1
|
Biological functions and therapeutic potential of SHCBP1 in human cancer. Biomed Pharmacother 2023; 160:114362. [PMID: 36739763 DOI: 10.1016/j.biopha.2023.114362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The incidence of cancer is increasing globally, and it is the most common cause of death. The identification of novel cancer diagnostic and prognostic biomarkers is important for developing cancer treatment strategies and reducing mortality. SHCSH2 domain-binding protein 1 (SHCBP1) is a protein that specifically binds to the SH2 domain of Src homology-collagen. It participates in the regulation of a variety of signal transduction pathways and can activate a variety of signaling molecules to perform a series of physiological functions. SHCBP1 is expressed in a variety of human tissues, but its abnormal expression in various systems is associated with cancer. SHCBP1 is abnormally expressed in a variety of tumors, and plays roles in almost all aspects of cancer biology (such as cell proliferation, apoptosis prevention, invasion, and metastasis) through various possible mechanisms. Its expression level is related to the clinicopathological characteristics of patients. In addition, the SHCBP1 expression pattern is closely related to cancer type, stage, and other clinical variables. Therefore, SHCBP1 is a promising tumor biomarker for cancer diagnosis and prognosis and a potential therapeutic target. This article reviews the expression, biological functions, mechanisms, and potential clinical significance of SHCBP1 in various human tumors to provide a new theoretical basis for clinical molecular diagnosis, molecular targeted therapy, and scientific research on cancer.
Collapse
|
2
|
Abstract
The function of histone deacetylase 2 (HDAC2) in transcriptional regulation and its role in oncogenesis have been well established. Here we discuss a transcription-independent HDAC2 pathway controlling cancer-related protein stability via the mouse double minute 2 homolog (MDM2) ubiquitin ligase. In synovial sarcoma, HDAC2 inactivation demonstrates significant therapeutic effect by degradation of the SS18-SSX driver oncoprotein.
Collapse
Affiliation(s)
| | - Le Su
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- CONTACT Le Su HudsonAlpha Institute for Biotechnology, 601 Genome Way, Huntsville, AL35806, USA
| |
Collapse
|
3
|
Innis SM, Cabot B. GBAF, a small BAF sub-complex with big implications: a systematic review. Epigenetics Chromatin 2020; 13:48. [PMID: 33143733 PMCID: PMC7607862 DOI: 10.1186/s13072-020-00370-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022] Open
Abstract
ATP-dependent chromatin remodeling by histone-modifying enzymes and chromatin remodeling complexes is crucial for maintaining chromatin organization and facilitating gene transcription. In the SWI/SNF family of ATP-dependent chromatin remodelers, distinct complexes such as BAF, PBAF, GBAF, esBAF and npBAF/nBAF are of particular interest regarding their implications in cellular differentiation and development, as well as in various diseases. The recently identified BAF subcomplex GBAF is no exception to this, and information is emerging linking this complex and its components to crucial events in mammalian development. Furthermore, given the essential nature of many of its subunits in maintaining effective chromatin remodeling function, it comes as no surprise that aberrant expression of GBAF complex components is associated with disease development, including neurodevelopmental disorders and numerous malignancies. It becomes clear that building upon our knowledge of GBAF and BAF complex function will be essential for advancements in both mammalian reproductive applications and the development of more effective therapeutic interventions and strategies. Here, we review the roles of the SWI/SNF chromatin remodeling subcomplex GBAF and its subunits in mammalian development and disease.
