The C-terminal Kinase Domain of the P34cdc2-Related PITSLRE Protein Kinase (p110c) Associates with P21-Activated Kinase 1 and Inhibits Its Activity During Anoikis

She Chen,Xianglei Yin,Xiaoyu Zhu,Jun Yan,Shuying Ji,Chun Chen,Mingmei Cai,Songwen Zhang,Hongliang Zong,Yun Hu,Zhenghong Yuan,Zonghou Shen,Jianxin Gu
DOI: https://doi.org/10.1074/jbc.m300818200
IF: 5.485
2003-01-01
Journal of Biological Chemistry
Abstract:The PITSLRE protein kinases are parts of the large family of p34cdc2-related kinases. During apoptosis induced by some stimuli, specific PITSLRE isoforms are cleaved by caspase to produce a protein that contains the C-terminal kinase domain of the PITSLRE proteins (p110C). The p110C induces apoptosis when it is ectopically expressed in Chinese hamster ovary cells. In our study, similar induction of this p110C was observed during anoikis in NIH3T3 cells. To investigate the molecular mechanism of apoptosis mediated by p110C, we used the yeast two-hybrid system to screen a human fetal liver cDNA library and identified p21-activated kinase 1 (PAK1) as an interacting partner of p110C. The association of p110C with PAK1 was further confirmed by in vitro binding assay, in vivo coimmunoprecipitation, and confocal microscope analysis. The interaction of p110C with PAK1 occurred within the residues 210–332 of PAK1. Neither association between p58PITSLRE or p110PITSLRE and PAK1 nor association between p110C and PAK2 or PAK3 was observed. Anoikis was increased and PAK1 activity was inhibited when NIH3T3 cells were transfected with p110C. Furthermore, the binding of p110C with PAK1 and inhibition of PAK1 activity were also observed during anoikis. Taken together, these data suggested that PAK1 might participate in the apoptotic pathway mediated by p110C. The PITSLRE protein kinases are parts of the large family of p34cdc2-related kinases. During apoptosis induced by some stimuli, specific PITSLRE isoforms are cleaved by caspase to produce a protein that contains the C-terminal kinase domain of the PITSLRE proteins (p110C). The p110C induces apoptosis when it is ectopically expressed in Chinese hamster ovary cells. In our study, similar induction of this p110C was observed during anoikis in NIH3T3 cells. To investigate the molecular mechanism of apoptosis mediated by p110C, we used the yeast two-hybrid system to screen a human fetal liver cDNA library and identified p21-activated kinase 1 (PAK1) as an interacting partner of p110C. The association of p110C with PAK1 was further confirmed by in vitro binding assay, in vivo coimmunoprecipitation, and confocal microscope analysis. The interaction of p110C with PAK1 occurred within the residues 210–332 of PAK1. Neither association between p58PITSLRE or p110PITSLRE and PAK1 nor association between p110C and PAK2 or PAK3 was observed. Anoikis was increased and PAK1 activity was inhibited when NIH3T3 cells were transfected with p110C. Furthermore, the binding of p110C with PAK1 and inhibition of PAK1 activity were also observed during anoikis. Taken together, these data suggested that PAK1 might participate in the apoptotic pathway mediated by p110C. Apoptosis is a form of altruistic cell suicide, which is involved in many physiological processes, including tissue homeostasis, embryonic development, and immune response (1Schwartzman R.A. Cidlowski J.A. Endocr. Rev. 1993; 14: 133-151Crossref PubMed Scopus (898) Google Scholar, 2Maller J.L. Curr. Opin. Cell Biol. 1991; 3: 269-275Crossref PubMed Scopus (96) Google Scholar). It is becoming increasingly clear that cell cycle regulators such as the p34cdc2 gene family could influence apoptosis (3Yonish-Rouach E. Grunwald D. Wilder S. Kimchi A. May E. Lawrence J.J. May P. Oren M. Mol. Cell. Biol. 1993; 13: 1415-1423Crossref PubMed Scopus (354) Google Scholar, 4Weinberg R.A. Cell. 1995; 81: 323-330Abstract Full Text PDF PubMed Scopus (4326) Google Scholar, 5Berry D.E. Lu Y. Schmidt B. Fallon P.G. O'Connell C. Hu S.X. Xu H.J. Blanck G. Oncogene. 