TIP30, also called CC3 or Htatip2, is a putative metastasis suppressor that promotes apoptosis and inhibits angiogenesis. Although TIP30 has several characteristic features of a tumor suppressor in in vitro analyses, tumor development as a result of TIP30 inactivation has not been demonstrated in vivo, and abnormal expression of TIP30 in human cancer has not been reported. Using genetically engineered mice and cells deficient in TIP30, we show that TIP30-deficient mice have a high incidence of hepatocellular carcinoma and other tumors, and loss of TIP30 enhances susceptibility of fibroblasts to transformation by the SV40 large T antigen. Furthermore, immunohistochemical analysis indicates that reduced TIP30 expression is associated with 33% of human hepatocellular carcinomas. Some of these carcinomas harbor missense mutations in the Tip30 gene, which cause abnormal expression of TIP30. Together, these results demonstrate that the Tip30 gene is a tumor susceptibility gene playing an important role in the suppression of hepatocarcinogenesis.

HIV-1 Tat-interacting protein TIP30, identical to the putative metastasis suppressor CC3, has been implicated in the regulation of tumor cell growth and metastasis (1, 2, 3, 4). Recent experiments demonstrated that a reduced level of TIP30 mRNA has been found in a number of tumor cell lines that include variant-small cell lung carcinoma (V-SCLC), classic-SCLC, neuroblastoma, colon cancer, and melanoma cell lines (2, 3, 4, 5, 6). Ectopic expression of the TIP30/CC3 gene in a variant SCLC line lacking TIP30/CC3 expression led to a significant suppression of the metastatic potential of these cells in SCID-hu mice. In addition, metastases in melanoma-bearing mice are significantly reduced after i.v. treatment with the TIP30/CC3 gene-cationic liposome-DNA complex (6). Subsequent work has led to the proposal that the suppression of cell growth and metastasis by ectopic expression of TIP30 may result, in part, from a propensity of cells to undergo apoptosis (2, 3, 4).

TIP30 was believed to be a transcription cofactor because it was demonstrated to potentiate Tat-mediated transcription in cooperation with components of elongation factor P-TEFb in transient transfection assays and is essential for Tat-mediated transcription in cell-free transcription assays (1, 3). TIP30 displays a serine-threonine kinase activity that can phosphorylate the carboxyl terminal domain of RNA polymerase II in a Tat-dependent manner in vitro(3). The intrinsic kinase activity is essential for TIP30 to enhance Tat-activated transcription and to sensitize NIH3T3 and v-SCLC cells to apoptosis (3). Consistent with the role of TIP30 in the suppression of tumor growth and metastasis via transcription mechanisms, studies revealed that the ectopic expression of TIP30 in v-SCLC cells and other tumor cell lines up-regulates the expression of proapoptotic factors (3) and angiogenic inhibitors, and down-regulates expression of angiogenic stimulators (4). Therefore, TIP30 may function as a tumor suppressor or tumor modifier that controls expression of genes involved in tumor growth and metastasis.

Although TIP30 has several characteristic features of a tumor suppressor, tumor development as a result of TIP30 inactivation has not been established in vivo, and abnormal expression of TIP30 in human cancer has not been reported. In this study, the generation of mice lacking Tip30 has allowed us to demonstrate a role of TIP30 in the suppression of tumor development in vivo. In addition, we have identified missense mutations in the TIP30 gene in the clinical human hepatocellular carcinomas (HCCs), which resulted in aberrant expression of TIP30.

Constructions of the Tip30 Targeting Vector and TIP30-Expressing Plasmids.

To isolate the mouse Tip30 gene, a mouse 129SvJ genomic library (Stratagene, La Jolla, CA) was screened with a mouse Tip30 cDNA probe. Fourteen overlapping clones contained a 49-kb genomic region that included five coding exons of the Tip30 gene locus. The HindIII-SacI 2.7-kb genomic fragment was replaced by LacZ in-frame and a phosphoglycerate kinase neo-cassette (Fig. 1 A). This replacement ablated the two exons that encode the NH2-terminal portion of TIP30, except for 10 amino acids after the translation start site. The targeting vector (7) included a 5.2-kb upstream homologous region and a 6.5-kb downstream region. Mutated Tip30 cDNA were generated by using pRSET-TIP30 (1) as templates and a site-directed mutagenesis kit (Stratagene). Plasmids containing Flag tag at the NH2 terminal of TIP30 were generated by cloning wild-type and mutant Tip30 coding regions between NdeI and BamHI pFlag-7 (8). pCIN4-flag-TIP30, pCIN4-flag-TIP30R106H, and pCIN4-flag-TIP30G134V expressing flag TIP30 proteins were generated by cloning Flag-tag TIP30 cDNA between EcoRI and BamHI sites of pCIN4 (1). Plasmids expressing green fluorescent protein-tagged wild-type TIP30 fusion protein (GFP-TIP30), GFP-TIP30R106H, and GFP-TIP30G134V fusion proteins were generated by cloning wild-type and mutant TIP30 coding regions between EcoRI and BamHI sites of pEGFP-C2 (Clontech Laboratories, Palo Alto, CA).

