当前位置: 首页 > 医学版 > 期刊论文 > 基础医学 > 病菌学杂志 > 2005年 > 第23期 > 正文
编号:11202066
Activation of p21-Activated Kinase 2 by Human Immu
     Department of Internal Medicine, Division of Infectious Diseases, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390

    Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111

    ABSTRACT

    The accessory human immunodeficiency virus type 1 (HIV-1) protein Nef activates the autophosphorylation activity of p21-activated kinase 2 (PAK2). Merlin, a cellular substrate of PAK2, is homologous to the ezrin-radixin-moesin family and plays a critical role in Rac signaling. To assess the possible impact on host cell metabolism of Nef-induced PAK2 activation, we investigated the phosphorylation of merlin in Nef expressing cells. Here we report that Nef induces merlin phosphorylation in multiple cell lines independently of protein kinase A. This intracellular phosphorylation of merlin directly correlates with in vitro assay of the autophosphorylation activity of Nef-activated PAK2. Importantly, merlin phosphorylation induced by Nef was also observed in human primary T cells. The finding that Nef induces phosphorylation of the key signaling molecule merlin suggests several possible roles for PAK2 activation in HIV pathogenesis.

    TEXT

    Nef is a small, multifunctional protein encoded by human immunodeficiency virus type 1 (HIV-1), HIV-2, and simian immunodeficiency virus. The in vivo simian model indicated that Nef is a major determinant of viral pathogenicity (11). Four of Nef's numerous known functions include down-regulation of cell surface CD4 and major histocompatibility complex (MHC) class I, enhancement of the infectivity of viral particles and activation of p21-activated kinase 2 (PAK2) (3, 19, 22, 25). The significance of Nef's ability to activate PAK2 is controversial since it is not clear whether the level of activation is sufficient to alter cellular metabolism (14, 20).

    PAK2 is a serine/threonine kinase activated by the GTPases Rac1 and Cdc42 through the CRIB (Cdc42-Rac1 interactive binding) domain. Merlin, the product of the neurofibromatosis 2 tumor suppressor gene (24, 29), is a key signaling protein involved in the Rac signal transduction pathway (27, 33). It has recently been shown to be phosphorylated at serine-518 by PAK2 and cyclic AMP-dependent protein kinase A (PKA) (1, 12, 23, 32). Merlin is a 595-amino-acid protein that resembles other members of the band 4.1 family, such as ezrin, radixin, and moesin (ERM), in its N-terminal half (24, 29). It is a negative regulator of cell proliferation, cell motility, and Rac1 signaling (33). Phosphorylation of merlin at serine-518 results in an inactive molecule thereby releasing its inhibitory effects (13, 34).

    Nef induces merlin phosphorylation. If Nef significantly alters metabolism of the infected cell by activating PAK2, then it would be expected that increased phosphorylation of merlin would result. We first looked for Nef-induced phosphorylation of merlin in human CEM (T) and U937 (monocytic) cells which express high levels of merlin and are often used in HIV research. We stably transduced these cell lines with either a control retroviral vector (LXSN) or a retroviral vector expressing HIV-1 SF2 Nef (LnefSNSF2) (4). Cells were lysed, and supernatants of whole cell lysates were analyzed by Western blotting (Fig. 1A). Merlin from control CEM and U937 cells migrates in sodium dodecyl sulfate-polyacrylamide gels as two bands with similar intensities. As reported by others, the slower migrating species represents a hyperphosphorylated form of merlin, and the faster migrating species represents a hypophosphorylated form (16, 27, 32). Interestingly, in the cells that expressed Nef, an increase in the amount of the slower migrating form (top band) was observed, and it became the dominant species (Fig. 1A). These experiments indicated that Nef expression induced merlin phosphorylation.

