To identify potential oncogenes that contribute to the development of uterine leiomyosarcoma, we conducted a cDNA microarray analysis between normal uterine smooth muscle and uterine leiomyosarcoma. We found that acrogranin (also named PCDGF or progranulin) is overexpressed in uterine leiomyosarcoma. With immunohistochemical staining of 12 leiomyosarcoma cases, we verified acrogranin expression in tumor cells. Furthermore, the intensity of acrogranin expression correlated with high histologic grade and poor prognosis. To directly analyze the oncogenic properties of acrogranin, we established an immortalized uterine smooth muscle cell line by transfection of human telomerase reverse transcriptase into primary culture. This cell line retained the original characteristics of uterine smooth muscle cells, including spindle-shaped extension as well as expression of vimentin, estrogen receptor α, progesterone receptor, and α smooth muscle actin. Transfection of acrogranin into the immortalized uterine smooth muscle cells resulted in colony formation in soft agar, but the diameter of the colonies did not exceed 100 μm. Transfection of both acrogranin and SV40 early region (SV40ER) into the immortalized uterine smooth muscle cells resulted in an increased number of colonies and increased colony size in soft agar versus transfection of SV40ER alone. We show that only immortalized uterine smooth muscle cells expressing both acrogranin and SV40ER are capable of tumor formation in nude mice. Thus, acrogranin is overexpressed in uterine leiomyosarcoma cells, particularly in high-grade cases, and forced expression of acrogranin in immortalized uterine smooth muscle cells contributes to malignant transformation, which suggest that acrogranin plays an important role in the pathogenesis of uterine leiomyosarcoma.

Leiomyosarcoma comprises only 1% of all gynecologic malignancies and has an extremely poor prognosis (1). Although the majority of leiomyosarcomas are diagnosed at an early and resectable stage, 40% of these cases recur after therapy. Eighty-one percent of women with stage III tumors will have tumor recurrence, and only 8% of women with stages II to IV tumors will survive 5 years (2, 3). As uterine leiomyosarcoma is resistant to chemotherapy and radiotherapy (4, 5), developing an efficient adjuvant therapy is expected to improve the prognosis of the disease.

Because a significant effect was achieved for breast cancer using antibody therapy directed against HER2, molecular targeting is regarded as a promising strategy for treatment of malignant tumors (6). In this regard, it is becoming increasingly important to identify targets that play an important role in the pathogenesis of specific tumors.

Our objective was to identify molecules involved in the pathogenesis of leiomyosarcoma that might serve as therapeutic targets. For this purpose, we conducted a cDNA microarray analysis and from these results we focused on acrogranin. Acrogranin (Genbank accession no. AF055008; National Cancer Institute of Canada abbreviation, GRN), also called PCDGF, progranulin, or proepithelin, is an 88 kDa glycoprotein that was identified as a growth factor secreted from prostate cancer cell, a human teratoma cell (7, 8). Physiologically, acrogranin is known to play important roles in development and wound repair (9, 10). It has also been recently reported that acrogranin is overexpressed in various malignant tumors, including gliomas; breast, ovarian, and prostatic cancers; and hepatocellular carcinomas (1116). In these tumors, the expression level of acrogranin was shown to be a prognostic factor. Experimentally, overexpression of acrogranin in SW-13 adrenal carcinoma cells and Madin-Darby canine kidney cells nontransformed renal epithelia results in the transfection-specific secretion of acrogranin, acquisition of clonogenicity in semisolid agar, and increased mitosis in monolayer culture (17). Acrogranin overproduction in SW-13 cells also increased tumorigenicity in nude mice (17). Another study showed diminution of acrogranin gene expression-impaired tumorigenicity of the breast cancer cell line MDA-MB-468 in immunodeficient mice (18). Thus far, it is unknown whether overexpression of acrogranin can transform human primary cells.

Development of malignant tumor is a complicated process involving multiple genetic and epigenetic alterations. Analyzing the process of malignant transformation that is induced experimentally in cell culture is undoubtedly a powerful approach for uncovering the basic biological and biochemical principles underlying development of malignant tumor. Because the first report of in vitro malignant transformation of primary human cells by transfection of defined genetic elements (19), several groups, including us, reported in vitro generation of human cancer cells in various organs (2025). However, there has been no report about in vitro malignant transformation of uterine smooth muscle cells.

Herein, we show that acrogranin is overexpressed in uterine leiomyosarcoma compared with uterine smooth muscles and leiomyomas and its expression is correlated with prognosis of uterine leiomyosarcoma. We also show malignant transformation of uterine smooth muscle cells by transfection of human telomerase reverse transcriptase (hTERT), acrogranin, and SV40 early region (SV40ER). These results suggest that acrogranin may play an important role in the pathogenesis of uterine leiomyosarcoma and further indicate that acrogranin may be a useful target of molecular therapy.

cDNA microarray analysis. cDNA microarray analysis was conducted using tissues extracted from two cases of uterine leiomyosarcoma and three cases of uterine smooth muscle as described previously (26). Briefly, from histologically diagnosed tissues, polyadenylate+ RNA was extracted from tissues using an mRNA purification kit (Amersham Biosciences Corp., Piscataway, NJ). To analyze the average expression values from different cases in one assay, the same amount of RNA extracted from leiomyosarcoma cases and uterine smooth muscle cases, respectively, was mixed individually and served for further analysis. Then, RNA was labeled with Cy3-dCTP (leiomyosarcoma RNA) and Cy5-dCTP (uterine smooth muscle RNA). Labeled probes were mixed with microarray hybridization solution version 2 (Amersham Biosciences) and formamide to yield a final concentration of 50%. Each sample was hybridized onto the UniGEM V cDNA microarray (Incyte Genomics, Inc., Saint Louis, MO). The UniGEM V array contains 7,800 unique and sequence-verified cDNA or expressed sequence tag elements. The Cy3/Cy5 signal ratio (leiomyosarcoma/uterine smooth muscle) was calculated and analyzed by GEM tools 2.4 (Incyte Genomics, Palo Alto, CA).

Construction of retroviral vectors and production of retroviruses. The plasmid containing hTERT cDNA (pcDNA3-hTERTn2; ref. 20) was cut to produce the hTERT cDNA fragment and this was inserted into the retroviral vector pPGS-CITEneo. The plasmid containing acrogranin cDNA (pCMV-SPORT6-acrogranin) was obtained from Invitrogen Corporation (Carlsbad, CA: clone ID 3457813). We converted pLHCX retroviral vector (Clontech, Franklin Lakes, NJ) to the Gateway destination vector by using the Gateway Vector Converting System (Invitrogen). Then pLHCX-acrogranin plasmid was constructed using Gateway technology. The retroviral vector containing the SV40ER (pWB-SV40ER) was kindly provided by Dr. Jean J. Zhao (Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; ref. 22). The final construct was transduced by lipofection into the packaging cell line, Amphopack 293 (Clontech). Forty-eight hours later, the supernatants containing amphotropic viruses were collected. Virus was used to infect uterine smooth muscle cells with 5 μg/mL 1,5-dimethyl-1,5-diazaundecamethylene polymethobromide (Nacalai Tesque; Kyoto, Japan).

