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.
Jung JR  Kim H  Jeun SS  Lee JY  Koh EJ  Ji C 《Molecules and cells》2005,20(2):196-200
The neurofibromatosis type2 (NF2) tumor suppressor gene product, merlin, is structurally related to the ezrin-radixin-moesin (ERM) family of proteins that anchor the actin cytoskeleton to specific membrane proteins and participate in cell signaling. However, the basis of the tumor suppressing activity of merlin is not well understood. Previously, we identified a role of merlin as an inhibitor of the Ras-ERK signaling pathway. Recent studies have suggested that phosphorylation of merlin, as of other ERM proteins, may regulate its function. To determine whether phosphorylation of merlin affects its suppression of Ras-ERK signaling, we generated plasmids expressing full-length merlin with substitutions of serine 518, a potential phosphorylation site. A substitution that mimics constitutive phosphorylation (S518D) abrogated the ability of merlin to suppress effects of the Ras-ERK signaling pathway such as Ras-induced SRE transactivation, Elk-mediated SRE transactivation, Ras-induced ERK phosphorylation and Ras-induced focus formation. On the other hand, an S518A mutant, which mimics nonphosphorylated merlin, acted like wild type merlin. These observations show that mimicking merlin phosphorylation impairs not only growth suppression by merlin but also its inhibitory action on the Ras-ERK signaling pathway.  相似文献   

2.
3.
Ras-induced cell transformation is mediated through distinct downstream signaling pathways, including Raf, Ral-GEFs-, and phosphatidylinositol 3-kinase (PI 3-kinase)-dependent pathways. In some cell types, strong activation of the Ras-Raf-MEK-extracellular signal-regulated kinase (ERK) cascade leads to cell cycle arrest rather than cell division. We previously reported that constitutive activation of this pathway induces sustained proliferation of primary cultures of postmitotic chicken neuroretina (NR) cells. We used this model system to investigate the respective contributions of Ras downstream signaling pathways in Ras-induced cell proliferation. Three RasV12 mutants (S35, G37, and C40) which differ by their ability to bind to Ras effectors (Raf, Ral-GEFs, and the p110 subunit of PI 3-kinase, respectively) were able to induce sustained NR cell proliferation, although none of these mutants was reported to transform NIH 3T3 cells. Furthermore, they all repressed the promoter of QR1, a neuroretina growth arrest-specific gene. Overexpression of B-Raf or activated versions of Ras effectors Rlf-CAAX and p110-CAAX also induced NR cell division. The mitogenic effect of the RasC40-PI 3-kinase pathway appears to involve Rac and RhoA GTPases but not the antiapoptotic Akt (protein kinase B) signaling. Division induced by RasG37-Rlf appears to be independent of Ral GTPase activation and presumably requires an unidentified mechanism. Activation of either Ras downstream pathway resulted in ERK activation, and coexpression of a dominant negative MEK mutant or mKsr-1 kinase domain strongly inhibited proliferation induced by the three Ras mutants or by their effectors. Similar effects were observed with dominant negative mutants of Rac and Rho. Thus, both the Raf-MEK-ERK and Rac-Rho pathways are absolutely required for Ras-induced NR cell division. Activation of these two pathways by the three distinct Ras downstream effectors possibly relies on an autocrine or paracrine loop, implicating endogenous Ras, since the mitogenic effect of each Ras effector mutant was inhibited by RasN17.  相似文献   

4.
We report here that immediate early gene pip92 is expressed during anisomycin-induced cell death in fibroblast NIH3T3 cells. To determine the mechanism by which this occurs and to identify downstream signaling pathways, we investigated the induction of the pip92 promoter. The activation of pip92 by anisomycin is mediated by the activation of MAP kinases, such as JNK and p38 kinase, but not ERK. Deletion analysis of the pip92 promoter indicated that pip92 activation occurs primarily within the region containing a serum response element (SRE). Further analysis of the SRE using a heterologous thymidine kinase promoter showed that both an Ets and CArG-like site are required for anisomycin-induced pip92 expression. Elk1, which binds to the Ets site, was phosphorylated by the JNK- and p38-dependent pathways and the phosphorylation of Elk1-GAL4 fusion proteins by these pathways was sufficient for the transactivation. Overall, this study suggested that different MAPK pathways are involved in the expression of immediate early gene pip92 by growth factors and environmental stresses.  相似文献   