1996; 12: 1809-1819PubMed Google Scholar, 6Shan B. Lee W.H. Mol. Cell. Biol. 1994; 14: 8166-8173Crossref PubMed Scopus (474) Google Scholar, 7Shi L. Nishioka W.K. Th'ng J. Bradbury E.M. Litchfield D.W. Greenberg A.H. Science. 1994; 263: 143-145Crossref Scopus (572) Google Scholar). The PITSLRE protein kinases, which are coded by the gene localized to human chromosome 1p36.3 and a syntenic region of mouse chromosome 4, are parts of the large family of p34cdc2-related kinases (8Bunnell B.A. Heath L.S. Adams D.E. Lahti J.M. Kidd V.J. Proc. Natl. Acad. Sci. U. S. A. 1990; 87: 7467-7471Crossref PubMed Scopus (126) Google Scholar, 9Eipers P.G. Barnoski B.L. Han J. Carroll A.J. Kidd V.J. Genomics. 1991; 11: 621-629Crossref PubMed Scopus (42) Google Scholar, 10Meyerson M. Enders G.H. Wu C.L. Su L.K. Gorka C. Nelson C. Harlow E. Tsai L.H. EMBO J. 1992; 11: 2909-2917Crossref PubMed Scopus (788) Google Scholar, 11Xiang J. Lahti J.M. Grenet J. Easton J. Kidd V.J. J. Biol. Chem. 1994; 269: 15786-15794Abstract Full Text PDF PubMed Google Scholar, 12Mock B.A. Padlan C. Kozak C.A. Kidd V. Mamm. Genome. 1994; 5: 191-192Crossref PubMed Scopus (10) Google Scholar, 13Lahti J.M. Valentine M. Xiang J. Jones B. Amann J. Grenet J. Richmond G. Look A.T. Kidd V.J. Nat. Genet. 1994; 7: 370-375Crossref PubMed Scopus (111) Google Scholar, 14Gururajan R. Lahti J.M. Grenet J. Easton J. Gruber I. Ambros P.F. Kidd V.J. Genome. Res. 1998; 8: 929-939Crossref PubMed Scopus (52) Google Scholar). There are at least 20 PITSLRE protein kinase isoforms, which are differentially expressed in mammalian tissues and regulate diverse cellular functions, including the cell cycle control, tumorigenesis, the regulation of RNA splicing or transcription, and so on (8Bunnell B.A. Heath L.S. Adams D.E. Lahti J.M. Kidd V.J. Proc. Natl. Acad. Sci. U. S. A. 1990; 87: 7467-7471Crossref PubMed Scopus (126) Google Scholar, 10Meyerson M. Enders G.H. Wu C.L. Su L.K. Gorka C. Nelson C. Harlow E. Tsai L.H. EMBO J. 1992; 11: 2909-2917Crossref PubMed Scopus (788) Google Scholar, 11Xiang J. Lahti J.M. Grenet J. Easton J. Kidd V.J. J. Biol. Chem. 1994; 269: 15786-15794Abstract Full Text PDF PubMed Google Scholar,13Lahti J.M. Valentine M. Xiang J. Jones B. Amann J. Grenet J. Richmond G. Look A.T. Kidd V.J. Nat. Genet. 1994; 7: 370-375Crossref PubMed Scopus (111) Google Scholar, 14Gururajan R. Lahti J.M. Grenet J. Easton J. Gruber I. Ambros P.F. Kidd V.J. Genome. Res. 1998; 8: 929-939Crossref PubMed Scopus (52) Google Scholar, 15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 16Beyaert R. Kidd V.J. Cornelis S. Van de Craen M. Denecker G. Lahti J.M. Gururajan R. Vandenabeele P. Fiers W. J. Biol. Chem. 1997; 272: 11694-11697Abstract Full Text Full Text PDF PubMed Scopus (124) Google Scholar, 17Tang D. Kidd V.J. J. Biol. Chem. 1998; 273: 28549-28552Abstract Full Text Full Text PDF PubMed Scopus (154) Google Scholar, 18Ariza M.E. Broome-Powell M. Lahti J.M. Kidd V.J. Nelson M.A. J. Biol. Chem. 1999; 274: 28505-28513Abstract Full Text Full Text PDF PubMed Scopus (62) Google Scholar, 19Nelson M.A. Ariza M.E. Yang J.M. Thompson F.H. Taetle R. Trent J.M. Wymer J. Massey-Brown K. Broome-Powell M. Easton J. Lahti J.M. Kidd V.J. Cancer. Genet. Cytogenet. 1999; 108: 91-99Abstract Full Text Full Text PDF PubMed Scopus (47) Google Scholar, 20Dave B.J. Pickering D.L. Hess M.M. Weisenburger D.D. Armitage J.O. Sanger W.G. Cancer. Genet. Cytogenet. 