Journal of Cancer Genetics And Biomarkers

Journal Of Cancer Genetics And Biomarkers

Journal of Cancer Genetics and Biomarkers

Current Issue Volume No: 1 Issue No: 1

Research Article Open Access Available online freely Peer Reviewed Citation

The Identification of Somatic Mutations in Interferon-G Signal Molecules in Human Uterine Leiomyosarcoma

1Dept. of Obstetrics and Gynecology, Shinshu University School of Medicine, Japan,

2Dept. of Medical Technology, International University of Health and Welfare, Japan,

3Dept. of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Japan,

4Dept. of Health Science, Kyushu University Graduate School of Medicine, Japan,

5Picower Institute for learning and memory, Massachusetts Institute of Technology, USA,

7The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo,

8Dept. of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan,

9National Hospital Organization Kyoto Medical Center, Japan,

10Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Japan,

11The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST).

Abstract

Human uterine leiomyosarcoma (LMS) is neoplastic malignancy that typically arises in tissues of mesenchymal origin. The identification of novel molecular mechanism leading to human uterine LMS formation and the establishment of new therapies has been hampered by several critical points. We earlier reported that mice with a homozygous deficiency for proteasome beta subunit 9 (PSMB9)/b1i, an interferon (IFN)-g inducible factor, spontaneously develop uterine LMS. The use of research findings of the experiment with mouse model has been successful in increasing our knowledge and understanding of how alterations, in relevant oncogenic, tumour suppressive, and signaling pathways directly impact sarcomagenesis. The IFN-g pathway is important for control of tumour growth and invasion and, has been implicated in several malignant tumours. In this study, experiments with human tissues revealed a defective PSMB9/b1i expression in human uterine LMS that was traced to the IFN-g pathway and the specific effect of somatic mutations of Janus kinase (JAK1) molecule or promoter region on the transcriptional activation of PSMB9/b1i gene. Understanding the molecular mechanisms of human uterine LMS may lead to identification of new diagnostic candidates or therapeutic targets in human uterine LMS.

Author Contributions
Received August 29 ; 2016; AcceptedPublished 11 Oct 2016; 15 Oct 2016;

Academic Editor: Xi Zhang, Co-founder & Scientist SinoScript LL

Checked for plagiarism: Yes

Review by: Single-blind

Copyright ©  2016 Takuma Hayashi, et al.

License
Creative Commons License     This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Competing interests

The authors have declared that no competing interests exist.

Citation:

Takuma Hayashi, Tomoyuki Ichimura, Hirofumi Ando, Koichi Ida, Miki Kawano et al. (2016) The Identification of Somatic Mutations in Interferon-G Signal Molecules in Human Uterine Leiomyosarcoma. Journal Of Cancer Genetics And Biomarkers - 1(1):29-45. https://doi.org/10.14302/issn.2572-3030.jcgb-16-1276

Download as RIS, BibTeX, Text (Include abstract )

DOI 10.14302/issn.2572-3030.jcgb-16-1276

Introduction

Uterine mesenchymal tumours have been traditionally divided into benign tumour leiomyomas (LMA) and malignant tumour leiomyosarcomas (LMS) based on cytological atypia, mitotic activity and other criteria. Uterine LMS, which are some of the most common neoplasms of the female genital tract, are relatively rare uterine mesenchymal tumour, having an estimated annual incidence of 0.64 per 100,000 women 1. They account for approximately one-third of uterine sarcomas, of only 53% for tumours confined to the uterus 2, 3. Generally, patients with uterine LMS typically present with vaginal bleeding, pain, and a pelvic mass. Gynecological cancer, for instance breast cancer and endometrial carcinomas, are strongly promoted by female hormones, but the rate of hormone receptor expression is reported to be significantly less in human uterine LMS compared with normal myometrium. These low receptor expressions were found to not correlate with the promotion of initial disease development or with the overall survival of patients with uterine LMS.

As uterine LMS is resistant to chemotherapy and radiotherapy, and thus surgical intervention is virtually the only means of treatment for this disease 4, 5, 6, however, molecular targeting therapies against tumours have recently shown remarkable achievements 7, 8. It is noteworthy that, when adjusting for stage and mitotic count, uterine LMS has a significantly worse prognosis than carcinosarcoma 9; developing an efficient

adjuvant therapy is expected to improve the prognosis of the disease. A trend towards prolonged disease-free survival is seen in patients with matrix metalloproteinase (MMP)-2-negative tumours 10. Although typical presentations with hypercalcemia or eosinophilia have been reported, this clinical abnormality is not an initial risk factor for uterine LMS. To the best of our knowledge, little is known regarding the biology of uterine LMS; therefore, the risk factors that promote the initial development of uterine LMS and regulate their growth in vivo remain poorly understood.

The mice with a targeted disruption of proteasome beta subunit 9 (PSMB9)/b1i, which is interferon (IFN)-g -inducible proteasome subunit, exhibited a defect in tissue- and substrate- dependent proteasome function, and female PSMB9-deficient mice shown to develop uterine LMS, with a disease prevalence of 37% by 14 months of age 11, 12. Defective PSMB9/b1i expression is likely to be one of the risk factors for the development of human uterine LMS, as it is in PSMB9/b1i-deficient mice 12 Recent report shows that stable PSMB9/b1i expression contributes to cell proliferation, which directly correlates to the progressive deterioration with increasing stage and grade of the tumour. As the importance and involvement of the IFN-g signal pathway in the transcriptional regulation of the transporter associated with antigen processing (TAP1) and PSMB9/b1i promoter have been established, it is demonstrated that the defective PSMB9/b1i expression was attributable to G871E somatic mutation in the ATP-binding region of JAK1 molecule in SKN cell line, which is established from patient with uterine LMS. It is furthermore likely that the expressions of PSMB9/b1i are down-regulated in human uterine LMS tissues such like human uterine LMS cell line. We demonstrate that there are serious mutational defects in the factors on the IFN-γ signal pathway, which is the key cell-signaling pathway for PSMB9/b1i expression and promoter region of PSMB9/b1i gene, in human uterine LMS tissues. The somatic mutational defects in the IFN-γ signal molecules may induce the initial development of human uterine LMS. Recent advances in our understanding of the biological characters of uterine LMS have concentrated on the impaired IFN-γ signal pathway. It is clear that somatic mutations in key regulatory factors (tumour suppressors and proto-oncogenes) alter the behavior of cells and can potentially lead to the unregulated growth seen in malignant tumour. Therefore, continued improvement of our knowledge of the molecular biology of uterine LMS may ultimately lead to novel therapies and improved outcome.