5.
6.
7.
The neurofibromatosis-2 (NF2) tumor suppressor protein, merlin or schwannomin, inhibits cell proliferation by modulating the growth activities of its binding partners, including the cell surface glycoprotein CD44, membrane-cytoskeleton linker protein ezrin and PIKE (PI 3-kinase Enhancer) GTPase etc. Merlin exerts its growth suppressive activity through a folded conformation that is tightly controlled through phosphorylation by numerous protein kinases including PAK, PKA and Akt. Merlin inhibits PI 3-kinase activity through binding to PIKE-L. Now, we show that merlin is a physiological substrate of Akt, which phosphorylates merlin on both T230 and S315 residues. This phosphorylation abolishes the folded conformation of merlin and inhibits its association with PIKE-L, provoking merlin polyubiquitination and proteasome-mediated degradation. This finding demonstrates a negative feed-back loop from merlin/PIKE-L/PI 3-kinase to Akt in tumors. The proliferation repressive activity of merlin is also partially regulated by S518 phosphorylation. Thus, Akt-mediated merlin T230/S315 phosphorylation, combined with S518 phosphorylation by PAK and PKA, provides new insight into abrogating merlin function in the absence of merlin mutational inactivation.  相似文献   

8.
9.
10.
Constitutive activation of M-Ras has previously been reported to cause morphologic and growth transformation of murine cells, suggesting that M-Ras plays a role in tumorigenesis. Cell transformation by M-Ras correlated with weak activation of the Raf/MEK/ERK pathway, although contributions from other downstream effectors were suggested. Recent studies indicate that signaling events distinct from the Raf/MEK/ERK cascade are critical for human tumorigenesis. However, it is unknown what signaling events M-Ras triggers in human cells. Using constitutively active M-Ras (Q71L) containing additional mutations within its effector-binding loop, we found that M-Ras induces MEK/ERK-dependent and -independent Elk1 activation as well as phosphatidylinositol 3 kinase (PI3K)/Akt and JNK/cJun activation in human MCF-7 breast cancer cells. Among several human cell lines examined, M-Ras-induced MEK/ERK-independent Elk1 activation was only detected in MCF-7 cells, and correlated with Rlf/M-Ras interaction and Ral/JNK activation. Supporting a role for M-Ras signaling in breast cancer, EGF activated M-Ras and promoted its interaction with endogenous Rlf. In addition, constitutive activation of M-Ras induced estrogen-independent growth of MCF-7 cells that was dependent on PI3K/Akt, MEK/ERK, and JNK activation. Thus, our studies demonstrate that M-Ras signaling activity differs between human cells, highlighting the importance of defining Ras protein signaling within each cell type, especially when designing treatments for Ras-induced cancer. These findings also demonstrate that M-Ras activity may be important for progression of EGFR-dependent tumors.  相似文献   

11.
12.
Activation of Ras signaling by growth factors has been associated with gene regulation and cell proliferation. Here we characterize the contributory role of cytosolic phospholipase A(2) in the oncogenic Ha-Ras(V12) signaling pathway leading to activation of c-fos serum response element (SRE) and transformation in Rat-2 fibroblasts. Using a c-fos SRE-luciferase reporter gene, we showed that the transactivation of SRE by Ha-Ras(V12) is mainly via a Rac-linked cascade, although the Raf-mitogen-activated protein kinase cascade is required for full activation. In addition, Ha-Ras(V12)-induced DNA synthesis was significantly attenuated by microinjection of recombinant Rac(N17), a dominant negative mutant of Rac1. To identify the mediators downstream of Rac in the Ha-Ras(V12) signaling, we investigated the involvement of cytosolic phospholipase A(2). Oncogenic Ha-Ras(V12)-induced SRE activation was significantly inhibited by either pretreatment with mepacrine, a phospholipase A(2) inhibitor, or cotransfection with the antisense oligonucleotide of cytosolic phospholipase A(2). We also found cytosolic phospholipase A(2) to be situated downstream of Ha-Ras(V12) in a signal pathway leading to transformation. Together, these results are indicative of mediatory roles of Rac and cytosolic phospholipase A(2) in the signaling pathway by which Ha-Ras(V12) transactivates c-fos SRE and transformation. Our findings point to cytosolic phospholipase A(2) as a novel potential target for suppressing oncogenic Ha-Ras(V12) signaling in the cell.  相似文献   

