TAB1beta (Transforming Growth Factor-beta -activated Protein Kinase 1-binding Protein 1beta ), a Novel Splicing Variant of TAB1 That Interacts with p38alpha but Not TAK1*

Baoxue Ge, Xinsheng Xiong, Qing Jing, Jennifer L. Mosley, Angela Filose, Dafang Bian, Shuang Huang, and Jiahuai HanDagger

From the Department of Immunology, The Scripps Research Institute, La Jolla, California 92037

Received for publication, October 25, 2002

    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

The mitogen-activated protein kinases (MAPKs) play an important role in a variety of biological processes. Activation of MAPKs is mediated by phosphorylation on specific regulatory tyrosine and threonine sites. We have recently found that activation of p38alpha MAPK can be carried out not only by its upstream MAPK kinases (MKKs) but also by p38alpha autophosphorylation. p38alpha autoactivation requires an interaction of p38alpha with TAB1 (transforming growth factor-beta -activated protein kinase 1-binding protein 1). The autoactivation mechanism of p38alpha has been found to be important in cellular responses to a number of physiologically relevant stimuli. Here, we report the characterization of a splicing variant of TAB1, TAB1beta . TAB1 and TAB1beta share the first 10 exons. The 11th and 12th exons of TAB1 were spliced out in TAB1beta , and an extra exon, termed exon beta , downstream of exons 11 and 12 in the genome was used as the last exon in TAB1beta . The mRNA of TAB1beta was expressed in all cell lines examined. The TAB1beta mRNA encodes a protein with an identical sequence to TAB1 except the C-terminal 69 amino acids were replaced with an unrelated 27-amino acid sequence. Similar to TAB1, TAB1beta interacts with p38alpha but not other MAPKs and stimulates p38alpha autoactivation. Different from TAB1, TAB1beta does not bind or activate TAK1. Inhibition of TAB1beta expression with RNA interference in MDA231 breast cancer cells resulted in the reduction of basal activity of p38alpha and invasiveness of MDA231 cells, suggesting that TAB1beta is involved in regulating p38alpha activity in physiological conditions.

    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

The mitogen-activated protein kinase (MAPK)1 signaling pathways have been shown to function in a wide variety of cellular processes, including cell growth, cell migration, cell differentiation, cell death, and immune cell activation (1-8). To date four distinct subfamilies of MAPKs have been identified: extracellular signal-regulated kinases (ERKs), c-Jun N-terminal or stress-activated protein kinases (JNK/SAPK), ERK5/big MAPK1 (BMK1), and p38 MAPKs (3, 9-14). Whereas the ERK family is activated by growth factors and is involved in cell proliferation, JNK and p38 are predominantly activated in response to pro-inflammatory cytokines and various types of environmental stresses (1, 3-5, 15).

Activation of MAPK is mediated through phosphorylation of specific tyrosine and threonine residues by upstream MAPK kinase (MAPKK), which is activated through phosphorylation of serine/threonine residues by MAPKKK further upstream (4, 16-18). ERK family kinases are activated by the MEK1 and MEK2, JNK by MKK4 and MKK7, and p38 by MKK3 and MKK6 (3, 4, 10, 19). Further upstream, Raf-1, A-Raf, B-Raf, and MOS function as MAPKKKs in the ERK activation pathway (20, 21). A subgroup of MAPKKK, including MEKK1-4, MTK1, MLK1-3, DLK/MUK, ASK1, Tpl-2/Cot, and TAK1, activates the MAPKKs that phosphorylate JNK and perhaps p38 as well (4, 7, 17, 22). Biochemical and genetic evidence have supported the above three-kinase module of MAPK activation (1, 7, 18). In addition, the MAPK cascades are evolutionarily conserved (1, 18). However, we have recently demonstrated an alternative activation pathway of p38alpha MAPK that did not require upstream kinases. We found that TAB1 interacts with p38alpha and leads to the activation of p38alpha (23). TAB1 is a protein that was initially described as an activator of a member of MAPKKK, TAK1, in response to stimulation of transforming growth factor-beta (24). When ectopically expressed together with TAK1, TAB1 interacts with TAK1 and leads to enhancement of TAK1 activation (24-29). The C-terminal 68-amino acid portion of TAB1 is sufficient for binding to and activation of TAK1 (24, 25, 28). Activation of TAK1 by TAB1 is mediated via autophosphorylation of Ser-192 within its kinase activation loop (26). We found that TAB1-mediated p38alpha activation also occurs through an autoactivation mechanism (23). However, the portion of the TAB1 protein that is responsible for p38alpha interaction and activation resides within amino acids 373-418, which is N-terminal to the TAK1 binding site (23, 24).

In this report we describe the characterization of a splicing variant of TAB1, TAB1beta . TAB1beta is an alternative splicing product of the TAB1 gene. TAB1beta and TAB1 have identical amino acid sequences with the exception of the C terminus. Similar to TAB1, TAB1beta interacts with p38alpha and induces p38alpha autoactivation. Different from TAB1, TAB1beta did not bind with TAK1 and had no effect on TAK1 activation. The present findings suggest that the alternative splicing of TAB1 has a role in intracellular signaling transduction.

    EXPERIMENTAL PROCEDURES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

cDNA of TAB1beta -- We completely sequenced two EST clones that showed homologues to TAB1 (clone identification numbers: 50210 and 3049027). One of the clones showed 100% homology with the 5'-end portion of TAB1 cDNA but different 3'-end sequence. This cDNA was termed as TAB1beta , and the cDNA sequence has been deposited in GenBankTM (AF425640).

Construction of Expression Vectors-- The TAB1beta coding region was subcloned from a cDNA clone (clone number 50210) into the mammalian expression vector pcDNA3 (Invitrogen, San Diego, CA) and the pET14 vector (Novagen, Madison, WI), respectively. Other expression vectors, including FLAG-p38, -p38beta , -p38gamma , -p38delta , -p38(KM), -JNK1, -ERK2, MKK6(E), MKK6(A), MKK3(A), and His-p38, were constructed as described previously (23, 30-34).

RT-PCR Analysis-- Total RNA was isolated from HEK293 (293), HPEG2, MDA-231, and RPMI8226 cells using the RNeasy Mini kit (Qiagen, Valencia, CA) following the manufacturer's instructions and treated with RNase-free DNase I. Reverse transcription was performed using SuperScript II RT (Invitrogen, Rockville, MA) according to the supplier's specifications. cDNA (1 µl) was amplified by PCR using the Taq Clontech 50xAdvantage 2 DNA polymerase (94 °C for 2 min, 58 °C for 10 s, 68 °C for 30 s, 45 cycles). The upstream oligonucleotide primer used for TAB1 and TAB1beta was: 5'-CAATCATCGCAGAGCCAGAAATC-3' located in exon-8. The downstream primers were: TAB1, 5'-ACGCTCCAGAGGCGGTAAAACTC-3' in exon-11; TAB1beta , 5'-CACTGAGGGATGGCTGTCAAATC-3' in exon-beta (Fig.1). The obtained RT-PCR fragments for TAB1 and TAB1beta were purified and sequenced to confirm their identity. Semi-quantitative RT-PCR was performed using competitive quantitative RT-PCR kits from Ambion (Austin, TX).

