand (Figure 1). A key feature of the model is based on recent evidence that substrate binding to CRLs can significantly reduce CSN access and CRL deneddylation8,9. When substrates are exhausted, accelerated deneddylation shifts the active SCF complex into a deneddylated transition state, which can either bind new substrate and become reactivated by Nedd8 conjugation, or enter the exchange state. The latter is characterized by a Cand1-bound transition complex that controls dissociation and association of FboxP-Skp1 complexes. This concept extends the previous neddylation cycle model based on a strong biochemical foundation and provides a hypothesis for dynamic remodeling of the SCF landscape by substrate demand. Pierce et al.7 support this biochemical concept with findings in vivo demonstrating significant shifts in the SCF landscape when Cand1 is absent.The importance of Cand1 as a F-box protein exchange factor is reinforced by two recent studies in yeast. Zelma et al.10 demonstrate the role of Cand1 in remodeling the SCF repertoire in response to changing growth conditions, and Wu et al.11 provide additional evidence for Cand1 as an F-box protein exchange factor in vivo. Clearly there are more challenges ahead to understanding CRL dynamics, but the significance of these findings may reach beyond ubiquitin biology as it introduces the concept of protein exchange factors that govern association of protein binding platforms with large numbers of interactors.  相似文献   

18.
19.
During cell proliferation, protein degradation is strictly regulated by the cell cycle and involves two complementary ubiquitin ligase complexes, the SCF (Skp, Cullin, F-box) and APC/C (Anaphase Promoting Complex/Cyclosome) ubiquitin ligases. SCF ligases are constitutively active and generally target only proteins after they have been selected for degradation, usually by phosphorylation. In contrast, APC/C complexes are themselves activated by phosphorylation and their substrates contain a targeting signal known as degron, a consensus amino acid sequence such as a D-Box. SCF complexes degrade proteins during the G1 phase. However, as DNA synthesis begins, the SCF complexes are degraded and APC/C complexes are activated. APC-2, a protein crucial to cell division, initiates anaphase by triggering the degradation of multiple proteins. This study explores an unexpected interaction between APC-2 and SCFFBG1. We found that FBG1 is a promiscuous ubiquitin ligase with many partners. Immunoprecipitation experiments demonstrate that FBG1 and APC2 interact directly. Mutagenesis-based experiments show that this interaction requires a D-Box found within the FBG1 F-box domain. Unexpectedly, we demonstrate that co-expression with FBG1 increases total APC2 levels. However, free APC2 is decreased, inhibiting cell proliferation. Finally, FACS analysis of cell populations expressing different forms of FBG1 demonstrate that this ubiquitin ligase induces S-phase arrest, illustrating the functional consequences of the interaction described. In summary, we have discovered a novel APC2 inhibitory activity of FBG1 independent from its function as ubiquitin ligase, providing the basis for future studies of FBG1 in aging and cancer.  相似文献   

20.
F-box proteins are the substrate recognition subunits of SCF (Skp1, Cul1, F-box protein) ubiquitin ligase complexes. Skp2 is a nuclear F-box protein that targets the CDK inhibitor p27 for ubiquitin- and proteasome-dependent degradation. In G0 and during the G1 phase of the cell cycle, Skp2 is degraded via the APC/CCdh1 ubiquitin ligase to allow stabilization of p27 and inhibition of CDKs, facilitating the maintenance of the G0/G1 state. APC/CCdh1 binds Skp2 through an N-terminal domain (amino acids 46-94 in human Skp2). It has been shown that phosphorylation of Ser69 and Ser72 in this domain dissociates Skp2 from APC/C. More recently, it has instead been proposed that phosphorylation of Skp2 on Ser72 by Akt/PKB allows Skp2 binding to Skp1, promoting the assembly of an active SCFSkp2 ubiquitin ligase, and Skp2 relocalization/retention into the cytoplasm, promoting cell migration via an unknown mechanism. According to these reports, a Skp2 mutant in which Ser72 is substituted with Ala is unable to promote cell proliferation and loses its oncogenic potential. Given the contrasting reports, we revisited these results and conclude that phosphorylation of Skp2 on Ser72 does not control Skp2 binding to Skp1 and Cul1, has no influence on SCFSkp2 ubiquitin ligase activity, and does not affect the subcellular localization of Skp2.  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