13.
14.
15.
Tang X  Jang SW  Wang X  Liu Z  Bahr SM  Sun SY  Brat D  Gutmann DH  Ye K 《Nature cell biology》2007,9(10):1199-1207
The neurofibromatosis-2 (NF2) tumour-suppressor gene encodes an intracellular membrane-associated protein, called merlin, whose growth-suppressive function is dependent on its ability to form interactions through its intramolecular amino-terminal domain (NTD) and carboxy-terminal domain (CTD). Merlin phosphorylation plays a critical part in dictating merlin NTD/CTD interactions as well as in controlling binding to its effector proteins. Merlin is partially regulated by phosphorylation of Ser 518, such that hyperphosphorylated merlin is inactive and fails to form productive intramolecular and intermolecular interactions. Here, we show that the protein kinase Akt directly binds to and phosphorylates merlin on residues Thr 230 and Ser 315, which abolishes merlin NTD/CTD interactions and binding to merlin's effector protein PIKE-L and other binding partners. Furthermore, Akt-mediated phosphorylation leads to merlin degradation by ubiquitination. These studies demonstrate that Akt-mediated merlin phosphorylation regulates the function of merlin in the absence of an inactivating mutation.  相似文献   

16.
The p38 pathway provides negative feedback for Ras proliferative signaling   总被引:15,自引:0,他引:15  
Ras activates three mitogen-activated protein kinases (MAPKs) including ERK, JNK, and p38. Whereas the essential roles of ERK and JNK in Ras signaling has been established, the contribution of p38 remains unclear. Here we demonstrate that the p38 pathway functions as a negative regulator of Ras proliferative signaling via a feedback mechanism. Oncogenic Ras activated p38 and two p38-activated protein kinases, MAPK-activated protein kinase 2 (MK2) and p38-related/activated protein kinase (PRAK). MK2 and PRAK in turn suppressed Ras-induced gene expression and cell proliferation, whereas two mutant PRAKs, unresponsive to Ras, had little effect. Moreover, the constitutive p38 activator MKK6 also suppressed Ras activity in a p38-dependent manner whereas arsenite, a potent chemical inducer of p38, inhibited proliferation only in a tumor cell line that required Ras activity. MEK was required for Ras stimulation of the p38 pathway. The p38 pathway inhibited Ras activity by blocking activation of JNK, without effect upon ERK, as evidenced by the fact that PRAK-mediated suppression of Ras-induced cell proliferation was reversed by coexpression of JNKK2 or JNK1. These studies thus establish a negative feedback mechanism by which Ras proliferative activity is regulated via signaling integrations of MAPK pathways.  相似文献   

17.
Sef was recently identified as a negative regulator of fibroblast growth factor (FGF) signaling in a genetic screen of zebrafish and subsequently in mouse and humans. By inhibiting FGFR1 tyrosine phosphorylation and/or Ras downstream events, Sef inhibits FGF-mediated ERK activation and cell proliferation as well as PC12 cell differentiation. Here we show that Sef and a deletion mutant of Sef lacking the extracellular domain (SefIC) physically interact with TAK1 (transforming growth factor-beta-associated kinase) and activate JNK through a TAK1-MKK4-JNK pathway. Sef and SefIC overexpression also resulted in apoptotic cell death, while dominant negative forms of MKK4 and TAK1 blocked Sef-mediated JNK activation and attendant 293T cell apoptosis. These investigations reveal a novel activating function of Sef that is distinct from its inhibitory effect on FGF receptor signaling and ERK activation.  相似文献   