Generation of uterine smooth muscle cell lines. Uterine smooth muscle tissue was obtained from a 48-year-old patient with regular menstrual cycles who underwent surgery for uterine leiomyomas at Kyoto University Hospital. Informed consent was obtained from the patient before surgery. Uterine smooth muscle tissue was minced and digested in DMEM (Nikken Biomedicals, Kyoto, Japan) containing 0.02% collagenase (Wako Pure Chemical Industries Ltd., Osaka, Japan) for 4 hours at 37°C with agitation. The dispersed cells were plated in DMEM containing penicillin-streptomycin (2%, v/v; Nacalai Tesque) and fetal bovine serum (10%, v/v, Asahi Technoglass, Funabashi, Japan) and maintained in a 37°C incubator ventilated with 5% CO2 in-room air. The following day, uterine smooth muscle cells were infected for 24 hours with the amphotropic hTERT retroviral vectors followed by supplementation 48 hours later with G418 (500 μg/mL). G418 was used for 6 weeks to eliminate all noninfected cells and select for colony formation of the hTERT-infected cells. Cell lines were generated by clonal selection and were maintained in DMEM with 10% fetal bovine serum and antibiotic. Among multiple colonies generated, one was isolated and maintained. It was defined as “population = 1” when cells become confluent in a 100 mm culture dish. When confluent, the cells were trypsinized and seeded at a 1:4 split ratio. The pLHCX retroviral vector containing acrogranin cDNA or the pLHCX retroviral vector only were transfected into the immortalized uterine smooth muscle cell line in the same way and selected by hygromycin B (200 μg/mL; Invitrogen). The pWB retroviral vector containing the SV40ER was similarly transfected into these cells and transfectants were selected and maintained using blastcidine (2 μg/mL; InvivoGen, San Diego, CA).

Immunostaining of cultured cells. The hTERT-transfected uterine smooth muscle cells were cultured for 3 days on multichambered culture slides (BD Biosciences, San Jose, CA) in a 37°C incubator ventilated with 5% CO2 in room air. Cells were fixed and permeabilized by immersion in −20°C methanol for 15 minutes and −20°C acetone for 1 minute. Blocking was done with normal rabbit serum (Nichirei, Tokyo, Japan). Samples were incubated with primary antibodies at 4°C overnight and washed. The cells were then incubated for 30 minutes with a 1:40 dilution of FITC-conjugated rabbit anti-mouse IgG secondary antibody (DakoCytomation, Glostrup, Denmark) and washed. Propidium iodide (Nacalai Tesque) was used for nuclear staining. Samples were soaked with glycerol (DakoCytomation) and coverslipped. Photographs were taken with a Zeiss microscope (Carl Zeiss MicroImaging, Inc., Thornwood, NY) equipped for epifluorescence. Primary antibodies used to stain cultured cells were all anti-human mouse monoclonal antibodies: anti–estrogen receptor α (ERα; Nichirei), anti–progesterone receptor (PR; Nichirei), anti–α smooth muscle actin (α-SMA; DakoCytomation), anticytokeratin (Biomeda, Foster City, CA), and antivimentin (Santa Cruz Biotechnology, Santa Cruz, CA).

Immunostaining of tissue samples. Tissues were fixed in 10% buffered formalin, embedded in paraffin, and sectioned. Sections were stained with H&E. For the immunohistochemical staining, they were deparaffinized in xylene and ethanol. Sections were blocked for endogenous peroxidase with 0.3% H2O2 followed by sequential incubation with normal goat serum, primary antibody, secondary antibody, and peroxidase-conjugated streptavidin. Slides were then stained with 3,3′-diaminobenzidine and hematoxylin. We used the following antibodies as primary antibodies: a goat polyclonal antiacrogranin antibody (Santa Cruz Biotechnology), a rabbit polyclonal antivimentin antibody (Santa Cruz Biotechnology), a mouse monoclonal anti-SV40 large T-antigen antibody (Santa Cruz Biotechnology), and a mouse monoclonal anticytokeratin antibody (Biomeda). For acrogranin and vimentin staining, samples were retrieved with Target Retrieval Solution high pH (DakoCytomation) for 15 minutes at 95°C. Then, SAB-PO kit (Nichirei), which contains blocking serum, secondary antibodies, and peroxidase-conjugated streptavidin, was used. For SV40 large T-antigen and cytokeratin staining, samples were soaked in citrate buffer and retrieved in a microwave for 15 minutes. To stain xenograft tumor tissues formed in nude mice with murine monoclonal antibodies, Mouse Stain kit was used according to the protocol of the manufacturer (Nichirei).

Evaluation of acrogranin expression in human tissue samples. The level of acrogranin protein expression was evaluated by immunohistochemical study and defined as follows: +, stronger than smooth muscle but weaker than endometrial glands; ++, equivalent to endometrial glands; +++, stronger than endometrial glands. Immunostaining of all human tissue samples was done simultaneously and under the same conditions. Three persons (gynecologic pathologists) examined each slide independently in a blind manner with final assignment of expression levels determined by consensus.

Reverse transcription-PCR. Total RNA was extracted from cultured uterine smooth muscle cell lines using Trizol reagent (Invitrogen). Reverse transcription-PCR (RT-PCR) was done with the One-Step RT-PCR kit (Qiagen, Hilden, Germany) according to the instructions of the manufacturer. ERα, PR, hTERT, acrogranin, and actin cDNAs were amplified using specific primers for each transcript. PCR amplification was done as follows: denaturation at 94°C for 1 minute, annealing at primer-specific temperatures (ERα, PR, 52°C; acrogranin, hTERT, actin, 55°C) for 1 minute, and extension at 72°C for 2 minutes. The constitutively expressed actin transcript was used as a reference to evaluate transcription of ER, hTERT, and PR. For evaluation of acrogranin mRNA expression, actin was used as an internal control by multiplexing the acrogranin- and actin-specific primers in each reaction. PCRs for ERα and PR were terminated at 30 cycles, PCR for hTERT at 28 cycles, and PCR for acrogranin at 26 cycles. PCR for actin was terminated at 25 cycles except for where actin was used as an internal control. Primers used were as follows: ERα, forward 5′-ACAAGCGCCAGAGAGATGAT-3′, reverse 5′-CAGATTCATCATGCGGAACC-3′; PR, forward 5′-GTTGCTCTCCCACAGCCATT-3′, reverse 5′-TACAGCATCTGCCCACTGAC-3′; hTERT, forward 5′-CGGAAGAGTGTCTGGAGCAA-3′, reverse 5′-GGATGAAGCGGAGTCTGGA-3′, acrogranin, forward 5′-TGGTTCACACCCGCTGCA-3′, reverse 5′-GTTGGGCATTGGGCAGCA-3′, actin: forward 5′-CCGCAAAGACCTGTACGCCA-3′, reverse 5′-TGGACTTGGGAGAGGACTGG-3′.

Assay for anchorage independence. A 2-fold concentration of DMEM supplemented with fetal bovine serum and antibiotics was made from powdered medium (Life Technologies), and 1% agarose was made with low-melting-temperature agarose (Invitrogen). Then, 0.5% agarose and 0.33% agarose in 1× DMEM was made by their mixture. For uterine smooth muscle cells transfected with hTERT and acrogranin or empty vector, colony formation in semisolid agar was assayed by suspending 4 × 104 cells in 800 μL of 0.33% agarose and placing this suspension on the top of 800 μL solidified 0.5% agarose. Cultures were maintained at 37°C in a 5% CO2 atmosphere. Colonies larger than 50 μm in diameter were counted 3 weeks later. For cells further transfected with SV40ER, colonies larger than 100 μm were counted 10 days later. The experiments were repeated four times individually.

