1
|
Bacchetti R, Yuan S, Rainero E. ADAMTS Proteases: Their Multifaceted Role in the Regulation of Cancer Metastasis. DISEASES & RESEARCH 2024; 4:40-52. [PMID: 38948119 PMCID: PMC7616120 DOI: 10.54457/dr.202401004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Cancer leads to nearly 10 million deaths worldwide per year. The tumour microenvironment (TME) is fundamental for tumour growth and progression. A key component of the TME, the extracellular matrix (ECM) has recently become a focus of interest in cancer research. Dysregulation of ECM synthesis and proteolysis leads to uncontrolled tumour growth and metastasis. Matrix remodelling enzymes, secreted by cancer cells and stromal cells, modify the overall structure and organisation of ECM proteins, therefore influencing biochemical interactions, tissue integrity and tissue turnover. While A Disintegrin and Metalloproteinases (ADAMs)' and matrix metalloproteinases' role in cancer has been deeply investigated, other proteolytic enzymes, like ADAMs with thrombospondin(-like) motifs (ADAMTSs) have been gaining interest due to their roles in modulating cancer cell-ECM interactions and oncogenic signalling pathways. In this review, we will discuss the dysregulation of ADAMTSs in cancer and their roles in regulating cancer development and progression, via ECM remodelling and cell signalling modulation.
Collapse
Affiliation(s)
- Rachele Bacchetti
- School of Biosciences, Department of Biomedical science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Shengnan Yuan
- School of Biosciences, Department of Biomedical science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Elena Rainero
- School of Biosciences, Department of Biomedical science, Firth Court, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
2
|
Xu B, Zhang J, Ye L, Yuan C. Chinese herbal compound SanHuang decoction reverses axitinib resistance in ccRCC through regulating immune cell infiltration by affecting ADAMTS18 expression. Am J Cancer Res 2023; 13:2841-2860. [PMID: 37560000 PMCID: PMC10408491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/20/2023] [Indexed: 08/11/2023] Open
Abstract
This investigation aims to study the reversal effect of the Chinese herbal compound SanHuang decoction on axitinib resistance in clear cell renal cell carcinoma (ccRCC) cells and its mechanistic role by employing cellular and mouse models. Axitinib-resistant ccRCC cell lines (A498-DR and 786-O-DR) were cultured and treated with SanHuang decoction. The apoptosis and migration of tumor cells were observed by flow cytometry and wound healing assays, respectively, and the expression of a disintegrin-like and metalloprotease with thrombospondin type 1 motif 18 (ADAMTS18) was evaluated by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting (WB). In addition, A498-DR cells were inoculated into mice to establish tumorigenic models, and the models were treated with normal saline, axitinib, or different concentrations of SanHuang decoction plus axitinib. Then, the tumor diameter in each group was measured, and the expression of ADAMTS18 was evaluated by RT-PCR, WB and immunohistochemistry. In addition, the distribution of T cells (CD45+, CD4+, CD8+) and PD-L1 expression was analyzed by flow cytometry to evaluate the level of immune cell infiltration. SanHuang decoction significantly reduced the proliferative activity of axitinib-resistant tumor cells and enhanced the sensitivity of tumors to axitinib in vitro (cell lines) and in mice. In the SanHuang decoction group, the expression level of ADAMTS18 was increased to some extent, and several phenomena were observed, including (1) subcutaneous transplanted tumors grew slower, (2) the CD45+/PD-L1 ratio was decreased and (3) the proportions of CD8+ and CD4+ T cells were increased. Overexpression of ADAMTS18 was synergistic with SanHuang decoction treatment to jointly improve tumor immune infiltration and inhibit immune escape. Pearson correlation analysis of sample data showed that there was a negative correlation between the expression of ADAMTS18 and PD-L1 in tumor tissues. In conclusion, the Chinese herbal compound SanHuang decoction can reverse axitinib resistance in ccRCC cells by regulating immune cell infiltration and affecting ADAMTS18 expression.