Collapse
Affiliation(s)
- Sarah M Innis
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Birgit Cabot
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
4
|
Xu N, Wu YP, Yin HB, Chen SH, Li XD, Xue XY, Gou X. SHCBP1 promotes tumor cell proliferation, migration, and invasion, and is associated with poor prostate cancer prognosis. J Cancer Res Clin Oncol 2020; 146:1953-1969. [PMID: 32447485 DOI: 10.1007/s00432-020-03247-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Prostate cancer (PCa) is an aggressive tumor. SHC SH2-domain-binding protein 1 (SHCBP1) has been identified frequently upregulated in various cancers, in addition to PCa. The aims of this study were to determine the relationships between SHCBP1 and clinicopathological characteristics of PCa and to explore the role of SHCBP1 in PCa proliferation and progression. METHODS Tissue microarray and immunohistochemistry were used to determine the prognostic significance of SHCBP1. The relationship between clinicopathological characteristics of PCa and SHCBP1 was then analyzed using Cox regression analyses. To investigate SHCBP1 functions in vitro and in vivo, we knocked down SHCBP1 in PCa cell lines and established xenograft mice models. A series of cytological function assays were utilized to determine the role of SHCBP1 in cell proliferation, migration, invasion, and apoptosis. RESULTS SHCBP1 was significantly upregulated in PCa tissues compared with BPH tissues. Patients with a higher expression of SHCBP1 were associated with poor survival outcomes than those with a lower expression of SHCBP1. Lentivirus-mediated shRNA knockdown of SHCBP1 in prostate cancer cell lines diminished cell growth, migration, and invasion dramatically both in vitro and in vivo, accompanied by an enhanced expression of large tumor suppressor 1 (LATS1) and tumor protein P53 (TP53) and inhibition of MDM2 proto-oncogene (MDM2), which suggested that SHCBP1 may promote proliferation and invasion in vitro via the LATS1-MDM2-TP53 pathway. The results of cycloheximide (CHX) and MG-132 assays indicated that SHCBP1 knockdown could attenuate the degradation of TP53 by the proteasome, prolong the half-life of TP53, and enhance the stabilization of TP53. CONCLUSION These findings suggest that SHCBP1 overexpression contributes to PCa progression and that targeting SHCBP1 might be therapeutically beneficial to patients with PCa.
Collapse
Affiliation(s)
- Ning Xu
- Departments of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Yuzhong District, Chongqing, 400016, China.,Departments of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Yu-Peng Wu
- Departments of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Hu-Bin Yin
- Departments of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Yuzhong District, Chongqing, 400016, China
| | - Shao-Hao Chen
- Departments of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiao-Dong Li
- Departments of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xue-Yi Xue
- Departments of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xin Gou
- Departments of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
5
|
Current Approaches for Personalized Therapy of Soft Tissue Sarcomas. Sarcoma 2020; 2020:6716742. [PMID: 32317857 PMCID: PMC7152984 DOI: 10.1155/2020/6716742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/27/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
Soft tissue sarcomas (STS) are a highly heterogeneous group of cancers of mesenchymal origin with diverse morphologies and clinical behaviors. While surgical resection is the standard treatment for primary STS, advanced and metastatic STS patients are not eligible for surgery. Systemic treatments, including standard chemotherapy and newer chemical agents, still play the most relevant role in the management of the disease. Discovery of specific genetic alterations in distinct STS subtypes allowed better understanding of mechanisms driving their pathogenesis and treatment optimization. This review focuses on the available targeted drugs or drug combinations based on genetic aberration involved in STS development including chromosomal translocations, oncogenic mutations, gene amplifications, and their perspectives in STS treatment. Furthermore, in this review, we discuss the possible use of chemotherapy sensitivity and resistance assays (CSRA) for the adjustment of treatment for individual patients. In summary, current trends in personalized management of advanced and metastatic STS are based on combination of both genetic testing and CSRA.
Collapse
|
6
|
Patel N, Wang J, Shiozawa K, Jones KB, Zhang Y, Prokop JW, Davenport GG, Nihira NT, Hao Z, Wong D, Brandsmeier L, Meadows SK, Sampaio AV, Werff RV, Endo M, Capecchi MR, McNagny KM, Mak TW, Nielsen TO, Underhill TM, Myers RM, Kondo T, Su L. HDAC2 Regulates Site-Specific Acetylation of MDM2 and Its Ubiquitination Signaling in Tumor Suppression. iScience 2019; 13:43-54. [PMID: 30818224 PMCID: PMC6393697 DOI: 10.1016/j.isci.2019.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/10/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylases (HDACs) are promising targets for cancer therapy, although their individual actions remain incompletely understood. Here, we identify a role for HDAC2 in the regulation of MDM2 acetylation at previously uncharacterized lysines. Upon inactivation of HDAC2, this acetylation creates a structural signal in the lysine-rich domain of MDM2 to prevent the recognition and degradation of its downstream substrate, MCL-1 ubiquitin ligase E3 (MULE). This mechanism further reveals a therapeutic connection between the MULE ubiquitin ligase function and tumor suppression. Specifically, we show that HDAC inhibitor treatment promotes the accumulation of MULE, which diminishes the t(X; 18) translocation-associated synovial sarcomagenesis by directly targeting the fusion product SS18-SSX for degradation. These results uncover a new HDAC2-dependent pathway that integrates reversible acetylation signaling to the anticancer ubiquitin response.
