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 15 毫秒
1.
The protease HAUSP is a critical component of the p53–Mdm2 pathway and acts as a specific deubiquitinase for both p53 and Mdm2 and thus is important for p53 regulation. In knock-down and knock-out cellular systems it was observed that ablation of HAUSP induces profound stabilization of p53 due to enhanced degradation of Mdm2. Thus, inhibiting HAUSP by small compound interference has been proposed as a rational therapeutic strategy to activate p53 in p53 wild type tumors. However, HAUSP-mediated effects in the p53–Mdm2 axis are highly complex and non-linear and to date the role of HAUSP in tumor suppression in vivo remains unexplored. Here we investigate the effect of HAUSP up- and downregulation on cell proliferation, apoptosis and tumor growth in vitro and in a xenograft model in vivo, using an inducible isogenic human colon carcinoma cell system. Importantly, in the absence of stress, both HAUSP up- and downregulation inhibit cell proliferation in vitro and tumor growth in vivo due to constitutively elevated p53 levels. Moreover, tumors with HAUSP up- and downregulation respond to radiotherapy with further growth inhibition. However, HAUSP downregulation causes resistance to Camptothecin- and irradiation-induced apoptosis, which correlates with suppressed mitochondrial translocation of p53. Our data suggest that changes in HAUSP modulate tumor growth and apoptotic sensitivity in vivo.  相似文献   

2.
A dynamic role of HAUSP in the p53-Mdm2 pathway   总被引:12,自引:0,他引:12  
Li M  Brooks CL  Kon N  Gu W 《Molecular cell》2004,13(6):879-886
Our previous study showed that ubiquitination of p53 is reversible and that the ubiquitin hydrolase HAUSP can stabilize p53 by deubiquitination. Here, we found that partial reduction of endogenous HAUSP levels by RNAi indeed destabilizes endogenous p53; surprisingly, however, nearly complete ablation of HAUSP stabilizes and activates p53. We further show that this phenomenon occurs because HAUSP stabilizes Mdm2 in a p53-independent manner, providing an interesting feedback loop in p53 regulation. Notably, HAUSP is required for Mdm2 stability in normal cells; in HAUSP-ablated cells, self-ubiquitinated-Mdm2 becomes extremely unstable, leading to indirect p53 activation. Furthermore, this feedback regulation is specific to Mdm2; in HeLa cells, where p53 is preferentially degraded by viral E6-dependent ubiquitination, depletion of HAUSP fails to activate p53. This study provides an example of an ubiquitin ligase (Mdm2) that is directly regulated by a deubiquitinase (HAUSP) and also reveals a dynamic role of HAUSP in the p53-Mdm2 pathway.  相似文献   

3.
4.
5.
Resistance to DNA-damaging agents is one of the main reasons for the low survival of cervical cancer patients. Previous reports have suggested that the Cdc25A oncoprotein significantly affects the level of susceptibility to DNA-damaging agents, but the molecular mechanism remains unclear. In this study, we used Western blot and flow cytometry analyses to demonstrate that the deubiquitinating enzyme HAUSP stabilizes Cdc25A protein level. Furthermore, in a co-immunoprecipitation assay, we found that HAUSP interacts with and deubiquitinates Cdc25A both exogenously and endogenously. HAUSP extends the half-life of the Cdc25A protein by circumventing turnover. HAUSP knockout in HeLa cells using the CRISPR/Cas9 system caused a significant delay in Cdc25A-mediated cell cycle progression, cell migration, and colony formation and attenuated tumor progression in a mouse xenograft model. Furthermore, HAUSP-mediated stabilization of the Cdc25A protein produced enhanced resistance to DNA-damaging agents. Overall, our study suggests that targeting Cdc25A and HAUSP could be a promising combinatorial approach to halt progression and minimize antineoplastic resistance in cervical cancer.  相似文献   

6.
Hu M  Gu L  Li M  Jeffrey PD  Gu W  Shi Y 《PLoS biology》2006,4(2):e27
Herpesvirus-associated ubiquitin-specific protease (HAUSP, also known as USP7), a deubiquitylating enzyme of the ubiquitin-specific processing protease family, specifically deubiquitylates both p53 and MDM2, hence playing an important yet enigmatic role in the p53–MDM2 pathway. Here we demonstrate that both p53 and MDM2 specifically recognize the N-terminal tumor necrosis factor–receptor associated factor (TRAF)–like domain of HAUSP in a mutually exclusive manner. HAUSP preferentially forms a stable HAUSP–MDM2 complex even in the presence of excess p53. The HAUSP-binding elements were mapped to a peptide fragment in the carboxy-terminus of p53 and to a short-peptide region preceding the acidic domain of MDM2. The crystal structures of the HAUSP TRAF-like domain in complex with p53 and MDM2 peptides, determined at 2.3-Å and 1.7-Å resolutions, respectively, reveal that the MDM2 peptide recognizes the same surface groove in HAUSP as that recognized by p53 but mediates more extensive interactions. Structural comparison led to the identification of a consensus peptide-recognition sequence by HAUSP. These results, together with the structure of a combined substrate-binding-and-deubiquitylation domain of HAUSP, provide important insights into regulation of the p53–MDM2 pathway by HAUSP.  相似文献   