Collapse
Affiliation(s)
- Ben Xu
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center Beijing, China
| | - Jiaen Zhang
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center Beijing, China
| | - Lin Ye
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center Beijing, China
| | - Changwei Yuan
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center Beijing, China
| |
Collapse
|
3
|
Zhu S, Xu H, Chen R, Shen Q, Yang D, Peng H, Tong J, Fu Q. DNA methylation and miR-92a-3p-mediated repression of HIP1R promotes pancreatic cancer progression by activating the PI3K/AKT pathway. J Cell Mol Med 2023; 27:788-802. [PMID: 36811277 PMCID: PMC10002968 DOI: 10.1111/jcmm.17612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/27/2022] [Accepted: 10/15/2022] [Indexed: 02/24/2023] Open
Abstract
Pancreatic cancer (PAAD) is a highly malignant tumour characterized of high mortality and poor prognosis. Huntingtin-interacting protein 1-related (HIP1R) has been recognized as a tumour suppressor in gastric cancer, while its biological function in PAAD remains to be elucidated. In this study, we reported the downregulation of HIP1R in PAAD tissues and cell lines, and the overexpression of HIP1R suppressed the proliferation, migration and invasion of PAAD cells, while silencing HIP1R showed the opposite effects. DNA methylation analysis revealed that the promoter region of HIP1R was heavily methylated in PAAD cell lines when compared to the normal pancreatic duct epithelial cells. A DNA methylation inhibitor 5-AZA increased the expression of HIP1R in PAAD cells. 5-AZA treatment also inhibited the proliferation, migration and invasion, and induced apoptosis in PAAD cell lines, which could be attenuated by HIP1R silencing. We further demonstrated that HIP1R was negatively regulated by miR-92a-3p, which modulates the malignant phenotype of PAAD cells in vitro and the tumorigenesis in vivo. The miR-92a-3p/HIP1R axis could regulate PI3K/AKT pathway in PAAD cells. Taken together, our data suggest that targeting DNA methylation and miR-92a-3p-mediated repression of HIP1R could serve as novel therapeutic strategies for PAAD treatment.
Collapse
Affiliation(s)
- Sixian Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huiting Xu
- Department of Abdominal Oncology, Hubei Cancer HospitalWuhanChina
| | - Runzhi Chen
- Department of Abdominal Oncology, Hubei Cancer HospitalWuhanChina
| | - Qian Shen
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dongmei Yang
- Department of Abdominal Oncology, Hubei Cancer HospitalWuhanChina
| | - Hui Peng
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jin Tong
- Department of PICC, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qiang Fu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
4
|
Curcumin Inhibits Proliferation of Renal Cell Carcinoma in vitro and in vivo by Regulating miR-148/ADAMTS18 through Suppressing Autophagy. Chin J Integr Med 2022:10.1007/s11655-022-3690-9. [PMID: 36477451 DOI: 10.1007/s11655-022-3690-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To explore the effect of curcumin on the proliferation of renal cell carcinoma and analyze its regulation mechanism. METHODS In RCC cell lines of A498 and 786-O, the effects of curcumin (2.5, 5, 10 µ mo/L) on the proliferation were analyzed by Annexin V+PI staining. Besides, A498 was inoculated into nude mice to establish tumorigenic models, and the model mice were treated with different concentrations of curcumin (100, 200, and 400 mg/kg), once daily for 30 days. Then the tumor diameter was measured, the tumor cells were observed by hematoxylin-eosin staining, and the protein expressions of miR-148 and ADAMTS18 were detected by immunohistochemistry. In vitro, after transfection of miR-148 mimics, miR-148 inhibitor or si-ADAMTS18 in cell lines, the expression of ADAMTS18 was examined by Western blotting and the cell survival rate was analyzed using MTT. Subsequently, Western blot analysis was again used to examine the autophagy phenomenon by measuring the relative expression level of LC3-II/LC3-I; autophagy-associated genes, including those of Beclin-1 and ATG5, were also examined when miR-148 was silenced in both cell lines with curcumin treatment. RESULTS Curcumin could inhibit the proliferation of RCC in cell lines and nude mice. The expression of miR-148 and ADAMTS18 was upregulated after curcumin treatment both in vitro and in vivo (P<0.05). The cell survival rate was dramatically declined upon miR-148 or ADAMTS18 upregulated. However, si-ADAMTS18 treatment or miR-148 inhibitor reversed these results, that is, both of them promoted the cell survival rate. CONCLUSION Curcumin can inhibit the proliferation of renal cell carcinoma by regulating the miR-148/ ADAMTS18 axis through the suppression of autophagy in vitro and in vivo. There may exist a positive feedback loop between miR-148 and ADAMTS18 gene in RCC.