Collapse
Affiliation(s)
- Nikita Patel
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Juehong Wang
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Kumiko Shiozawa
- Division of Rare Cancer Research, National Cancer Center, Tokyo 104-0045, Japan
| | - Kevin B Jones
- Department of Orthopaedics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Yanfeng Zhang
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Jeremy W Prokop
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | | | - Naoe T Nihira
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Zhenyue Hao
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Derek Wong
- Biomdical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Sarah K Meadows
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Arthur V Sampaio
- Biomdical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ryan Vander Werff
- Biomdical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Makoto Endo
- Genetic Pathology Evaluation Centre, Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kelly M McNagny
- Biomdical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Torsten O Nielsen
- Genetic Pathology Evaluation Centre, Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - T Michael Underhill
- Biomdical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center, Tokyo 104-0045, Japan
| | - Le Su
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA.
| |
Collapse
|
7
|
Ishii M, Suehara Y, Sano K, Kohsaka S, Hayashi T, Kazuno S, Akaike K, Mukaihara K, Kim Y, Okubo T, Takamochi K, Takahashi F, Kaneko K, Saito T. Proteomic signatures corresponding to the SS18/SSX fusion gene in synovial sarcoma. Oncotarget 2018; 9:37509-37519. [PMID: 30680066 PMCID: PMC6331019 DOI: 10.18632/oncotarget.26493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 12/10/2018] [Indexed: 01/04/2023] Open
Abstract
Synovial sarcoma (SS) is a malignant soft tissue lesion and most commonly arises in young adults. Chromosomal translocation t(X;18)(p11;q11) results in the formation of SS18/SSX by gene fusion of the SS18 gene on chromosome 18 to either SSX1, SSX2, or SSX4 gene located on chromosome X, which is detected in more than 95% of SSs. Although multiple lines of evidence suggest that the SS18/SSX fusion is the oncogene in this tumor, the protein expression profiles associated with SS18/SSX have yet to be elucidated. In this study, we conducted proteomic studies using SS18/SSX knockdown in three SS cell lines to identify the regulated proteins associated with SS18/SSX in SS. Isobaric tags for relative and absolute quantitation (i-TRAQ) analyses identified approximate 1700–2,000 proteins regulated by the SS18/SSX fusion in each SS cell line. We also analyzed the three profiles to identify proteins that were similarly altered in all 3 cell lines and found 17 consistently upregulated and 18 consistently downregulated proteins, including TAGLN and ACTN4. In addition, network analyses identified several critical pathways including RUNX2 and SMARCA4. RUNX2 and SMARCA4 had the highest ranking in these identified pathways. In addition, we found that expression of TAGLN inhibited cell viability in SS cell lines. Our data suggest that the differentiation and cell growth of SS may be enhanced by the identified proteins induced by SS18/SSX. We believe that the findings obtained in the present functional analyses will help to improve our understanding of the relationship between SS18/SSX and malignant behavior in SS.
Collapse
Affiliation(s)
- Midori Ishii
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kei Sano
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Shinji Kohsaka
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Keisuke Akaike
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenta Mukaihara
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Youngji Kim
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Taketo Okubo
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuo Kaneko
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Peng C, Song Y, Chen W, Wang X, Liu X, Wang F, Wu D, Ma S, Wang X, Gao C. FLVCR1 promotes the proliferation and tumorigenicity of synovial sarcoma through inhibiting apoptosis and autophagy. Int J Oncol 2018. [PMID: 29532854 DOI: 10.3892/ijo.2018.4312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Feline leukemia virus subgroup C receptor 1 (FLVCR1) has been reported to have a crucial role in variety of biological processes, including cell proliferation, cell death, apoptosis, oxidative stress response, cellular differentiation and metabolism. However, little is known about its role in synovial sarcoma (SS). In the current study, FLVCR1 expression was analyzed in two SS cell lines (SW982 and HS-SY-II), and in eight SS tissues and paired adjacent non-tumor tissues using reverse transcription-quantitative polymerase chain reaction, western blot analysis and immunohistochemistry. Lentivirus-mediated short hairpin RNAs were used to knock down FLVCR1 expression in SW982 and HS-SY-II cells. The effects of FLVCR1 knockdown on the cell proliferation, clonogenicity, cell cycle and apoptosis in SS cells were evaluated by MTT, colony formation assay, flow cytometry analysis, western blotting and in vivo tumorigenesis in nude mice. In the current study, gene expression of FLVCR1 was upregulated in SS cell lines (SW982 and HS-SY-II) and SS tissues from patients. The protein levels of FLVCR1 in SS tissues were also significantly higher than in adjacent non-tumor tissues. Furthermore, suppressing the expression of FLVCR1 in SS cells using short hairpin RNA effectively attenuated cell proliferation, colony formation and impaired the cell cycle, and also significantly induced apoptosis and autophagy. In accordance with this, an in vivo tumorigenicity assay in mice demonstrated that suppression of FLVCR1 expression inhibited the growth of SS tumors implanted subcutaneously. Collectively, these results demonstrated that FLVCR1 may act as an oncoprotein, and have key roles in promoting proliferation and tumorigenicity of SS, and this may shed new light on finding novel therapeutic strategies against SS.