18.
The neurofibromatosis-2 (NF2) tumor suppressor protein, merlin or schwannomin, inhibits cell proliferation by modulating the growth activities of its binding partners, including the cell surface glycoprotein CD44, membrane-cytoskeleton linker protein ezrin and PIKE (PI 3-kinase enhancer) GTPase, etc. Merlin exerts its growth suppressive activity through a folded conformation that is tightly controlled through phosphorylation by numerous protein kinases including PAK, PKA and Akt. Merlin inhibits PI 3-kinase activity through binding to PIKE-L. Now, we show that merlin is a physiological substrate of Akt, which phosphorylates merlin on both T230 and S315 residues. This phosphorylation abolishes the folded conformation of merlin and inhibits its association with PIKE-L, provoking merlin polyubiquitination and proteasome-mediated degradation. This finding demonstrates a negative feed-back loop from merlin/PIKE-L/PI 3-kinase to Akt in tumors. The proliferation repressive activity of merlin is also partially regulated by S518 phosphorylation. Thus, Akt-mediated merlin T230/S315 phosphorylation, combined with S518 phosphorylation by PAK and PKA, provides new insight into abrogating merlin function in the absence of merlin mutational inactivation.Key Words: Akt, merlin, neurofibromatosis, phosphorylation, cell invasion and migrationNeurofibromatosis 2 (NF2) is a dominantly inherited disorder characterized by bilateral occurrence of vestibular schwannomas and other brain tumors, including meningiomas and ependymomas.1 The NF2 tumor suppressor protein merlin belongs to the band 4.1 family of cytoskeleton-associated proteins.2,3 Merlin isoform I possesses a “closed” conformation via an NTD (N-terminal domain)/CTD (Carboxy terminal domain) intramolecular interaction. In contrast, the alternatively spliced merlin isoform II exists in an “open” conformation that cannot function as a negative growth regulator.4 Merlin with NF2 patient missense mutations in the NTD or CTD exhibit an “open” conformation and do not suppress cell growth.5 Merlin plays a key role in regulating cell proliferation and cell migration. Merlin exerts its growth suppressive activity through intramolecular folding that dictates its binding affinities to various cellular partners including HRS (hepatocyte growth factor regulated tyrosine kinase substrate), CD44 cell surface glycoprotein, schwannomin interacting protein-1 (SCHIP1), βII-spectrin or fodrin, PIKE-L GTPase and other ERM proteins.610 For instance, CD44 preferentially associates with hypophosphorylated merlin, and relatively little phosphorylated merlin binds CD44. Interference with merlin binding to CD44 impairs merlin growth suppression in RT4 rat schwannoma cells.11We have previously shown that the PIKE/PI 3-kinase signaling pathway is negatively regulated by protein 4.1N, a neuronal selective isoform of band 4.1 superfamily.12 Recently, we show that PIKE-L is an important mediator of merlin growth suppression. We show that merlin blocks cell proliferation by inhibiting PI 3-kinase through binding to PIKE-L. Interestingly, wild-type merlin, but not patient-derived mutant (L64P), binds PIKE-L and inhibits PI 3-kinase activity. This suppression of PI 3-kinase activity results from merlin disrupting the binding of PIKE-L to PI 3-kinase. Mutation of PIKE-L with Proline 187 into Leucine disrupts its interaction with merlin. Accordingly, merlin suppression of PI 3-kinase activity as well as schwannoma cell growth is abrogated by a single PIKE-L point mutation (P187L).10Merlin is phosphorylated on S518 by members of the PAK family of kinases, including PAK1 and PAK2,1315 which mislocates merlin from the plasma membrane to the cytoplasm. A merlin mutant that mimics S518 phosphorylation (S518D) cannot suppress cell growth or motility in RT4 rat schwannoma cells, and leads to dramatic changes in cell morphology and actin cytoskeleton organization.16 S518 phosphorylation results in impaired merlin NTD/CTD folding as well as altered interactions with critical merlin associated proteins, including CD44 and HRS.17 Recently, Alfthan and colleagues demonstrated that Protein Kinase-A (PKA) induces merlin phosphorylation on both N-and C-terminal residues.18 In addition to S518 phosphorylation, PKA can phosphorylate merlin at S66 in the N-terminal domain (Fig. 1). When PAK activity is suppressed, merlin can still be phosphorylated by PKA in cells, indicating that these two kinases function