7.
The p53 tumor suppressor protein has a major role in protecting the integrity of the genome. In unstressed cells, p53 is maintained at low levels by the ubiquitin-proteasome pathway. A balance between ubiquitin ligase activity (Hdm2, COP1, and Pirh2) and the ubiquitin protease activity of the Herpes virus-associated ubiquitin-specific protease (HAUSP) determines the half-life of p53. HAUSP also modulates p53 stability indirectly by deubiquitination and stabilization of Hdm2. The Hdmx protein affects p53 stability as well through its interaction with and regulation of Hdm2. Vice versa, Hdmx is a target for Hdm2-mediated ubiquitination and degradation. Here, we show that HAUSP also interacts with Hdmx, resulting in its direct deubiquitination and stabilization. HAUSP activity is required to maintain normal Hdmx protein levels. Therefore, the balance between HAUSP and Hdm2 activity determines Hdmx protein stability. Importantly, impaired deubiquitination of Hdmx/Hdm2 by HAUSP contributes to the DNA damage-induced degradation of Hdmx and transient instability of Hdm2.  相似文献   

8.
Herpesvirus-associated ubiquitin-specific protease (HAUSP, also known as USP7), a deubiquitylating enzyme of the ubiquitin-specific processing protease family, specifically deubiquitylates both p53 and MDM2, hence playing an important yet enigmatic role in the p53–MDM2 pathway. Here we demonstrate that both p53 and MDM2 specifically recognize the N-terminal tumor necrosis factor–receptor associated factor (TRAF)–like domain of HAUSP in a mutually exclusive manner. HAUSP preferentially forms a stable HAUSP–MDM2 complex even in the presence of excess p53. The HAUSP-binding elements were mapped to a peptide fragment in the carboxy-terminus of p53 and to a short-peptide region preceding the acidic domain of MDM2. The crystal structures of the HAUSP TRAF-like domain in complex with p53 and MDM2 peptides, determined at 2.3-Å and 1.7-Å resolutions, respectively, reveal that the MDM2 peptide recognizes the same surface groove in HAUSP as that recognized by p53 but mediates more extensive interactions. Structural comparison led to the identification of a consensus peptide-recognition sequence by HAUSP. These results, together with the structure of a combined substrate-binding-and-deubiquitylation domain of HAUSP, provide important insights into regulation of the p53–MDM2 pathway by HAUSP.  相似文献   

9.
Inhibition of Mdm2 function is a validated approach to restore p53 activity for cancer therapy; nevertheless, inhibitors of Mdm2 such as Nutlin-3 have certain limitations, suggesting that additional targets in this pathway need to be further elucidated. Our finding that the Herpesvirus-Associated Ubiquitin-Specific Protease (HAUSP, also called USP7) interacts with the p53/Mdm2 protein complex, was one of the first examples that deubiquitinases (DUBs) exhibit a specific role in regulating protein stability. Here, we show that inhibitors of HAUSP and Nutlin-3 can synergistically activate p53 function and induce p53-dependent apoptosis in human cancer cells. Notably, HAUSP can also target the N-Myc oncoprotein in a p53-independent manner. Moreover, newly synthesized HAUSP inhibitors are more potent than the commercially available inhibitors to suppress N-Myc activities in p53 mutant cells for growth suppression. Taken together, our study demonstrates the utility of HAUSP inhibitors to target cancers in both a p53-depdentent and -independent manner.  相似文献   

10.
Stability of the 'guardian of the genome' tumor suppressor protein p53 is regulated predominantly through its ubiquitination. The ubiquitin-specific protease HAUSP plays an important role in this process. Recent experiments showed that p53 demonstrates a differential response to changes in HAUSP which nature and significance are not understood yet. Here a data-driven mathematical model of the Mdm2-mediated p53 ubiquitination network is presented which offers an explanation for the cause of such a response. The model predicts existence of the HAUSP-regulated switch from auto- to p53 ubiquitination by Mdm2. This switch suggests a potential role of HAUSP as a downstream target of stress signals in cells. The model accounts for a significant amount of experimental data, makes predictions for some rate constants, and can serve as a building block for the larger model describing a complex dynamic response of p53 to cellular stresses.  相似文献   