Tumorigenicity assays. Six-week-old female BALB/C nude mice were purchased from CLEA Japan (Tokyo, Japan). The animals received proper care according to the rules of the Kyoto University Committee on Animal Care. Animal experiments were done in compliance with the United Kingdom Coordinating Committee on Cancer Research Guidelines. In nude mice, 1 × 107 cells were injected s.c. per animal. If a s.c. mass was confirmed, the mice were sacrificed and the tumors were resected. Cells were considered nontumorigenic if the mouse failed to form a tumor within 4 months of injection.

Statistical analysis. For analysis of between-group differences, P values were determined using Mann-Whitney U test. P < 0.05 was considered significant.

cDNA microarray analysis.Table 1 lists highly expressed genes in leiomyosarcoma from a cDNA microarray analysis of 7,800 genes between leiomyosarcoma and uterine smooth muscle. Acrogranin was the focus of the present study because of (a) the high ratio of acrogranin expression in leiomyosarcoma compared with smooth muscle, (b) a previous report concerning pathogenesis of cancers (17, 18), and (c) the fact that acrogranin may be a useful molecular tumor marker and therapeutic target in leiomyosarcoma, for which sensitive tumor markers and effective cytotoxic agents do not currently exist.

Table 1.

cDNA microarray analysis showing stronger acrogranin expression in uterine leiomyosarcoma than in smooth muscle

Gene nameSymbolGenbank accession no.Ratio
Chitinase 3-like 1 (cartilage glycoprotein-39) CHI3L1 Y08374 84.35 
Neuromedin B GPNMB X76534 55.36 
Cathepsin K (pycnodysostosis) CTSK X82153 37.7 
Hexabrachion (tenascin C, cytotactin) TNC X78565 15.87 
Brain-specific polypeptide PEP-19 PCP4 U52969 13.42 
Regulator of G-protein signaling 5 RGS5 AB008109 11.16 
Cadherin 3, P-cadherin (placental) CDH3 X63629 7.6 
Cerebellin 1 precursor CBLN1 M58583 7.41 
Thrombospondin 2 THBS2 L12350 7.13 
Ferritin, light polypeptide FTL M11147 6.9 
Human scaffold protein Pbp1 SYCL AF000652 6.29 
Cathepsin B CTSB L04288 6.24 
Human mRNA for cysteine protease LGMN D55696 5.7 
Protein regulating cytokinesis 1 PRC1 AF044588 5.5 
DNA-damage-inducible transcript 1 GADD45A L24498 5.46 
Homo sapiens exportin t XPO1 AF039022 5.46 
Transforming growth factor, β3 TGFB3 X14885 5.06 
Integrin, α8 ITGA8 L36531 4.96 
Human mRNA for pM5 protein NOMO X57398 4.84 
Tropomodulin TMOD1 M77016 4.64 
Microfibrillar-associated protein 4 MFAP4 L38486 4.62 
Acrogranin GRN AF055008 4.56 
Uridine phosphorylase UPP1 X90858 4.55 
Superoxide dismutase 2, mitochondrial SOD2 X07834 4.5 
Myosin VA (heavy polypeptide 12, myoxin) MYO5A Y07759 4.48 
TYRO protein tyrosine kinase binding protein TYROBP AF019562 4.25 
Follistatin FST M19481 4.2 
Clathrin-associated/assembly/adaptor protein, large β1 AP2B1 M34175 4.06 
Ferritin, heavy polypeptide 1 FTH1 L20941 4.05 
Ribonucleotide reductase M1 polypeptide RRM1 X59543 4.03 
Transketolase (Wernicke-Korsakoff syndrome) TKT L12711 3.96 
CD24 CD24 L33930 3.96 
Collagen, type VI, α3 COL6A3 X52022 3.96 
Protease, serine, 11 (IGF binding) PRSS11 D87258 3.92 
5-Hydroxytryptamine (serotonin) receptor 2B HTR2B X77307 3.92 
Ubiquitin-specific protease 9 USP9X X98296 3.9 
Chemokine (C-C motif) receptor 1 CCR1 D10925 3.9 
Phosphoglucomutase 1 PGM1 M83088 3.85 
Tubulin, γ polypeptide TUBG1 M61764 3.78 
Serine/threonine kinase 2 STK2 L20321 3.78 
Hect domain and RLD 2 HERC2 AB002391 3.75 
3-Phosphoglycerate dehydrogenase PHGDH AF006043 3.74 
BH-protocadherin (brain-heart) PCDH7 AB006757 3.64 
APK1 antigen COVA1 S72904 3.61 
Matrix metalloproteinase 2 MMP2 M55593 3.52 
Ubiquitin-specific protease 9 USP9Y AF000986 3.5 
Integrin, α5 ITGA5 X06256 3.45 
Roundabout homologue 1 ROBO1 AF040990 3.38 
p21 (CDKN1A)-activated kinase 2 PAK2 U24153 3.36 
Chloride channel 4 CLCN4 X77197 3.36 
Ribosomal protein S11 IDH2 X69433 3.3 
Inositol phosphate 5′-phosphatase 2 (synaptojanin 2) SYNJ2 AB002346 3.3 
ATP citrate lyase ACLY X64330 3.25 
IFN-γ-inducible protein 16 IFI16 M63838 3.23 
Insulin-like growth factor binding protein 3 precursor IGFBP3 M35878 3.2 
Enolase 1 (α) ENO1 M55914 3.19 
Peptidyl-prolyl cis-trans isomerase A PPIA X52851 3.15 
Equilibrative nucleoside transporter 1 SLC29A1 U81375 3.12 
Protein S (α) PROS1 X12892 3.1 
Human leukemia virus receptor 1 (GLVR1) SLC20A1 L20859 3.08 
Apolipoprotein C-I APOC1 X00570 3.08 
Calpastatin CAST D16217 3.08 
Ubiquitin-specific protease 11 USP11 U44839 3.08 
Human arginine-rich protein (ARP) ARMET M83751 3.06 
TrkB NTRK2 U12140 
T-complex protein 1, α subunit TCP1 X52882 
Thyroid hormone receptor, β THRB X04707 
Gene nameSymbolGenbank accession no.Ratio
Chitinase 3-like 1 (cartilage glycoprotein-39) CHI3L1 Y08374 84.35 
Neuromedin B GPNMB X76534 55.36 
Cathepsin K (pycnodysostosis) CTSK X82153 37.7 
Hexabrachion (tenascin C, cytotactin) TNC X78565 15.87 
Brain-specific polypeptide PEP-19 PCP4 U52969 13.42 
Regulator of G-protein signaling 5 RGS5 AB008109 11.16 
Cadherin 3, P-cadherin (placental) CDH3 X63629 7.6 
Cerebellin 1 precursor CBLN1 M58583 7.41 
Thrombospondin 2 THBS2 L12350 7.13 
Ferritin, light polypeptide FTL M11147 6.9 
Human scaffold protein Pbp1 SYCL AF000652 6.29 
Cathepsin B CTSB L04288 6.24 
Human mRNA for cysteine protease LGMN D55696 5.7 
Protein regulating cytokinesis 1 PRC1 AF044588 5.5 
DNA-damage-inducible transcript 1 GADD45A L24498 5.46 
Homo sapiens exportin t XPO1 AF039022 5.46 
Transforming growth factor, β3 TGFB3 X14885 5.06 
Integrin, α8 ITGA8 L36531 4.96 
Human mRNA for pM5 protein NOMO X57398 4.84 
Tropomodulin TMOD1 M77016 4.64 
Microfibrillar-associated protein 4 MFAP4 L38486 4.62 
Acrogranin GRN AF055008 4.56 
Uridine phosphorylase UPP1 X90858 4.55 
Superoxide dismutase 2, mitochondrial SOD2 X07834 4.5 
Myosin VA (heavy polypeptide 12, myoxin) MYO5A Y07759 4.48 
TYRO protein tyrosine kinase binding protein TYROBP AF019562 4.25 
Follistatin FST M19481 4.2 
Clathrin-associated/assembly/adaptor protein, large β1 AP2B1 M34175 4.06 
Ferritin, heavy polypeptide 1 FTH1 L20941 4.05 
Ribonucleotide reductase M1 polypeptide RRM1 X59543 4.03 
Transketolase (Wernicke-Korsakoff syndrome) TKT L12711 3.96 
CD24 CD24 L33930 3.96 
Collagen, type VI, α3 COL6A3 X52022 3.96 
Protease, serine, 11 (IGF binding) PRSS11 D87258 3.92 
5-Hydroxytryptamine (serotonin) receptor 2B HTR2B X77307 3.92 
Ubiquitin-specific protease 9 USP9X X98296 3.9 
Chemokine (C-C motif) receptor 1 CCR1 D10925 3.9 
Phosphoglucomutase 1 PGM1 M83088 3.85 
Tubulin, γ polypeptide TUBG1 M61764 3.78 
Serine/threonine kinase 2 STK2 L20321 3.78 
Hect domain and RLD 2 HERC2 AB002391 3.75 
3-Phosphoglycerate dehydrogenase PHGDH AF006043 3.74 
BH-protocadherin (brain-heart) PCDH7 AB006757 3.64 
APK1 antigen COVA1 S72904 3.61 
Matrix metalloproteinase 2 MMP2 M55593 3.52 
Ubiquitin-specific protease 9 USP9Y AF000986 3.5 
Integrin, α5 ITGA5 X06256 3.45 
Roundabout homologue 1 ROBO1 AF040990 3.38 
p21 (CDKN1A)-activated kinase 2 PAK2 U24153 3.36 
Chloride channel 4 CLCN4 X77197 3.36 
Ribosomal protein S11 IDH2 X69433 3.3 
Inositol phosphate 5′-phosphatase 2 (synaptojanin 2) SYNJ2 AB002346 3.3 
ATP citrate lyase ACLY X64330 3.25 
IFN-γ-inducible protein 16 IFI16 M63838 3.23 
Insulin-like growth factor binding protein 3 precursor IGFBP3 M35878 3.2 
Enolase 1 (α) ENO1 M55914 3.19 
Peptidyl-prolyl cis-trans isomerase A PPIA X52851 3.15 
Equilibrative nucleoside transporter 1 SLC29A1 U81375 3.12 
Protein S (α) PROS1 X12892 3.1 
Human leukemia virus receptor 1 (GLVR1) SLC20A1 L20859 3.08 
Apolipoprotein C-I APOC1 X00570 3.08 
Calpastatin CAST D16217 3.08 
Ubiquitin-specific protease 11 USP11 U44839 3.08 
Human arginine-rich protein (ARP) ARMET M83751 3.06 
TrkB NTRK2 U12140 
T-complex protein 1, α subunit TCP1 X52882 
Thyroid hormone receptor, β THRB X04707 