Collapse
Affiliation(s)
- Changliang Peng
- Department of Orthopaedics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan Song
- Department of Nephrology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Wei Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, P.R. China
| | - Xiaoying Wang
- Department of Pathology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Fang Wang
- Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Dongjin Wu
- Department of Orthopaedics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Shengzhong Ma
- Department of Orthopaedics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiuwen Wang
- Department of Orthopaedics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Chunzheng Gao
- Department of Orthopaedics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
9
|
Peng C, Zhao H, Chen W, Song Y, Wang X, Li J, Qiao Y, Wu D, Ma S, Wang X, Gao C. Identification of SHCBP1 as a novel downstream target gene of SS18-SSX1 and its functional analysis in progression of synovial sarcoma. Oncotarget 2018; 7:66822-66834. [PMID: 27572315 PMCID: PMC5341840 DOI: 10.18632/oncotarget.11651] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/21/2016] [Indexed: 01/10/2023] Open
Abstract
The SS18-SSX1 fusion gene has been shown to play important roles in the development of synovial sarcoma (SS), but the underlying molecular mechanisms and its downstream target genes are still not clear. Here SHC SH2-domain binding protein 1 (SHCBP1) was identified and validated to be a novel downstream target gene of SS18-SSX1 by using microarray assay, quantitative real-time (qPCR) and western blot. Expression of SHCBP1 was firstly confirmed in SS cell line and SS tissues. The effects of SHCBP1 overexpression or knockdown on SS cell proliferation and tumorigenicity were then studied by cell proliferation, DNA replication, colony formation, flow cytometric assays, and its in vivo tumorigenesis was determined in the nude mice. Meanwhile, the related signaling pathways of SHCBP1 were also examined in SS cells. The results indicated that SHCBP1 was significantly increased in SS cells and SS tissues compared with adjacent noncancerous tissues. The expression of SHCBP1 was demonstrated to be positively correlated with the SS18-SSX1 level. Overexpression and ablation of SHCBP1 promoted and inhibited, respectively, the proliferation and tumorigenicity of SS cells in vitro. SHCBP1 knockdown also significantly inhibited SS cell growth in nude mice, and lowered the MAPK/ERK and PI3K/AKT/mTOR signaling pathways and cyclin D1 expression. Our findings disclose that SHCBP1 is a novel downstream target gene of SS18-SSX1, and demonstrate that the oncogene SS18-SSX1 promotes tumorigenesis by increasing the expression of SHCBP1, which normally acts as a tumor promoting factor.