Generation of the Tip30 Knockout Mice.

E14 embryotic stem (ES) cells were electroporated with the linearized targeting vector and selected with geneticin on embryonic fibroblast feeder cells as described previously (7). In total, 348 G418-resistant clones were screened by Southern blot analysis using the 5′ external probe, and 65 clones displayed evidence for the homologous recombination of the disrupted Tip30 gene. Ten ES clones were microinjected into blastocysts of C57BL6/J female mice. Germ-line chimeras were bred to C57BL6J mice to generate heterozygous mutant F1 mice. All of the animal experimentation was performed according to the NIH guidelines in the Rockefeller University Laboratory Animal Research Center and the University of Nebraska Medical Center.

For genotyping, the genomic DNA isolated from ES cells or mouse tails was subjected to Southern blot analysis with a 5′ or 3′ external probe for lacZ or used for PCR analysis (primer sequences available on request). Mouse embryotic fibroblasts (MEFs) were prepared from E14.5 embryos obtained by heterozygous crossings of mice that had been backcrossed 10 times with C57BL6/J mice.

Northern Blot Analysis.

Northern blot analysis was performed as described previously (7). The full-length TIP30 cDNA was used as a probe, and labeled with [α32p]dCTP and a random primer DNA labeling system (Invitrogen, Carlsbad, CA).

Growth and Soft Agar Assays of Immortalized MEFs.

Primary Tip30+/+, Tip30+/−, and Tip30−/− MEFs in passage 2 were used to introduce the SV40 T-antigen expression vector by LipofectAMINE reagent (Invitrogen) and be immortalized after 20 passages. For growth curves, immortalized cells (104 per well of 12-well plates) were seeded, and cells were stained with trypan blue and counted daily. For soft agar assays, immortalized MEFs (5 × 103) were grown in DMEM supplemented with 10% fetal bovine serum and 0.53% agarose on top of a layer containing 0.4% agarose for 21 days. The clones were stained with crystal violet, and the clones with diameters >1 mm were counted.

Histology and Immunohistochemistry.

Tissues were fixed with 10% buffered-formalin and embedded in paraffin blocks. Tissue sections were deparaffinized, rehydrated, and stained with H&E. For immunohistochemistry, rehydrated sections were incubated overnight at 4°C with affinity-purified antihuman TIP30 antibody. Staining was developed using VECTASTAIN Elite ABC kit (Vector Laboratories, Burlingame, CA).

Identification of Tip30 Mutations.

Genomic DNA was prepared from paraffin tissue blocks with the EX-WAX DNA extraction kit for paraffin-embedded tissue according to the manufacturer’s instructions (Intergen Co., Purchase, NY). Exon 3 of the Tip30 gene was amplified with Vent polymerase (New England Biolab. Inc., Beverly, MA). The primers used for amplification were 5′-GGCCTCCCAGCCTGCTAC-3′ and 5′-GTACTGCCAAAATAGCTAG-3′. The primers for the nesting PCR were 5′-GCTACCCAAGCCAAGTAGTAATC-3′ and 5′-GCTAGGGTTCTCCAAAGTG-3′. The first reactions were carried out for 25 cycles of 94°C, 30 s; 60°C, 30; and 72°C, 30 s. The nesting PCR reactions were carried out for 32 cycles of 94°C, 30 s; 60°C, 30; and 72°C, 30 s. The PCR products were incubated with Taq polymerase for 10 min at 72°C for adding A to the ends, and purified with Qiagen PCR purification columns (Qiagen, Valencia, CA), then cloned to the TA vector pCR2.1 (Invitrogen). Ten clones for each sample were sequenced for both DNA strands by the Eppley core facility at the University of Nebraska Medical Center using M13 reversal and universal primers. At least three clones containing identical mutations were sequenced for each patient. The HepG2 cell line was purchased from the American Type Culture Collection (Manassas, VA). Experiments in this study have been approved by the Institutional Review Board for the protection of human subjects at the University of Nebraska Medical Center.

Western Blot Analysis and Fluorescence Microscopy.