independently. The N-terminus of ezrin strongly binds to a PKA-phosphorylated, but not unphosphorylated, merlin CTD. In contrast, PAK2-induced S518 phosphorylation has a minimal effect on the interaction between full-length merlin and full-length ezrin.17 Besides regulation of cell growth, merlin also mediates cell motility presumably through directly binding to actin cytoskeleton.19 Depletion of merlin in normal fibroblast results in enhanced cell invasion. Nevertheless, expression of merlin attenuates Y397 phosphorylation on FAK, an essential player in cell migration and invasion. This observation might provide a molecular mechanism accounting for merlin inhibitory activity in cell motility.20Open in a separate windowFigure 1Merlin phosphorylation sites by various kinases.In addition to PAK and PKA, we show that Akt potently phosphorylates merlin at both T230 and S315 residues. Blocking one site phosphorylation abolishes the other site phosphorylation by Akt, indicating that these two phosphorylation sites are mutually regulated.21 The physiological significance of the tight control on merlin phosphorylation by Akt remains incompletely understood. Presumably, only when mitogenic signal or oncogenic stress is strongly enough to provoke cell proliferation or migration, does Akt simultaneously phosphorylate both sites. Akt phosphorylation of merlin attenuates the NTD/CTD interaction and inhibits its binding activity to PIKE-L, CD44 and ezrin. Further, phosphorylation mediates the biological activities of merlin, as expression of a phosphomimetic merlin mutant (T230DS315D) increases cell motility and proliferation in a rat schwannoma cell line (Fig. 2). By contrast, expression of a mutant (T230A/S315A) that was unable to undergo phosphorylation inhibited cell growth and motility. The F1 motif in FERM proteins including merlin exhibits an ubiquitin-like structure. This domain facilitates MDM2 degradation and stimulates the ubiquitination and degradation of TRBP, a double-stranded RNA binding protein. Surprisingly, inhibition of the proteasome does not affect total merlin protein levels in human glioblastoma cells, but leads to a marked increase of phospho-S315 merlin. Simultaneous treatment with MG132, which blocks proteasome-mediated degradation and PI 3-kinase inhibitor, wortmannin, which inhibits Akt phosphorylation of merlin, substantially enhances merlin levels. Coimmunoprecipitation studies demonstrate that Akt-phosphorylated merlin is rapidly ubiquitinated, presumably by spectrin, which binds to merlin and possesses ubiquitin-conjugating and ubiquitin E3 ligase function. However, S518 phosphorylation fails to trigger merlin ubiquitination, suggesting that Akt, but not PAK or PKA, phosphorylation selectively elicits merlin ubiquitination. Using a panel of human primary nervous system tumors, we found that merlin phosphorylation by Akt also mediates its degradation in primary tumors. Accordingly, tumors that possess high levels of phospho-Akt exhibited low levels of merlin. Therefore, our data suggest a novel role for Akt in promoting phosphorylation and subsequent degradation of merlin. Loss of merlin has been linked to schwannomas and other nervous system tumors, and these results indicate that inhibitors for PI 3-kinase/Akt pathway might restore merlin function in tumors.Open in a separate windowFigure 2The model for Akt interaction with merlin and its phosphorylation.  相似文献   

19.
20.
L-ascorbic acid (LAA) shows cytotoxicity and induces apoptosis of malignant cells in vitro, but the mechanisms by which such effects occur have not been elucidated. In the present study, we provide evidence that the ERK MAP kinase pathway is activated in response to LAA (< 1 mM) in acute myeloid leukemia cell lines. LAA treatment of cells induces a dose-dependent phosphorylation of extracellular signal-regulated kinases (ERK) and results in activation of its catalytic domain. Our data also demonstrate that the small G protein Raf1 and MAPK-activated protein kinase 2 are activated by LAA as an upstream and a downstream regulator of ERK, respectively. Although the ERK pathway has been known to activate cell proliferation, pharmacologic inhibition of ERK reduces LAA-dependent apoptosis and growth inhibitory response of acute myeloid leukemia cell lines, suggesting that this signaling cascade positively regulates induction of apoptotic response by LAA.  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