    In the cell, merlin can be phosphorylated by either PKA or p21-activated kinases at S518 (1, 12, 32). To investigate whether PKA activity is involved in Nef-mediated merlin phosphorylation, we treated control and Nef-expressing CEM and U937 cells with H89 (25 μM), a selective PKA inhibitor, for 5 h (7). H89 eliminated the hyperphosphorylated (slower migrating) merlin species in the control samples (Fig. 1B). These results indicated that in the absence of Nef, PKA is responsible for the basal level of merlin phosphorylation. However, when Nef-expressing cells were treated with H89, merlin shifted to the slower migrating species (Fig. 1B). Similar results were obtained in 293T cells (Fig. 1C, lanes 5 and 6). We then determined if a mutant merlin replacing a serine at position 518 with alanine was phosphorylated in response to Nef in transient transfection experiments by using hemagglutinin (HA)-tagged merlin in 293T cells. The S518A merlin mutant migrated in the hypophosphorylated (faster migrating) form (Fig. 1C, lane 2). Nef expression did not alter its migration, suggesting that S518 is the main site of phosphorylation induced by Nef (Fig. 1C, lane 4). We further used an affinity-purified phosphoserine-518-merlin antibody to confirm that serine-518 is phosphorylated in the presence of Nef. We transfected 293T cells with a control vector, a vector expressing S518A mutant merlin, a vector expressing wild-type merlin, or vectors expressing wild-type merlin and SF2 Nef. The cell lysates were analyzed by Western blotting using either anti-merlin antibody or affinity purified anti-phosphoserine-518-merlin antibody. As shown in Fig. 1D, the S518A mutant merlin migrated in the hypophosphorylated form (lane 2, middle panel), while wild-type merlin migrated in both hyper- and hypophosphorylated forms (lane 3, middle panel). However, in the presence of Nef, wild-type merlin migrated mainly in the hyperphosphorylated form (lane 4, middle panel). The phosphoserine-518-merlin antibody detected only the hyperphosphorylated merlin (Fig. 1D, lane 3 and 4, top panel). Collectively, the experiments described above indicate that Nef-induced merlin phosphorylation at serine-518 is independent of PKA activity in CEM, U937, or 293T cells.

    Phosphorylation of merlin is dependent on PAK2 activation. We have previously demonstrated that a conserved phenylalanine in the C-terminal region of Nef (position 195 of SF2 Nef) is essential for PAK2 activation but not for CD4 downregulation, MHC class I downregulation, or the enhancement of viral infectivity (9). When the SF2 Nef F195 was mutated to isoleucine or arginine, the mutant lost its ability to activate PAK2 but retained the activities of CD4 or MHC class I downregulation and enhancement of infectivity (9) (data not shown). When SF2 Nef or the F195R mutant was coexpressed in 293T cells with merlin, Western blot analysis revealed that in cells expressing Nef F195R, there was no shift of the hypophosphorylated band, whereas in SF2 Nef expressing cells, there was a significant shift of this band (Fig. 2). Furthermore, HIV-1 D88-11 Nef, a primary isolate which was previously shown to be defective in PAK2 activation (9) did not alter merlin phosphorylation levels. However, replacing R189 with serine in D88-11 Nef, an alteration that restores the ability of D88-11 to activate PAK2 (9), also restored merlin phosphorylation (Fig. 2). These results strongly link PAK2 activation to Nef-induced merlin phosphorylation.

    Nef-associated PAK2 phosphorylates GST-merlin in vitro. In order to establish whether Nef-associated PAK2 phosphorylates merlin directly, we immunoprecipitated the Nef/PAK2 complex from 293T cells transfected with SF2 Nef or D88-11 Nef. We then incubated the immunoprecipitates with glutathione S-transferase (GST)-tagged merlin in the presence of [-32P]ATP. The GST-merlin fusion protein contained amino acids 478 to 535 of merlin and was expressed in Escherichia coli and purified on glutathione-Sepharose beads (12). Phosphorylation of GST-merlin was visualized by autoradiography. As shown in Fig. 3A, only immunoprecipitates from SF2 Nef transfected cells showed PAK2 kinase activity and trans-phosphorylation of GST-tagged merlin. To determine if merlin S518 can serve as a substrate of PAK2 activated by SF2 Nef outside the context of the cellular milieu, we also used a recently described cell-free system to translate Nef mRNA in the presence of canine microsomal membranes to produce the Nef/PAK2 activated complex (21). As shown in Fig. 3B, wild-type GST-merlin, but not GST-merlin A518 was phosphorylated in immunoprecipitates from translation reactions containing Nef mRNA. Taken together, these in vitro results serve to substantiate the conclusion that Nef activated PAK2 phosphorylates merlin.