11.
Lee HJ  Kim MS  Kim YK  Oh YK  Baek KH 《FEBS letters》2005,579(21):4867-4872
The tumor suppressor protein p53 is ubiquitinated and neddylated by MDM2 and then degraded by 26S proteasome. However, p53 is stabilized by the HAUSP (Herpes-virus-associated ubiquitin-specific protease) deubiquitinating enzyme. In this study, we discovered that rat HAUSP (rHAUSP) is polyubiquitinated, polyneddylated, and dimerized using co-immunoprecipitation assays. This suggests that rHAUSP may function as a dimer or multimer and is also degraded through the proteasome-mediated degradation. Transfection of rHAUSP into RGC-Lac-Z cell line with the integrated p53 response element revealed that rHAUSP contributed to p53 stabilization, and a rHAUSP (C224S) mutant contributed to p53 destabilization in a dose-dependent manner.  相似文献   

12.
Herpes simplex virus type 1 immediate-early protein Vmw110 stimulates the onset of virus infection and is required for efficient reactivation from latency. In transfection assays, Vmw110 is a potent activator of gene expression, but its mode of action has yet to be determined. Previous work has shown that Vmw110 localizes to specific intranuclear structures known as ND10, PML bodies, or PODs and causes the disruption of these domains. The ability of Vmw110 to disrupt ND10 correlates with its biological activities in infected and transfected cells. It has also been found that Vmw110 binds strongly and specifically to a ubiquitin-specific protease known as HAUSP, itself a component of a subset of ND10. In this study we have investigated the role of HAUSP in Vmw110 activity; single amino acid residues of Vmw110 required for the interaction were identified, and the effects of mutation of these residues in infected and transfected cells were then assayed. The results indicate that the ability to bind to HAUSP contributes to the functional activities of Vmw110.  相似文献   

13.
Hu M  Li P  Li M  Li W  Yao T  Wu JW  Gu W  Cohen RE  Shi Y 《Cell》2002,111(7):1041-1054
The ubiquitin-specific processing protease (UBP) family of deubiquitinating enzymes plays an essential role in numerous cellular processes. HAUSP, a representative UBP, specifically deubiquitinates and hence stabilizes the tumor suppressor protein p53. Here, we report the crystal structures of the 40 kDa catalytic core domain of HAUSP in isolation and in complex with ubiquitin aldehyde. These studies reveal that the UBP deubiquitinating enzymes exhibit a conserved three-domain architecture, comprising Fingers, Palm, and Thumb. The leaving ubiquitin moiety is specifically coordinated by the Fingers, with its C terminus placed in the active site between the Palm and the Thumb. Binding by ubiquitin aldehyde induces a drastic conformational change in the active site that realigns the catalytic triad residues for catalysis.  相似文献   

14.
15.
This study investigated the efficacy of Toxoplasma GRA16, which binds to herpes virus‐associated ubiquitin‐specific protease (HAUSP), in anticancer treatment, and whether the expression of GRA16 in genetically modified hepatocellular carcinoma (HCC) cells (GRA16‐p53‐wild HepG2 and GRA16‐p53‐null Hep3B) regulates PTEN because alterations in phosphatase and tensin homologue (PTEN) and p53 are vital in liver carcinogenesis and the abnormal p53 gene appears in HCC. For this purpose, we established the GRA16 cell lines using the pBABE retrovirus system, assessed the detailed mechanism of PTEN regulation in vitro and established the anticancer effect in xenograft mice. Our study showed that cell proliferation, antiapoptotic factors, p‐AKT/AKT ratio, cell migration and invasive activity were decreased in GRA16‐stable HepG2 cells. Conversely, the apoptotic factors PTEN and p53 and apoptotic cells were elevated in GRA16‐stable HepG2 cells but not in Hep3B cells. The change in MDM2 was inconspicuous in both HepG2 and Hep3B; however, the PTEN level was remarkably elevated in HepG2 but not in Hep3B. HAUSP‐bound GRA16 preferentially increased p53 stabilization by the nuclear localization of PTEN rather than MDM2‐dependent mechanisms. These molecular changes appeared to correlate with the decreased tumour mass in GRA16‐stable‐HepG2 cell‐xenograft nude mice. This study establishes that GRA16 is a HAUSP inhibitor that targets the nuclear localization of PTEN and induces the anticancer effect in a p53‐dependent manner. The efficacy of GRA16 could be newly highlighted in HCC treatment in a p53‐dependent manner.  相似文献   