Transfection of Cells-- HEK293 or MDA231 cells were maintained in Dulbecco's modified Eagle's medium supplemented with 5% fetal bovine serum, 2 mM glutamine, and 100 µg/ml penicillin and streptomycin. Cells on six-well plates were transiently transfected with 1 µg (total) of plasmid DNA using LipofectAMINE. After 36 h, the cells were treated with stimuli as described in the text. The plasmid pCMVbeta (Clontech, Palo Alto, CA) was co-transfected, and transfection efficiency was normalized by quantifying beta -galactosidase activity.

Western Blot and Immunoprecipitation Analysis-- Total cell lysates were prepared using a lysis buffer A: 20 mM Tris-HCl, pH 7.5, 120 mM NaCl, 10% glycerol, 1 mM Na3VO4, 2 mM EDTA, 1 mM phenylmethanesulfonyl fluoride, 1% Triton X-100. Equal protein loading of cell extracts in SDS-PAGE was determined by Bio-Rad protein assay solution (Bio-Rad, Hercules, CA) and by staining the transferred nitrocellulose membrane with Ponceau S solution (Sigma, St. Louis, MO). Standard Western blot methods were used (35). Rabbit polyclonal antibodies raised against bacterially expressed recombinant His-p38 protein, rabbit polyclonal antibodies raised against bacterially expressed recombinant His-TAB1beta protein, polyclonal goat anti-TAB1 (N-19) (Santa Cruz Biotechnology, Santa Cruz, CA), anti-FLAG M2 mAb (Sigma, St. Louis, MO), anti-HA mAb (12CA5) (Roche Molecular Biochemicals, Indianapolis, IN), and anti-phospho p38 (New England BioLabs, Beverley, MA) were used in immunoblotting. Selective viewing of ectopically overexpressed TAB1 and TAB1beta by Western blotting was achieved by adjusting exposure time. For co-immunoprecipitation, cell lysates prepared as described above were incubated with anti-FLAG M2 beads (Sigma) and gently shaken for 4 h at 4 °C. The beads were washed three times with lysis buffer and 1 time with 50 mM Tris, pH 6.8. Then, the SDS sample buffer (50 µl) was added and heated for 5 min at 100 °C. The supernatant was applied to SDS-PAGE and detected by immunoblotting.

Reporter Gene Assay-- Cells were grown on 35-mm-diameter multiwell plates and transiently transfected with GAL4-responsive luciferase plasmid or the NF-kappa B-dependent luciferase reporter plasmid (36, 37). A beta -galactosidase expression plasmid (pCMV-beta -gal, Clontech, Palo Alto, CA) was used to control for transfection efficiency. The total amount of DNA for each transfection was kept constant by using the empty vector pcDNA3. Cell extracts were prepared 36 h later, and beta -galactosidase and luciferase activities were measured.

Preparation of Recombinant Proteins-- GST fusion proteins of TAB1, TAB1beta , and MKK4 were expressed in Escherichia coli strain BL21 and purified using glutathione-Sepharose 4B beads (Amersham Biosciences, Uppsala, Sweden). All of the His6-tagged recombinant proteins were expressed in the BL21(DE3) strain and purified using the nickel-nitrilotriacetic acid purification system (Qiagen, Valencia, CA). Myelin basic protein (MBP) was purchased from Sigma.

Protein Kinase Assays-- In vitro kinase assays were carried out at 37 °C for 30 min using ~0.2 µg of recombinant kinase or recombinant protein, 5 µg of kinase substrate, 250 µM ATP, and 10 µCi of [gamma -32P]ATP in 20 µl of kinase reaction buffer. Reactions were terminated by the addition of Laemmli sample buffer. Reaction products were resolved by 12% SDS-PAGE, and the extent of protein phosphorylation was visualized by autoradiography. For the transiently expressed protein, FLAG-p38alpha protein in 293 cells was immunoprecipitated with anti-M2 beads. After washing three times with lysis buffer and one time with kinase reaction buffer, the beads were further used in the analysis of kinase activity.

RNAi-- RNAi constructs were constructed using pSuper vector (38). The hairpin RNA encoded by R1 and R1(M) constructs is shown in Fig. 2C. The sequences of other RNAi constructs, which cannot decrease in endogenous genes, are available upon request. Each of the pSuper constructs was co-transfected with pSVNeo into breast cancer cell line MDA231, and stable lines were selected by G418 at 1 mg/ml concentration.

Matrigel Invasion Assay-- Cell invasion was analyzed using BIOCOAT Matrigel invasion chambers (BD Biosciences) according to the manufacturer's instructions. Briefly, 2 × 105 cells in 300 µl were added to each chamber and allowed to invade Matrigel for 20 h at 37 °C and 5% CO2 atmosphere. The non-invading cells on the upper surface of the membrane were removed from the chambers, and the invading cells on the lower surface of membrane were stained with a Diff-Quick stain kit (BD Biosciences). After two washes with water, the chambers were allowed to air dry. The number of invading cells was then counted using a phase-contrast microscope.

    RESULTS
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Identification of TAB1beta -- In our study of TAB1, we have sequenced a couple of EST clones that have sequence homology with TAB1. The cDNA of one of the EST clones appeared to be a splicing variant of TAB1; its sequence was 100% identical to TAB1 in the 5'-portion but different at the 3'-end. The protein encoded by this cDNA was termed TAB1beta and shown is in Fig. 1A in comparison with TAB1. The N-terminal sequence of amino acids 1-435 of TAB1beta is identical to TAB1. Different from TAB1, TAB1beta lacks the C-terminal 69 amino acids found in TAB1 and has an unrelated 29-amino acid sequence instead (Fig. 1A). Because the C-terminal 68-amino acid portion of TAB1 was mapped as a TAK1 binding domain (25, 28), TAB1beta does not have the TAK1 binding domain.


View larger version (18K):
[in this window]
[in a new window]
 
Fig. 1.   TAB1beta is a splicing isoform of TAB1. A, amino acid sequences of TAB1beta and TAB1. B, structure of exon and intron in TAB1beta and TAB1.

To determine whether TAB1 and TAB1beta are splicing variants, we analyzed the gene structure in the TAB1 locus. We found that the TAB1 gene is composed of 12 exons. TAB1beta shares 1-10 exons with TAB1. Exons 11 and 12 of TAB1 were spliced out in TAB1beta , and an exon further downstream termed exon beta  was the 11th exon of TAB1beta (Fig. 1B). Thus, the difference between TAB1 and TAB1beta is indeed generated by alternative splicing.

Expression of TAB1beta -- To investigate the expression of TAB1beta in different cells, total RNAs were extracted from HEK293, HPEG2, MDA-231, and RPMI 8226 cells. The existence of TAB1beta and TAB1 transcripts was examined by RT-PCR using their specific primers. Both TAB1 and TAB1beta transcripts were found in all cell lines tested (Fig. 2A), suggesting that TAB1 and TAB1beta are concurrently expressed.