Collapse
|
5
|
Chen Z, Li Z, Li C, Li B, Wang H, Nong D, Li X, Huang G, Lin J, Li W. Speckle-type POZ protein could play a potential inhibitory role in human renal cell carcinoma. BMC Cancer 2022; 22:1277. [PMID: 36474188 PMCID: PMC9727862 DOI: 10.1186/s12885-022-10340-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Speckle-type POZ protein(SPOP), a substrate adaptor of Cul3 ubiquitin ligase, plays crucial roles in solid neoplasms by promoting the ubiquitination and degradation of substrates. Limited studies have shown that SPOP is overexpressed in human renal cell carcinoma (RCC) tissue. However, the exact role of SPOP in RCC remains unclear and needs to be further elucidated. The present study showed that SPOP was expressed at different levels in different RCC cell lines. The purpose of this study was to explore the roles of SPOP in the biological features of RCC cells and the expression levels of SPOP in human tissue microarray (TMA) and kidney tissues. METHODS Here, SPOP was overexpressed by lentiviral vector transfection in ACHN and Caki-1 cells, and SPOP was knocked down in Caki-2 cells with similar transfection methods. The transfection efficiency was evaluated by quantitative PCR and western blotting analyses. The role of SPOP in the proliferation, migration, invasion and apoptosis of cell lines was determined by the MTT, wound-healing, transwell and flow cytometry assays. Moreover, the cells were treated with different drug concentrations in proliferation and apoptosis assays to investigate the effect of sunitinib and IFN-α2b on the proliferation and apoptosis of SPOP-overexpressing cells and SPOP-knockdown RCC cells. Finally, immunohistochemical staining of SPOP was performed in kidney tissues and TMAs, which included RCC tissues and corresponding adjacent normal tissues. RESULTS Overexpression of SPOP inhibited cell proliferation, migration and invasion and increased cell apoptosis. Interestingly, sunitinib and IFN-α2b at several concentrations increased the proliferation inhibitory rate and total apoptosis rate of cells overexpressing SPOP. The findings of the present study showed that the SPOP protein was significantly expressed at low levels in most clear cell RCC (ccRCC) tissues and at relatively high levels in the majority of adjacent normal tissues and kidney tissues. Kaplan-Meier survival analysis showed that there was no statistically significant difference in cumulative survival based on the data of different SPOP expression levels in TMA and patients. CONCLUSIONS In contrast to previous studies, our findings demonstrated that overexpression of SPOP might suppress the progression of RCC cells, which was supported by cell experiments and immunohistochemical staining. SPOP could be a potential tumour inhibitor in RCC.
Collapse
Affiliation(s)
- Zhi Chen
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Zuan Li
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Chunlin Li
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Bingcai Li
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Haojian Wang
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Deyong Nong
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Ximing Li
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Guihai Huang
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Junhao Lin
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| | - Wei Li
- grid.410652.40000 0004 6003 7358Department of Urology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy Of Medical Sciences, Nanning, China
| |
Collapse
|
6
|
Xiao W, Tang P, Sui Z, Han Y, Zhao G, Wu X, Yang Y, Zhu N, Gong L, Yu Z, Zhang H. Establishment of a risk model by integrating hypoxia genes in predicting prognosis of esophageal squamous cell carcinoma. Cancer Med 2022; 12:2117-2133. [PMID: 35789548 PMCID: PMC9883439 DOI: 10.1002/cam4.5002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/03/2022] [Accepted: 06/11/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) has a dismal prognosis, and hypoxia plays a key role in metastasis and proliferation of ESCC. Thus, we aimed to develop a hypoxia-based gene signature to assist in the treatment decisions and prognosis. METHODS We performed consensus clustering analysis on samples from GSE53625 dataset from the Gene Expression Omnibus (GEO) database and used weighted gene co-expression network analysis to filter out candidate modules, which were then intersected with differentially expressed genes from clustered subgroups to obtain hypoxia-related genes (HRGs). After that, the aforementioned genes were used to construct risk score models and validated in The Cancer Genome Atlas (TCGA) database and Cox regression analysis were used to construct a nomogram. Immunohistochemical was used to detect protein expression levels of relevant genes. Moreover, the relationship between risk scores and tumor microenvironment was explored. RESULTS A hypoxia risk model containing six genes (PNPLA1, CARD18, IL-18, SLC37A2, ADAMTS18, and FAM83C) was constructed by screening key HRGs. Poorer prognosis in the high-risk group than in the low-risk group. And Cox regression analysis showed that risk score was an independent prognostic factor. The nomogram based on risk scores could well predict 1-, 3-, and 5-year survival. P53, Wnt, and hypoxia signaling pathways may be some regulatory mechanisms of hypoxia associated with the tumor microenvironment. In addition, we confirmed the high expression of BGN and low expression of IL-18 in ESCC tissues. CONCLUSIONS Our study determined the prognostic value of a 6-hypoxia gene signature and a prognostic model, providing potential prognostic predictors and therapeutic targets for ESCC.