For Western blot analysis in Fig. 3 B, HepG2 cells were transfected with pCIN4-flag-TIP30 and pCIN4-flag-TIP30G134V using SuperFect reagents (Qiagen). After 48-h transfection, cells were harvested and whole cell extracts were prepared as described previously (3). Whole cell extracts were incubated with anti-Flag M2-agarose (Sigma, St. Louis, MO) at 4°C overnight. After brief centrifugation, supernatants were removed for Western blots with anti-β-actin antibodies. Precipitated proteins were washed and subjected to Western blots with anti-TIP30 antibodies. Fluorescence microscopy was performed as described previously (9). Before being fixed with 4% buffered paraformaldehyde, cells were grown in DMEM supplemented with 10% fetal bovine serum in the presence or absence of 20 nm of Leptomycin B (LMB; Sigma) for 3 h.

Protein Stability Assay.

HepG2 cells were cultured in 150-mm dishes and transfected with pCIN4-flag-TIP30 and pCIN4-flag-TIP30G134V using FuGene 6 reagents (Roche Diagnostics Corp., Indianapolis, IN) according to the manufacturer’s instruction. To inhibit protein synthesis, cycloheximade (100 μg/ml) was added to the medium, and cells were then harvested at the indicated time points. Cell lysates were subjected to immunoprecipitation and Western blot analysis as described above. Alexa Fluor 680 goat antirabbit IgG and goat antimouse IgG (Molecular Probes) were used for quantitative Western blot analysis. Li-Cor system was used to visualize and quantitate TIP30 protein and β-actin protein.

To investigate the roles of TIP30 in both cultured cells and living animals, we generated mice carrying inactivated Tip30 gene by homologous recombination. ES cell clones with a disrupted Tip30 locus were used to obtain germ-line chimeras that, in turn, were used to generate heterozygous F1 mutant 129SvJ/C57BL6/J hybrid mice (Fig. 1,A). Crosses between mice heterozygous for the Tip30 gene led to the expected Mendelian ratios of live born progeny, thus indicating that the Tip30 gene is not essential for development. The loss of TIP30 expression was confirmed by Southern blot analyses of tail genomic DNAs, and by Northern blot analyses of liver and stomach total RNAs (Fig. 1, B and C). Histological examinations of the Tip30−/− mice before the age of 1 year did not reveal obvious gross alterations or developmental abnormalities in organs other than the mammary glands.5 Both Tip30−/− males and females were fertile and generated normal litter sizes.

To determine whether TIP30 is a tumor suppressor, we have kept a cohort of TIP30-deficient and wild-type animals (50% of C57BL6/J and 50% 129SvJ of genetic background) for long-term observation of the development of spontaneous malignancy. At the age of 18–20 months, 31 females and 16 males of the F2 littermates were sacrificed. Autopsy and histological analyses revealed tumor development in 9 of 18 (50%) TIP30-deficient female mice and 2 of 9 TIP30-deficient male mice. In contrast, none of the 13 female and 7 male wild-type mice displayed tumors (Table 1,A; wild-type mice versus mutant mice; P < 0.005; X2 = 11.3). The spectrum of tumor types in Tip30-mutant mice (Table 1 B) is distinct from that seen in older wild-type mice described by others (10, 11, 12), as many of the tumors found in TIP30-deficient mice were carcinomas. Among those tumors, HCC showed the highest incidence, constituting 30% of the total tumors. The relatively high incidence of HCC indicates that TIP30 has a prevalent role in tumor suppression in hepatocytes. Interestingly, 1 Tip30+/− mouse had two tumors, a lipoid cell tumor at the right ovary and a neuroblastoma that arose at the right adrenal gland, with multiple metastases to tissues that included spleen, thymus, and salivary glands. The metastases were also found in the neck, mesenterium, and mediastinum. In addition, 1 mouse developed a transitional cell carcinoma in the urinary bladder, and exhibited metastases along the epithelium that involved the ureters and kidneys, whereas 2 male mice developed sarcomas that metastasized to the liver, spleen, and pancreas. These data demonstrate that TIP30-deficient mice are prone to tumor development.

To reveal the basis for the susceptibility of Tip30-mutant mice to tumor development, we analyzed Tip30 wild-type and mutant MEFs. In an examination of the growth rates of MEFs, no significant difference was detected (data not shown). We then asked if TIP30-deficient MEFs may be more susceptible to transformation by SV40 large T antigen, which is able to inactivate p53 and retinoblastoma protein (reviewed in Ref. 13). These cells were immortalized by ectopic expression of the T antigen, and growth rates were measured. Immortalized Tip30−/− and Tip30+/− MEFs grew much faster than immortalized Tip30+/+ MEFs (Fig. 2,A). Tip30−/− and Tip30+/− MEFs also grew in a more disorganized fashion thanTip30+/+ MEFs (data not shown). Therefore, we assessed the ability of each type of MEF to grow in soft agar. Tip30−/− and Tip30+/− MEFs showed a marked increase in the number and size of colonies as compared with Tip30+/+ MEFs (Fig. 2, B and C). Western blot analysis revealed similar levels of T-antigen expression in Tip30+/+ and Tip30−/− MEFs (Fig. 2 D), and slightly higher expression of T antigen in Tip30+/− MEFs. These results demonstrate that a lack of TIP30 in cells enhances transformation by the SV40 T antigen.