View larger version (36K):
[in this window]
[in a new window]
 
Fig. 2.   Expression of TAB1beta . A, expression of TAB1beta and TAB1 mRNA in different cells. Total RNA was isolated from different cells, and RT-PCR was performed using a common 5' primer and TAB1beta - or TAB1-specific 3' primer. B, proteins expression of TAB1 isoforms in different cells. Total cell lysates of different cells as indicated were resolved by SDS-PAGE and immunoblotted with antibody against the N terminus of TAB1 and TAB1beta . The ectopically overexpressed TAB1 and TAB1beta in 293 cells are shown in the right panel. A short exposure time was used to selectively show the overexpressed TAB1 and TAB1beta . C, the 19-bp hairpin RNAs (hpRNA) encoded by R1 and R1(M) RNAi constructs. D, the expression of TAB1beta protein in MDA231 cells that were stably transfected with RNAi constructs for TAB1beta . Lysates of the cells stably transfected with nothing (-), empty pSuper vector, R1, R1(M), R2, or R3 were subjected to immunoblotting using antibody against the N terminus of TAB1 and TAB1beta . E, the expression of TAB1 and TAB1beta mRNA in the stably transfected MDA231 cell lines. Competitive quantitative RT-PCR was performed using the total RNA isolated from the cell lines indicated.

To examine the protein expression of TAB1 and TAB1beta , total cell lysates were prepared from HEK293, HEPG2, MDA231, RPMI8226, and MCF-7 cells. The expression of TAB1 or TAB1beta was analyzed by Western blotting using anti-TAB1 antibodies. The antibody against a peptide, found in the N terminus of both TAB1 and TAB1beta , detected two major protein bands in Western blotting analysis (Fig. 2B, left panel). The same result was obtained with an antibody that was raised by using recombinant TAB1beta protein (data not shown). We overexpressed TAB1 and TAB1beta in 293 cells and detected the ectopically expressed TAB1 and TAB1beta by Western blotting (Fig. 2B, right panel). Comparison of the migrations of endogenous proteins detected by the anti-TAB1/TAB1beta antibodies with the ectopically expressed TAB1 and TAB1beta gives no indication which protein band or bands are TAB1 or TAB1beta , because there was a higher molecular weight band of endogenous protein that could be post-translationally modified TAB1 or TAB1beta or could even be another TAB1 isoform. We have tried to raise TAB1- and TAB1beta -specific antibodies using C-terminal peptides from TAB1 and TAB1beta . Because the difference between TAB1 and TAB1beta is very limited (29 and 69 amino acids), there were not many peptide sequences that could be chosen to generate antibodies. As a result, we have not generated any antibodies that can selectively detect TAB1 or TAB1beta . We cannot formally conclude whether TAB1 and/or TAB1beta were expressed as protein in cells by the Western blotting analysis shown in Fig. 2B, but the protein expression of at least one of TAB1 isoforms was confirmed.

RNA interference (RNAi) is the process of gene silencing whereby double-stranded RNA induces the homology-dependent degradation of its cognate mRNA. We designed RNAi constructs to target TAB1 and TAB1beta using the method of Brummelkamp et al. (38) and stably transfected these constructs into MDA231 cells. Stable transfection of the RNAi constructs, which target the common region of TAB1 and TAB1beta , and the specific region of TAB1 did not affect the expression of TAB1s in MDA231 cells (data not shown). Transfection of R1 (Fig. 2C), one of the RNAi constructs that were designed to target TAB1beta , resulted in reduction of the expression of the upper-band protein detected by anti-TAB1 antibody (Fig. 2D). In contrast, transfection of R1(M), which had a single point mutant in comparison with R1 (Fig. 2C), did not produce any effect, indicating the specificity of the RNAi effect. The other two RNAi constructs that were designed to target TAB1beta also had no effect on TAB1beta expression. These results indicated that the upper band is TAB1beta . Because the apparent molecular weight of endogenous TAB1beta is higher than ectopically expressed TAB1beta (Fig. 2, B and D), the endogenous TAB1beta is most likely being post-translationally modified. The reduction of TAB1beta protein by R1 is specific, because the protein level of the lower band, which is most likely to be TAB1, and p38alpha were not affected by R1 (Fig. 2D). To further confirm that R1 selectively targeted TAB1beta mRNA, we examined mRNA expression of TAB1 and TAB1beta by competitive quantitative RT-PCR using their specific primers. The cells stably transfected with R1, but not with R1(M) or empty vector, had reduced TAB1beta mRNA (Fig. 2E), confirming that R1 specifically mediated a reduction of TAB1beta mRNA. Thus, TAB1beta protein is expressed in cells.

TAB1beta Interacts with p38alpha but Not with TAK1-- Our previous studies have demonstrated that the TAB1 protein interacts with p38alpha (23). To find out if the TAB1beta interacts with p38alpha , expression plasmids encoding TAB1beta , TAB1, and FLAG-p38alpha were co-transfected into 293 cells. The FLAG-p38alpha was precipitated with agarose beads conjugated with anti-FLAG antibody (M2), and the existence of TAB1 in the immunoprecipitation complexes was detected by immunoblotting with anti-TAB1 antibody. The results showed that both TAB1beta and TAB1 bind with p38alpha (Fig. 3A).


View larger version (32K):
[in this window]
[in a new window]
 
Fig. 3.   TAB1beta interacts with p38alpha but not TAK1. A, FLAG-p38alpha was co-expressed with TAB1 or TAB1beta in 293 cells. Cells were lysed 24 h after transfection, and immunoprecipitation was performed with ant-FLAG antibody. The immunoprecipitates and cell lysates were analyzed by immunoblotting with antibody against TAB1 or FLAG as indicated. TAB1beta was immunoprecipitated with p38alpha . B, the interaction of TAB1beta with p38beta , p38gamma , or p38delta was examined as in A. These three p38 family members had no or very weak interaction with TAB1beta . C, the interaction of TAB1beta with JNK1 or ERK2 was examined as in A. No interaction was detected. D, TAB1 or TAB1beta was co-expressed with HA-TAK1. TAK1 was immunoprecipitated with anti-HA antibody. The immunoprecipitates and cell lysates were analyzed by immunoblotting with antibody to TAB1 or HA as indicated. TAB1beta was not co-immunoprecipitated with TAK1. The experiments were performed three times with comparable results.

To investigate the specificity of the interaction between TAB1beta and p38alpha , we first tested if TAB1beta could interact with three other p38 isoforms. As shown in Fig. 3B, a small amount of TAB1beta was pulled down by p38gamma but not by p38beta or p38delta . The expression level of p38gamma was higher than any other p38 subfamily members (Fig. 3B, middle panel, and data not shown), but the amount of TAB1beta pulled down with p38gamma was very little in comparison with p38alpha (Fig. 3, A and B), so the affinity of TAB1beta and p38gamma should be much smaller than TAB1beta and p38alpha . We next evaluated whether there is interaction between TAB1beta with the other two MAPKs, JNK1 and ERK2. The results demonstrated that TAB1 bound strongly with p38alpha but not with ERK2 or JNK1 (Fig. 3C).