NOTE: cDNA microarray analysis was conducted between leiomyosarcoma and uterine smooth muscle tissues. The table lists genes for which expression ratios of leiomyosarcoma/uterine smooth muscle are >3. Expressed sequence tags are excluded.

Immunohistochemical study for acrogranin expression. An immunohistochemical study was done among uterine leiomyosarcoma, leiomyoma, and smooth muscle tissues using an antiacrogranin antibody. Acrogranin protein expression was observed in uterine leiomyosarcoma, but expression was weak or undetectable in smooth muscles and leiomyomas (Fig. 1A-D). Strong expression was observed in endometrial glands, which were used as an internal control. Uterine leiomyosarcoma was diagnosed using standard criteria (27). Furthermore, histologic grading was done based on previously established criteria (28). Among leiomyosarcoma cases, the histologically high-grade group showed significantly stronger expression of acrogranin than the low-grade group and myxoid type (P < 0.05, χ2 test; Fig. 1E-G; Table 2). Specifically, strong expression was observed in five of six high-grade cases (case 11 is of high grade but with low acroganin expression). In contrast, strong expression was observed in only one (case 4) of six low-grade/myxoid cases. Prognoses of high-grade cases were extremely poor (Table 2).

Fig. 1.

Immunohistochemical study on acrogranin protein expression in uterine smooth muscle, leiomyoma, and various-grade leiomyosarcoma. An immunohistochemical study was done among uterine leiomyosarcoma, leiomyoma, and smooth muscle tissues using an antiacrogranin antibody. A, leiomyosarcoma. B, leiomyosarcoma (high magnification). C, smooth muscle (arrow, endometrial gland). D, leiomyoma. Strong expression was observed in endometrial glands, which was used as an internal control. Among leiomyosarcoma cases, the histologically high-grade group showed stronger expression of acrogranin than the low-grade group and myxoid type. E, high grade. F, low grade. G, myxoid. Bar, 50 μm.

Fig. 1.

Immunohistochemical study on acrogranin protein expression in uterine smooth muscle, leiomyoma, and various-grade leiomyosarcoma. An immunohistochemical study was done among uterine leiomyosarcoma, leiomyoma, and smooth muscle tissues using an antiacrogranin antibody. A, leiomyosarcoma. B, leiomyosarcoma (high magnification). C, smooth muscle (arrow, endometrial gland). D, leiomyoma. Strong expression was observed in endometrial glands, which was used as an internal control. Among leiomyosarcoma cases, the histologically high-grade group showed stronger expression of acrogranin than the low-grade group and myxoid type. E, high grade. F, low grade. G, myxoid. Bar, 50 μm.

Close modal
Table 2.