Result and Discussion

Defective PSMB9/b1i Expression of Human Uterine LMS

The effects of IFN-γ on expression of PSMB9/b1i was examined using five cell lines 13. The expression of PSMB9/b1i were not markedly induced by IFN-γ treatment in human uterine LMS cell lines, although cervical epithelial adenocarcinoma cell lines and normal human uterus smooth muscle cells underwent strong induction of PSMB9/b1i following IFN-γ treatment 13. Furthermore, the immunohistochemistry (IHC) experiments revealed a serious loss in the ability to induce expression of PSMB9/b1i in human uterine LMS tissues in comparison with normal myometrium tissues located in same tissue sections and other 4 mesenchymal tumour types (Figure 1). of 58 uterine LMS, 50 cases (86.2%) were negative for PSMB9/b1i, 4 cases (6.9%) were focally positive, 2 cases (3.4%) were weakly positive, and 2 cases (3.4%) were positive (Figure 1B). IHC analyses showed positivity for Ki-67/MIB1 and differential expression of estrogen receptor (ER), progesterone receptor (PR), tumour protein (TP53), and CALPONIN h1 (Supplemental Table 1). In addition, PSMB9/b1i expression level was also examined in the skeletal muscle metastasis from uterine LMS, the histological diagnosis was consistent with metastatic LMS for skeletal muscle lesions. Pathological examination of surgical samples showed presence of a mass measuring 3 cm at largest diameter in lumbar quadrate muscle without a fibrous capsule. All lymphnodes were negative. In western blotting experiments and RT-PCR experiments, PSMB9/b1i was expressed in normal myometrium, LMA, and IFN-g-treated HeLa cells, but not in human uterine LMS (Figure 1C). The both research experiments strongly supported the research findings obtained from IHC experiments.

Supplemental Table 1. Expression of ER, PR, Ki-67, TP53, PSMB9/b1i, and CALPONIN h1, and somatic mutations of cathartic domains of JAK1, JAK2, STAT1, and the promoter region of PSMB9/b1i gene in human uterine leiomyosarcoma. Immunohistochemistry of ER, PR, Ki-67, TP53, PSMB9/b1i, and CALPONIN h1 in normal uterine smooth muscle and uterine leiomyosarcoma tissues located in same tissue section was performed. The isolating of gemonic DNA of uterine leiomyosarcoma tissues and direct sequencing for cathartic domains of JAK1, JAK2, STAT1, and the promoter region of PSMB9/b1i gene in human uterine leiomyosarcoma was carried out.
Expression of ER, PR, Ki-67, p53, PSMB9, Calponin hi and Senescence staining in human uterine leiomyosarcoma  
patient Age in No. yrs TNN stage MF CCN Immunohi stochemi cal staining TP53 PSMB9 CAL. Somatic mutation Follow up (■..the) Senescence
ER PR Ki-67 JAK1 JAK2 STAT1 PSMB9pro. p53 p-gal. PML
1 37 T4N1M0 97 + - - 3000 +++ - - ND ND ND ND SM D(1) - -
2 58 T3N0M3 24 + - - 3500 * +/- - ND ND ND SM SM D(23) +/- +/-
3 45 T2N0MEI 32 + +1- +/- 2150 +++ - - SM ND SM SM SM D(24) - -
4 65 T1N0M3 30 + +/- +/- 1700 +++ - - SM ND ND ND SM D(20) - -
5 52 TINEIMa 107 + - + 2600 ++ + - SM ND ND ND ND D(13) + +/-
6 49 T1N0M0 46 + - - 4300 + - - ND ND ND ND ND D(24) - -
7 55 T1N0M3 75 + - - 4000 +++ - - ND ND SM SM ND D(!8) - -
8 43 T3N0M0 57 + + - 2000 - +/- +/- ND ND ND ND ND D(10) +/- +/-
9 67 T1N0M0 13 + - +1- 1430 - - - SM ND ND ND ND A(34) - -
10 67 T1N0M0 37 + - - 2100 - - - ND ND SM SM ND A(15) - -
11 51 T1N0M0 93 + - - 4500 - - - ND ND SM ND ND A(94) - -
12 48 T1N0M0 14 + - - 900 ++ + 1- ND ND ND ND ND A(58) +/- +/-
13 51 TIMM@ 22 + +/- + 450 + - - SM ND ND SM ND A(34) - -
14 67 T1N0M0 64 + - + 1450 ++ - - ND ND ND ND ND A(15) - -
15 52 T1N0M0 65 + - - 1780 ++ - - SM ND SM ND ND D(23) - -
16 42 T3N0M0 73 + - - 2130 ++ - - ND ND ND SM ND A(21) - -
17 80 T1N0M0 98 + - - 1980 +++ - - SM ND ND ND ND D(19) - -
18 56 T1N0M0 78 + - - 1860 ++ - - ND ND ND ND ND A(11) - -
19 58 T1N0M0 40 + - - 1750 ++ - - ND ND ND ND ND A(10) - -
20 65 T2N0M0 67 + - - 780 +++ - - SM ND ND SM ND A(12) - -
21 45 T1N0M0 52 + - - 1045 ++ - - SM ND ND SM ND A(13) - -
22 57 T2N0M0 62 + - - 980 ++ +/- - SM ND ND SM ND A(11) +/- +/-
23 54 UNDO 54 + - - 860 +++ - - ND ND ND SM ND A(02) - -

Somatic Mutations in IFN-γ Signal Pathway in Human uterine LMS Tissues

IFN-γ treatment markedly increased the expression of PSMB9/b1i, a subunit of the immunoproteasome, which alters the proteolytic specificity of proteasomes. After binding of IFN-γ to the type II IFN receptor, which is constructed by two components, IFN-γ receptor subunit 1 (IFNGR1) and IFN-γ receptor subunit 2 (IFNGR2), Janus-activated kinase 1 (JAK1) and JAK2 are activated and phosphorylate the signal transducer and activator of transcription 1(STAT1) on the tyrosine residue at position 701 (Tyr701) and the serine residue at position 727 (Ser727) 14, 15 (Supplemental Figure 1). Tyrosine phosphorylated STAT1 forms homodimers that translocate to the nucleus and bind GAS (IFN-γ-activated site) elements in the promoters of IFN-γ-regulated genes 14, 15 (Supplemental Figure 1). The phosphorylation of Ser727 is not essential for the translocation of STAT1 to the nucleus or for the binding of STAT1 to enhancer/promoter region of targeted DNA, but it is required for full transcriptional activation 16, 17 (Supplemental Figure 1).