1999; 108: 120-126Abstract Full Text Full Text PDF PubMed Scopus (36) Google Scholar, 21Loyer P. Trembley J.H. Lahti J.M. Kidd V.J. J. Cell Sci. 1998; 111: 1505-1506Crossref Google Scholar, 22Trembley J.H. Hu D. Hsu L.C. Yeung C.Y. Slaughter C. Lahti J.M. Kidd V.J. J. Biol. Chem. 2002; 277: 2589-2596Abstract Full Text Full Text PDF PubMed Scopus (70) Google Scholar, 23Dickinson L.A. Edgar A.J. Ehley J. Gottesfeld J.M. J. Biol. Chem. 2002; 277: 25465-25473Abstract Full Text Full Text PDF PubMed Scopus (95) Google Scholar, 24Zhang S.W. Xu S.L. Cai M.M. Yan J. Zhu X.Y. Hu Y. Gu J.X. Mol. Cell. Biochem. 2001; 221: 161-168Crossref PubMed Scopus (14) Google Scholar, 25Zhang S.W. Cai M.M. Zhang S. Xu S.L. Chen S. Chen X.N. Chen C. Gu J.X. J. Biol. Chem. 2002; 277: 35314-35322Abstract Full Text Full Text PDF PubMed Scopus (72) Google Scholar, 26Cai M.M. Zhang S.W. Zhang S. Chen S. Yan J. Zhu X.Y. Hu Y. Chen C. Gu J.X. Mol. Cell. Biochem. 2002; 238: 49-55Crossref PubMed Scopus (8) Google Scholar). Studies indicated that several of the PITSLRE protein kinase isoforms might serve as effectors in apoptotic signaling pathways (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 16Beyaert R. Kidd V.J. Cornelis S. Van de Craen M. Denecker G. Lahti J.M. Gururajan R. Vandenabeele P. Fiers W. J. Biol. Chem. 1997; 272: 11694-11697Abstract Full Text Full Text PDF PubMed Scopus (124) Google Scholar, 17Tang D. Kidd V.J. J. Biol. Chem. 1998; 273: 28549-28552Abstract Full Text Full Text PDF PubMed Scopus (154) Google Scholar, 18Ariza M.E. Broome-Powell M. Lahti J.M. Kidd V.J. Nelson M.A. J. Biol. Chem. 1999; 274: 28505-28513Abstract Full Text Full Text PDF PubMed Scopus (62) Google Scholar). During apoptosis induced by Fas or tumor necrosis factor, the action of p110C, a novel processed PITSLRE isoform resulting from the proteolysis of specific larger PITSLTR isoforms, significantly increased (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 18Ariza M.E. Broome-Powell M. Lahti J.M. Kidd V.J. Nelson M.A. J. Biol. Chem. 1999; 274: 28505-28513Abstract Full Text Full Text PDF PubMed Scopus (62) Google Scholar). Previous studies of our group and others (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 26Cai M.M. Zhang S.W. Zhang S. Chen S. Yan J. Zhu X.Y. Hu Y. Chen C. Gu J.X. Mol. Cell. Biochem. 2002; 238: 49-55Crossref PubMed Scopus (8) Google Scholar) showed that apoptosis was increased when p110C was ectopically expressed in Chinese hamster ovary cells or SMMC 7721 hepatocarcinoma cells. These data suggested that p110C might play an important role in mediating apoptosis. In this study, we also detected the expression of p110C in NIH3T3 cells during anoikis, which is a form of apoptosis induced by the disruption of cell-matrix interaction. In agreement with the previous studies, a similar induction of p110C was observed during anoikis in NIH3T3 cells. It seems that p110C could participate in apoptosis induced by different factors. However, little is known about the mechanism of p110C-mediated apoptotic signaling pathway. Previous studies of the p58PITSLRE or p110PITSLRE isoforms showed that they could interact with different proteins and mediate different cellular functions. For example, p110PITSLRE isoform interacted with the RNA-binding protein RNPS1, RNA polymerase II, and multiple transcriptional elongation factors, regulating some aspects of RNA splicing or transcription in proliferating cells (21Loyer P. Trembley J.H. Lahti J.M. Kidd V.J. J. Cell Sci. 1998; 111: 1505-1506Crossref Google Scholar, 22Trembley J.H. Hu D. Hsu L.C. Yeung C.Y. Slaughter C. Lahti J.M. Kidd V.J. J. Biol. Chem. 2002; 277: 2589-2596Abstract Full Text Full Text PDF PubMed Scopus (70) Google Scholar), whereas the p58PITSLRE isoform interacted with cyclin D3 in the G2/M phase of the cell cycle, regulating G2/M phase cell cycle progression (24Zhang S.W. Xu S.L. Cai M.M. Yan J. Zhu X.Y. Hu Y. Gu J.X. Mol. Cell. Biochem. 