Collapse
Affiliation(s)
- Changliang Peng
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Hui Zhao
- Department of Orthopaedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wei Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yan Song
- Nephrology Research Institute, Shandong University Second Hospital, Jinan, China
| | - Xiaoying Wang
- Department of Pathology, Shandong University Second Hospital, Jinan, China
| | - Ji Li
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Yong Qiao
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Dongjin Wu
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Shengzhong Ma
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Xiuwen Wang
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| | - Chunzheng Gao
- Department of Orthopaedics, Shandong University Second Hospital, Jinan, China
| |
Collapse
|
10
|
Vlenterie M, Hillebrandt-Roeffen MHS, Schaars EWM, Flucke UE, Fleuren EDG, Navis AC, Leenders WPJ, Versleijen-Jonkers YMH, van der Graaf WTA. Targeting Cyclin-Dependent Kinases in Synovial Sarcoma: Palbociclib as a Potential Treatment for Synovial Sarcoma Patients. Ann Surg Oncol 2016; 23:2745-52. [PMID: 27334220 PMCID: PMC4972869 DOI: 10.1245/s10434-016-5341-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Indexed: 11/18/2022]
Abstract
Background In synovial sarcomas alterations in the cyclin D1-CDK4/6-Rb axis have been described. Also, β-catenin, a cyclin D1 regulator, is often overexpressed. Additionally, studies have shown that the t(X;18) translocation influences tumor behavior partly through cyclin D1 activation. We investigated how alterations in the cyclin D1-CDK4/6-Rb axis impact prognosis and studied effects of targeting this axis with the CDK4/6 inhibitor palbociclib. Methods Synovial sarcoma samples (n = 43) were immunohistochemically stained for β-catenin, cyclin D1, p16, p21, p27, Rb, and phospho-Rb. Fluorescent in situ hybridization (FISH) was performed to detect CCND1 amplification or translocation. In 4 synovial sarcoma cell lines sensitivity to palbociclib was investigated using cell viability assays, and effects on the sensitive cell lines were evaluated on protein level and by cell cycle arrest. Results Expression of nuclear phospho-Rb and nuclear β-catenin in the patient samples was associated with poor survival. FISH showed a sporadic translocation of CCND1 in a subset of tumors. An 8-fold CCND1 amplification was found in 1 cell line, but not in the patient samples investigated. Palbociclib effectively inhibited Rb-phosphorylation in 3 cell lines, resulting in an induction of a G1 arrest and proliferation block. Conclusions In this series nuclear phospho-Rb and nuclear β-catenin expression were negative prognostic factors. In vitro data suggest that palbociclib may be a potential treatment for a subset of synovial sarcoma patients. Whether this effect can be enhanced by combination treatment deserves further preclinical investigations. Electronic supplementary material The online version of this article (doi:10.1245/s10434-016-5341-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Myrella Vlenterie
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | - Esther W M Schaars
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Uta E Flucke
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emmy D G Fleuren
- The Institute of Cancer Research London and the Royal Marsden NHS Foundation Trust, London, UK
| | - Anna C Navis
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,The Institute of Cancer Research London and the Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
11
|
Carmody Soni EE, Schlottman S, Erkizan HV, Uren A, Toretsky JA. Loss of SS18-SSX1 inhibits viability and induces apoptosis in synovial sarcoma. Clin Orthop Relat Res 2014; 472:874-82. [PMID: 23716114 PMCID: PMC3916608 DOI: 10.1007/s11999-013-3065-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Most synovial sarcomas contain a chromosomal translocation t(X;18), which results in the formation of an oncoprotein SS18-SSX critical to the viability of synovial sarcoma. QUESTIONS/PURPOSES We (1) established and characterized three novel synovial sarcoma cell lines and asked (2) whether inhibition of SS18-SSX1 decreases cell viability in these cell lines; and (3) whether reduction in viability after SS18-SSX1 knockdown is caused by apoptosis. After identifying a specific posttranscriptional splice variant in our cell lines, we asked (4) whether this provides a survival benefit in synovial sarcoma. METHODS Cells lines were characterized. SS18-SSX1 knockdown was achieved using a shRNA system. Cell viability was assessed by WST-1 analysis and apoptosis examined by caspase-3 activity. RESULTS We confirmed the SS18-SSX1 translocation in all cell lines and identified a consistent splicing variant. We achieved successful knockdown of SS18-SSX1 and with this saw a significant reduction in cell viability. Decreased viability was a result of increased apoptosis. Reintroduction of the exon 8 sequence into cells reduced cell viability in all cell lines. CONCLUSIONS We confirmed the presence of the SS18-SSX1 translocation in our cell lines and its importance in the survival of synovial sarcoma. We have also demonstrated that reduction in cell viability is related to an increase in apoptosis. In addition, we have identified a potential mediator of SS18-SSX function in exon 8. CLINICAL RELEVANCE SS18-SSX represents a tumor-specific target in synovial sarcoma. Exploitation of SS18-SSX and its protein partners will allow us to develop potent tumor-specific therapeutic agents.