Supplemental Figure 1.The interferon-g signaling pathway and mutations in its components found in human uterine leiomyosarcoma. After binding of interferon-g (IFN- g) to the type II IFN receptor, Janus activated kinase 1 (JAK1) and JAK2 are activated and phosphorylate signal transducer and activator of transcription 1 (STAT1) on the tyrosine residue at position 701 (Tyr701). The tyrosine-phosphorylated form of STAT1 forms homodimers that translocate to the nucleus and bind GAS (IFN-g-activated site) elements, which are present in the promoters of IFN-γ-regulated genes. The IFN-g-activated JAKs also regulate, through as-yet-unknown intermediates, activation of the catalytic subunit (p110) of phosphatidylinositol 3-kinase (PI3K). The activation of PI3K ultimately results in downstream activation of protein kinase C-δ (PKC-δ), which in turn regulates phosphorylation of STAT1 on the serine residue at position 727 (Ser727). The phosphorylation of Ser727 is not essential for the translocation of STAT1 to the nucleus or for the binding of STAT1 to DNA, but it is required for full transcriptional activation. IFNGR1; IFN-g receptor subunit 1, IFNGR2; IFN-g receptor subunit 2.
 The interferon-g signaling pathway and mutations in its components found in human uterine leiomyosarcoma. After binding of interferon-g (IFN- g) to the type II IFN receptor, Janus activated kinase 1 (JAK1) and JAK2 are activated and phosphorylate signal transducer and activator of transcription 1 (STAT1) on the tyrosine residue at position 701 (Tyr701). The tyrosine-phosphorylated form of STAT1 forms homodimers that translocate to the nucleus and bind GAS (IFN-g-activated site) elements, which are present in the promoters of IFN-γ-regulated genes. The IFN-g-activated JAKs also regulate, through as-yet-unknown intermediates, activation of the catalytic subunit (p110) of phosphatidylinositol 3-kinase (PI3K). The activation of PI3K ultimately results in downstream activation of protein kinase C-δ (PKC-δ), which in turn regulates phosphorylation of STAT1 on the serine residue at position 727 (Ser727). The phosphorylation of Ser727 is not essential for the translocation of STAT1 to the nucleus or for the binding of STAT1 to DNA, but it is required for full transcriptional activation. IFNGR1; IFN-g receptor subunit 1, IFNGR2; IFN-g receptor subunit 2.

The defect was localized to JAK1 activation, which acts upstream in the IFN-γ signal pathway since IFN-γ treatment could not strongly induce JAK1 kinase activity in human uterine LMS cell lines. Sequence analysis demonstrated that the loss of IFN-γ responsiveness in the human uterine LMS cell line was attributable to the inadequate kinase activity of JAK1 due to a G781E somatic mutation in the ATP-binding region 13 Genetic alterations in tyrosine kinases have previously been firmly implicated in tumourigenesis, but only a few serine/threonine kinases are known to be mutated in human cancers 18, 19, 20, 21, 22. For instance, mice carrying homozygous deletion of Pten alleles developed wide spread smooth muscle cell hyperplasia and abdominal leiomyosarcomas 21, and JUN proto-oncogene amplification and over-expression block adipocytic differentiation in highly aggressive sarcomas 22.

Most frequently, LMS have appeared in the uterus, retroperitoneum or extremities, and although histologically indistinguishable, they have different clinical courses and chemotherapeutic responses. The molecular basis for these differences remains unclear. Therefore, the examination of human uterine LMS tissues 23 LMS tissue sections and normal tissue sections located in the same tissue) was performed to detect somatic mutations in the IFN-γ signaling molecules cascade, JAK1, JAK2, STAT1 and promoter region of PSMB9/b1i gene. As the catalytic domains of these molecules are most likely to harbour mutations that activate the gene product, we focused on stretches (exons) containing the kinase domains, transcriptional activation domains and enhancer/promoter region. Over all, nearly 43.5% (10/23) of uterine LMS tissues had serious mutations in the ATP binding region or kinase-specific active site of JAK1; furthermore, 43.5% (10/23) of uterine LMS tissues had serious mutations in essential sites of the promoter region of PSMB9/b1i gene, which is required for transcriptional activation of PSMB9/b1i gene (Table 1, Table 2). No somatic mutation in essential sites, Tyr701 and Ser727, which are required for transcriptional activation of STAT1, was elucidated in human uterine LMS. Nearly 21.7% (5/23) of human uterine LMS tissues unexpectedly had mutations in the STAT1 intermolecular region, which is not yet reported to be important for biological function as transcriptional activation. No somatic mutation in the ATP-binding region and kinase-active site of JAK2 was detected in human uterine LMS (Table 1, Table 2). MOTIF Search profiling 23 and NCBI's Conserved Domain Database and Search Service, v2.17 analysis also revealed that somatic mutations, which were identified in the catalytic domains of these molecules, resulted in impaired activations of tyrosine kinases or transcriptional factor 24.

Table 1. Identification of somatic (tumour-specific) mutations in the cataclytic domains of JAK1, JAK2 kinases, STAT1 or activation region of the promoter region of PSMB9/b1i gene in human uterine leiomyosarcoma (total 23 cases). The gemonic DNA was extracted from the human uterine leiomyosarcoma tissues and normal uterine smooth muscle tissues using the protocol indicated in the materials and methods section of this manuscript. The restricted DNA fragments for the direct sequence analysis were amplified by PCR procedure with the appropriate primers for the ATP-binding region and kinase activation domain of the JAK1 molecule, the promoter region of PSMB9/b1i gene, the Tyr701 and Ser727 aminoacid of STAT1 molecule, and the ATP-binding region and kinase activation domain of the JAK2 molecule. Information of human uterine leiomyosarcomas and the primer sets was indicated in Supplementary Table 1 and the materials and methods section of Supplementary material.
Hayashi ct al. Table 1        
Mutations in JAK1 kinase, PSMB9 promoter region, and STAT1 in human uterine leiomyosarcoma  
Patient # JAK1 kinase PSMB9 promoter region STAT1(701Y,727S)5 JAK2 kinase PSMB9?
#1 wt wt wt wt Neg
#2 wt A210G, C214T(IRF-E)3 wt wt P.Posi
#3 Q986P(active) R995S(active)1 C214T, G219A(IRF-E) (S710A) 6 wt Neg
#4 G876R(ATP)2 wt wt wt Neg
#5 C881 F (ATP) wt wt wt P.posi
#6 wt wt wt wt Neg
#7 wt A216G(IRF-E) (L693R) 6 wt Neg
#8 wt wt wt wt F.pos
#9 Y987S(active) wt wt wt Neg
#10 wt A217G(IRF-E) (R716S) 6 wt Neg
#11 wt wt (1702L) 6 wt Neg
#12 wt wt wt wt F.Posi.
#13 Y987S(active) A216G(IRF-E) wt wt Neg
#14 wt wt wt wt Neg
#15 G871E(ATP) wt (1702L) 6 wt Neg
#16 wt G239A(HSF)'1 wt wt Neg
#17 C881 F(ATP) wt wt wt Neg
#18 wt wt wt wt Neg
#19 wt wt wt wt F.Posi.
#20 G873D(ATP) A210G(IRF-E) wt wt Neg
#21 C881Stop(TGC-TGA) G209T(IRF-E) wt wt Neg
#22 Q986P(active) G215A(IRF-E) wt wt F.Posi.
#23 wt C213A(IRF-E) wt wt Neg