2001; 221: 161-168Crossref PubMed Scopus (14) Google Scholar, 25Zhang S.W. Cai M.M. Zhang S. Xu S.L. Chen S. Chen X.N. Chen C. Gu J.X. J. Biol. Chem. 2002; 277: 35314-35322Abstract Full Text Full Text PDF PubMed Scopus (72) Google Scholar). Thus, the identification of cellular proteins that do interact with p110C, physically and/or functionally, should be a useful approach for defining the cellular functions and regulatory mechanisms of p110C in apoptotic signaling pathways. To investigate this issue, the yeast two-hybrid system with p110C as bait was used to screen a human fetal liver cDNA library. As a result, PAK1 1The abbreviations used are: PAK1, p21-activated kinase 1; HA, influenza hemagglutinin monoclonal antibody epitope; GST, glutathione S-transferase; ECL, enhanced chemiluminescence; EGFP, enhanced green fluorescent protein; PBS, phosphate-buffered saline; MBP, myelin basic protein; aa, amino acid(s); PMSF, phenylmethylsulfonyl fluoride; PVDF, polyvinylidene difluoride; FITC, fluorescein isothiocyanate; DMEM, Dulbecco's modified Eagle's medium; PI, propidium iodide; PS, phosphatidylserine; JNK, c-Jun N-terminal kinase; poly-HEMA, poly(2-hydroxyethyl methacrylate). (p21-activated kinase 1) was identified as a p110C-associated protein via the domain (aa 210–332). This association of p110C with PAK1 was specific, as verified by the inability of PAK2 or PAK3 to associate with p110C and the inability of PAK1 to associate with p58PITSLRE or p110PITSLRE. Anoikis was increased and PAK1 activity was inhibited when NIH3T3 cells were transfected with p110C. Furthermore, the association of p110C, with PAK1 and inhibition of PAK1 activity, was also observed when anoikis was induced in NIH3T3 cells. Taken together, the data suggested that PAK1 might play a role in the regulation of p110C-mediated apoptotic signaling pathway. Cell Lines and Reagents—The NIH3T3 cells and SMMC 7721 cells were obtained from the Institute of Cell Biology, Academic Sinica. The mammalian expression vector pcDNA3 was from Invitrogen (Carlsbad, CA). MATCHMAKER LexA two-hybrid system and human fetal liver MATCHMAKER LexA cDNA library were products of Clontech (Palo Alto, CA). The rabbit polyclonal anti-PITSLRE antibody and the goat polyclonal anti-γ-PAK antibody were purchased from Santa Cruz Biotechnology. The rabbit polyclonal anti-PAK1 and the anti-rabbit-horseradish peroxidase secondary antibodies were purchased from New England BioLabs. The rabbit polyclonal anti-PAK3 antibody was from Calbiochem. Protein G-agarose, glutathione-Sepharose beads, and the mouse monoclonal anti-HA (12CA5) antibody were purchased from Roche Applied Science. Leupeptin, aprotinin, PMSF, poly-HEMA, and MBP were purchased from Sigma Chemical Co. [γ-32P]ATP (>3000 Ci/mm), [35S]methionine, Hybond PVDF membrane, and the enhanced chemiluminescence (ECL) assay kit were purchased from Amersham Biosciences. Annexin V-FITC recombinant protein was purchased from Bender MedSystems. Other reagents were commercially available in China. Cell Culture, Transient Transfection, and Induction of Anoikis— NIH3T3 cells were cultured with DMEM supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. SMMC 7721 cells were cultured with RPMI 1640 supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. They were maintained in a 37 °C humidified atmosphere containing 5% CO2 in air. For transient transfection assays, the cells (5 × 105) were grown on 60-mm tissue culture dishes. The following day, the cells were transfected with relevant plasmids. The transfection was performed with LipofectAMINETM according to the manufacturer's recommendations. After 48 h, the cells were harvested for further analysis. For induction of anoikis (27Ibuki Y. Goto R. Biochem. Biophys. Res. Commun. 