    Nef induces merlin phosphorylation in human primary T cells. We next investigated whether Nef induced activation of PAK2 in human primary T cells resulted in merlin phosphorylation. Three types of vesicular stomatitis virus G protein (VSV-G)-pseudotyped env-negative HIV-1 with an intact SF2 nef, a frameshift mutant nef, or nef F195R were used to infect activated primary human T cells. After infection cells were harvested and the expression of merlin, Nef, and Gag from the cell extracts was determined by Western blot analysis. As in the cell lines evaluated above, in activated human T cells, merlin was present as two species (Fig. 4, lane 1). Also, as observed with cell lines, the hyperphosphorylated merlin band was sensitive to the PKA inhibitor H89 (Fig. 4, lane 2). When T cells were infected with a virus containing an intact nef in the presence of H89, we observed the expected shift from the hypophosphorylated form to the hyperphosphorylated form (Fig. 4, lane 3). T cells infected by viruses with either a frameshift mutation (Nef) or Nef F195R resulted in no shift to the hyperphosphorylated form (Fig. 4, lanes 4 and 5). These results show that merlin is a bona fide target for Nef activated PAK2 in human primary T cells.

    The pathogenic significance of the activation of PAK2 by Nef has been difficult to establish. We attribute this situation, in part, to the extremely low level of the Nef/PAK2 activation complex in the cell. Here we report that merlin is strongly regulated by Nef through activation of PAK2. Our results suggest that the main site of phosphorylation on merlin in response to Nef is S518. These data are consistent with the results previously published by several other laboratories that identified S518 as the major site of merlin phosphorylation by PAK2 (12, 23, 27, 28, 32, 34). The multiple reported roles for merlin in cellular signaling, which include cell proliferation, cell motility, and Rac signaling (33), suggest several possible alterations of host cell metabolism that could be advantageous for viral replication.

    Cytoskeletal remodeling is critical in several steps following the stimulation of T-cell receptors (2, 8, 31, 35). Since merlin interacts with actin and ERM proteins, some of these steps might involve activated merlin. In the presence of HIV-1 Nef, merlin was hyperphosphorylated. As a result, this modification may promote actin assembly and positively regulate T-cell activation (17). A possible connection between merlin and Nef's function in T-cell activation is also supported by the demonstration that PAK2 is a positive regulator for T-cell activation and that this effect is dependent on its kinase activity (6). Although the downstream networks of PAK2 leading to T-cell activation were not examined by these authors, it is conceivable that merlin plays a role as a downstream mediator after PAK2.

    Nef increases T-cell activation by lowering the T-cell activation threshold (26). Furthermore, expression of Nef in primary resting cells following exposure to HIV-1 results in enhanced activation and enhanced viral production (26, 30). Nef-induced merlin phosphorylation may play a role in promoting T-cell activation and proliferation. Although HIV-1 preferentially infects activated/proliferating T cells, Johnson and Kaur (10) recently suggested that "resting" and "activated" may be oversimplification of the range of cellular states exhibited by T cells. Two recent studies found highly efficient depletion of memory CD4+ T cells from mucosal surfaces within a few days of infecting macaques with simian immunodeficiency virus even though these cells are noncycling (15, 18). Though these cells are immunophenotypically "resting," their susceptibility to HIV-1 infection suggests that they are not truly resting T cells (15, 18). Enhancing viral production in these cells through the proliferative signal of merlin phosphorylation is a possible mechanism for enhancing viral production. Early, massive production of virus is thought to set the parameters of the subsequent chronic disease (5). Future studies will assess a possible role for Nef-induced phosphorylation of merlin at this early juncture of HIV-1 infection.

    ACKNOWLEDGMENTS

    We thank A. Shaw for the preparation of expression plasmids and L. Baugh for expert assistance with flow cytometry analysis, T. Jacks for the GST-merlin fusion plasmids, and J. Allan for mouse anti-p24 monoclonal antibody (AG3.0) through the AIDS Research and Reference Program.

    This work was supported by National Institutes of Health grants AI-33331 (J.V.G.) and CA-45745 (J.R.T.). Alexa Raney was supported in part by National Institutes of Health training grant 5 F32 AI058541-02.

    REFERENCES

    Alfthan, K., L. Heiska, M. Gronholm, G. H. Renkema, and O. Carpen. 2004. Cyclic AMP-dependent protein kinase phosphorylates merlin at serine 518 independently of p21-activated kinase and promotes merlin-ezrin heterodimerization. J. Biol. Chem. 279:18559-18566.

    Allenspach, E. J., P. Cullinan, J. Tong, Q. Tang, A. G. Tesciuba, J. L. Cannon, S. M. Takahashi, R. Morgan, J. K. Burkhardt, and A. I. Sperling. 2001. ERM-dependent movement of CD43 defines a novel protein complex distal to the immunological synapse. Immunity 15:739-750.

    Arora, V. K., B. L. Fredericksen, and J. V. Garcia. 2002. Nef: agent of cell subversion. Microbes Infect. 4:189-199.