Summary of correlation among histologic grading, expression of acrogranin, and prognosis in uterine leiomyosarcoma cases

Case no.Age (y)Size (cm)OperationAdjuvant therapyClinical stageGradeFollow-upAcrogranin expression
51 12 TAH ND Low grade NED 60+ mo 
49 TAH + BSO CAP-F (CDDP-ADR-CPM-5FU) ×2 Low grade NED 60+ mo 
49 16 TAH + BSO ND Low grade NED 60+ mo 
39 15 TAH + BSO ND Low grade NED 60+ mo ++ 
65 10 TAH + BSO ND Myxoid NED 51 mo 
22 20 TAH + BSO CDDP-THP-IFO ×3 Myxoid NED 50 mo 
52 12 TAH + LSO ND High grade NED 60+ mo ++ 
52 8.5 TAH + BSO CDDP-ADR ×2, VP-16 ×2 High grade DOD 8 mo ++ 
76 13 TAH + BSO ND II High grade DOD 14 mo +++ 
10 44 TAH + BSO CDDP-THP-IFO ×3 IV High grade DOD 24 mo ++ 
11 47 Tumor resection + TAH + BSO CDDP-ADR-VCR ×2, CAP-F (CDDP-ADR-CPM-5FU) ×2 III High grade DOD 24 mo 
12 60 Tumor resection+ TAH + BSO CAP-F (CDDP-ADR-CPM-5FU) ×2 III High grade DOD 60 mo +++ 
Case no.Age (y)Size (cm)OperationAdjuvant therapyClinical stageGradeFollow-upAcrogranin expression
51 12 TAH ND Low grade NED 60+ mo 
49 TAH + BSO CAP-F (CDDP-ADR-CPM-5FU) ×2 Low grade NED 60+ mo 
49 16 TAH + BSO ND Low grade NED 60+ mo 
39 15 TAH + BSO ND Low grade NED 60+ mo ++ 
65 10 TAH + BSO ND Myxoid NED 51 mo 
22 20 TAH + BSO CDDP-THP-IFO ×3 Myxoid NED 50 mo 
52 12 TAH + LSO ND High grade NED 60+ mo ++ 
52 8.5 TAH + BSO CDDP-ADR ×2, VP-16 ×2 High grade DOD 8 mo ++ 
76 13 TAH + BSO ND II High grade DOD 14 mo +++ 
10 44 TAH + BSO CDDP-THP-IFO ×3 IV High grade DOD 24 mo ++ 
11 47 Tumor resection + TAH + BSO CDDP-ADR-VCR ×2, CAP-F (CDDP-ADR-CPM-5FU) ×2 III High grade DOD 24 mo 
12 60 Tumor resection+ TAH + BSO CAP-F (CDDP-ADR-CPM-5FU) ×2 III High grade DOD 60 mo +++ 

NOTE: The strength of acrogranin expression was evaluated as described in Materials and Methods. Twelve cases of uterine leiomyosarcoma were treated in Kyoto University Hospital during the last 15 years.

Abbreviations: TAH, total abdominal hysterectomy; BSO, bilateral salpingo-oophorectomy; LSO, left salpingo-oophorectomy; ND, not done; CDDP, Cisplatin; ADR, Adriamycin; CPM, Cyclophosphamide; 5FU, 5-Fluorouracil; THP, Therarubicin; IFO, Ifosfamide; VP-16, Etoposide; VCR, Vincristin; NED, no evidence of disease; DOD, died of disease.

Generation of a uterine smooth muscle cell line. To study the effect of acrogranin overexpression in uterine smooth muscle cells, we generated a uterine smooth muscle cell line by hTERT gene transfection into primary culture cells (Fig. 2). It was previously reported by several groups that uterine smooth muscle cells could be immortalized by hTERT gene transfection (2931). The cell line we generated maintained characteristics of uterine smooth muscle cells (Fig. 2B and C). RT-PCR analysis was done to confirm the expression of ERα, PR, and forced expression of hTERT (data not shown). This cell line did not show acrogranin gene expression as a primary culture (Fig. 3A). On the contrary, SKN (uterine leiomyosarcoma cell line) showed acrogranin gene expression (Fig. 3A).

Fig. 2.

Generation of a uterine smooth muscle cell line retaining the phenotype of uterine smooth muscle cells by hTERT gene transfection. Uterine smooth muscle cells were transfected with hTERT cDNA and selected with G418. A colony was isolated and subcultured. A, proliferation of the hTERT gene–transfected uterine smooth muscle cells continued over 300 days. One hundred population doublings of these cells were counted and, therefore, this cell line is immortalized. B, these cells showed spindle-shaped extension under phase-contrast microscopy. C, immunostaining of immortalized smooth muscle cells observed in confocal microscopy [top left: propidium iodide (PI) staining] showed expression of vimentin, α-SMA, ERα, and PR, but did not show expression of cytokeratin.

Fig. 2.

Generation of a uterine smooth muscle cell line retaining the phenotype of uterine smooth muscle cells by hTERT gene transfection. Uterine smooth muscle cells were transfected with hTERT cDNA and selected with G418. A colony was isolated and subcultured. A, proliferation of the hTERT gene–transfected uterine smooth muscle cells continued over 300 days. One hundred population doublings of these cells were counted and, therefore, this cell line is immortalized. B, these cells showed spindle-shaped extension under phase-contrast microscopy. C, immunostaining of immortalized smooth muscle cells observed in confocal microscopy [top left: propidium iodide (PI) staining] showed expression of vimentin, α-SMA, ERα, and PR, but did not show expression of cytokeratin.

Close modal
Fig. 3.

Effect of transfection of with acrogranin and SV40ER genes on anchorage-independent growth of the immortalized uterine smooth muscle cells. A, acrogranin mRNA expression in SKN, uterine leiomyosarcoma cells, and in the immortalized uterine smooth muscle cells. I-SM, immortalized uterine smooth muscle cell line by hTERT transfection. Acrogranin mRNA expression was analyzed by RT-PCR (26 cycles). As an internal control, actin primers were also included in the reactions. SKN (uterine leiomyosarcoma cell line) showed acrogranin gene expression. Acrogranin was not detected in the immortalized uterine smooth muscle cell line and in the uterine smooth muscle cells in primary culture. The acrogranin gene was retrovirally transfected into the immortalized smooth muscle cell line and transfected cells were selected with hygromycin B. Forced expression of acrogranin was confirmed in the immortalized uterine smooth muscle cells by RT-PCR. B, anchorage-independent growth of acrogranin-transfected immortalized uterine smooth muscle cell line. Immortalized symmetrical cells (5 × 104/mL) were seeded in soft agar and colonies larger than 50 μm were counted 20 days later. C, number of colonies in soft agar in (B); the data are shown as the sum of colony numbers in 800 μL (eight wells of a 96-well plate). This experiment was done four times independently. Column, mean; bar, SE. D, accelerated anchorage-independent growth of the immortalized uterine smooth muscle cells transfected with acrogranin and SV40ER genes. The uterine smooth muscle cells used in (B) were further transfected with SV40ER using a retroviral vector and transfected cells were selected by blastcidine. Cells (5 × 104/mL) were seeded in soft agar and colonies larger than 100 μm were counted 10 days later. A, phase-contrast microscopy of colonies in soft agar. E, number of formed colonies in soft agar in (D). Data are colony numbers in 100 μL (one well of a 96-well plate). This experiment was done four times independently. Columns, mean; bars, SE. *, P < 0.001.

Fig. 3.