TAB1 was originally identified as a binding protein of TAK1. It promotes the TAK1 kinase activation through the binding with TAK1. The sequence data shown in Fig. 1A suggest that TAB1beta does not have a TAK1 binding domain, however, whether TAB1beta can interact with TAK1 needs to be examined. We transfected the HA-TAK1, TAB1, and TAB1beta expression plasmids into 293 cells. Immunoprecipitation was performed with agarose beads conjugated with anti-HA antibody. The immunoprecipitates were immunoblotted with anti-TAB1 antibody. We found that immunoprecipitation of TAK1 pulled down TAB1 but not TAB1beta (Fig. 3D), indicating that there was no interaction between TAB1beta and TAK1. These data agree with domain mapping of TAB1 reported by other groups and indicate that TAB1beta only interacts with p38alpha and not TAK1.

Activation of p38alpha by TAB1beta -- It has been shown that interaction of TAB1 with p38alpha resulted in autoactivation of p38alpha (23). We next evaluated the ability of TAB1beta to activate p38alpha . The expression plasmids of TAB1beta , TAB1, and p38alpha were co-transformed into 293 cells in different combinations, and the extent of p38alpha dual phosphorylation was examined by immunoblotting using anti-phospho-p38 antibody. As we reported previously, co-expression of TAB1 with p38alpha in cells led to p38alpha phosphorylation; expression of TAB1beta also led to the phosphorylation of p38alpha (Fig. 4A). To ensure that the phosphorylation of p38alpha detected by anti-phospho-p38 antibody indeed represented p38alpha activity, FLAG-p38alpha protein was immunoprecipitated by using anti-FLAG M2 beads and further analyzed by in vitro kinase assay using MBP as a substrate. The increase of p38alpha dual phosphorylation by TAB1 or TAB1beta was correlated with the increase of p38alpha activity (Fig. 4B).


View larger version (19K):
[in this window]
[in a new window]
 
Fig. 4.   TAB1beta promotes p38alpha activation. A, FLAG-p38alpha was co-expressed with TAB1beta or TAB1 in 293 cells. FLAG-p38alpha was immunoprecipitated with anti-FLAG antibody. The immunoprecipitates and cell lysates were analyzed by immunoblotting with anti-phospho-p38, anti-FLAG, or anti-TAB1 as indicated. Co-expression of TAB1beta or TAB1 increased p38alpha phosphorylation. B, the experiments were performed as in A, and the immunoprecipitates were used as kinase in the in vitro kinase assay using MBP as a substrate. TAB1beta and TAB1 increased p38alpha kinase activity. C, expression vectors of a luciferase reporter gene under the control of 5xGal4 binding site (5xGAL), GAL4 binding domain fused with ATF2 activation domain (GAL-ATF2), TAB1beta , TAB1, and FLAG-p38alpha were transfected into 293 cells in different combinations as indicated. Luciferase activity was measured 24 h later. TAB1 and TAB1beta enhanced ATF2-dependent luciferase expression when co-expressed with p38alpha . Experiments were performed three times with comparable results.

It is known that p38alpha activation has an effect on expression of a number of genes. Previous studies showed that activation of p38alpha by MKK6 or MKK3 augments the transcriptional activity of ATF2 in transient transfection assays (19). To determine whether activation of p38alpha by TAB1beta interaction had a similar effect on ATF2, we used fusion proteins containing the transactivation domain of ATF2 fused with GAL4 DNA binding domain. Whether TAB1beta -mediated p38alpha phosphorylation can transduce signaling downstream was determined by whether TAB1beta -mediated activation of p38alpha could enhance the transcriptional activity of GAL4-ATF2 protein. The GAL4-driven luciferase reporter construct, the expression plasmid of GAL-ATF2 fusion protein, and the expression plasmids of p38alpha , TAB1, or TAB1beta were transfected into 293 cells in different combinations. Luciferase activity was examined 48 h after transfection. Luciferase expression was significantly increased in the cells co-expressing p38alpha and TAB1 or TAB1beta (Fig. 4C). Thus, TAB1beta -mediated p38alpha activation can transduce signaling downstream.

TAB1 but Not TAB1beta Activates TAK1-- Because TAB1 activates TAK1 (24), we investigated whether the TAB1beta had effect on TAK1. The expression vectors of TAB1beta or TAB1 were transfected into 293 cells with HA-TAK1. HA-TAK1 was immunoprecipitated by anti-HA beads, and kinase activity of TAK1 was examined by a coupled in vitro kinase assay using the GST-MKK4 and His-p38alpha (M) as substrates (Fig. 5A). The increase in kinase activity of TAK1 can be determined by activation of MKK4, which in turn phosphorylates p38alpha (M). As reported by others, co-expression of TAB1 increased TAK1 activity. In contrast, expression of TAB1beta had no effect TAK1 activity (Fig. 5A). These data are consistent with the data in Fig. 3D, showing that TAB1beta did interact with TAK1.


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 5.   TAB1beta does not activate TAK1. A, TAB1 or TAB1beta were co-expressed with HA-TAK1 in 293 cells. TAK1 was immunoprecipitated, and in vitro coupled kinase assay was performed using immunoprecipitates as kinase, GST-MKK4 as a substrate for TAK1, and His-p38alpha (M) as a substrate for MKK4. Only TAB1 increased TAK1 activity. B, NF-kappa B reporter was co-expressed with TAK1, TAB1, or TAB1beta in 293 cells as indicated. Luciferase activity was measured 24 h later. TAK1 and TAB1beta co-expression did not enhance NF-kappa B-dependent gene expression. The experiments were performed two times with comparable results.

It was reported that TAB1-mediated TAK1 activation could dramatically induce NF-kappa B reporter gene expression. We were able to reproduce the result of NF-kappa B activation by TAK1 and TAB1 co-expression (Fig. 5B). In contrast to TAB1 co-expression, TAB1beta co-expression did not have any effect on TAK1-mediated NF-kappa B-dependent reporter gene expression (Fig. 5B). These data further support the notion that TAB1beta does not have any effect on TAK1.