To determine whether our studies in mice are relevant to human disease, we analyzed 24 surgical specimens of human HCC and compared the expression of TIP30 in cancerous cells with expression in adjacent benign hepatocytes on formalin-fixed, paraffin-embedded tissues with antihuman TIP30 antibody (1). Undetectable or significantly decreased TIP30 expression was found in cancerous cells in 8 specimens (33%) in comparison with the adjacent tissues. An example of the immunohistochemical analyses is shown (Fig. 3,A). These data indicate that abnormal expression of TIP30 is implicated in human HCC. We next sought to examine whether these HCC cells harbor mutations in the TIP30 gene. Because the SNP database has listed four single nucleotide polymorphisms (SNPs) in the TIP30 coding regions, we first compared the TIP30 cDNA sequences in National Center for Biotechnology Information databases (14) to identify bp changes other than these SNPs in the human TIP30 coding region. Initially, we examined the sequences of TIP30 cDNA clones to identify insertion, deletion, or bp changes that will result in amino acid substitution. Besides those reported SNPs in the TIP30 gene that were found in both normal and cancer cells, we only identify missense mutations in exon 3. We found that none of the normal cells in the database (0 of 75) harbored a single base-pair substitution in exon 3, and 24% of various types of cancer cells (21 of 52) harbored at least a single base-pair substitution in the same region. Interestingly, the amino acid sequences encoded by Tip30 exon 3 are conserved among human, mouse, and Caenorhabditis elegans TIP30 proteins, and this region is important for the nuclear localization of TIP30.6 Therefore, we focused our mutational study on Tip30 exon 3 for those liver cancer patients who have abnormal TIP30 expression in their cancer cells. Three of 8 cancer samples contained at least a single bp change in the Tip30 gene (Table 1 C). These bp changes were not found in normal cells according to the National Center for Biotechnology Information database. In addition, whereas analyzing the sequences of cloned exon 3 DNA, we found that some of clones (20–80%) from the same cancer sample did not have any bp change, and individual clones from 1 HCC even harbored different bp changes. Importantly, we noted that 2 patients even had the same mutations in their carcinoma cells. Therefore, it is very likely that these base changes are somatic mutations that occurred during cancer development.

To explore the functional consequences of these amino acid substitutions in TIP30, we first investigated whether these mutations affected expression of TIP30 in cells. We transiently transfected a HCC cell line, HepG2, with mammalian expression plasmids encoding wild-type TIP30 and a mutant form of TIP30 with substitutions in G134V (TIP30G134V) identified from the HCC showing in Fig. 3,A. On Western blot analysis showed in Fig. 3,B, TIP30G134V was less expressed compared with the level of the wild-type TIP30. We then investigated whether this mutation affects the stability of the TIP30 protein in HepG2 cells. The relative level of TIP30 was monitored after addition of cycloheximide, a protein synthesis inhibitor. As shown in Fig. 3, C and D, G134V substitution markedly reduces the half-life of TIP30. This result suggests that G134V substitution reduces the stability of TIP30, thereby resulting in a lower level of TIP30 in cells. To investigate whether mutations affect the cellular localization of TIP30, we transfected HepG2 cells with mammalian expression vectors encoding GFP-TIP30 and mutant fusion proteins (GFP-TIP30R106H and GFP-TIP30G134V). The GFP tag had no influence on the cellular distribution of TIP30, because GFP-TIP30 had a subcellular distribution similar to endogenous TIP30 in HeLa and MCF7 cells (data not shown). In cells expressing wild-type GFP-TIP30 (Fig. 3,E, panel 1), ∼90% of transfected cells contained GFP-TIP30 in the cytoplasm, and 10% of transfected cells expressed GFP-TIP30 in the nucleus. When cells were treated with LMB, a CRM1-dependent nuclear export inhibitor (15), nuclear localization of GFP-TIP30 (Fig. 3,E, panel 2) was increased to ∼30% of transfected cells, suggesting that TIP30 is exported to the cytoplasm from the nucleus. However, cells expressing GFP-TIP30R106H showed an exclusively cytoplasmic staining (Fig. 3,E, panel 5), and LMB treatment did not increase the nuclear localization of GFP-TIP30R106H (Fig. 3,E, panel 6). This indicates that the R106H change may abrogate nuclear localization of TIP30 through inhibition of nuclear import. In contrast, GFP-TIP30G134V (Fig. 3,E, panel 3) was expressed in both the cytoplasm and nucleus similar to the expression of GFP alone, suggesting that this mutation may affect its nuclear export. However, LMB treatment did not increase accumulation of GFP-TIP30G134V in the nucleus (Fig. 3 E, panel 4), suggesting that the mutation may affect the nuclear import of TIP30. Therefore, it is possible that the mutation alters the protein structure, and results in abnormal nuclear import and export of TIP30. Given that TIP30 acts as a transcription cofactor in the nucleus and predisposes cells to apoptosis (1, 2, 3, 4, 5), it is conceivable that these mutations may impair the ability of TIP30 to regulate gene expression and apoptosis. Therefore, it is possible that TIP30 deficiency as a result of mutations in the TIP30 gene may inhibit its function in the liver cells and contribute to the pathogenesis of human HCC.