Collapse
Affiliation(s)
- Wanyi Xiao
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Peng Tang
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Zhilin Sui
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina,Department of Thoracic SurgeryNational Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and PeKing Union Medical CollegeShenzhenChina
| | - Youming Han
- Department of Respiratory MedicineBinhai Hospital of Tianjin Medical University General HospitalTianjinChina
| | - Gang Zhao
- Department of Gastrointestinal Cancer BiologyTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Xianxian Wu
- Department of Thoracic SurgeryNational Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and PeKing Union Medical CollegeShenzhenChina
| | - Yueyang Yang
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Ningning Zhu
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Lei Gong
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| | - Zhentao Yu
- Department of Thoracic SurgeryNational Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and PeKing Union Medical CollegeShenzhenChina
| | - Hongdian Zhang
- Department of Esophageal CancerTianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for CancerTianjinChina
| |
Collapse
|
7
|
Xu B, Zhu WJ, Peng YJ, Cheng SD. Curcumin reverses the sunitinib resistance in clear cell renal cell carcinoma (ccRCC) through the induction of ferroptosis via the ADAMTS18 gene. Transl Cancer Res 2022; 10:3158-3167. [PMID: 35116623 PMCID: PMC8797884 DOI: 10.21037/tcr-21-227] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/25/2021] [Indexed: 01/22/2023]
Abstract
Background To explore the possible mechanism by which curcumin reverses the sunitinib resistance in clear cell renal cell carcinoma (ccRCC). Methods A sunitinib-resistant ccRCC cell model was established. The MTT assay was used to determine the half maximal inhibitory concentration (IC50) and drug resistance (DR) index. The effects of curcumin plus sunitinib or sunitinib alone on drug-resistant cell lines were verified by the cell counting kit-8 (CCK-8)assay, colony formation assay, and apoptosis assay. The concentration of iron ions in the cell lines was analyzed using an Abcam Iron Assay Kit. The expressions of ADAMTS18 gene and ferroptosis-related proteins (NCOA4, FTH1 and p53) after curcumin plus sunitinib treatment were analyzed by reverse transcription polymerase chain reaction (RT-PCR) and Western blotting. After transfection of curcumin plus sunitinib/sunitinib alone-treated drug-resistant cell lines with si-ADAMTS18, cell proliferation activity was assessed by the CCK-8 assay, and the protein expression levels of ADAMTS18, NCOA1, FTH1 and p53 were analyzed by Western blotting. After treatment with ferroptosis-1 (Fer-1; a ferroptosis inhibitor), the cell proliferation activity of drug-resistant cell lines treated with curcumin plus sunitinib/sunitinib alone was reassessed using the CCK-8 assay. Results Curcumin plus sunitinib inhibited the proliferation of sunitinib-resistant ccRCC cells (P<0.05). Curcumin significantly decreased the concentration of iron ions and increased the expression of ADAMTS18 gene, while significantly inhibited ferroptosis-related protein expression (P<0.05). After silencing the ADAMTS18 gene, there was no significant difference in cell proliferation or ferroptosis-related protein expression between curcumin plus sunitinib and sunitinib-treated drug-resistant cell lines (P>0.05). Ferroptosis inhibitors reversed the inhibitory effect of curcumin on sunitinib-resistant ccRCC cell lines. Conclusions Curcumin can reverse the sunitinib resistance in ccRCC, possibly by upregulating the expression of the ADAMTS18 gene to induce ferroptosis.