2000; 279: 872-878Crossref PubMed Scopus (32) Google Scholar), NIH3T3 cells were seeded in 3 × 106 cells/100-mm tissue culture dish. After culturing for 6 h, the medium was changed to serum-free DMEM. Following a further culturing for 18 h, the cells were trypsinized and plated in cell suspension dishes previously coated with 10 mg/ml poly-HEMA. After indicated time, the cells were collected for further investigation. Apoptosis was evaluated by staining with propidium iodide (PI) to detect subdiploid DNA content and by using annexin V to detect cell surface phosphatidylserine (PS). For PI staining, the cells (106) were washed twice, collected, and then fixed with 70% ethanol at –20 °C overnight. After staining with 10 μg/ml propidium iodide, the cells were analyzed on a BD Biosciences fluorescence-activated cell sorter. The percentage of apoptosis was quantitated from sub-G1 events. To use annexin V to detect cell surface PS, the cells were washed twice in PBS and then resuspended in binding buffer (10 mm Hepes/NaOH, pH 7.4, 140 mm NaCl, 2.5 mm CaCl2). 195 μl of cell suspension containing 105 cells was taken, and 5 μl of annexin V-FITC was added, mixed, and incubated for 10 min in the dark. After that, the cells were wash twice in PBS and resuspended in 190 μl of binding buffer. 10 μl of 20 μg/ml PI was added and then analyzed by fluorescence-activated cell sorting. Apoptotic cells could be stained with annexin V but not PI. Western Blot Analysis—Western blot experiments were used to measure certain proteins. Briefly, the cells were lysed in lysis buffer (120 mm Tris (pH 7.4), 135 mm NaCl, 1 mm EDTA, 1% Nonidet P-40, 0.1% SDS, 1 mm Na3VO4, 1 mm aprotinin, 1 mm PMSF). A total of 50 μg of protein from each sample was electrophoresed by 12% SDS-PAGE and then transferred to a PVDF membrane. After blocking with PBS containing 5% nonfat milk and 0.1% Tween 20 overnight, the membrane was incubated with primary antibody at 4 °C overnight. After washing with PBS containing 0.1% Tween 20 three times, each for 5 min, the membrane was then incubated with horseradish peroxidase-labeled secondary antibody for another 2 h at room temperature. The membrane was then developed using the enhanced chemiluminescent (ECL) detection system. Yeast Two-hybrid cDNA Library Screening and Protein-Protein Interaction Assay—MATCHMAKER LexA two-hybrid system was used to perform yeast two-hybrid screening according to the manufacturer's instruction. The p110C was used as bait to screen by the reporter genes LEU2 and lacZ. The transformed yeast was grown on plates lacking both histidine to select the bait plasmid and tryptophan to select the prey plasmid, respectively. The individual cDNA of interest was purified from the positive clones and re-transformed into yeast to confirm its specific interaction with p110C. 14 clones could activate both the LEU2 and lacZ reporter genes only when they contain pLexA-p110C. All of these clones were revealed to encode the fragment of p21-activated kinase 1 (PAK1) by sequence analysis. The quantitative β-galactosidase assay was performed to detect the protein-protein interaction on at least three independent colonies from the same clone, using o-nitrophenyl-β-d-galactopyranoside as a substrate. Murine p53 fused with LexA as a bait and SV40 large T-antigen fused as a prey were used as a positive control. Construction of the p110C and the Mutants of PAK1—pLexA-p110C containing the C-terminal PITSLRE p110 coding sequence (GenBankTM accession number U04824, nucleotides 1357–2451) was constructed. The p110C was generated