Table 2. Somatic mutations in IFN-g signaling pathway in human uterine leiomyosarcoma. The data of somatic mutations in Table 1 was shown separately with respect to each gene, JAK1, JAK2, STAT1 and activation region of the promoter of PSMB9/b1i gene.
Hayashi et al. Table 2              
Mutations in the IFN-y pathway in human uterine Ieiomyosarcoma      
Gene Name Locus GenBank Accession MIM ID Tumor Nucleotide Amino Acid Domain Evolutionary4 conservation
JAK1 HUMPTKJAK1 M64174.1 147795 ULMS G2612A G2618A G781E ATP binding G873D ATP binding p,c,m,r,g,d
          G2626A G876R ATP binding  
          G2642T C881F ATP binding  
          G2643A A2957 C C881 Stop ATP binding Q986P active site  
          A2960 C Y987S active site  
          A2985T R995S active site  
JAK2 AF005216 AF005216.1 +147796 ULMS ND2 ND ND p,c,b,m,r,g,d
STAT1 NM_007315 NM_007315 +600555 ULMS A2104C 1702L NA3 c,b,m,r,g,d
          T2128G S710A NA  
          T2078 G L693R NA  
          A2148C R716S NA  
PSMB91 X62741 X62741.1 177045 ULMS A209T IRF-E site p,c,b,m,r,d
          A210G IRF-E site  
          C213A IRF-E site  
          C214T IRF-E site  
          G215A IRF-E site  
          A216G IRF-E site  
          A217G IRF-E site  
          G219 A IRF-E site  
          G239 A HSF site  

In a recent report, a comparative genomic hybridization (CGH)-based analysis of human LMS using a high resolution genome-wide array gave gene-level information about the amplified and deleted regions that may play a role in the development and progression of human uterine LMS. Other reports showed that among the most intriguing changes in genes were losses of JAK1 (1p31-p32) and PSMB9/b1i (6p21.3) 25, 26. It has also been demonstrated that a correlation exists between the development of malignant tumours and ethnic background, so we conducted CGH experiments with tissue samples obtained from Japanese patients in order to obtain gene-level information. Our results showed that human uterine LMS having a clear functional loss at JAK1 (1p31-p32) and PSMB9/b1i (6p21.3) also harbored one nonsense mutation and one deletion, suggesting a possible homozygous loss of function. The discovery of these mutational defects in a key cell-signaling pathway may be important in understanding the pathogenesis of human uterine LMS.

Gene Analyses of Human Uterine Lyomyosarcoma

Uterine LMS are relatively rare mesenchymal tumours, having an estimated annual incidence of 0.64 per 100 000 women. They account for approximately one-third of uterine sarcomas and 1.3% of all uterine malignancies. They are the disease with extremely poor prognosis, considering aggressive malignancies with a 5-year survival rate of only 50% for tumours confined to the uterus. At present, surgical intervention is virtually the only means of treatment for uterine LMS 4, 5, 6, 7, 8. Although adjuvant pelvic irradiation appears to decrease the rate of local recurrence, adjuvant therapy does not appear to significantly improve survival. Furthermore, gynaecological cancer, for instance breast cancer and endometrial carcinomas, are strongly promoted by female hormones, but the rate of estrogen receptor and progesterone receptor expression is reported to be significantly less in human uterine LMS compared with normal myometrium. These low receptor expressions were found to not correlate with the promotion of initial disease development or with the overall survival of patients with human uterine LMS; however, molecular targeting therapies against tumours have recently shown remarkable achievements 27. To improve the prognosis of human uterine LMS, research experiments were performed to identify the key role of pro- or anti-oncogenic factors that have an important function in their pathogenesis and that could serve as molecular targets for tumour treatment. For this purpose, several research facilities conducted a microarray procedure between human uterine LMS and normal myometrium and showed that several known pro-oncogenic factors, such as brain-specific polypeptide PEP-19 and a transmembrane tyrosine kinase receptor, c-kit, may be associated with the pathogenesis of human uterine LMS 28, 29, 30. However, in terms of the tumourigenesis of human uterine LMS, merely comparing the expression of potential pro-oncogenic factors 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. In addition, dysregulation of apoptotic mechanisms has also been implicated in many human malignancies. Although the significant differential expression of apoptotic and cell cycle regulatory factors in human uterine LMS, such as B-cell Lymphoma-2 (BCL-2), BCL-2-Associated X protein (BAX), P16 Inhibts CDK4 (P16/INK4a), P21 Cyclin-Dependent Kinase Inhibitor 1 (P21/CIP1), P27 Kinase Inhibitor Protein 1 (P27/KIP1), Cellular v-KIT Hardy-Zuckerman 4 Feline Sarcoma Viral Oncogene Homolog (c-KIT), Mitogen-Inducible Gene-2 (MIG-2), MDM2, Tumour Protein 53 (TP53), have all been reported and compared to normal myometrium, there exists no scientific evidence to show that abnormal expression of these factors directly correlates to the initiation and promotion of human uterine LMS. PSMB9/b1i-deficient mice were reported to be prone to the development of uterine LMS, but not in their parental mice, C57BL/6 mice 12. The percentage of mice with overt tumours increased with age after six months, with a cumulative prevalence of disease in female mice of 37% by 14 months of age and no apparent plateau at this late observation time. Histopathological examinations of PSMB9/b1i-deficient uterine neoplasms revealed common characteristic abnormalities of uterine LMS. In addition, recent research reports show the loss in the IFN-g-inducible ability of PSMB9/b1i expressions in SKN cell line and other primary human uterine LMS cells established from patients. The histopathological experiments demonstrated a serious loss in the ability to induce the expression of PSMB9/b1i in human uterine LMS tissues in comparison with normal myometrium tissues located in same tissue sections (Figure 1).

Figure 1.Differential expression of PSMB9/b1i expression in human normal myometrium and several mesenchymal tumour types. (A) Immunohistchemistry of PSMB9/b1i in normal myometrium, usual leiomyoma, Bizarre leiomyoma, smooth muscle tumour of uncertain malignant potential (STUMP) and uterine leiomyosarcoma tissues located in same tissue. For all samples, 5-mm sections of tissues specimens were stained with anti- PSMB9/b1i antibody revealed by peroxidase-cojugated anti-rabbit IgG antibody. (B) IHC experiments individually performed at several medical facilities revealed a marked loss in the ability to induce PSMB9/b1i expression in human uterine LMS tissues compared to that in normal human myometrium located in the same tissue section, as well as to that in LMA tissues. Normal total: 58 cases, LMA total: 50 cases, Bizarre Leiomyoma total: 3 cases, LMS total: 56 cases. The experiments were performed three times with similar results. (C) Examinations of mRNA expression for PSMB9/b1i and b-ACTIN in normal human myometrium (Myo.), uterine usual leiomyoma (LMA), uterine leiomyosarcoma (LMS) and IFN-g-treated HeLa cells by reverse transcription-polymerase chain reaction (RT-PCR). RT-PCR was performed with the appropriate primers indicated in the materials and methods section of this manuscript. The DNA products amplified by RT-PCR were loaded in agarose gel. The expression levels of PSMB9/b1i and b-ACTIN were examined by western blotting with appropriate antibodies. The cytosolic extracts were prepared from normal human myometrium, uterine usual leiomyoma (LMA) and uterine leiomyosarcoma (LMS), and IFN-g-treated HeLa cells, and 50 mg of cytosolic extracts were resolved by 10% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE). PSMB9/b1i levels were significantly decreased in human uterine LMS compared with normal human myometrium and human usual LMA. Student’s t-test. *P < 0.005. (n = 8 per group).
 Differential expression of PSMB9/b1i expression in human normal myometrium and several mesenchymal tumour types. (A) Immunohistchemistry of PSMB9/b1i in normal myometrium, usual leiomyoma, Bizarre leiomyoma, smooth muscle tumour of uncertain malignant potential (STUMP) and uterine leiomyosarcoma tissues located in same tissue. For all samples, 5-mm sections of tissues specimens were stained with anti- PSMB9/b1i antibody revealed by peroxidase-cojugated anti-rabbit IgG antibody. (B) IHC experiments individually performed at several medical facilities revealed a marked loss in the ability to induce PSMB9/b1i expression in human uterine LMS tissues compared to that in normal human myometrium located in the same tissue section, as well as to that in LMA tissues. Normal total: 58 cases, LMA total: 50 cases, Bizarre Leiomyoma total: 3 cases, LMS total: 56 cases. The experiments were performed three times with similar results. (C) Examinations of mRNA expression for PSMB9/b1i and b-ACTIN in normal human myometrium (Myo.), uterine usual leiomyoma (LMA), uterine leiomyosarcoma (LMS) and IFN-g-treated HeLa cells by reverse transcription-polymerase chain reaction (RT-PCR). RT-PCR was performed with the appropriate primers indicated in the materials and methods section of this manuscript. The DNA products amplified by RT-PCR were loaded in agarose gel. The expression levels of PSMB9/b1i and b-ACTIN were examined by western blotting with appropriate antibodies. The cytosolic extracts were prepared from normal human myometrium, uterine usual leiomyoma (LMA) and uterine leiomyosarcoma (LMS), and IFN-g-treated HeLa cells, and 50 mg of cytosolic extracts were resolved by 10% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE). PSMB9/b1i levels were significantly decreased in human uterine LMS compared with normal human myometrium and human usual LMA. Student’s t-test. *P < 0.005. (n = 8 per group).