Collapse
Affiliation(s)
- Ben Xu
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Wei-Jie Zhu
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Yi-Ji Peng
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Si-Da Cheng
- Department of Urology, Peking University First Hospital and Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| |
Collapse
|
8
|
Wu G, Li J, Xu Y, Che X, Chen F, Wang Q. A New Survival Model Based on ADAMTSs for Prognostic Prediction in Clear Cell Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:2606213. [PMID: 34603444 PMCID: PMC8486512 DOI: 10.1155/2021/2606213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022]
Abstract
The main purpose of this study was to explore the genetic variation, gene expression, and clinical significance of ADAMTSs (a disintegrin and metalloprotease domains with thrombospondin motifs) across cancer types. Analysis of data from the TCGA (The Cancer Genome Atlas) database showed that the ADAMTSs have extensive CNV (copy number variation) and SNV (single nucleotide variation) across cancer types. Compared with normal tissues, the methylation of ADAMTSs in cancer tissues is also significantly different, which affects the expression of ADAMTS gene and the prognosis of cancer patients. Through gene expression analysis, we found that ADAMTS family has significant changes in gene expression across cancer types and is closely related to the prognosis of carcinoma, especially in ccRCC (clear cell renal cell carcinoma). LASSO regression analysis was used to establish a prognostic model based on the ADAMTSs to judge the prognosis of patients with ccRCC. Multiple Cox regression analysis suggested that age, grade, stage, and risk score of the prognostic model of ccRCC were independent prognostic factors in patients with renal clear cell carcinoma. These findings indicate that the ADAMTSs-based survival model can accurately predict the prognosis of patients with ccRCC and suggest that ADAMTSs are a potential prognostic biomarker and therapeutic target in ccRCC.
Collapse
Affiliation(s)
- Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jianyi Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory on Organ Donation and Transplant Immunology, Guangzhou, China
| | - Yingkun Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
9
|
Curcumin Inhibits Viability of Clear Cell Renal Cell Carcinoma by Down-Regulating ADAMTS18 Gene Methylation though NF-κ B and AKT Signaling Pathway. Chin J Integr Med 2021; 28:419-424. [PMID: 33997938 DOI: 10.1007/s11655-021-3445-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the effect of curcumin on viability of clear cell renal cell carcinoma (ccRCC) and analyze its possible mechanism. METHODS In cell lines of A498 and 786-O, the effects of curcumin (1.25, 2.5, 5 and 10 μ mol/L) on the viability of ccRCC were analyzed at 24, 48 and 72 h by MTT assay. The protein expression levels of ADAMTS18 gene, p65, phosphorylation p65 (pp65), AKT, phosphorylation AKT (pAKT) and matrix metallopeptidase 2 (MMP-2) before and after curcumin (10 μ mol/L) treatment were examined by Western blotting. Real-time PCR and methylation specific PCR (MSP) were applied to analyze the expression and methylation level of ADAMTS18 gene before and after curcumin treatment (10 μ mol/L). RESULTS Curcumin significantly inhibited the viability of A498 and 786-O cell lines in a dose- and time-dependent manner (P<0.01). Up-regulation of ADAMTS18 gene expression with down-regulation of ADAMTS18 gene methylation was reflected after curcumin treatment, accompanied by down-regulation of nuclear factor κ B (NF-κ kB) related protein (p65 and pp65), AKT related protein (AKT and pAKT), and NF-κ B/AKT common related protein MMP-2. With ADAMTS18 gene overexpressed, the expression levels of p65, AKT and MMP2 were downregulated, of which were conversely up-regulated in silenced ADAMTS18 (sh-ADAMTS18). The expression of pp65, pAKT and MMP2 in sh-ADAMTS18 was down-regulated after being treated with PDTC (NF-κ B inhibitor) and LY294002 (AKT inhibitor). CONCLUSIONS Curcumin could inhibit the viability of ccRCC by down-regulating ADAMTS18 gene methylation though NF-κ B and AKT signaling pathway.