16.
DNA damage can occur through diverse stimulations such as toxins, drugs, and environmental factors. To respond to DNA damage, mammalian cells induce DNA damage response (DDR). DDR signal activates a rapid signal transduction pathway, regulating the cell fate based on the damaged cell condition. Moreover, serious damaged cells have to be eliminated by the macrophage to maintain homeostasis. Because the DDR induces genomic instability followed by tumor formation, targeting the DDR signaling can be applied for the cancer therapy. Herpes virus-associated ubiquitin-specific protease (HAUSP/USP7) is one of the well-known deubiquitinating enzymes (DUBs) owing to its relevance with Mdm2-p53 complex. The involvement of HAUSP in DDR through p53 led us to investigate novel substrates for HAUSP, which is related to DDR or apoptosis. As a result, we identified annexin-1 (ANXA1) as one of the putative substrates for HAUSP. ANXA1 has numerous roles in cellular systems including anti-inflammation, damage response, and apoptosis. Several studies have demonstrated that ANXA1 can be modified in a post-translational manner by processes such as phosphorylation, SUMOylation, and ubiquitination. In addition, DNA damage gives various functions to ANXA1 such as stress response or cleavage-mediated apoptotic cell clearance. In the current study, our proteomic analysis using two-dimensional electrophoresis, matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF-MS) and nano LC-MS/MS, and immunoprecipitation revealed that ANXA1 binds to HAUSP through its HAUSP-binding motif (P/AXXS), and the cleavage and damage-responsive functions of ANXA1 upon UV-induced DNA damage may be followed by HAUSP-mediated deubiquitination of ANXA1. Intriguingly, the UV-induced damage responses via HAUSP-ANXA1 interaction in HeLa cells were different from the responses shown in the Jurkat cells, suggesting that their change of roles may depend on the cell types.Most proteins follow the ubiquitin-proteasome pathway (UPP) to degradation; this involves successive enzymatic activities of the E1, E2, and E3 enzymes. In addition to proteasomal degradation, the proteins obtain or alter their functions through mono- or polyubiquitination.1 Thus, the ‘ubiquitin tag'' is considered as an important feature for intracellular homeostasis. Deubiquitination is a reversible process against ubiquitination that detaches ubiquitin molecules from ubiquitinated proteins, and the process of deubiquitination is mediated by specific enzymes called deubiquitinating enzymes (DUBs). To date, almost ~100 DUBs have been identified, and they are involved in various cellular functions through their capability by which they deubiquitinate and thereby stabilize or alter the functions of their target proteins.2 DUBs are composed of at least six subfamilies: ubiquitin-specific proteases (USPs), ubiquitin C-terminal hydrolases (UCHs), ovarian tumor (OTU), Machado-Josephin domain papain-like cysteine proteases (MJDs), JAB1/MPN/Mov34 metalloenzyme (JAMM) domain zinc-dependent metalloprotease family, and monocyte chemotactic protein-induced proteases (MCPIPs).3 In addition, DUBs share specific regions including Cys, Asp/Asn, and His boxes for their deubiquitinating activities.4 The USP family has the most number among DUBs (~58 USPs),5 and many studies have demonstrated that human USPs have important roles in a broad range of cellular systems.6 In particular, their involvement in cell proliferation, signal transduction, and apoptosis emphasizes that abnormal or deregulated functions of USPs can be related to severe diseases including immune disorders and cancers.2, 6, 7 Accordingly, USPs have been widely targeted for the therapy of several diseases; however, a clear understanding of the molecular details underlining USPs and other DUBs has not yet been obtained.HAUSP, also known as USP7, is a member of the USP family of DUBs. The importance of HAUSP in cells was demonstrated by its ability to specifically recognize and deubiquitinate both the tumor suppressor p53 and Mdm2, a p53-specific E3 ligase. In the normal state, HAUSP specifically binds to and deubiquitinates Mdm2, thereby stabilizing Mdm2 and subsequently inducing the proteasomal degradation of p53 through Mdm2 activity. Upon DNA damage, HAUSP is dephosphorylated by PPM1G. In this state, the deubiquitinating activity of HAUSP for Mdm2 decreases and HAUSP prefers p53 for its substrate instead of Mdm2. Such altered affinity of HAUSP to p53 leads to DNA repair and tumor-suppressive functions of p53.8, 9, 10 In addition to Mdm2 and p53, further studies have revealed that HAUSP can regulate various substrates, including ataxin-1, Chfr, claspin, Daxx, FOXO4, histone H2B, PTEN, NF-κB, Tip60, UbE2E1, and UVSSA.2 These findings suggest that HAUSP has diverse roles in the cell through the regulation of different substrates and other additional proteins. In a present study, we performed two-dimensional gel electrophoresis (2-DE) and other proteomics-based experiments using HeLa cells to identify putative substrates regulated by HAUSP. We found several putative substrates, some of which are known to be involved in apoptosis or DNA damage response (DDR). Annexin a1, also known as ANXA1 and lipocortin 1, was also found as a putative binding partner for HAUSP, suggesting that ANXA1 may possibly be regulated by HAUSP-mediated deubiquitination.Annexins consist of 13 annexin members and have four conserved repeated domains, which are responsible for Ca2+ and phospholipid binding. In most annexins, the conserved annexin domains enable them to bind the phospholipid of the membranes in a Ca2+-dependent manner, resulting in subsequent activities such as membrane trafficking, signal transduction, and exocytosis.11 However, major differences of annexins derive from their unique N-terminal regions. The N-terminus of each annexin member, which is responsible for specific functions, varies.12 ANXA1, the first member of the annexin superfamily, is a 37-kDa protein abundant in cells. Like other annexin proteins, ANXA1 binds to phospholipid in the presence of Ca2+.13 The biological functions of ANXA1 are extensively studied: anti-inflammatory mediator,14, 15 relationship with tumorigenesis,16 DDR,17, 18 and involvement in apoptosis and apoptotic cell clearance.19, 20 Another important feature of ANXA1 activity is the cleavage of the N-terminal region of ANXA1. When DNA damage or stress occurs, ANXA1 is cleaved by several proteases, resulting in the generation of the N-terminal fragment (Ac2-26) and cleaved form of ANXA1 (33 kDa). Importantly, both the full-length ANXA1 and Ac2-26 can be translocated to the cell membrane and induce apoptotic cell clearance by recruiting monocytes via chemoattraction.20 Thus, the ANXA1 cleavage process is considered essential for cell phagocytosis, as also revealed in neutrophil apoptosis and phagocytosis during inflammation.14 Otherwise, in response to cell damage, ANXA1 functions as a stress protein or a protective protein for DNA damage, resulting in nuclear localization of ANXA1.18, 21, 22 Overall, it is evident that ANXA1 participates in various cellular responses.In the current study, we have identified ANXA1 as a novel substrate for HAUSP. HAUSP can bind to, deubiquitinate, and co-localize with ANXA1. Surprisingly, upon UV-induced DNA damage, the binding and the deubiquitinating activity of HAUSP to ANXA1 are increased. In addition, ANXA1 in HAUSP-deficient cells showed different localization and altered expression level and cleavage. Moreover, HAUSP-mediated regulation of ANXA1 shown in HeLa cells was different from the one in Jurkat cells. We found that apoptosis and transmigrative ratio of monocytes in HAUSP-depleted Jurkat cells coincides with the regulation of ANXA1 protein level and cleavage. Taken together, we suggest that ANXA1 functions of UV-induced DDR are regulated by the deubiquitinating activity of HAUSP.  相似文献   