IFN-g treatment markedly increased the expression of PSMB9/b1i, a subunit of the proteasome, which alters the proteolytic specificity of proteasomes. Sequence analysis demonstrated that the loss of IFN- g responsiveness in the human uterine LMS cell line was attributable to the inadequate kinase activity of JAK1 due to a G781E somatic mutation in the ATP-binding region 13. The defect was localized to JAK1 activation, which acts upstream in the IFN-g signal pathway since IFN-g treatment could not strongly induce JAK1 kinase activity in human uterine LMS cell lines. Genetic alterations in tyrosine kinases have previously been firmly implicated in tumourigenesis, but only a few serine/threonine kinases are known to be mutated in human cancers 31, 32, 33, 34. For instance, mice carring homozygous deletion of Phosphatase and Tensin Homolog Deleted from Chromosome 10 (Pten) alleles developed widespread smooth muscle cell hyperplasia and abdominal LMS 35, and JUN oncogene amplification and over-expression block adipocytic differentiation in highly aggressive sarcomas 22. Most frequently, LMS have appeared in the uterus, retroperitoneum or extremities, and although histologically indistinguishable, they have different clinical courses and chemotherapeutic responses. The molecular basis for these differences remains unclear, therefore, the examination of human uterine LMS tissues 2 LMS tissue sections and normal tissue sections located in the same tissue) was performed to detect somatic mutations in the IFN-g signal molecules. In a recent report, high-resolution genomewide array comparative genomic hybrodization (CGH) analysis of LMS cases gave gene-level information about the amplified and deleted regions that may play a role in the development and progression of human uterine LMS. Among the most intriguing genes, whose copy number sequence was revealed by CGH, were loss of JAK1 (1p31-p32) and PSMB9/b1i (6p21.3) 25, 26. The discovery of these mutational defects in a key cell-signaling pathway may be an important development in the pathogenesis of human uterine LMS.

The growth of JAK1-deficient cell lines reportedly is unaffected; similarly, the cell cycle distribution pattern of freshly explanted tumour cells derived from JAK1-deficient tumours shows no response to IFN-g signaling 36. The growth of the original SKN cells, which had defective JAK1 activity, was unaffected by IFN-g treatment. In contrast, the growth of JAK1-transfected SKN cells, which had strong exogenous JAK1 activity, was prevented by IFN-g treatment. Interestingly, when PSMB9/b1i-transfected SKN cells, which have marked the expression of PSMB9/b1i, were analyzed, expression of exogenous PSMB9/b1i resulted in cell growth inhibition. Conversely, the growth of PSMB9/b1i-transfected SKN cells was unaffected by IFN-g signal pathway. Taken together, IFN-g response to cell growth inhibition may be attributable to the physiological significance of PSMB9/b1i.

The down regulation of major histocompatibility complex (MHC) expression, including the TAP1 and PSMB9/b1i genes, is one of the biological mechanisms tumour cells use to evade host immunosurveillance 37, 38, 39. Recently, the incidence of IFN-g unresponsiveness in human tumours was examined in several malignant tumours, and revealed that approximately 33% of each group exhibited a reduction in IFN-g sensitivity 40. Nevertheless, the expression of PSMB9/b1i, rather than providing an escape from immune surveillance, seems to play an important role in the negative regulation of human uterine LMS cell growth. Defective expression of PSMB9/b1i is likely to be one of the risk factors for the development of human uterine neoplasm, as it is in the PSMB9/b1i-deficient mouse. Thus, gene therapy with PSMB9/b1i expression vectors may be a new treatment for human uterine LMS that exhibits a defect in the expression of PSMB9/b1i. Because there is no effective therapy for unresectable human uterine LMS, our results may bring us to specific molecular therapies to treat this disease.

Materials and Methods

Tissue Collection

A total of 51 patients aged between 32 and 83 years who were diagnosed with smooth muscle tumours in the uterus were selected from pathological files. Serial sections were cut from at least 2 tissue blocks from each patient for hematoxylin and eosin staining and immunostaining. All tissues were used with the approval of the Ethical Committee of Shinshu University after obtaining written consent from each patient. The pathological diagnosis of human uterine mesenchymal tumours was performed using established criteria with some modifications 41, 42. Briefly, usual leiomyoma (usual LMA) was defined as a tumour showing typical histological features with a mitotic index (MI) (obtained by counting the total number of mitotic figures (MFs) in 10 high-power fields (HPFs)) of <5 MFs per 10 HPFs. Cellular leiomyoma (cellular LMA) was defined as a tumour with significantly increased cellularity (>2000 myoma cells/HPF) and a MI<5, but without cytologic atypia. Bizarre leiomyoma (BL) was defined as a tumour either with diffuse nuclear atypia and a MI<2 or with focal nuclear atypia and a MI<5 without coagulative tumour cell necrosis. A tumour of uncertain malignant potential (UMP) was defined as a tumour with no mild atypia and a MI<10, but with coagulative tumour cell necrosis. Leiomyosarcoma (LMS) was diagnosed in the presence of a MI>10 with either diffuse cytologic atypia, coagulative tumour cell necrosis, or both. Of the 113 uterine mesenchymal tumours, 52 cases were diagnosed as uterine LMA, 3 cases were Bizarre LMA, and 58 cases were uterine LMS. Protein expression studies with cervical epithelium and carcinoma tissues were performed using tissue arrays (Uterus cancer tissues, AccuMax Array, Seoul, Korea). Details regarding tissue sections are indicated in the manufacturer’s literature (AccuMax Array).