Collapse
|
10
|
Diez-Calzadilla NA, Noguera Salvá R, Soriano Sarrió P, Martínez-Jabaloyas JM. Genetic profile and immunohistochemical study of clear cell renal carcinoma: Pathological-anatomical correlation and prognosis. Cancer Treat Res Commun 2021; 27:100374. [PMID: 33932757 DOI: 10.1016/j.ctarc.2021.100374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) accounts for 2-3% of all tumors being the most frequent solid lesion in the kidney. OBJECTIVE To determine what genetic alterations and immunohistochemical (IHC) of clear cell renal carcinoma (ccRCC) are associated with prognosis and tumor aggressiveness. PATIENTS AND METHODS Experimental analytical study with 57 patients who underwent radical and partial nephrectomy between 2005 and 2011, all with diagnosis of ccRCC and minimum post-operative follow-up of 36 months. The pathological study included IHC determination of biomarkers associated (CAIX, CAM 5.2, CD10, c-erbB-2, EGFR, HIF-1a, Ki67, MDM2, PAX-2 y 8, p53, survivin and VEGFR 1 and 2). Genetic analysis was carried out using multiplex ligation-dependent probe amplification (MLPA). Clinical data were collected and summarized using an access-type database, adding genetic analysis and IHC data of each patient's tumor sample. IHC statistical analysis included Chi-square, Kruskal-Wallis and multivariate analysis. The genetic analysis was performed using multivariate logistic regression (normal/deletion-duplication). Significance level p<0.05. RESULTS Pathologic stage was: pT1 (61.8%), pT2 (32.7%); pT3-T4 (5.4%); 16.3% were pN+ and 19.3% M1. 23.6% recurred being predominantly to distance in 83.3%. 27.3% of patients died (73.3% ccCCR). CAIX (Carbonic anhydrase IX) and tumor size were associated with worse Fuhrman grade (p = 0.035; p = 0.001 respectively). Deletion-duplication of genes increased the likelihood: of death (APC, Bcl-2 and CDKN2A by 11, 7 and 4 respectively and SMAD4 reduced the probability by 88%); tumor recurrence (CDKN2A by fifteen fold and VHL reduced the probability by 87%); pT greater than 2 (CCND2, MDM2 and WT1 multiplied by 6, 7 and 9); risk of N+ (CDK4 and EBF1 by 13); distant metastases (BRCA2 and DLEU1 by 5); Fuhrman grade ≥3 (BRCA1, BRCA2 and p53 by 40, 75 and 34 respectively, while that FHIT reduced by 96%). Deletion-duplication of CDK4 and DCC increased survival by a factor of 13 and 16, while that DLEU1 and RUNX1 decreased survival time by 80%. CONCLUSION CAIX and tumor size are associated with increased aggressiveness. The mutations to level 5q, 9p, 11p, 12, 13q, 17, 18q and 21q are associated with more aggressive tumors and with worse survival rate.
Collapse
Affiliation(s)
- N A Diez-Calzadilla
- Department of Urology. Hospital de Sagunto, Av. Ramón y Cajal, s/n, 46520. Sagunto, Valencia-España.
| | - R Noguera Salvá
- Department of Pathology. Faculty of Medicine. University of Valencia, Av. Blasco Ibáñez, 13. 46010. Valencia-España
| | - P Soriano Sarrió
- Department of Pathology. Faculty of Medicine. University of Valencia, Av. Blasco Ibáñez, 13. 46010. Valencia-España
| | - J M Martínez-Jabaloyas
- Department of Urology. Hospital Clínico Universitario de Valencia, Av. Blasco Ibáñez, 17, 46010. Valencia- España.