Collapse
Affiliation(s)
- Emily E. Carmody Soni
- MedStar Georgetown Orthopaedic Institute, 110 Irving Street, NW C-2173, Washington, DC 20010 USA ,Department of Oncology, Lombardi Comprehensive Cancer Institute, Washington, DC USA
| | - Silke Schlottman
- Department of Oncology, Lombardi Comprehensive Cancer Institute, Washington, DC USA
| | - Hayriye V. Erkizan
- Department of Oncology, Lombardi Comprehensive Cancer Institute, Washington, DC USA
| | - Aykut Uren
- Department of Oncology, Lombardi Comprehensive Cancer Institute, Washington, DC USA
| | - Jeffrey A. Toretsky
- Department of Oncology, Lombardi Comprehensive Cancer Institute, Washington, DC USA
| |
Collapse
|
12
|
The clinical implication of SS18-SSX fusion gene in synovial sarcoma. Br J Cancer 2013; 109:2279-85. [PMID: 24022186 PMCID: PMC3798954 DOI: 10.1038/bjc.2013.547] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/16/2013] [Accepted: 08/16/2013] [Indexed: 02/07/2023] Open
Abstract
Background: The aim of this study is to evaluate distribution and clinical impact of the SS18-SSX fusion gene in patients with synovial sarcoma in China. Methods: We collected and analysed the clinical data of 88 patients using univariate and multivariate survival analysis. HEK 293T and NIH 3T3 cell lines were transfected with the SS18-SSX1 or SS18-SSX2 gene to determine the respective involvement of these fusion genes in cell proliferation and invasion. Results: Overall survival was significantly better among SS18-SSX2 cases (P=0.001), FNCLCC grade 2 cases (P<0.001), and UICC stage 1 or 2 (P<0.001) by univariate and multivariate survival analysis. SS18-SSX1-positive cells were more proliferative and invasive than SS18-SSX2-positive cells. Conclusion: SS18-SSX fusion type is a significant prognostic factor for patients with synovial sarcoma.
Collapse
|
13
|
Molecular detection and targeting of EWSR1 fusion transcripts in soft tissue tumors. Med Oncol 2013; 30:412. [PMID: 23329308 PMCID: PMC3586390 DOI: 10.1007/s12032-012-0412-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 11/30/2012] [Indexed: 12/11/2022]
Abstract
Soft tissue tumors are a heterogeneous group of tumors, traditionally classified according to morphology and histogenesis. Molecular classification divides sarcomas into two main categories: (a) sarcomas with specific genetic alterations and (b) sarcomas showing multiple complex karyotypic abnormalities without any specific pattern. Most chromosomal alterations are represented by translocations which are increasingly detected. The identification of fusion transcripts, in fact, not only support the diagnosis but also provides the basis for the development of new therapeutic strategies aimed at blocking aberrant activity of the chimeric proteins. One of the genes most susceptible to breakage/translocation in soft tissue tumors is represented by Ewing sarcoma breakpoint region 1 (EWSR1). This gene has a large number of fusion partners, mainly associated with the pathogenesis of Ewing's sarcoma but with other soft tissue tumors too. In this review, we illustrate the characteristics of this gene/protein, both in normal cellular physiology and in carcinogenesis. We describe the different fusion partners of EWSR1, the molecular pathways in which is involved and the main molecular biology techniques for the identification of fusion transcripts and for their inhibition.
Collapse
|
14
|
Downstream and intermediate interactions of synovial sarcoma-associated fusion oncoproteins and their implication for targeted therapy. Sarcoma 2012; 2012:249219. [PMID: 22550415 PMCID: PMC3329658 DOI: 10.1155/2012/249219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022] Open
Abstract
Synovial sarcoma (SS), an aggressive type of soft tissue tumor, occurs mostly in adolescents and young adults. The origin and molecular mechanism of the development of SS remain only partially known. Over 90% of SS cases are characterized by the t(X;18)(p11.2;q11.2) translocation, which results mainly in the formation of
SS18-SSX1 or SS18-SSX2 fusion genes. In recent years, several reports describing direct and indirect interactions of SS18-SSX1/SSX2 oncoproteins have been published. These reports suggest that the fusion proteins particularly affect the cell growth, cell proliferation, TP53 pathway, and chromatin remodeling mechanisms, contributing to SS oncogenesis. Additional research efforts are required to fully explore the protein-protein interactions of SS18-SSX oncoproteins and the pathways that are regulated by these partnerships for the development of effective targeted therapy.
Collapse
|
15
|
Kekeeva TV, Ryazantseva AA, Zavalishina LE, Andreeva YY, Babenko OV, Zaletaev DV, Frank GA. Analysis of SYT/SSX1 and SYT/SSX2 fusion genes in synovial sarcoma. Mol Biol 2011. [DOI: 10.1134/s0026893311040078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|