by PCR using p110PITSLRE cDNA (a gift from Dr. Sigrid Cornelis) as template and using primer 1 (5′-GATGAATTCATGTGCCGGAGCGTCGAGGAG-3′, EcoRI site underlined), primer 2 (5′-GATGTCGACTTAGAACTTGAGGCTGAA-3′, SalI site underlined). pcDNA3-HA-p110C and pcNDA3-GST-p110C with a GST fusion at the N-terminal were constructed by inserting the same PCR product of p110C in-frame into the EcoRI and XhoI sites of pcDNA3-HA vector or pcDNA3-GST vector. The full-length of p110 was cut from pSV-sport-p110PITSLRE by EcoRI and XbaI and cloned into pcDNA3-HA vector. The full-length cDNA and mutants of PAK1 were designated as FL (aa 1–545), N (aa 1–270), C (aa 271–545), F1 (aa 1–456), F2 (aa 1–394), F3 (aa 1–332), F4 (aa 150–332), F5 (aa 210–332), F6 (aa 240–332), and F7 (aa 210–300) and performed by PCR using pcMV6-myc-PAK1 (wt) (a gift from Dr. Gary Bokoh) as template. FL was generated from primer 3 (5′-GATGAATTCATGTCAAATAACGGCCTAGAC-3′) and primer 4 (5′-GATCTCGAGTTAGTGATTGTTCTTTGT-3′), N from primers 3 and 5 (5′-GATGTCGACTTATTTCTTCTTAGGATCGCC-3′), C from primer 6 (5′-GATGAATTCATGTATACACGGTTTGAGAAG-3′) and primer 4, F1 from primer 3 and primer 7 (5′-GATCTCGAGTTATTCGATGGCCATGATGCC-3′), F2 from primer 3 and primer 8 (5′-GATCTCGAGTTAATTGTCACTCTTGATGTC-3′), F3 from primer 3 (5′-GATCTCGAGTTAGTCCAAGTAATTCACAAT) and primer 9, F4 from primer 10 (5′-GATGAATTCATGGCTGAGGATTACAATTCT-3′) and primer 9, F5 from primer 11 (5′-GATGAATTCATGCCTGTCACTCCAACTCGG-3′) and primer 9, F6 from primer 12 (5′-GATGAATTCATGGAGAAGCAGAAGAAG-3′) and primer 9, and F7 from primer 11 and primer 13 (5′-GATCTCGAGTTACTGCTTAATGGCCTC-3′) by PCR separately. The EcoRI site underlined in the forward primer and the XhoI site underlined in the reverse primer were used to clone these fragments into pB42AD plasmid or pcDNA3 vector. The PAK2 was cloned into pB42AD plasmid or pcDNA3 vector by PCR using PAK2 cDNA (a gift from Dr. Gray Bokoch) as template, and using primer 14 (5′-GATGAATTCATGTCTGATAACGGAGAACTG-3′, EcoRI site underlined) and primer 15 (5′-GATCTCGAGTTAACGGTTACTCTTCATTGC-3′, XhoI site underlined). The PAK3 was cloned into pB42AD plasmid or pcDNA3 vector by PCR using mPAK3 cDNA (a gift from Dr. Richard A. Cerione) as template and using primer 16 (5′-GATGAATTCATGTCTGACAGCTTGGATAAC-3′, EcoRI site underlined) and primer 17 (5′-GATCTCGAGCTAACGGCTACTGTTCTTAAT-3′, XhoI site underlined). In Vitro Binding Analysis—p110C was cloned into pcDNA3-GST vector. PAK1 and its mutants, PAK2, and PAK3 were cloned into pcDNA3 individually. Their transcripts were produced by using an in vitro transcription kit (Promega). Rabbit reticulocyte lysates from Promega were used to generate [35S]Met-labeled proteins, which were used immediately for binding studies. The binding reactions were performed in protein binding buffer (20 mm Tris, pH 7.5, 50 mm NaCl, 10% glycerol, 10 mm NaF, 1% Nonidet P-40, 1 mm NaVO4, 10 μg/ml aprotinin, 10 μg/ml leupeptin, 1 mm PMSF) at 4 °C for 2 h with constant mixing (28Bagheri-Yarmand R. Mandal M. Taludker A.H. Wang R.A. Vadlamudi R.K. Kung H.J. Kumar R. J. Biol. Chem. 2001; 276: 29403-29409Abstract Full Text Full Text PDF PubMed Scopus (106) Google Scholar). The beads were washed three times with the same buffer, and the bound proteins were subjected to 12% SDS-PAGE analysis. The gel was then dried and autoradiographed. In Vivo Interaction Assay—48 h after transfection, the cells were washed three times with ice-cold PBS and solubilized with 1 ml of lysis buffer (50 mm Tris-HCl (pH 7.5), 150 mm NaCl, 0.1% Nonidet P-40, 5 mm EDTA, 5 mm EGTA, 15 mm MgCl2, 60 mm β-glycerophosphate, 0.1 mm sodium orthovanadate, 0.1 mm NaF, 0.1 mm benzamide, 10 μg/ml aprotinin, 10 μg/ml leupeptin, 1 mm PMSF). Detergent-insoluble materials were removed by