Cellular gene products can function as tumor suppressors or tumor modifiers to regulate tumorigenesis (16, 17). In this study, we showed that TIP30-deficient mice spontaneously develop tumors in their second year of life. Some tumors were carcinomas such as HCC that rarely occurs in 129SvJ/C57BL6/J mice (18). In addition, 1 Tip30 mutant mouse even had two different types of tumor. These data clearly represent a significant enhancement of susceptibility to tumorigenesis in TIP30-deficient mice. The same incidences of tumors in Tip30−/− and Tip30+/− mice is similar to the observations described in previous studies with DMP1, an Arf transcription activator, and p27kip1, a potent tumor suppressor, that are haploinsufficient for tumor suppression (10, 19). We found that TIP30 mRNA and protein were expressed in both primary and metastatic sites of the neuroblastoma and hemangiosarcoma arisen in Tip30+/− mice by reverse transcription-PCR and immunohistochemical analyses (data not shown). Although we are not sure that the second allele in the tumors of Tip30+/− mice encodes a normal TIP30, based on the similar tumor incidence and latency in Tip30+/− and Tip30−/− mice, we suggest that TIP30 is haploinsufficient for tumor suppression.

The role of TIP30 in tumorigenesis is also suggested by the observation of reduced expression of TIP30 in human HCC and Tip30 mutations in those HCC. One mutation results in significantly decreased expression of TIP30, and another mutation changes cellular localization of TIP30. Therefore, it is possible that Tip30 deficiency as a result of mutations in the Tip30 gene may contribute to the pathogenesis of human HCC. Together, these results strengthen a conclusion that TIP30 plays an important role in tumorigenesis.

Grant support: Grants for Beginning Investigators RSG0216501GMC from American Cancer Society, and from the University of Nebraska Medical Center (H. X.), and by the NIH grant (R. G. R.). M. I. was supported by a Human Frontier Science Program (HFSP) long-term fellowship. C. J. is partly supported by a breast cancer research training fellowship DAMD17-00-1-0361 from Department of Defense. X. Z., J. Z., and J. G. were supported in part by Grants from Natural Science Foundation of China, and Shanghai Commission of Science and Technology.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Notes: Drs. Ito and Jiang contributed equally to this work.

Requests for reprints: Hua Xiao, Eppley Institute for Cancer Research, University of Nebraska Medical Center, 987696 Nebraska Medical Center, Omaha, NE 68198-7696. Phone: (402) 559-3323; Fax: (402) 559-3739; E-mail: [email protected]

5

Jill Pecha and Hua Xiao, unpublished observations.

6

C. Jiang and H. Xiao, unpublished observations.

Fig. 1.

Spontaneous development of tumors in TIP30-deficient mice. A, disruption of the Tip30 gene. Wild-type allele of the mouse Tip30 gene (top), the targeting vector with LacZ and phosphoglycerate kinase neo cassettes (middle), and the predicted mutant allele resulting from homologous recombination (bottom) are presented. B, Southern blot analysis of offspring obtained by a heterozygous cross. Genomic DNAs from wild-type (+/+), heterozygous (+/−), and homozygous (−/−) F2 tails, digested with EcoRI or HindIII, were hybridized with the 5′ or 3′ external probe or lacZ. C, Northern blot analysis of Tip30 mRNA in liver and stomach of the indicated genotypes. The signals of two heterozygous mice represent a maternal or paternal allele each. The actin is shown as a control.

Fig. 1.

Spontaneous development of tumors in TIP30-deficient mice. A, disruption of the Tip30 gene. Wild-type allele of the mouse Tip30 gene (top), the targeting vector with LacZ and phosphoglycerate kinase neo cassettes (middle), and the predicted mutant allele resulting from homologous recombination (bottom) are presented. B, Southern blot analysis of offspring obtained by a heterozygous cross. Genomic DNAs from wild-type (+/+), heterozygous (+/−), and homozygous (−/−) F2 tails, digested with EcoRI or HindIII, were hybridized with the 5′ or 3′ external probe or lacZ. C, Northern blot analysis of Tip30 mRNA in liver and stomach of the indicated genotypes. The signals of two heterozygous mice represent a maternal or paternal allele each. The actin is shown as a control.