Immunohitochemistry (IHC)

Immunohistochemical staining for PSMB9/b1i, EstrogenReceptor (ER), Progesterone Receptor (PR), TP53, and Ki-67/MIB1 was performed on the serial human uterine LMS sections. Antibodies for ER(ER1D5), PR(PR10A), TP53(DO-1), and Ki-67(MIB-1) were purchased from Immunotech (Marseille, France). Anti-human PSMB9 antibody was produced by SIGMA-Aldrich collaboration Laboratory (SIGMA-Aldrich, Japan Science and Technology Agency (JST) and Shinshu University). IHC was performed using the avidin-biotin complex method previously described. Briefly, one representative 5-mm tissue section was cut from a paraffin-embedded sample of the radical hysterectomy specimen from patients with uterine LMS. Sections were deparaffinized and rehydrated in graded alcohols and then incubated with normal mouse serum for 20 min. Sections were incubated at room temperature for 1 h with primary antibody. Afterwards, sections were incubated with a biotinylated secondary antibody (Dako, Carpinteria, CA, USA) and then exposed to a streptavidin complex (Dako). Complete reaction was revealed by 3, 3¢-diaminobenzidine, and the slide was counterstained with hematoxylin. Normal USM portions in the specimens were used as positive controls. Negative controls consisted of tissue sections also incubated with normal rabbit IgG instead of the primary antibody. These studies are registered, at Shinshu University in accordance with local guidelines (approval no. M192)

Reverse Transcription-Polymerase Chain Reaction Analysis (RT-PCR)

The expressions PSMB9/b1i and b-ACTIN transcripts were examined using RT-PCR. Total RNA was prepared from human uetine LMS tissues and normal myometrim tissues using TRIzol reagent according to the manufacturer’s protocol (Invitrogen Co., CA). The RNA was reverse-transcribed with Superscript II enzyme (Invitrogen), the single strand cDNA was used to amplify. PSMB9/b1i and b-ACTIN transcripts using PCR analysis with the appropriate primer sets following a program of 35 cycles of 94°C for 30s, 60°C for 30s and 72°C for 1.5min with additional 5min for extention of transcripts 13, 43, 44. The primers used for PCR are as follows:PSMB9/b1i; 5’-GGGATAGA ACTGGAGGAACC-3’ and 5’-AGATGACACCCCCGCTTGAG-3’, b2-MICROGLOBULIN; 5’-T GAAGCTGACAGCATTCG-3’ and 5’-TGCGGCATCTTCAAACCTCC-3’, b-ACTIN; 5’-TCCGG AGACGGGGTCA-3’ and 5’-CCTGCTTGCTGATCCA-3’. These studies are registered, at Shinshu University in accordance with local guidelines (approval no. 4737, no. 150 and no. M192)

Western Blotting

Equal amounts of proteins (20 mg) were size fractionated on 7.5% SDS–polyacrylamide gel electrophoresis and transferred onto a poly-vinylidene difluoride membrane (PVDF). The blots were allowed to air dry and then placed in blocking buffer (1% BSA in 10 mM Tris buffer with 100 mM NaCl, 0.1% Tween-20, pH 7.5) for 1 h at room temperature. The blots were then incubated with specific primary antibodies for 1 h at room temperature. All the primary antibodies were mouse monoclonal or rabbit polyclonal, obtained from several industries, and were used at different final dilutions (1:1000~1:500) in the blocking buffer. These antibodies were raised using the following proteins as immunogens: PSMB9/b1i (23.4 kDa protein), PSMB8/b5i (30.3 kDa protein), b-ACTIN (41.7 kDa protein). The blots were washed three times for 30 min each with wash buffer (10 mM Tris, 100 mM NaCl, 0.1% Tween-20, pH 7.5) and then incubated with alkaliphosphatase conjugated goat-anti-mouse IgG antibody or anti-rabbit IgG antibody (Promega, Madison, WI) diluted in 5% non-fat milk in wash buffer. The PVDF membranes were washed with wash buffer three times for 30 min, and Western Blue Stabilized Substrate (Promega Co. Madison, WI) was added and incubated as previously reported 45, 46.

Sequencing of the Catalytic Domains of JAK1, STAT1, JAK2, and PSMB9/b1i Promoter Region

To demonstrate whether the somatic mutations in the ATP-binding region and kinase activation domain of the JAK1 molecule, promoter region of PSMB9/b1i gene, the Tyr701 and Ser727 aminoacid of STAT1 molecule, and the ATP-binding region and kinase activation domain of the JAK2 molecule playing in human uterine LMS were identified, the isolating of gemonic DNA and direct sequencing was carried out. The gemonic DNA was extracted from the human uterine LMS tissues and normal USM tissues using the standard protocol. The gemonic DNA was subjected to PCR procedure. The restricted DNA fragments for the direct sequence analysis were amplified by PCR according to the published sequencing oligonuclrotide primers. Polymerase chain reaction products were directly sequenced using DYEnamic Terminator Cycle sequencing Kit (Amersham-Biosciences, Piscataway, NJ, USA) by ABI Prism 3100 Genetic Analyzer (Applied Biosystem, Foster City, CA, USA). The sequences of mutant JAK1, STAT1, and promoter region of PSMB9/b1i gene derived from the individual human uterine LMS tissue sections were registered at DDBJ (Accession: AB219242, DJ055380, DJ055379, DJ055378, DJ055377, DJ055376). Details of direct sequencing are indicated in Supplementary material.

Acknowledgments

We sincerely appreciate the generous donation of PSMB9/b1i-deficient breeding mice and technical comments by Dr. Susumu Tonegawa, Massachusetts Institute of Technology. We thank Isamu Ishiwata for his generous gift of the uterine LMS cell lines. This work was supported by grants from the Ministry of Education, Culture, Science and Technology, the Japan Science and Technology Agency, the Foundation for the Promotion of Cancer Research, Kanzawa Medical Research Foundation, and The Ichiro Kanehara Foundation.

Supplementary Material

Sequencing of the Catalytic Domains of the JAK1, STAT1, JAK2 Genes and the Promoter Region of PSMB9/b1i Gene.

To determine whether somatic mutations exist in the ATP-binding region or kinase activation domain of JAK 1 and JAK2, in the promoter region of PSMB9/b1i gene at Tyr701 or Ser727 of STAT1, or in the ATP-binding region and kinase activation domain of JAK2 in human uterine LMS, genomic DNA was isolated and direct sequencing was carried out. Genomic DNA was extracted from consecutive paraffin-embedded human uterine LMS tissue and normal myometrium tissue sections using the microwave-based DNA extraction method for PCR amplification(1). To avoid contamination of normal myometrium or inflammatory cells, the tumour areas were confirmed using a hematoxylin and eosin-stained glass slide as a template. The tumour tissues were scraped by razor-micro dissection from paraffin-embedded consecutive tissue sections. The genomic DNA was subjected to PCR, and restricted DNA fragments for direct sequencing analysis were amplified using published oligonucleotide primers. PCR products were directly sequenced using a DYEnamic Terminator Cycle Sequencing Kit (Amersham-Biosciences, Piscataway, NJ) with an ABI Prism 3100 Genetic Analyzer (Applied Biosystem, Foster City, CA). The sequences of mutant JAK1, STAT1, and the promoter region of PSMB9/b1i gene derived from individual uterine LMS tissue sections are registered in the DDBJ (Accession: AB219242, DJ055380, DJ055379, DJ055378, DJ055377, DJ055376).