    Arora, V. K., R. P. Molina, J. L. Foster, J. L. Blakemore, J. Chernoff, B. L. Fredericksen, and J. V. Garcia. 2000. Lentivirus Nef specifically activates Pak2. J. Virol. 74:11081-11087.

    Barouch, D. H., S. Santra, J. E. Schmitz, M. J. Kuroda, T. M. Fu, W. Wagner, M. Bilska, A. Craiu, X. X. Zheng, G. R. Krivulka, K. Beaudry, M. A. Lifton, C. E. Nickerson, W. L. Trigona, K. Punt, D. C. Freed, L. Guan, S. Dubey, D. Casimiro, A. Simon, M. E. Davies, M. Chastain, T. B. Strom, R. S. Gelman, D. C. Montefiori, M. G. Lewis, E. A. Emini, J. W. Shiver, and N. L. Letvin. 2000. Control of viremia and prevention of clinical AIDS in rhesus monkeys by cytokine-augmented DNA vaccination. Science 290:486-492.

    Chu, P. C., J. Wu, X. C. Liao, J. Pardo, H. Zhao, C. Li, M. K. Mendenhall, E. Pali, M. Shen, S. Yu, V. C. Taylor, G. Aversa, S. Molineaux, D. G. Payan, and E. S. Masuda. 2004. A novel role for p21-activated protein kinase 2 in T cell activation. J. Immunol. 172:7324-7334.

    Davies, S. P., H. Reddy, M. Caivano, and P. Cohen. 2000. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem. J. 351:95-105.

    Dustin, M. L., and J. A. Cooper. 2000. The immunological synapse and the actin cytoskeleton: molecular hardware for T cell signaling. Nat. Immunol. 1:23-29.

    Foster, J. L., R. P. Molina, T. Luo, V. K. Arora, Y. Huang, D. D. Ho, and J. V. Garcia. 2001. Genetic and functional diversity of human immunodeficiency virus type 1 subtype B Nef primary isolates. J. Virol. 75:1672-1680.

    Johnson, R. P., and A. Kaur. 2005. HIV: viral blitzkrieg. Nature 434:1080-1081.

    Kestler, H. W., D. J. Ringler, K. Mori, D. L. Panicali, P. K. Sehgal, M. D. Daniel, and R. C. Desrosiers. 1991. Importance of the nef gene for maintenance of high virus loads and for development of AIDS. Cell 65:651-662.

    Kissil, J. L., K. C. Johnson, M. S. Eckman, and T. Jacks. 2002. Merlin phosphorylation by p21-activated kinase 2 and effects of phosphorylation on merlin localization. J. Biol. Chem. 277:10394-10399.

    Kissil, J. L., E. W. Wilker, K. C. Johnson, M. S. Eckman, M. B. Yaffe, and T. Jacks. 2003. Merlin, the product of the Nf2 tumor suppressor gene, is an inhibitor of the p21-activated kinase, Pak1. Mol. Cell 12:841-849.

    Lang, S. M., A. J. Iafrate, C. Stahl-Hennig, E. M. Kuhn, T. Nisslein, F. J. Kaup, M. Haupt, G. Hunsmann, J. Skowronski, and F. Kirchhoff. 1997. Association of simian immunodeficiency virus Nef with cellular serine/threonine kinases is dispensable for the development of AIDS in rhesus macaques. Nat. Med. 3:860-865.

    Li, Q., L. Duan, J. D. Estes, Z. M. Ma, T. Rourke, Y. Wang, C. Reilly, J. Carlis, C. J. Miller, and A. T. Haase. 2005. Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells. Nature 434:1148-1152.

    Maeda, M., T. Matsui, M. Imamura, and S. Tsukita. 1999. Expression level, subcellular distribution and rho-GDI binding affinity of merlin in comparison with ezrin/radixin/moesin proteins. Oncogene 18:4788-4797.

    Manchanda, N., A. Lyubimova, H. Y. Ho, M. F. James, J. F. Gusella, N. Ramesh, S. B. Snapper, and V. Ramesh. 2005. The NF2 tumor suppressor Merlin and the ERM proteins interact with N-WASP and regulate its actin polymerization function. J. Biol. Chem. 280:12517-12522.

    Mattapallil, J. J., D. C. Douek, B. Hill, Y. Nishimura, M. Martin, and M. Roederer. 2005. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 434:1093-1097.