TAB1beta -induced p38alpha Activation Is Dependent on p38alpha Autophosphorylation-- We uncovered recently that TAB1-mediated p38alpha activation is through p38alpha autophosphorylation (23). The same approaches were used here to determine whether TAB1beta -mediated p38alpha phosphorylation is also autophosphorylation. First, we examined whether TAB1beta -mediated p38alpha phosphorylation can be inhibited by dominant negative MKK6 or TAK1, the known upstream kinases of p38alpha . Mutation of regulatory phosphorylation sites in MKK6 to alanine (MKK6(A)) resulted in the loss of kinase activity, and this mutant has been successfully used as a dominant negative mutant in a number of studies (19, 31, 39). Mutation in the ATP binding site of TAK1 (TAK1(K63W)) leads to kinase death, and this mutant can be used as a dominant negative mutant (24, 27). We co-transfected MKK6(A) or TAK1(K63W) with TAB1beta and p38alpha and compared the level of p38alpha phosphorylation. Expression of MKK6(A) or TAK1(K63W) did not interfere with TAB1beta -induced p38alpha activation (Fig. 6A). Similar results were obtained when MKK3(A) or MKK4(A) was expressed (data not shown). These results agreed with the report that TAB1-mediated p38alpha phosphorylation is independent from the known upstream kinases of p38alpha and indicated that TAB1beta -mediated p38alpha occurs most likely through a similar mechanism. Second, we investigated whether TAB1beta -induced p38alpha activation is also dependent on p38alpha intrinsic enzymatic activity. Two methods were used to diminish the p38alpha intrinsic activity. One was to use kinase-dead p38alpha mutants. A p38alpha mutant with an impaired ATP binding site (Lys-53 to Met mutant termed p38alpha (M)) was used in the experiments. The other method was to treat the cells with p38 inhibitor SB203580 (40, 41). As shown in Fig. 6B, phosphorylation of the kinase-dead mutant of p38alpha was observed in the cells expressing MKK6(E) but not in the cells expressing TAB1beta ; activation of p38alpha by TAB1beta was inhibited by treatment of cells with SB203580. These data indicate that TAB1beta -mediated p38alpha phosphorylation requires intrinsic kinase activity of p38alpha . Third, we examined TAB1beta -mediated p38alpha activation in vitro. p38alpha and TAB1beta were synthesized in and purified from bacteria as His-tagged and GST fusion proteins, respectively. GST-TAB1beta was incubated with His-p38alpha in kinase reaction buffer with cold ATP. The activity of p38alpha was examined by using myelin basic protein (MBP) as a substrate. TAB1beta significantly increased p38alpha activity toward MBP. Moreover, TAB1beta -mediated p38alpha activity was reduced by SB203580 (Fig. 6C). Collectively, our data demonstrate that TAB1beta -mediated p38alpha activation is p38alpha autoactivation.


View larger version (25K):
[in this window]
[in a new window]
 
Fig. 6.   TAB1beta -mediated p38alpha dual phosphorylation is a p38alpha autophosphorylation. A, 293 cells were co-transfected with different expression vectors as indicated. 2- and 4-fold plasmid DNA of MKK6(A) and TAK1(K63W) were used in some samples to increase the expression of these dominant negative proteins. Immunoprecipitation and Western blotting were performed as in Figs. 2 and 3. Overexpression of dominant negative MKK6 (MKK6(A)) or TAK1 (TAK1(K63W)) had no effect on TAB1beta -mediated p38alpha dual phosphorylation. B, co-expression of FLAG-p38alpha or FLAG-p38alpha (M) with TAB1 or MKK6(E) was performed as in A. SB203580 (5 µM) was added into cell culture medium 4 h after transfection in the sample indicated. Immunoprecipitation and Western blotting were performed as in A. TAB1-mediated p38alpha dual phosphorylation requires intrinsic p38alpha activity. C, TAB1-medtated p38alpha phosphorylation in vitro requires intrinsic p38alpha kinase activity. GST-TAB1beta , His-p38alpha , and MBP were incubated in kinase reaction buffer containing [32P]ATP in the presence of different concentrations of SB203580. The kinase reaction was stopped by SDS-sample buffer, and phosphorylated proteins were resolved on SDS-PAGE and exposed to x-ray film. SB203580 inhibited both autophosphorylation and activity of p38alpha . Data shown are representative of two to three independent experiments.

Extracellular Stimuli Affects TAB1beta and p38alpha Association-- We reported previously that TAB1-dependent activation of p38alpha was used differently by different extracellular stimuli (23). Due to the identification of the similar effects of TAB1beta on p38alpha activation to those of TAB1, we test whether TAB1beta could selectively be involved in p38alpha activation induced with different stimuli. The 293 cells were transfected with TAB1beta and stimulated with different stimuli. As shown in Fig. 7A, overexpression of TAB1beta significantly enhanced peroxynitrite and TNF-induced p38alpha phosphorylation. The effects of TAB1beta on anisomycin- and arsonite-induced p38alpha phosphorylation were modest. Overexpression of TAB1beta did not effect high osmolar sorbital-induced p38alpha phosphorylation. The selective involvement of TAB1beta in enhancing p38alpha phosphorylation induced by different stimuli fit well with the profiles of kinase cascade-independent p38alpha activation reported previously (23). Because the association of TAB1beta with p38alpha can induce p38alpha activation, we next investigated whether the extracellular stimuli selectively affected the association of TAB1beta with p38alpha . 293 cells were transfected with expression plasmids of TAB1 and p38alpha for 24 h, and then were stimulated with several extracellular stimuli. The association of TAB1beta with p38alpha was examined by using a co-immunoprecipitation assay. We observed that TNF and peroxynitrite significantly increased the binding of TAB1beta with p38alpha and anisomycin or arsonite also induced this association but to a lesser extent. Sorbitol failed to induce this association (Fig. 7B). These data correlated with the enhancement of p38alpha phosphorylation by TAB1beta overexpression in cells treated with different stimuli (Fig. 7, A and B). The time course of TAB1beta -p38alpha interactions induced by TNF is shown in Fig. 7C. These results suggest that enhanced p38alpha and TAB1 interactions are differentially involved in different extracellular stimuli-induced cellular activation.


View larger version (31K):
[in this window]
[in a new window]
 
Fig. 7.   TAB1beta selectively involves p38alpha activation induced by different extracellular stimuli. A, FLAG-p38alpha was co-expressed with or without TAB1beta in 293 cells. 24 h later, the cells were treated with TNF-alpha (100 ng/ml), peroxynitrite (500 µM), anisomycin (50 ng/ml), arsonite (200 µM), and sorbitol (0.4 M) for 30, 5, 30, and 30 min, respectively. The levels of p38alpha and phospho-p38alpha were determined by immunoprecipitation and immunoblotting as in Figs. 2 and 3. Expression of TAB1beta enhanced p38alpha phosphorylation induced by some stimuli but not others. B, TAB1beta and FLAG-p38alpha were expressed in 293 cells, and the cells were treated with different stimuli as indicated. FLAG-p38alpha was immunoprecipitated, and the co-precipitated TAB1beta was determined. The amount of TAB1beta co-precipitated varied when the cells were treated with different stimuli. C, TAB1beta and FLAG-p38alpha were expressed in 293 cells, and the cells were treated with TNF for different periods of time. FLAG-p38alpha was immunoprecipitated, and co-precipitated TAB1beta was determined. A time-dependent co-precipitation of TAB1beta was observed in TNF-treated cells. Data shown are representative of two to three independent experiments.

Reduction of TAB1beta by RNAi Reduced Basal Activity of p38alpha and Invasiveness of MDA231 Cells-- MDA231 is an invasive breast cancer cell line. We showed previously that the invasiveness of MDA231 is dependent on, at least partially, the high basal activity of p38alpha (42). Because the RNAi construct R1 can specifically inhibit TAB1beta expression in MDA231 cells (Fig. 2D), we evaluated the role of TAB1beta in p38alpha activity and invasion of MDA231 cells. As shown in Fig. 8A, the basal level of p38alpha phosphorylation was significantly reduced in R1 transfected cells in comparison with non-transfected, vector-transfected, and R1(M)-transfected cells. Therefore, TAB1beta has a role in regulating basal activity of p38alpha . Stimulation of MDA231 cells with TNF can lead to a 2- to 3-fold increase in p38alpha phosphorylation in vector- and R1(M)-transfected cells (Fig. 8B). The p38alpha phosphorylation in R1-transfected cells was significantly lower in comparison with controls before and after TNF stimulation (Fig. 8B), and the -fold induction of p38alpha phosphorylation by TNF was also lower. We used Matrigel invasion assays to evaluate whether reduction of TAB1beta by RNAi had an effect on invasion of MDA231 cells. As shown in Fig. 8C, invasion of MDA231 cells was inhibited ~50% when TAB1beta was knocked down by RNAi.