Effect of transfection of with acrogranin and SV40ER genes on anchorage-independent growth of the immortalized uterine smooth muscle cells. A, acrogranin mRNA expression in SKN, uterine leiomyosarcoma cells, and in the immortalized uterine smooth muscle cells. I-SM, immortalized uterine smooth muscle cell line by hTERT transfection. Acrogranin mRNA expression was analyzed by RT-PCR (26 cycles). As an internal control, actin primers were also included in the reactions. SKN (uterine leiomyosarcoma cell line) showed acrogranin gene expression. Acrogranin was not detected in the immortalized uterine smooth muscle cell line and in the uterine smooth muscle cells in primary culture. The acrogranin gene was retrovirally transfected into the immortalized smooth muscle cell line and transfected cells were selected with hygromycin B. Forced expression of acrogranin was confirmed in the immortalized uterine smooth muscle cells by RT-PCR. B, anchorage-independent growth of acrogranin-transfected immortalized uterine smooth muscle cell line. Immortalized symmetrical cells (5 × 104/mL) were seeded in soft agar and colonies larger than 50 μm were counted 20 days later. C, number of colonies in soft agar in (B); the data are shown as the sum of colony numbers in 800 μL (eight wells of a 96-well plate). This experiment was done four times independently. Column, mean; bar, SE. D, accelerated anchorage-independent growth of the immortalized uterine smooth muscle cells transfected with acrogranin and SV40ER genes. The uterine smooth muscle cells used in (B) were further transfected with SV40ER using a retroviral vector and transfected cells were selected by blastcidine. Cells (5 × 104/mL) were seeded in soft agar and colonies larger than 100 μm were counted 10 days later. A, phase-contrast microscopy of colonies in soft agar. E, number of formed colonies in soft agar in (D). Data are colony numbers in 100 μL (one well of a 96-well plate). This experiment was done four times independently. Columns, mean; bars, SE. *, P < 0.001.

Close modal

Impact of forced expression of acrogranin in uterine smooth cells on anchorage-independent cell growth.Acrogranin was retrovirally transfected into the immortalized smooth muscle cell line and transfected cells were selected with hygromycin B. Forced expression of acrogranin in the immortalized uterine smooth muscle cells was confirmed by RT-PCR (Fig. 3A). The immortalized uterine smooth muscle cell line (transfected only with hTERT gene) did not show increased cell proliferation compared with early primary culture of uterine smooth muscle cells as detected by WST-1 assay (data not shown). When this cell line was further transfected with acrogranin, it showed no change in cell proliferation, but showed anchorage-independent growth (Fig. 3B and C) to some extent. However, in this case, the colony size did not exceed 100 μm. The immortalized uterine smooth muscle cell line was also transfected with SV40ER (eventually transfected with hTERT + SV40ER). Transfection of SV40ER resulted in slight increase in cell proliferation and in more apparent anchorage-independent growth (some of the colonies exceeded 100 μm in size as shown in Fig. 3D). When the cell was eventually transfected with hTERT + SV40ER + acrogranin, anchorage-independent growth was markedly accelerated (Fig. 3D and E), but further stimulation of cell growth was not observed. Cells (5 × 104/mL) were seeded in soft agar and colonies larger than 100 μm were counted 10 days later. The cells transfected with hTERT + SV40ER + acrogranin formed significantly increased number of colonies in soft agar compared with cells transfected with hTERT + SV40ER (Fig. 3E).

Impact of forced expression of acrogranin in uterine smooth cells on tumor formation in nude mice. Cells used in Fig. 3 were inoculated s.c. into nude mice at 6 to 8 weeks of age (1 × 107 per animal). Tumor formation and growth was assessed until at least 4 months after the inoculation. Tumor formation in nude mice was observed when uterine smooth muscle cells transfected with hTERT, acrogranin, and SV40ER were inoculated (Fig. 4A; Table 3). In histologic analysis, tumor cells were arranged around tumor vessels and caused sudden massive hemorrhage necrosis, which are typical features of mesenchymal tumors (Fig. 4B). The mesenchymal character of the tumor was also confirmed by the immunohistochemical analysis: vimentin (+) and cytokeratin (−) (Fig. 4C). However, ERα, PR, and α-SMA were negative (data not shown). Acrogranin and SV40 expression were confirmed in all tumor cells (Fig. 4D).

Fig. 4.

Tumor formation in nude mice by uterine smooth muscle cells transfected with hTERT, acrogranin, and SV40ER. A, a representative photograph of tumor-bearing mice. Arrow, a representative tumor. B, histologic analysis of the tumor formed in mice. In H&E staining, histologically massive hemorrhage and coagulative necrosis existed in tumor tissues (left). In high magnification (right), tumor cells were arranged around tumor vessels. C, immunohistochemical study of tumors. Tumors were positive for vimentin and negative for cytokeratin, showing that the tumor originated from mesenchymal cells. D, forced expression of acrogranin and SV40 in tumor cells was confirmed by the immunohistochemical study. Bar, 50 μm.

Fig. 4.

Tumor formation in nude mice by uterine smooth muscle cells transfected with hTERT, acrogranin, and SV40ER. A, a representative photograph of tumor-bearing mice. Arrow, a representative tumor. B, histologic analysis of the tumor formed in mice. In H&E staining, histologically massive hemorrhage and coagulative necrosis existed in tumor tissues (left). In high magnification (right), tumor cells were arranged around tumor vessels. C, immunohistochemical study of tumors. Tumors were positive for vimentin and negative for cytokeratin, showing that the tumor originated from mesenchymal cells. D, forced expression of acrogranin and SV40 in tumor cells was confirmed by the immunohistochemical study. Bar, 50 μm.

Close modal
Table 3.

Summary of tumor formation in nude mice

Smooth muscle cells + hTERT + empty vector 0/3 
Smooth muscle cells + hTERT + acrogranin 0/4 
Smooth muscle cells + hTERT + empty vector + SV40ER 0/7 
Smooth muscle cells + hTERT + acrogranin + SV40ER 6/8 
Smooth muscle cells + hTERT + empty vector 0/3 
Smooth muscle cells + hTERT + acrogranin 0/4 
Smooth muscle cells + hTERT + empty vector + SV40ER 0/7 
Smooth muscle cells + hTERT + acrogranin + SV40ER 6/8 

NOTE: 1× 107 cells were inoculated s.c. in nude mice at 6 to 8 weeks of age. Tumor formation and growth was not assessed until at least 4 months postinoculation.

Uterine leiomyosarcoma is a disease with extremely poor prognosis, is highly aggressive, and is resistant to chemotherapies. At present, surgical intervention is virtually the only means of treatment for uterine leiomyosarcoma (4, 5). However, molecular-targeting therapies against malignant tumors have recently shown remarkable achievements (6). To improve the prognosis of uterine leiomyosarcoma, we aimed to search for key oncogenes that play an important role in their pathogenesis and that could serve as a molecular target for treatment. For this purpose, we conducted a cDNA microarray analysis between uterine leiomyosarcoma and normal uterine smooth muscle and showed that several factors, such as brain-specific polypeptide PEP-19, may be associated with the pathogenesis of leimyosarcoma (26). However, in terms of tumorigenesis of leiomyosarcoma, merely comparing the expression of potential oncogenes between normal and malignant tissues is not sufficient because the results obtained may be the consequence of malignant transformation and, therefore, not necessarily the cause. To obtain more direct evidence for a genetic contribution to tumor occurrence, we established a method in which a candidate oncogene was transfected into phenotypically normal smooth muscle cells and oncogenicity was determined using soft agar assays or tumor formation in nude mice.