Close modal
Fig. 2.

Deletion of the TIP30 gene enhances fibroblast transformation by the SV40 large T antigen A, TIP30−/− and TIP30+/− mouse embryotic fibroblasts (MEFs) proliferate faster than wild-type MEFs after immortalization by SV40 T antigen. Immortalized Tip30+/+, Tip30+/−, and Tip30−/− MEFs were counted each day. Each value represents the mean of a representative experiment performed in triplicate. B, T antigen-immortalized Tip30−/− and Tip30+/− MEFs develop more colony foci than TIP30+/+ immortalized MEFs in soft agars. Each value represents the mean of a representative experiment performed in triplicate; bars, ± SD. C, an example of a colony from each MEF genotype is shown. D, Western blot analysis of T-antigen expression in Tip30+/+, Tip30+/−, and Tip30−/− MEFs. Equivalent amounts of whole cell extracts from each of the indicated MEFs were analyzed by immunoblotting with an anti-T-antigen monoclonal antibody and antiactin monoclonal antibody.

Fig. 2.

Deletion of the TIP30 gene enhances fibroblast transformation by the SV40 large T antigen A, TIP30−/− and TIP30+/− mouse embryotic fibroblasts (MEFs) proliferate faster than wild-type MEFs after immortalization by SV40 T antigen. Immortalized Tip30+/+, Tip30+/−, and Tip30−/− MEFs were counted each day. Each value represents the mean of a representative experiment performed in triplicate. B, T antigen-immortalized Tip30−/− and Tip30+/− MEFs develop more colony foci than TIP30+/+ immortalized MEFs in soft agars. Each value represents the mean of a representative experiment performed in triplicate; bars, ± SD. C, an example of a colony from each MEF genotype is shown. D, Western blot analysis of T-antigen expression in Tip30+/+, Tip30+/−, and Tip30−/− MEFs. Equivalent amounts of whole cell extracts from each of the indicated MEFs were analyzed by immunoblotting with an anti-T-antigen monoclonal antibody and antiactin monoclonal antibody.

Close modal
Fig. 3.

Abnormal expression of TIP30 in human hepatocellular carcinoma. A, a representative immunohistochemical stain of human hepatocellular carcinoma with anti-TIP30 serum is presented in the left. It shows much decreased staining of TIP30 in tumor cells (bottom red arrow) compared with the strongly positive staining of TIP30 in the adjacent hepatocytes (top green arrow). A control analysis of a consecutive tissue section with preimmune serum is presented in the right. B, the levels of ectopic expression of wild-type Tip30 and Tip30 mutants in HepG2 cells. HepG2 cells were transiently transfected with plasmids expressing Flag-TIP30 and Flag-TIP30G134V. Equivalent amounts of whole cell extracts made from these cells were subjected to immunoprecipitation with the anti-Flag M2 agarose. The precipitated proteins were then immunoblotted with anti-TIP30 antibodies. The supernatants were immunoblotted with anti-β-actin antibodies. C and D, G134V substitution reduces the half-life of TIP30 protein. HepG2 cells were transfected with plasmids expressing Flag-TIP30 and Flag-TIP30G134V, treated with cycloheximade (CHX) and then harvested at various times. Cell extracts were subjected to immunoprecipitation with the anti-Flag M2 agarose. Precipitated TIP30 proteins and β-actin proteins in the supernatants were detected by Western blot analysis. Quantitation of the results was shown in D. E, cellular localization of GFP-TIP30, GFP-TIP30R106H, and GFP-TIP30G134V in HepG2 cells. Transfected HepG2 cells were treated with or without Leptomycin B (LMB), fixed on slide, and stained with 4′,6-diamidino-2-phenylindole. The representative images for both green fluorescent protein and DNA staining are presented as indicated. Panels 1, 3, and 5 are the untreated cells, and panels 2, 4, and 6 are LMB-treated cells.

Fig. 3.