    Nunn, M. F., and J. W. Marsh. 1996. Human immunodeficiency virus type 1 Nef associates with a member of the p21-activated kinase family. J. Virol. 70:6157-6161.

    Pulkkinen, K., G. H. Renkema, F. Kirchhoff, and K. Saksela. 2004. Nef associates with p21-activated kinase 2 in a p21-GTPase-dependent dynamic activation complex within lipid rafts. J. Virol. 78:12773-12780.

    Raney, A., L. S. Kuo, L. L. Baugh, J. L. Foster, and J. V. Garcia. 2005. Reconstitution and molecular analysis of an active human immunodeficiency virus type 1 Nef/p21-activated kinase 2 complex. J. Virol. 79:12732-12741.

    Renkema, G. H., A. Manninen, D. A. Mann, M. Harris, and K. Saksela. 1999. Identification of the Nef-associated kinase as p21-activated kinase 2. Curr. Biol. 9:1407-1410.

    Rong, R., E. I. Surace, C. A. Haipek, D. H. Gutmann, and K. Ye. 2004. Serine 518 phosphorylation modulates merlin intramolecular association and binding to critical effectors important for NF2 growth suppression. Oncogene 23:8447-8454.

    Rouleau, G. A., P. Merel, M. Lutchman, M. Sanson, J. Zucman, C. Marineau, K. Hoang-Xuan, S. Demczuk, C. Desmaze, B. Plougastel, S. Pulst, P. Lenior, E. Bijlsma, R. Fashold, J. Dumanski, P. deJong, D. Parry, R. Eldrige, A. Aurias, O. Delattre, and G. Thomas. 1993. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature 363:515-521.

    Sawai, E. T., A. Baur, H. Struble, B. M. Peterlin, J. A. Levy, and C. Cheng-Mayer. 1994. Human immunodeficiency virus type 1 Nef associates with a cellular serine kinase in T lymphocytes. Proc. Natl. Acad. Sci. USA 91:1539-1543.

    Schrager, J. A., and J. W. Marsh. 1999. HIV-1 Nef increases T cell activation in a stimulus-dependent manner. Proc. Natl. Acad. Sci. USA 96:8167-8172.

    Shaw, R. J., J. G. Paez, M. Curto, A. Yaktine, W. M. Pruitt, I. Saotome, J. P. O'Bryan, V. Gupta, N. Ratner, C. J. Der, T. Jacks, and A. I. McClatchey. 2001. The Nf2 tumor suppressor, merlin, functions in Rac-dependent signaling. Dev. Cell 1:63-72.

    Surace, E. I., C. A. Haipek, and D. H. Gutmann. 2004. Effect of merlin phosphorylation on neurofibromatosis 2 (NF2) gene function. Oncogene 23:580-587.

    Trofatter, J. A., M. M. MacCollin, J. L. Rutter, J. R. Murrell, M. P. Duyao, D. M. Parry, R. Eldridge, N. Kley, A. G. Menon, K. Pulaski, V. H. Haase, C. A. Ambrose, D. Munroe, C. Bove, J. H. Haines, R. L. Martuza, M. E. McDonald, B. R. Seizinger, M. P. Short, A. J. Buckler, and J. F. Gusella. 1993. A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell 72:791-800.

    Wu, Y., and J. W. Marsh. 2001. Selective transcription and modulation of resting T cell activity by preintegrated HIV DNA. Science 293:1503-1506.

    Wulfing, C., and M. M. Davis. 1998. A receptor/cytoskeletal movement triggered by costimulation during T cell activation. Science 282:2266-2269.

    Xiao, G. H., A. Beeser, J. Chernoff, and J. R. Testa. 2002. p21-activated kinase links Rac/Cdc42 signaling to merlin. J. Biol. Chem. 277:883-886.

    Xiao, G. H., J. Chernoff, and J. R. Testa. 2003. NF2: the wizardry of merlin. Genes Chromosomes Cancer 38:389-399.

    Xiao, G. H., R. Gallagher, J. Shetler, K. Skele, D. A. Altomare, R. G. Pestell, S. Jhanwar, and J. R. Testa. 2005. The NF2 tumor suppressor gene product, merlin, inhibits cell proliferation and cell cycle progression by repressing cyclin D1 expression. Mol. Cell. Biol. 25:2384-2394.

    Yu, H., D. Leitenberg, B. Li, and R. A. Flavell. 2001. Deficiency of small GTPase Rac2 affects T cell activation. J. Exp. Med. 194:915-926.(Bangdong L. Wei, Vivek K.)