Among the various genes that exhibited differential expression between normal and malignant uterine smooth muscles, acrogranin, which is an autocrine/paracrine growth factor, was chosen as our research target. This was largely because, thus far, clinically successful molecular targets include growth factors or their receptors, such as HER2 or vascular endothelial growth factor. Acrogranin is a pluripotent growth factor that mediates cell cycle progression and cell motility (32, 33). Structurally, it bears no homology to the well-established growth factor families (34). In vivo, acrogranin is expressed at a high level in epithelial cells that undergo rapid turnover, notably in the intestinal deep crypt and epidermal keratinocytes, suggesting a role of acrogranin in the regulation of epithelial proliferation (35). It is scarce in connective tissue, endothelia, and muscle in healthy adult tissues.

First, we examined the expression of acrogranin in uterine smooth muscle tumors and showed that it is highly expressed in uterine leiomyosarcoma and in the leiomyosarcoma cell line SKN (Figs. 1 and 3A; Table 2) In immunohistochemical studies, the intensity of its expression correlated with the histologic grade and prognosis although the number of samples analyzed are relatively few (Table 2). In other types of malignancies, acrogranin is highly expressed in aggressive cancer cell lines and clinical specimens, including breast, hepatocellular, ovarian, and prostatic cancers, as well as gliomas (1116, 18). Accordingly, we hypothesized that acrogranin may play a critical role in tumorigenesis in uterine leiomyosarcoma.

We attempted to evaluate the oncogenic property of acrogranin in a more direct fashion, namely by transfecting its gene into phenotypically normal uterine smooth muscle cells. It has been reported that uterine smooth muscle cells are immortalized by transfection of hTERT cDNA and these immortalized cells retain their original characteristics (2931). However, to our knowledge, there have been no attempts to transform immortalized uterine smooth muscle cells by transfecting potential oncogenes. We first transfected hTERT cDNA into uterine smooth muscle cells and one of the selected colonies was maintained. These cells expressed ERα, PR, and α-SMA (Fig. 2C), showing the original character of primary uterine smooth muscle cells. This cell line did not show anchorage-independent growth or tumorigenicity in nude mice and, therefore, was suitable to examine the oncogenic function of acrogranin.

Although it was reported that overexpression of acrogranin in leiomyosarcoma cell line promote proliferation (35), it did not promote cell proliferation in immortalized uterine smooth muscle cells as detected by WST-1 assay. However, it resulted in colony formation in soft agar, which was only microscopically detected; three to four colonies of 4 × 104 cells were obtained that were only ∼50 μm in diameter and never exceeded 100 μm (Fig. 3B). These cells did not form tumors in nude mice (Table 3). These results may show a potential oncogenic function of acrogranin, but also suggest that coexpression of hTERT and acrogranin is not sufficient to transform uterine smooth muscle cells.

We then transfected the SV40ER into these uterine smooth muscle cells. When transfected with the SV40ER, hTERT-immortalized uterine smooth muscle cells showed obvious anchorage-independent growth but did not form tumors in nude mice (Fig. 3D; Table 3). On the other hand, coexpression of hTERT, acrogranin, and SV40ER resulted in more extensive anchorage-independent growth and tumor formation in nude mice (Figs. 3D and 4A; Table 3). This result indicates that coexpression of these three genes are necessary and sufficient to transform uterine smooth muscle cells. A number of nonmitogenic actions of acrogranin relevant to cancer are known, e.g., decreased susceptibility to anoikis, increased angiogenesis, invasion, and motility (10, 11, 18). As the expression of acrogranin did not accelerate cell proliferation in monolayer (data not shown), our results suggest oncogenic, but nonproliferative, actions of acrogranin.

Studies on in vitro malignant transformation have revealed that to transform primary human cells, the following four signals are thought to be necessary: (a) elongation of telomere length by hTERT expression, (b) perturbation of p53 and retinoblastoma by SV40LT expression or other methods, (c) perturbation of PP2A by SV40 small tumor antigen expression or other methods, and (d) constitutive RAS signaling by transfection of the oncogenic allele of RAS (3639). In general, signals from growth factors are transmitted through tyrosine kinase type receptors to RAS (40). As such, constitutively activated RAS signaling can be used to substitute for persistent growth factor signaling. Actually, as we previously reported, ovarian surface epithelial cells immortalized by coexpression of hTERT and SV40 are transformed by c-ERBB2 instead of oncogenic RAS (20). However, there has been no report that overexpression of growth factors can substitute for transfection of oncogenic RAS.

Signals generated by acrogranin are thought to be strong. Embryonic fibroblasts from mice in which the insulin-like growth factor-I receptor was deleted (i.e., R− cells) fail to divide when stimulated by well-established growth factors. They do, however, proliferate when exposed to acrogranin (41). Acrogranin activates the mitogen-activated protein kinase and phosphatidylinositol-3 kinase signaling cascades (42). Activated phosphatidylinositol-3 kinase can substitute, at least in part, for oncogenic RAS signal in de novo transformation (22). Hence, it is one of the possible mechanisms that acrogranin exert oncogenicity via phosphatidylinositol-3 kinase activation by substituting RAS signaling. However, we do not have direct evidence for that yet and other possibilities cannot be excluded.

In summary, we showed that acrogranin was overexpressed in uterine leiomyosarcoma and their higher histologic grade was correlated with higher expression of acrogranin. Transfection of hTERT, SV40ER, and acrogranin in uterine smooth muscle cells resulted in transformation of primary cultured cells. These data indicate that acrogranin may be a promising target for developing effective molecular targeting therapy for treatment of uterine leiomyosarcoma. In addition, acrogranin may also serve as a specific diagnostic marker to identify leiomyosarcomas of the uterus, the diagnosis of which is clinically difficult. Moreover, our study established a new method to evaluate the oncogenic property of specific genes in the process of malignant transformation of hTERT-immortalized and SV40-activated human smooth muscle cells.

Grant support: Grants-in-aid for Scientific Research 15209057 (S. Fujii), 17659514 (K. Takakura), and 17591729 (T. Higuchi) from Japan Society for the Promotion of Science.

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.

We thank Dr. Susan Murphy at Duke University Medical Center for her useful advice in the preparation of the manuscript and Michiko Muraoka for her great contribution as a laboratory technician.