1. Banerjee SK, Makdisi WF, Weston AP, et al. Microwave-based DNA extraction from paraffin-embedded tissue for PCR amplification. BioTechniques 1995; 18: 768-774.

Primer Sets for Direct Sequence Analysis.

JAK1:(F, 5’-caccaaatctttaaaccggaccccagcctt-3’, R, 5’-tacgatggggcttccctgataacagcacat-3’),(F, 5’-atggcttt ctgtgctaaaatgaggagctcc-3’, R, 5’-tccatcctgctcggtcttggggtctcgaat-3’), (F, 5’-attcgagaccccaagaccgagcagga tgga-3’, R, 5’-tccactggattccaagattcccagtcacca-3’), (F, 5’-tggtgactgggaatcttggaatccagtgga-3’, R, 5’-ggcg gctcatgaggtctcccaagctgggga-3’), (F, 5’-tccccagcttgggagacctcatgagccacc-3’, R, 5’-ccgtaatggggatgccggg gtcactgagct-3’), and (F, 5’-agctcagtgaccccggcatccccattacgg-3’, R, 5’-cagatcagctatgtggttacctccactctc-3’)

JAK2:(F, 5’-cagattatgggtaatgattaaaggctccca-3’, R, 5’-cacagcatttctccaacatctgacaaccaaacc-3’), (F, 5’-ga cagtctgctaattccagctactagaa-3’, R, 5’-gcctctccctctgggcattggcataagtcc-3’), and F, 5’-atgaagcaaccgtgttga agtagacattag-3’, R, 5’-cccacgtggactataaccatgactataagacc-3’), Primer sets for the nested-PCR: (F, 5’-gaa actatttgagtttccctgtatcatttag-3’, R, 5’-ctacaagcactccttaaaatgttgtagaaag-3’),(F, 5’-gtaatttgccttgaaaactggt atttcc-3’, R, 5’-gcataagtccagatcgttaagacattgtac-3’), and (F, 5’-gaagtagacattaggaaatcatctagacg-3’, R, 5’-cactgttactgtaaatatagaaatggcaaac-3’)

STAT1: (Ser727 F, 5’-cacttattgagagctacacacaggccagcc-3’, R, 5’-ggctggggacatgagaatcccatgagctgt-3’) and (Tyr701 F, 5’-tgctgataggcagtaacacggggatctcaa-3’, R, 5’-aggaggctaagctgtct agaaacacagtag-3’) Primer sets for the nested-PCR:

(Ser727 F, 5’-ttgagagctacacacaggccagccgtggta-3’, R, 5’-gggacatgagaatcccat gagctgtacttt-3’) and (Tyr701 F, 5’-tgctgataggcagtaacacggggatctcaa-3’, R, 5’-gtctagaaacacagtagaacttt aatcccc-3’)

The promoter region of PSMB9/b1i gene: (F, 5’-cgagaagctcagccatttaggggaaagcga-3’, R, 5’-cgcccgcagc atccctgcaaggcaccgctc-3’). Primer sets for the nested-PCR: (F, 5’-aagcgaaatcgaaagcggccgcctgctcac-3’, R, 5’-ctctcctcgccgcctggggcactggtttcc-3’)