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 8.   TAB1beta RNAi reduces basal activity of p38alpha and invasiveness of MDA231 cells. A, basal level of p38alpha phosphorylation in MDA231 cell lines stably transfected with TAB1beta RNAi constructs. Lysates of the cells were immunoblotted with anti-phospho-p38 and anti-p38alpha antibody. B, p38alpha phosphorylation in the stably transfected MDA231 cell lines stimulated with or without TNF. C, Matrigel invasion assay of the stably transfected MDA231 cell lines. Quantification of cells invading the undersurface of the Matrigel chamber is shown.


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

TAB1beta is a splicing variant of TAB1 based on its gene structure (Fig. 1B). The expression of TAB1beta was demonstrated by combined results from RT-PCR, Western blotting, and RNAi (Fig. 2). TAB1beta has a different C-terminal sequence from that of TAB1 and does not have a TAK1 interacting domain that is harbored at the C terminus of TAB1. TAB1beta retained the p38alpha interacting domain located in the N-terminal side to the TAK1 interacting domain in TAB1. In addition to the interacting domains for p38alpha and TAK1, computer analysis indicated that there is a protein phosphatase 2C (PP2C)-like domain in the N-terminal of TAB1 and TAB1beta . The domain structure of TAB1 and TAB1beta is shown in Fig. 9. Like TAB1, TAB1beta interacts with p38alpha , and this interaction leads to autoactivation of p38alpha . Unlike TAB1, TAB1beta cannot interact with TAK1 and has no effect on TAK1 activity. Specific inhibition of TAB1beta expression by RNAi reduced basal activity of p38alpha in MDA231 cells and decreased the invasiveness of MDA231 cells, indicating that TAB1beta is involved in the regulation of p38alpha activity in a physiological setting.


View larger version (10K):
[in this window]
[in a new window]
 
Fig. 9.   Domain structure of TAB1 and TAB1beta .

Our data show that TAB1- and TAB1beta -mediated p38alpha activation is an autophosphorylation of p38alpha . However, how an interaction with TAB1 or TAB1beta leads to p38alpha autoactivation remains to be elucidated. It is well established that activation of MAPKs by their corresponding MAPKKs is through dual phosphorylation on specific threonine and tyrosine residues (4, 18), and we have shown in our previous work (23) that p38alpha autoactivation is also achieved by dual phosphorylation on the regulatory sites. MAPKs are serine/threonine kinases, but low level phosphorylation on their regulatory tyrosine residue has been observed in ERK1, ERK2, and p38alpha in vitro (23, 43, 44). It was demonstrated quite a long time ago that this phosphorylation is mediated by autophosphorylation (43-45). These early works suggested that autophosphorylation on one of the regulatory sites could occur in MAPKs, although the efficiency was low. It is possible that the binding of TAB1 or TAB1beta to p38alpha changes the confirmation of p38alpha and facilitates autophosphorylation of p38alpha on its regulatory sites. p38alpha has a similar overall structure to the ERK2 MAPK (46). Analysis of crystal structures of inactive and active forms of the ERK2 has indicated that phosphorylation of ERK2 results in a conformational change to an "energetically unfavorable state" (47). It is possible that autophosphorylation of p38alpha can be enhanced by stabilization of p38alpha in an "energetically unfavorable transient confirmation" by the binding of an "activator" protein like TAB1 or TAB1beta (48, 49). A recent study by Emrick et al. (50) shows that a double mutation of leucine 73 to proline and serine 151 to aspartic acid in ERK2 leads to ERK2 autophosphorylation and the mutant is constitutively active. By analyzing the three-dimensional structure of ERK2, authors of this work suggested that mutations of L73P and S151D permit a regulatory phosphorylation site Y185 to move within hydrogen bonding distance of the catalytic aspartate Asp-147, facilitating the phosphoryl transfer. This work supports the idea that conformational changes can lead to MAPK autophosphorylation and activation. The activation mechanism of p38alpha , after binding to TAB1 or TAB1beta , should be similar to the constitutive active ERK2 mutant, because both are activated by autophosphorylation. It is thus very likely that TAB1 and p38alpha interaction leads to conformational changes that are similar to that in ERK2(L73P,S151D) and that such changes relieve structural constraints in the catalytic site that suppress autophosphorylation and autoactivation.

Autophosphorylation is a common mechanism for the activation of a number of different kinases such as receptor tyrosine kinases (51, 52) and some of the MAPKKKs (53-56). TAB1-mediated TAK1 activation was shown to be a result of autoactivation (26, 29). Because TAK1 and p38alpha bind to different sites of TAB1 and both are activated by autophosphorylation after binding to TAB1, one would assume that TAB1 has a structural feature that triggers autophosphorylation of these two different kinases. Sequence analysis revealed that the N-terminal two-thirds of TAB1 and TAB1beta protein had low level sequence homology with phosphatase PP2C. However, phosphatase activity has not been detected in TAB1 or TAB1beta so far. Whether this PP2C-like domain has a function in regulating p38alpha and/or TAK1 activity is a subject for future studies. Because the C-terminal 68-amino acid peptide of TAB1 is sufficient to bind and activate TAK1, the N-terminal PP2C-like domain is dispensable for TAK1 activation. To date we were unable to obtain a protein that only contains the p38alpha interacting domain, so we were unable to determine whether this domain alone can lead to p38alpha activation. A common docking domain has been identified in MAPKs as a common site for binding to upstream MKK, downstream substrates, and dual phosphatase (57, 58). A gain-of-function mutant of a Drosophila melanogaster MAPK was found to be resulting from disrupted interactions between MAPKs and phosphatase by a mutation in a common docking domain (D334N) (59). It is possible that TAB1 prevents p38alpha and phosphatase interactions to enhance p38alpha activity. However, this possibility was excluded, because purified TAB1 can cause p38alpha autophosphorylation in the absence of any phosphatase in vitro (23). Furthermore, TAB1 did not compete with PP2C-mediated inactivation of p38alpha in co-expression experiments (data not shown). Genetic screening has isolated a gain-of-function mutant of p38alpha (49). This mutant (F327L or S) can gain kinase activity in the absence of tyrosine phosphorylation, suggesting the driven force for converting p38alpha to active conformation in this mutant is different from that of TAB1-mediated p38alpha activation.