centrifugation at 13,000 rpm for 15 min at 4 °C. Whole cell lysates were incubated with mouse normal IgG (Santa Cruz Biotechnology) or relevant antibody at 4 °C for 2 h. Pre-equilibrated protein G-agarose beads (Roche Applied Science) were then added and collected by centrifugation after 2 h of incubation and then gently washed three times with the lysis buffer. The bound proteins were eluted by boiling in SDS sample buffer and resolved on a 12% SDS-PAGE gel. The proteins were transferred onto a PVDF membrane and analyzed using Western blots. Fluorescence Imaging of Living Cells—The enhanced GFP segment (Clontech) was attached to the C terminus of human p110C by standard recombinant techniques. Briefly, the DNA fragments of p110C was generated by PCR (primer 18: 5′-GATGAATTCATGTGCCGGAGCGTCGAGGAG-3′, EcoRI site underlined; primer 19: 5′-GATGTCGACGAACTTGAGGCTGAAGCC-3′, SalI site underlined), digested with EcoRI and SalI, and then cloned into pEGFPN3 (Clontech) to create pEG-FPN3-p110C. Full-length PAK1 was cloned into the pDsRed1-C1 vector (Clontech) so as to be expressed as a fusion with a red fluorescent protein (DsRed). The fragment of PAK1 was prepared by PCR (primer 20: 5′-GATGTCGACATGTCAAATAACGGCCTAGAC-3′, SalI site underlined; primer 21: 5′-GATGGATCCTTAGTGATTGTTCTTTGT-3′, BamHI site underlined), digested with SalI and BamHI, and then ligated into the pDsRed1-C1 vector to create pDsRed1-C1-PAK1. SMMC 7721 hepatocellular carcinoma cells were transiently co-transfected with pDsRed1-C1-PAK1 and pEGFPN3-p110C. After 48 h, the cells were observed under a fluorescence microscope and digitally photographed. PAK1 Kinase Assay—As described by Chant et al. (29Brown J.L. Stowers L. Baer M. Trejo J. Coughlin S. Chant J. Curr. Biol. 1996; 6: 598-605Abstract Full Text Full Text PDF PubMed Scopus (228) Google Scholar), the cells were washed twice with ice-cold PBS, and lysed at 4 °C in lysis buffer (50 mm Tris-HCl (pH 7.5), 150 mm NaCl, 0.1% Nonidet P-40, 5 mm EDTA, 5 mm EGTA, 15 mm MgCl2, 60 mm β-glycerophosphate, 0.1 mm sodium orthovanadate, 0.1 mm NaF, 0.1 mm benzamide, 10 μg/ml aprotinin, 10 μg/ml leupeptin, 1 mm PMSF). Equal total protein was immunoprecipitated with rabbit polyclonal anti-PAK1 (1:50) at 4 °C overnight. Immune complexes were covered by adding protein G beads and then centrifuged. The beads were washed twice in lysis buffer and twice in kinase buffer (100 mm NaCl, 50 mm Tris (pH 7.5), 10 mm MgCl2, and 1 mm MnCl2). Kinase assays were performed in 50-μl reactions with 50 mm ATP, 10 μCi of [32P]ATP, and 5 μg of MBP. After 30 min at 30 °C, the reactions were terminated by addition of 10 μl of 6× sample buffer, boiled, separated by 15% SDS-PAGE, and analyzed using autoradiography. Quantitation of MBP phosphorylation by PAK1 was determined by phosphorimaging. Induction of p110C Protein during Anoikis in NIH3T3 Cells—Previous studies indicated that transfection of p110C in Chinese hamster ovary cells did not influence mitosis and cell growth but could induce apoptosis (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar). When the apoptosis of cells was induced by treating with Fas antibody or tumor necrosis factor, p110C protein was induced, which was not due to new protein synthesis but processing of some PITSLRE isoforms (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 18Ariza M.E. Broome-Powell M. Lahti J.M. Kidd V.J. Nelson M.A. J. Biol. Chem. 1999; 274: 28505-28513Abstract Full Text Full Text PDF PubMed Scopus (62) Google Scholar). These suggested that p110C might have an important role in apoptotic signaling pathway. To test whether the induction of p110C isoform was in general in response to different apoptotic inducers, NIH3T3 cell apoptosis was induced by