17.
The ubiquitin-specific protease USP7/HAUSP regulates p53 and MDM2 levels, and cellular localization of FOXO4 and PTEN, and hence is critically important for their role in cellular processes. Here we show how the 64 kDa C-terminal region of USP7 can positively regulate deubiquitinating activity. We present the crystal structure of this USP7/HAUSP ubiquitin-like domain (HUBL) comprised of five ubiquitin-like (Ubl) domains organized in 2-1-2 Ubl units. The last di-Ubl unit, HUBL-45, is sufficient to activate USP7, through binding to a "switching" loop in the catalytic domain, which promotes ubiquitin binding and increases activity 100-fold. This activation can be enhanced allosterically by the metabolic enzyme GMPS. It binds to the first three Ubl domains (HUBL-123) and hyperactivates USP7 by stabilization of the HUBL-45-dependent active state.  相似文献   

18.
19.
20.
p53 ubiquitination: Mdm2 and beyond   总被引:12,自引:0,他引:12  
Brooks CL  Gu W 《Molecular cell》2006,21(3):307-315
Although early studies have suggested that the oncoprotein Mdm2 is the primary E3 ubiquitin ligase for the p53 tumor suppressor, an increasing amount of data suggests that p53 ubiquitination and degradation are more complex than once thought. The discoveries of MdmX, HAUSP, ARF, COP1, Pirh2, and ARF-BP1 continue to uncover the multiple facets of this pathway. There is no question that Mdm2 plays a pivotal role in downregulating p53 activities in numerous cellular settings. Nevertheless, growing evidence challenges the conventional view that Mdm2 is essential for p53 turnover.  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