References

  1. 1.Zaloudek C, Hendrickson M R. (2002) Mesenchymal tumors of the uterus. in Kurman RJ. (ed): Blaustein‘s Pathology of the Female Genital Tract (ed 5) , New York, Springer-Verlag 561-578.
  1. 2.Gadducci A, Landoni F, Sartori E, Zola P, Maggino T et al. (1996) Uterine leiomyosarcoma:analysis of treatment failures and survival.Gynecol.Oncol. 62, 25-32.
  1. 3.Nordal R, Thoresen S. (1997) Uterine sarcomas in Norway 1956–1992: incidence, survival and mortality. , Eur. J. Cancer 33, 307-311.
  1. 4.Brooks S E, Zhan M, Cote T, Baquet C R. (2004) Surveillance,epidemiology,and end results analysis of 2677 cases of uterine sarcoma 1989–1999. , Gynecol. Oncol 93, 204-208.
  1. 5.Dusenbery K E, Potish R A, Argenta P A, Judson P L. (2005) On the apparent failure of adjuvant pelvic radiotherapy to improve survival for women with uterine sarcomas confined to the uterus. , Am. J. Clin. Oncol 28, 295-300.
  1. 6.Wu T I, Chang T C, Hsueh S, Hsu K H, Chou H H et al. (2006) Prognostic factors and impact of adjuvant chemotherapy for uterine leiomyosarcoma. , Gynecol. Oncol 100, 166-172.
  1. 7. (2004) Leitao MM,Soslow RA,Nonaka D,Olshen AB,Aghajanian C,Sabbatini P, et al. Tissue microarray immunohistochemical expression of estrogen, progesterone, and androgen receptors in uterine leiomyomata and :leiomyosarcoma. Cancer. 101, 1455-1462.
  1. 8.Perez E A, Pusztai L, Vijver M Van de. (2004) Improving patient care through molecular diagnostics. , Semin. Oncol 31, 14-20.
  1. 9.Miettinen M, Fetsch J F. (2006) Evaluation of biological potential of smooth muscle tumours. Histopathology. 48, 97-105.
  1. 10.Bodner-Adler B, Bodner K, Czerwenka K, Kimberger O, Leodolter S et al. (2005) Expression of p16 protein in patients with uterine smooth muscle tumors: an immunohistochemical analysis. Gynecol Oncol. 96(1), 62-66.
  1. 11.L Van Kaer, Ashton-Rickardt P G, Eichelberger M, Gaczynska M, Nagashima K et al. (1994) Altered peptidase and viral-specifi c T cell response in LMP2 mutant mice. Immunity. 1, 533-541.
  1. 12.Hayashi T, Faustman D L. (2002) Development of spontaneous uterine tumors in low molecular mass polypeptide-2 knockout mice. Cancer Res. 62, 24-27.
  1. 13.Hayashi T, Kobayashi Y, Kohsaka S, Sano K. (2006) The mutation in the ATPbinding region of JAK1, identified in human uterine leiomyosarcomas, results in defective interferon-gamma inducibility of TAP1 and LMP2. Oncogene. 25, 4016-4026.
  1. 14.Parmar S, Platanias L C. (2005) . , Interferons.Cancer Treat.Res 126, 45-68.
  1. 15.Platanias L C. (2005) Mechanisms of type-I- and type-II-interferon-mediated signalling. , Nature Rev. :Immunol 5, 375-386.
  1. 16.Bardelli A, Parsons D W, Silliman N, Ptak J, Szabo S et al. (2003) Mutational analysis of the tyrosine kinome in colorectal cancers. Science. 300-949.
  1. 17.Futreal P A, Coin L, Marshall M, Down T, Hubbard T et al. (2004) A census of human cancer genes. Nature Rev. Cancer. 4, 177-183.
  1. 18.Hunter C, Bignell G, Edkins S, Davies H, Teague J et al. (2004) Lung cancer: intragenic ERBB2 kinase mutations in tumours. , Nature 431, 525-526.
  1. 19.Wang Z, Shen D, Parsons D W, Bardelli A, Sager J et al. (2004) Mutational analysis of the tyrosine phosphatome in colorectal cancers. Science. 304, 1164-1166.
  1. 20.Parsons D W, Wang T L, Samuels Y, Bardelli A, Cummins J M et al. (2005) at el. Colorectal cancer: Mutations in a signalling pathway. Nature. 436-792.
  1. 21.Hernando E, Charytonowicz E, Dudas M E, Menendez S, Matushansky I et al. (2007) The AKT-mTOR. pathway plays a critical role in the development of leiomyosarcomas. Nature Med. 13, 748-753.
  1. 22.Mariani O, Brennetot C, Coindre J M, Gruel N, Ganem C et al. (2007) Oncogene Amplification and Overexpression Block Adipocytic Differentiation in Highly Aggressive Sarcomas. Cancer Cell. 11, 361-374.
  1. 23.MOTIF Search profiling.. http://motif.genome.jp
  1. 24.NCBI’s Conserved Domain Database and Search Service, v2.17 analysis. http://www.ncbi.nlm.nih.gov/ Structure/cdd/cdd.shtml
  1. 25.Larramendy M L, Kaur S, Svarvar C, Bo¨hling T, Knuutila S. (2006) Gene copy number profiling of soft-tissue leiomyo- sarcomas by array comparative genome hybridization. , Cancer Genet. Cytogen 169, 94-101.
  1. 26.Svarvar C, Larramendy M L, Blomqvist C, Gentile M, Koivisto-Korander R et al. (2006) Do DNA copy number changes differentiate uterine from nonuterine leiomyosarcomas and predict metastasis? Modern Pathol. 19, 1068-1082.
  1. 27.Hayashi T, Sano K HoriuchiA, Hiraoka N, Ichimura T, Sudo T. (2014) Potential diagnostic biomarkers: LMP2/b1i and Cyclin B1 differential expression in human uterine mesenchymal tumors. , Tumori2014 100, 509-516.
  1. 28.Kanamori T, Takakura K, Mandai M, Kariya M, Fukuhara K et al. (2003) PEP-19 overexpression in human uterine leiomyoma. , Mol. Hum. Reprod 9, 709-717.
  1. 29.Wang L, Felix J C, Lee J L, Tan P Y, Tourgeman D E et al. (2003) The proto oncogene c-kit is expressed in leiomyosarcomas of the uterus. , Gynecol. Oncol 90, 402-406.
  1. 30.Ylisaukko-oja S K, Kiuru M, Lehtonen H J, Lehtonen R, Pukkala E et al. (2006) Analysis of fumarate hydratase mutations in a population- based series of early onset uterine leiomyosarcoma patients. , Int. J. Cancer 119, 283-287.
  1. 31.Futreal P A, Coin L, Marshall M, Down T, Hubbard T et al. (2004) A census of human cancer genes. Nature Rev. Cancer. 4, 177-183.
  1. 32.Jones P A. (2007) Baylin SB. The epigenomics of cancer. , Cell 128(4), 683-692.
  1. 33.Lengyel E, Sawada K, Salgia R.Tyrosine kinase mutations in human cancer. , Curr. Mol. Med 2007, 77-84.
  1. 34.Pajares M J, Ezponda T, Catena R, Calvo A, Pio R et al. (2007) Alternative splicing: an emerging topic in molecular and clinical oncology. Lancet Oncol. 8, 349-357.
  1. 35.Post S M. (2012) Mouse models of sarcomas: critical tools in our understanding of the pathology. Clinical Sarcoma Res. 2-20.
  1. 36.Sexl V, Kovacic B, Piekorz R, Moriggl R, Stoiber D et al. (2003) Jak1 deficiency leads to enhanced Abelson-induced B-cell tumor formation. Blood. 101, 4937-4943.
  1. 37.Singal D P, Ye M, Ni J, Snider D P. (1996) Markedly decreased expression of TAP1 and LMP2 genes in HLA class I-deficient human tumor cell lines. Immunol Lett. 50, 149-154.
  1. 38.Dovhey S E, Ghosh N S, Wright K L. (2000) Loss of interferon-gamma inducibility of TAP1 and LMP2 in a renal cell carcinoma cell line. Cancer Res. 60, 5789-5796.
  1. 39.Cabrera C M, Jimenez P, Cabrera T, Esparza C, Ruiz-Cabello F et al. (2003) Total loss of MHC class I in colorectal tumors can be explained by two molecular pathways: beta2-microglobulin inactivation in MSI-positive tumors and LMP7/TAP2 downregulation in MSI-negative tumors. Tissue Antigens. 61, 211-219.
  1. 40.Kaplan D H, Shankaran V, Dighe A S, Stockert E, Aguet M et al. (1998) Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci USA 95, 7556-7561.
  1. 41.Kurma R J. (2001) . Pathology of the Female Genital Tract, 4th ed. New York,Springer-Verlag 4, 499.
  1. 42. (2003) Diagnostic Criteria for LMS, Adapted from. WHO Guidelines:World Health Organization Classification of Tumours:Pathology and Genetics,Pathology and Genetics of Tumours of the Breast and Female Genital Organs.IARC Press,France .
  1. 43.Cabrera C M, Jiménez P, Cabrera T, Esparza C, Ruiz-Cabello F et al. (2003) loss of MHC class I in colorectal tumors can be explained by two molecular pathways: beta2-microglobulin inactivation in MSI-positive tumors and LMP7/TAP2 downregulation in MSI-negative tumors.Tissue Antigens. 61(3), 211-219.
  1. 44.Miyagi T, Tatsumi T, Takehara T, Kanto T, Kuzushita N et al. (2003) Impaired expression of proteasome subunits and human leukocyte antigens class I in human colon cancer cells. J Gastroenterol Hepatol. 18(1), 32-40.
  1. 45.Hayashi T, Ueno Y, Okamoto T. (1993) Oxidoreductive Regulation of Nuclear Factor-kB, Involvement of a cellular reducing catalyst thiredoxin. , J Biol Chem 268, 11380-11388.
  1. 46.Hayashi T, Horiuchi A, Sano K, Yaegashi N, Konishi I. (2015) Uterine Leiomyosarcoma Tumorigenesis in Lmp2-deficient Mice: Involvement of Impaired Anti-oncogenic Factor IRF1. Anticancer Res. 35(9), 4665-4679.