detachment from the cell matrix (also known as anoikis), and then the expression of the PITSLRE protein isoforms was detected. Cells undergoing apoptosis could be recognized by flow cytometry by detecting a sub-G1 peak using PI staining and phosphatidylserine (PS) on the outer leaflet of the cell membrane using an annexin V label. About 26% of the NIH3T3 cells cultured in a poly-HEMA-coated dish (detached from the extracellular matrix) under serum-free condition for 10 h underwent apoptosis (Fig. 1A). Western blot analysis using anti-PITSLRE antibody showed that p110C was markedly induced when the cells were induced during anoikis. This result was consistent with the previous studies (15Lahti J.M. Xiang J. Heath L.S. Campana D. Kidd V.J. Mol. Cell. Biol. 1995; 15: 1-11Crossref PubMed Scopus (191) Google Scholar, 18Ariza M.E. Broome-Powell M. Lahti J.M. Kidd V.J. Nelson M.A. J. Biol. Chem. 1999; 274: 28505-28513Abstract Full Text Full Text PDF PubMed Scopus (62) Google Scholar). In addition, it was found that the induction of p110C did not result from the processing of p110 PITSLRE isoform but largely results from cleavage of the p58PITSLRE isoform when anoikis was induced in NIH3T3 cells (Fig. 1B). Therefore, our study also showed the similar induction of p110C during anoikis in NIH3T3 cells. Identification of PAK1 as p110C-associated Protein—To identify proteins that interact with p110C, the yeast two-hybrid system was used with p110C as bait, and a human fetal liver cDNA library was screened. We plated a pool of cells that contained 5 × 107 primary library transformants at a multiplicity of 10 onto selection induction medium. Approximately 40 clones were obtained whose LEU2 and lacZ reporter genes were activated. Among them 14 clones could activate both the LEU2 and lacZ reporter genes only when they contained pLexA-p110C. The DNA of interest was isolated and sequenced. Sequence homology search identified the encoded proteins. All of these peptides were positioned in-frame to the p21-activated kinase (PAK1) and contained a partly N-terminal deletion portion of PAK1. PAK1 was first identified as a Rac-interacting protein (30Manser E. Leung T. Salihuddin H. Zhao Z.S. Lim L. Nature. 1994; 367: 40-46Crossref PubMed Scopus (1305) Google Scholar). There are at least six known PAK isoforms (PAK1–6) that are differentially expressed in mammalian tissues (31Jaffer Z.M. Chernoff J. Int. J. Biochem. Cell Biol. 2002; 34: 713-717Crossref PubMed Scopus (318) Google Scholar, 32Pandey A. Dan I. Kristiansen T.Z. Watanabe N.M. Voldby J. Kajikawa E. Khosravi-Far R. Blagoev B. Mann M. Oncogene. 2002; 21: 3939-3948Crossref PubMed Scopus (103) Google Scholar, 33Dan C. Nath N. Liberto M. Minden A. Mol. Cell. Biol. 2002; 22: 567-577Crossref PubMed Scopus (138) Google Scholar). Based on their conserved structure, PAK1–3 are classed together as the Group I PAKs (34Sells M.A. Chernoff J. Trends. Cell Biol. 1997; 7: 162-167Abstract Full Text PDF PubMed Scopus (265) Google Scholar, 35Knaus U.G. Bokoch G.M. Int. J. Biochem. Cell Biol. 1998; 30: 857-862Crossref PubMed Scopus (161) Google Scholar, 36Bagrodia S. Cerione R.A. Trends. Cell Biol. 1999; 9: 350-355Abstract Full Text Full Text PDF PubMed Scopus (343) Google Scholar). To investigate if PAK1 was the unique one in the Group I PAKs that could interact with p110C, a direct two-hybrid assay was performed. The other two Group I PAKs, PAK2 and PAK3, were co-transformed, respectively, with p110C into EGY48 (p8op-lacZ). Subsequent transformation results showed that neither of them permitted activation of the reporter genes, whereas PAK1, in the presence of p110C, did activate the two report genes. It suggested th
What problem does this paper attempt to address?