Abnormal expression of TIP30 in human hepatocellular carcinoma. A, a representative immunohistochemical stain of human hepatocellular carcinoma with anti-TIP30 serum is presented in the left. It shows much decreased staining of TIP30 in tumor cells (bottom red arrow) compared with the strongly positive staining of TIP30 in the adjacent hepatocytes (top green arrow). A control analysis of a consecutive tissue section with preimmune serum is presented in the right. B, the levels of ectopic expression of wild-type Tip30 and Tip30 mutants in HepG2 cells. HepG2 cells were transiently transfected with plasmids expressing Flag-TIP30 and Flag-TIP30G134V. Equivalent amounts of whole cell extracts made from these cells were subjected to immunoprecipitation with the anti-Flag M2 agarose. The precipitated proteins were then immunoblotted with anti-TIP30 antibodies. The supernatants were immunoblotted with anti-β-actin antibodies. C and D, G134V substitution reduces the half-life of TIP30 protein. HepG2 cells were transfected with plasmids expressing Flag-TIP30 and Flag-TIP30G134V, treated with cycloheximade (CHX) and then harvested at various times. Cell extracts were subjected to immunoprecipitation with the anti-Flag M2 agarose. Precipitated TIP30 proteins and β-actin proteins in the supernatants were detected by Western blot analysis. Quantitation of the results was shown in D. E, cellular localization of GFP-TIP30, GFP-TIP30R106H, and GFP-TIP30G134V in HepG2 cells. Transfected HepG2 cells were treated with or without Leptomycin B (LMB), fixed on slide, and stained with 4′,6-diamidino-2-phenylindole. The representative images for both green fluorescent protein and DNA staining are presented as indicated. Panels 1, 3, and 5 are the untreated cells, and panels 2, 4, and 6 are LMB-treated cells.

Close modal
Table 1

TIP30

A. Tumor frequency in TIP30-deficient mice
GenotypeTumor rate
TotalFemaleMale
Tip30              +/+ 0/20 (0%) 0/13 (0%) 0/7 (0%) 
Tip30              +/− or −/− 11/27 (41%) 9/18 (50%) 2/9 (22%) 
Tip30              +/− 7/13 6/8 1/6 
Tip30              −/− 4/13 3/10 1/3 
A. Tumor frequency in TIP30-deficient mice
GenotypeTumor rate
TotalFemaleMale
Tip30              +/+ 0/20 (0%) 0/13 (0%) 0/7 (0%) 
Tip30              +/− or −/− 11/27 (41%) 9/18 (50%) 2/9 (22%) 
Tip30              +/− 7/13 6/8 1/6 
Tip30              −/− 4/13 3/10 1/3 
B. Spectrum of tumors in TIP30-deficient mice
SexGenotypeHistological typeAnatomic site
+/− Hepatocellular carcinoma Liver 
+/− Hepatocellular carcinoma Liver 
−/− Hepatocellular carcinoma Liver 
+/− Thymoma Thymus 
+/− Transitional cell carcinoma, grade III Ureters, bladder, renal pelvis 
+/− Neuroblastoma lipoid cell tumor Adrenal gland ovary 
−/− Adenocarcinoma Duodenum 
−/− Leiomyoma Uterus 
+/− Hemangiosarcoma Retroperitoneum, liver, spleen, subcutis 
−/− Undifferentiated sarcoma Retroperitoneum, liver, spleen, pancreas 
B. Spectrum of tumors in TIP30-deficient mice
SexGenotypeHistological typeAnatomic site
+/− Hepatocellular carcinoma Liver 
+/− Hepatocellular carcinoma Liver 
−/− Hepatocellular carcinoma Liver 
+/− Thymoma Thymus 
+/− Transitional cell carcinoma, grade III Ureters, bladder, renal pelvis 
+/− Neuroblastoma lipoid cell tumor Adrenal gland ovary 
−/− Adenocarcinoma Duodenum 
−/− Leiomyoma Uterus 
+/− Hemangiosarcoma Retroperitoneum, liver, spleen, subcutis 
−/− Undifferentiated sarcoma Retroperitoneum, liver, spleen, pancreas 
C. Amino acid substitutions in TIP30
NameCodon/nucleotideBase changeAmino acid change
HLC 472 134/341 G → T Gly → Val 
HLC 485 109/325 C → T Arg → stop 
 106/316 C → A Arg → Ser 
 115/344 C → A Ser → Tyr 
HLC 583 106/316 C → A Arg → Ser 
 108/324 G → T Asp → Tyr 
 116/346 G → A Ala → Thr 
 144/430 C → A Leu → Ile 
C. Amino acid substitutions in TIP30
NameCodon/nucleotideBase changeAmino acid change
HLC 472 134/341 G → T Gly → Val 
HLC 485 109/325 C → T Arg → stop 
 106/316 C → A Arg → Ser 
 115/344 C → A Ser → Tyr 
HLC 583 106/316 C → A Arg → Ser 
 108/324 G → T Asp → Tyr 
 116/346 G → A Ala → Thr 
 144/430 C → A Leu → Ile 

We thank C. Yang and the Transgenic Facility of the Rockefeller University for help with ES cell manipulation and blastocyst injection, K. Ge for SV40 T-antigen expression vector, P-C. Cui, and K. Wagner for technical help and useful discussions, and C. Eischen and A. Diehl for critical reading of the manuscript.