The p38alpha MAPK is activated in response to a variety of extracellular stimuli, including pro-inflammatory cytokines and environmental stresses (4, 60). How these different stimuli via different receptors or other molecular sensors activate p38alpha is still not fully understood. Previous work by a number of investigators, including us, showed that there are at least two different mechanisms immediately upstream of p38alpha in regulating p38alpha activity (19, 23). One is dependent on upstream kinase MKK3 or MKK6; another is dependent on TAB1-mediated p38alpha autophosphorylation. The data presented in this report added TAB1beta into the regulation network of p38alpha . To date we were unable to generate isoform-specific antibodies for TAB1beta and TAB1, however, in combination with RNAi, we were able to identify the protein band of TAB1beta resolved on SDS-PAGE (Fig. 2D). The role of TAB1beta in controlling the basal activity of p38alpha has been documented by our experiments (Fig. 8). Whether TAB1 and TAB1beta have different roles in mediating p38alpha activation in cells has not been determined. Nevertheless, TAB1 and TAB1beta are clearly different in their functions, because TAB1 is capable of regulating both TAK1 and p38alpha whereas TAB1beta only activates p38alpha . The different functions of TAB1 splicing variants may be a mechanism required for precise regulation of intracellular signaling that controls p38alpha activation.

    ACKNOWLEDGEMENT

We thank Dr. Reuven Agami for the pSuper vector.

    FOOTNOTES

* This work was supported by United States Public Health Services Grant AI41637 from NIAID, National Institutes of Health.The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

The nucleotide sequence(s) reported in this paper has been submitted to the GenBankTM/EBI Data Bank with accession number(s) AF425640.

Dagger To whom correspondence should be addressed: Dept. of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037. Tel.: 858-784-8704; Fax: 858-784-8665; E-mail: jhan@scripps.edu.

Published, JBC Papers in Press, November 11, 2002, DOI 10.1074/jbc.M210918200

    ABBREVIATIONS

The abbreviations used are: MAPK, mitogen-activated protein kinase; MAPKK, MAPK kinase; MAPKKK, MAPK kinase kinase; TAB1, transforming growth factor-beta -activated protein kinase 1-binding protein 1; ERK, extracellular signal-regulated kinase; MEK1, 2, MAPK/ERK kinase 1 and 2; JNK, c-Jun N-terminal kinase; SAPK, stress-activated protein kinase; EST, expressed sequence tag; RT, reverse transcription; CMV, cytomegalovirus; mAb, monoclonal antibody; HA, hemagglutinin; GST, glutathione S-transferase; MBP, myelin basic protein; RNAi, RNA interference; TNF, tumor necrosis factor; PP2C, protein phosphatase 2C; MKK, MAPK kinase; BMK1, Big MAPK1.