1
Echt G, Jepson J, Steel J, et al. Treatment of uterine sarcomas.
Cancer
1990
;
66
:
35
–9.
2
Gadducci A, Landoni F, Sartori E, Zola P, Maggino T, Lissoni A. Uterine leiomyosarcoma: analysis of treatment failures and survival.
Gynecol Oncol
1996
;
62
:
25
–32.
3
Salazar OM, Bonfiglio TA, Patten SF, Keller BE, Feldstein M, Dunne ME. Uterine sarcomas: natural history, treatment and prognosis.
Cancer
1978
;
42
:
1152
–60.
4
Dusenbery KE, Potish RA, Judson P. Limitations of adjuvant radiotherapy for uterine sarcomas spread beyond the uterus.
Gynecol Oncol
2004
;
94
:
191
–6.
5
Giuntoli RL II, Bristow RE. Uterine leiomyosarcoma: present management.
Curr Opin Oncol
2004
;
16
:
324
–7.
6
Perez EA, Pusztai L, Van de Vijver M. Improving patient care through molecular diagnostics.
Semin Oncol
2004
;
31
:
14
–20.
7
Zhou J, Gao G, Crabb JW, Serrero G. Purification of an autocrine growth factor homologous with mouse epithelin precursor from a highly tumorigenic cell line.
J Biol Chem
1993
;
268
:
10863
–9.
8
Bhandari V, Palfree RG, Bateman A. Isolation and sequence of the granulin precursor cDNA from human bone marrow reveals tandem cysteine-rich granulin domains.
Proc Natl Acad Sci U S A
1992
;
89
:
1715
–9.
9
Daniel R, Daniels E, He Z, Bateman A. Progranulin (acrogranin/PC cell-derived growth factor/granulin-epithelin precursor) is expressed in the placenta, epidermis, microvasculature, and brain during murine development.
Dev Dyn
2003
;
227
:
593
–9.
10
He Z, Ong CH, Halper J, Bateman A. Progranulin is a mediator of the wound response.
Nat Med
2003
;
9
:
225
–9. Epub 2003 Jan 13.
11
Cheung ST, Wong SY, Leung KL, et al. Granulin-epithelin precursor overexpression promotes growth and invasion of hepatocellular carcinoma.
Clin Cancer Res
2004
;
10
:
7629
–36.
12
Davidson B, Alejandro E, Florenes VA, et al. Granulin-epithelin precursor is a novel prognostic marker in epithelial ovarian carcinoma.
Cancer
2004
;
100
:
2139
–47.
13
Pan CX, Kinch MS, Kiener PA, et al. PC cell-derived growth factor expression in prostatic intraepithelial neoplasia and prostatic adenocarcinoma.
Clin Cancer Res
2004
;
10
:
1333
–7.
14
Serrero G, Ioffe OB. Expression of PC-cell-derived growth factor in benign and malignant human breast epithelium.
Hum Pathol
2003
;
34
:
1148
–54.
15
Jones MB, Michener CM, Blanchette JO, et al. The granulin-epithelin precursor/PC-cell-derived growth factor is a growth factor for epithelial ovarian cancer.
Clin Cancer Res
2003
;
9
:
44
–51.
16
Liau LM, Lallone RL, Seitz RS, et al. Identification of a human glioma-associated growth factor gene, granulin, using differential immuno-absorption.
Cancer Res
2000
;
60
:
1353
–60.
17
He Z, Bateman A. Progranulin gene expression regulates epithelial cell growth and promotes tumor growth in vivo.
Cancer Res
1999
;
59
:
3222
–9.
18
Lu R, Serrero G. Inhibition of PC cell-derived growth factor (PCDGF, epithelin/granulin precursor) expression by antisense PCDGF cDNA transfection inhibits tumorigenicity of the human breast carcinoma cell line MDA-MB-468.
Proc Natl Acad Sci U S A
2000
;
97
:
3993
–8.
19
Hahn WC, Counter CM, Lundberg AS, Beijersbergen RL, Brooks MW, Weinberg RA. Creation of human tumour cells with defined genetic elements.
Nature
1999
;
400
:
464
–8.
20
Kusakari T, Kariya M, Mandai M, et al. C-erbB-2 or mutant Ha-ras induced malignant transformation of immortalized human ovarian surface epithelial cells in vitro.
Br J Cancer
2003
;
89
:
2293
–8.
21
Liu J, Yang G, Thompson-Lanza JA, et al. A genetically defined model for human ovarian cancer.
Cancer Res
2004
;
64
:
1655
–63.
22
Zhao JJ, Gjoerup OV, Subramanian RR, et al. Human mammary epithelial cell transformation through the activation of phosphatidylinositol 3-kinase.
Cancer Cell
2003
;
3
:
483
–95.
23
Lundberg AS, Randell SH, Stewart SA, et al. Immortalization and transformation of primary human airway epithelial cells by gene transfer.
Oncogene
2002
;
21
:
4577
–86.
24
MacKenzie KL, Franco S, Naiyer AJ, et al. Multiple stages of malignant transformation of human endothelial cells modelled by co-expression of telomerase reverse transcriptase, SV40 T antigen and oncogenic N-ras.
Oncogene
2002
;
21
:
4200
–11.
25
Rich JN, Guo C, McLendon RE, Bigner DD, Wang XF, Counter CM. A genetically tractable model of human glioma formation.
Cancer Res
2001
;
61
:
3556
–60.
26
Kanamori T, Takakura K, Mandai M, et al. PEP-19 overexpression in human uterine leiomyoma.
Mol Hum Reprod
2003
;
9
:
709
–17.
27
Blaustein's pathology of the female genital tract. 5th ed. New York: Springer-Verlag, Inc.; 2002. p. 561–615.
28
Bell SW, Kempson RL, Hendrickson MR. Problematic uterine smooth muscle neoplasms. A clinicopathologic study of 213 cases.
Am J Surg Pathol
1994
;
18
:
535
–58.
29
Condon J, Yin S, Mayhew B, et al. Telomerase immortalization of human myometrial cells.
Biol Reprod
2002
;
67
:
506
–14.
30
Carney SA, Tahara H, Swartz CD, et al. Immortalization of human uterine leiomyoma and myometrial cell lines after induction of telomerase activity: molecular and phenotypic characteristics.
Lab Invest
2002
;
82
:
719
–28.
31
Soloff MS, Jeng YJ, Ilies M, et al. Immortalization and characterization of human myometrial cells from term-pregnant patients using a telomerase expression vector.
Mol Hum Reprod
2004
;
10
:
685
–95. Epub 2004 Jul 8.
32
He Z, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis.
J Mol Med
2003
;
81
:
600
–12.
33
Serrero G. Autocrine growth factor revisited: PC-cell-derived growth factor (progranulin), a critical player in breast cancer tumorigenesis.
Biochem Biophys Res Commun
2003
;
308
:
409
–13.
34
Hrabal R, Chen Z, James S, Bennett HP, Ni F. The hairpin stack fold, a novel protein architecture for a new family of protein growth factors.
Nat Struct Biol
1996
;
3
:
747
–52.
35
Daniel R, He Z, Carmichael KP, Halper J, Bateman A. Cellular localization of gene expression for progranulin.
J Histochem Cytochem
2000
;
48
:
999
–1009.
36
Rangarajan A, Hong SJ, Gifford A, Weinberg RA. Species- and cell type-specific requirements for cellular transformation.
Cancer Cell
2004
;
6
:
171
–83.
37
Akagi T. Oncogenic transformation of human cells: shortcomings of rodent model systems.
Trends Mol Med
2004
;
10
:
542
–8.
38
Bocchetta M, Carbone M. Epidemiology and molecular pathology at crossroads to establish causation: molecular mechanisms of malignant transformation.
Oncogene
2004
;
23
:
6484
–91.
39
Zhao JJ, Roberts TM, Hahn WC. Functional genetics and experimental models of human cancer.
Trends Mol Med
2004
;
10
:
344
–50.
40
Campbell PM, Der CJ. Oncogenic Ras and its role in tumor cell invasion and metastasis.
Semin Cancer Biol
2004
;
14
:
105
–14.
41
Xu SQ, Tang D, Chamberlain S, et al. The granulin/epithelin precursor abrogates the requirement for the insulin-like growth factor 1 receptor for growth in vitro.
J Biol Chem
1998
;
273
:
20078
–83.
42
Zanocco-Marani T, Bateman A, Romano G, Valentinis B, He ZH, Baserga R. Biological activities and signaling pathways of the granulin/epithelin precursor.
Cancer Res
1999
;
59
:
5331
–40.