1
Xiao H., Tao Y., Greenblatt J., Roeder R. G. A cofactor, TIP30, specifically enhances HIV-1 Tat-activated transcription.
Proc. Natl. Acad. Sci. USA
,
95
:
2146
-2151,  
1998
.
2
Shtivelman E. A link between metastasis and resistance to apoptosis of variant small cell lung carcinoma.
Oncogene
,
14
:
2167
-2173,  
1997
.
3
Xiao H., Palhan V., Yang Y., Roeder R. G. TIP30 has an intrinsic kinase activity required for up-regulation of a subset of apoptotic genes.
EMBO J.
,
19
:
956
-963,  
2000
.
4
NicAmhlaoibh R., Shtivelman E. Metastasis suppressor CC3 inhibits angiogenic properties of tumor cells in vitro.
Oncogene
,
20
:
270
-275,  
2001
.
5
Whitman S., Wang X., Shalaby R., Shtivelman E. Alternatively spliced products CC3 and TC3 have opposing effects on apoptosis.
Mol. Cell. Biol.
,
20
:
583
-593,  
2000
.
6
Liu Y., Thor A., Shtivelman E., Cao Y., Tu G., Heath T. D., Debs R. J. Systemic gene delivery expands the repertoire of effective antiangiogenic agents.
J. Biol. Chem.
,
274
:
13338
-13344,  
1999
.
7
Ito M., Yuan C-X., Okano H. J., Darnell R. B., Roeder R. G. Involvement of the TRAP220 component of the TRAP/SMCC coactivator complex in embryonic development and thyroid hormone action.
Mol. Cell
,
5
:
683
-693,  
2000
.
8
Chiang C. M., Roeder R. G. Expression and purification of general transcription factors by FLAG epitope-tagging and peptide elution.
Pept. Res.
,
6
:
62
-64,  
1993
.
9
Alt J. R., Gladden A. B., Diehl J. A. p21 (Cip1) promotes cyclin D1 nuclear accumulation via direct inhibition of nuclear export.
J. Biol. Chem.
,
277
:
8517
-8523,  
2002
.
10
Inoue K., Zindy F., Randle D. H., Rehg J. E., Sherr C. J. Dmp1 is haplo-insufficient for tumor suppression and modifies the frequencies of Arf and p53 mutations in Myc-induced lymphomas.
Genes Dev.
,
15
:
2934
-2939,  
2001
.
11
Steele-Perkins G., Fang W., Yang X. H., Van Gele M., Carling T., Gu J., Buyse I. M., Fletcher J. A., Liu J., Bronson R., Chadwick R. B., de la Chapelle A., Zhang X., Speleman F., Huang S. Tumor formation and inactivation of RIZ1, an Rb-binding member of a nuclear protein-methyltransferase superfamily.
Genes Dev.
,
15
:
2250
-2262,  
2001
.
12
Hakem R., Mak T. W. Animal models of tumor-suppressor genes.
Annu. Rev. Genet.
,
35
:
209
-241,  
2001
.
13
Levine A. J. p53, the cellular gatekeeper for growth and division.
Cell
,
88
:
323
-331,  
1997
.
14
Zhang J., Madden T. L. Power BLAST: a new network BLAST application for interactive or automated sequence analysis and annotation.
Genome Res.
,
7
:
649
-656,  
1997
.
15
Harbers M., Nomura T., Ohno S., Ishii S. Intracellular localization of the ret finger protein depends on a functional nuclear export signal and protein kinase C activation.
J. Biol. Chem.
,
276
:
48596
-48607,  
2001
.
16
Balmain A. Cancer as a complex genetic trait: tumor susceptibility in humans and mouse models.
Cell
,
108
:
145
-152,  
2002
.
17
Van Dyke T., Jacks T. Cancer modeling in the modern era: progress and challenges.
Cell
,
108
:
135
-144,  
2002
.
18
Ward J. M., Mahler J. F., Maronpot R. R., Sundberg J. P., Frederickson R. M. Pathology of mice commonly used in genetic engineering (C57BL/6; 129; B6, 129; and FVB/N) Ward J. M. Mahler J. F. Maronpot R. R. Sundberg J. P. Frederickson R. M. eds. .
Pathology of Genetically Engineered Mice
, First Edition
161
-179, Iowa State University Press Ames  
2000
.
19
Philipp-Staheli J., Payne S. R., Kemp C. J. p27(Kip1): regulation and function of a haploinsufficient tumor suppressor and its misregulation in cancer.
Exp. Cell Res.
,
264
:
148
-168,  
2001
.