    REFERENCES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

1. Waskiewicz, A. J., and Cooper, J. A. (1995) Curr. Opin. Cell Biol. 7, 798-805[CrossRef][Medline] [Order article via Infotrieve]
2. Karin, M., and Hunter, T. (1995) Curr. Biol. 5, 747-757[Medline] [Order article via Infotrieve]
3. Ip, Y. T., and Davis, R. J. (1998) Curr. Opin. Cell Biol. 10, 205-219[CrossRef][Medline] [Order article via Infotrieve]
4. Pearson, G., Robinson, F., Beers Gibson, T., Xu, B. E., Karandikar, M., Berman, K., and Cobb, M. H. (2001) Endocr. Rev. 22, 153-183[Abstract/Free Full Text]
5. Guan, K. L. (1994) Cell Signal. 6, 581-589[CrossRef][Medline] [Order article via Infotrieve]
6. Dong, C., Davis, R. J., and Flavell, R. A. (2002) Annu. Rev. Immunol. 20, 55-72[CrossRef][Medline] [Order article via Infotrieve]
7. Schaeffer, H. J., and Weber, M. J. (1999) Mol. Cell. Biol. 19, 2435-2444[Free Full Text]
8. Cobb, M. H., and Goldsmith, E. J. (1995) J. Biol. Chem. 270, 14843-14846[Free Full Text]
9. Han, J., Lee, J.-D., Bibbs, L., and Ulevitch, R. J. (1994) Science 265, 808-811[Medline] [Order article via Infotrieve]
10. Cobb, M. H., Boulton, T. G., and Robbins, D. J. (1991) Cell Regul. 2, 965-978[Medline] [Order article via Infotrieve]
11. Lee, J. C., Laydon, J. T., McDonnell, P. C., Gallagher, T. F., Kumar, S., Green, D., McNulty, D., Blumenthal, M. J., Heys, J. R., Landvatter, S. W., Strickler, J. E., McLaughlin, M. M., Siemens, I. R., Fischer, S. M., Livi, G. P., White, J. R., Adams, J. L., and Young, P. R. (1994) Nature 372, 739-746[CrossRef][Medline] [Order article via Infotrieve]
12. Sugden, P. H., and Clerk, A. (1997) Cell Signal. 9, 337-351[CrossRef][Medline] [Order article via Infotrieve]
13. Kato, Y., Kravchenko, V. V., Tapping, R. I., Han, J., Ulevitch, R. J., and Lee, J. D. (1997) EMBO J. 16, 7054-7066[Abstract/Free Full Text]
14. Chao, H. H. J., Kwak, S. P., and Dixon, J. E. (1995) Mol. Biol. Cell 6, 736
15. Gutkind, J. S. (2000) Science's STKE http://stke.sciencemag.org/cgi/content/ full/OC_sigtrans;2000/40/re1
16. Ferrell, J. E., and Bhatt, R. R. (1997) J. Biol. Chem. 272, 19008-19016[Abstract/Free Full Text]
17. Fanger, G. R., Gerwins, P., Widmann, C., Jarpe, M. B., and Johnson, G. L. (1997) Curr. Opin. Genet. Dev. 7, 67-74[CrossRef][Medline] [Order article via Infotrieve]
18. Widmann, C., Gibson, S., Jarpe, M. B., and Johnson, G. L. (1999) Physiol. Rev. 79, 143-180[Abstract/Free Full Text]
19. Raingeaud, J., Whitmarsh, A. J., Barrett, T., Derijard, B., and Davis, R. J. (1996) Mol. Cell. Biol. 16, 1247-1255[Abstract]
20. Wu, X., Noh, S. J., Zhou, G., Dixon, J. E., and Guan, J.-L. (1996) J. Biol. Chem. 271, 3265-3271[Abstract/Free Full Text]
21. Zhang, B. H., and Guan, K. L. (2001) Exp. Lung Res. 27, 269-295[CrossRef][Medline] [Order article via Infotrieve]
22. Garrington, T. P., and Johnson, G. L. (1999) Curr. Opin. Cell Biol. 11, 211-218[CrossRef][Medline] [Order article via Infotrieve]
23. Ge, B., Gram, H., di Padova, F., Huang, B., New, L., Ulevitch, R. J., Luo, Y., and Han, J. (2002) Science 295, 1291-1294[Abstract/Free Full Text]
24. Shibuya, H., Yamaguchi, K., Shirakabe, K., Tonegawa, A., Gotoh, Y., Ueno, N., Irie, K., Nishida, E., and Matsumoto, K. (1996) Science 272, 1179-1182[Abstract]
25. Sakurai, H., Miyoshi, H., Mizukami, J., and Sugita, T. (2000) FEBS Lett. 474, 141-145[CrossRef][Medline] [Order article via Infotrieve]
26. Kishimoto, K., Matsumoto, K., and Ninomiya-Tsuji, J. (2000) J. Biol. Chem. 275, 7359-7364[Abstract/Free Full Text]
27. Ninomiya-Tsuji, J., Kishimoto, K., Hiyama, A., Inoue, J., Cao, Z., and Matsumoto, K. (1999) Nature 398, 252-256[CrossRef][Medline] [Order article via Infotrieve]
28. Ono, K., Ohtomo, T., Sato, S., Sugamata, Y., Suzuki, M., Hisamoto, N., Ninomiya-Tsuji, J., Tsuchiya, M., and Matsumoto, K. (2001) J. Biol. Chem. 276, 24396-24400[Abstract/Free Full Text]
29. Sakurai, H., Miyoshi, H., Toriumi, W., and Sugita, T. (1999) J. Biol. Chem. 274, 10641-10648[Abstract/Free Full Text]
30. Derijard, B., Raingeaud, J., Barrett, T., Wu, I., Han, J., Ulevitch, R. J., and Davis, R. J. (1995) Science 267, 682-685[Medline] [Order article via Infotrieve]
31. Han, J., Lee, J.-D., Jiang, Y., Li, Z., Feng, L., and Ulevitch, R. J. (1996) J. Biol. Chem. 271, 2886-2891[Abstract/Free Full Text]
32. Jiang, Y., Chen, C., Li, Z., Guo, W., Gegner, J. A., Lin, S., and Han, J. (1996) J. Biol. Chem. 271, 17920-17926[Abstract/Free Full Text]
33. Li, Z., Jiang, Y., Ulevitch, R. J., and Han, J. (1996) Biochem. Biophys. Res. Commun. 228, 334-340[CrossRef][Medline] [Order article via Infotrieve]
34. Jiang, Y., Gram, H., Zhao, M., New, L., Gu, J., Feng, L., di Padova, F., Ulevitch, R. J., and Han, J. (1997) J. Biol. Chem. 272, 30122-30128[Abstract/Free Full Text]
35. Han, J., Lee, J. D., Tobias, P. S., and Ulevitch, R. J. (1993) J. Biol. Chem. 268, 25009-25014[Abstract/Free Full Text]
36. Zhao, M., New, L., Kravchenko, V. V., Kato, Y., Gram, H., di Padova, F., Olson, E. N., Ulevitch, R. J., and Han, J. (1999) Mol. Cell. Biol. 19, 21-30[Abstract/Free Full Text]
37. Schindler, U., and Baichwal, V. R. (1994) Mol. Cell. Biol. 14, 5820-5831[Abstract]
38. Brummelkamp, T. R., Bernards, R., and Agami, R. (1929) Science 296, 550-553[Medline] [Order article via Infotrieve]
39. Wang, X., McGowan, C. H., Zhao, M., He, L., Downey, J. S., Fearns, C., Wang, Y., Huang, S., and Han, J. (2000) Mol. Cell. Biol. 20, 4543-4552[Abstract/Free Full Text]
40. Cuenda, A., Rouse, J., Doza, Y. N., Meier, R., Cohen, P., Gallagher, T. F., Young, P. R., and Lee, J. C. (1995) FEBS Lett. 364, 229-233[CrossRef][Medline] [Order article via Infotrieve]
41. Tong, L., Pav, S., White, D. M., Rogers, S., Crane, K. M., Cywin, C. L., Brown, M. L., and Pargellis, C. A. (1997) Nat. Struct. Biol. 4, 311-316[Medline] [Order article via Infotrieve]
42. Huang, S., New, L., Pan, Z., Han, J., and Nemerow, G. R. (2000) J. Biol. Chem. 275, 12266-12272[Abstract/Free Full Text]
43. Robbins, D. J., Zhen, E., Owaki, H., Vanderbilt, C. A., Ebert, D., Geppert, T. D., and Cobb, M. H. (1993) J. Biol. Chem. 268, 5097-5106[Abstract/Free Full Text]
44. Rossomando, A. J., Wu, J., Michel, H., Shabanowitz, J., Hunt, D. F., Weber, M. J., and Sturgill, T. W. (1992) Proc. Natl. Acad. Sci. U. S. A. 89, 5779-5783[Abstract]
45. Wu, J., Rossomando, A. J., Her, J. H., Del Vecchio, R., Weber, M. J., and Sturgill, T. W. (1991) Proc. Natl. Acad. Sci. U. S. A. 88, 9508-9512[Abstract]
46. Wang, Z., Harkins, P. C., Ulevitch, R. J., Han, J., Cobb, M. H., and Goldsmith, E. J. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 2327-2332[Abstract/Free Full Text]
47. Canagarajah, B. J., Khokhlatchev, A., Cobb, M. H., and Goldsmith, E. J. (1997) Cell 90, 859-869[Medline] [Order article via Infotrieve]
48. Jiang, Y., Li, Z., Schwarz, E. M., Lin, A., Guan, K., Ulevitch, R. J., and Han, J. (1997) J. Biol. Chem. 272, 11096-11102[Abstract/Free Full Text]
49. Bell, M., Capone, R., Pashtan, I., Levitzki, A., and Engelberg, D. (2001) J. Biol. Chem. 276, 25351-25358[Abstract/Free Full Text]
50. Emrick, M. A., Hoofnagle, A. N., Miller, A. S., Ten Eyck, L. F., and Ahn, N. G. (2001) J. Biol. Chem. 276, 46469-46479[Abstract/Free Full Text]
51. van der Geer, P., Hunter, T., and Lindberg, R. A. (1994) Annu. Rev. Cell Biol. 10, 251-337[CrossRef]
52. Heldin, C. H. (1995) Cell 80, 213-223[Medline] [Order article via Infotrieve]
53. Leung, I. W., and Lassam, N. (1998) J. Biol. Chem. 273, 32408-32415[Abstract/Free Full Text]
54. Gotoh, Y., and Cooper, J. A. (1998) J. Biol. Chem. 273, 17477-17482[Abstract/Free Full Text]
55. Deak, J. C., and Templeton, D. J. (1997) Biochem. J. 322, 185-192[Medline] [Order article via Infotrieve]
56. Posas, F., and Saito, H. (1998) EMBO J. 17, 1385-1394[Abstract/Free Full Text]
57. Tanoue, T., Maeda, R., Adachi, M., and Nishida, E. (2001) EMBO J. 20, 466-479[Abstract/Free Full Text]
58. Tanoue, T., Adachi, M., Moriguchi, T., and Nishida, E. (2000) Nat. Cell Biol. 2, 110-116[CrossRef][Medline] [Order article via Infotrieve]
59. Brunner, D., Oellers, N., Szabad, J., Biggs, W. H., III, Zipursky, S. L., and Hafen, E. (1994) Cell 76, 875-888[Medline] [Order article via Infotrieve]
60. Ono, K., and Han, J. (2000) Cell Signal. 12, 1-13[CrossRef][Medline] [Order article via Infotrieve]


Copyright © 2003 by The American Society for Biochemistry and Molecular Biology, Inc.