| |
Collapse
|
11
|
Diagnostic DNA Methylation Biomarkers for Renal Cell Carcinoma: A Systematic Review. Eur Urol Oncol 2021; 4:215-226. [DOI: 10.1016/j.euo.2019.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/23/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023]
|
12
|
Xu B, Peng YJ, Ma BL, Cheng SD. Aberrant methylation of the 16q23.1 tumor suppressor gene ADAMTS18 promotes tumorigenesis and progression of clear cell renal cell carcinoma. Genes Genomics 2021; 43:123-131. [DOI: 10.1007/s13258-021-01036-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022]
|
13
|
Prognostic Values for the mRNA Expression of the ADAMTS Family of Genes in Gastric Cancer. JOURNAL OF ONCOLOGY 2020; 2020:9431560. [PMID: 32884571 PMCID: PMC7455834 DOI: 10.1155/2020/9431560] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
The “A Disintegrin and Metalloproteinase with Thrombospondin Motif” (ADAMTS) family of genes is involved in the occurrence and development of different cancers. However, the prognostic value of these genes in gastric cancer (GC) has not been revealed. The present study was thus conducted to determine the prognostic value for the ADAMTS family of genes in GC. First, we evaluated the mRNA expression levels of the ADAMTS family in GC patients using a GEPIA dataset. Thereafter, we determined the prognostic value of these genes by analyzing their mRNA level using the Kaplan–Meier Plotter database. The mRNA expression level of ADAMTS12 was randomly validated by qRT-PCR and meta-analysis while its coexpression genes were derived using Coexpedia. Finally, we performed Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses using the OmicShare Tools. Compared to normal tissues, expression of ADAMTS2 and 12 was significantly higher while that of ADAMTS1, 13, and 15 was significantly lower in GC tissues. According to the RNA-seq and gene chip data, the ADAMTS family (6, 7, 12, 15, and 18) of genes was closely related to the prognosis of GC, and their high expression levels were associated with poor prognosis and survival time. In addition, ADAMTS12 was highly expressed in 20 pairs of GC tissues based on RT-PCR (P=0.016) and meta-analysis (SMD: 0.73, 95% CI: 0.32–1.14, P < 0.001). GO and KEGG pathway analyses indicated that the ADAMTS12 coexpressed genes were enriched in the pathways of extracellular matrix organization, extracellular matrix structural constituent, extracellular matrix, and protein digestion and absorption. Herein, we discovered the prognostic values and biological roles of the ADAMTS genes in GC.
Collapse
|
14
|
Zhang C, Wang F, Guo F, Ye C, Yang Y, Huang Y, Hou J, Tian F, Yang B. A 13-gene risk score system and a nomogram survival model for predicting the prognosis of clear cell renal cell carcinoma. Urol Oncol 2020; 38:74.e1-74.e11. [PMID: 31952997 DOI: 10.1016/j.urolonc.2019.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 12/18/2019] [Accepted: 12/24/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is the second common malignant tumor in the urinary system, and 85% of RCC cases are clear cell RCC (ccRCC). This study is designed to build a risk score system for ccRCC. METHODS The gene methylation and expression data of ccRCC samples were downloaded from The Cancer Genome Atlas database (training set) and ArrayExpress database (validation set). The differentially methylated genes (DMGs) and differentially expressed genes (DEGs) were identified by limma package, and their intersecting genes with negative Pearson correlation coefficients were remained using cor.test function. Prognosis-associated genes were identified by survival package, and the optimal DMGs were obtained using penalized package. After risk score system was built, nomogram survival model was constructed using rms package. Additionally, pathways were enriched for the DEGs between high- and low-risk groups using Gene Set Enrichment Analysis. RESULTS There were 3,638 DMGs and 2,702 DEGs between tumor and normal samples. Among the 312 intersecting genes, 43 prognosis-associated genes were identified. A total of 13 optimal DMGs (BTBD19, ADAM8, BGLAP, TNFRSF13C, JPH4, BEST1, GNRH2, UBE2QL1, CHODL, GDF9, UPB1, KCNH3; and ADAMTSL4) were obtained for building the risk score system. After pathological M, pathological T, platelet qualitative, and RS status were revealed to be independent prognostic factors, a nomogram survival model was constructed. For the 920 DEGs between the high- and low-risk samples, 6 significant pathways were enriched. CONCLUSION The 13-gene risk score system and the nomogram survival model might be used for prognostic prediction of ccRCC patients.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Fubo Wang
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Fei Guo
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Chen Ye
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Yue Yang
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Feng Tian
- Department of Urology, Shanghai Eighth People's Hospital, Shanghai, China.
| | - Bo Yang
- Department of Urology, Changhai Hospital, the Second Military Medical University, Shanghai, China.
| |
Collapse
|
15
|
Zhang Y, Xu H, Mu J, Guo S, Ye L, Li D, Peng W, He X, Xiang T. Inactivation of ADAMTS18 by aberrant promoter hypermethylation contribute to lung cancer progression. J Cell Physiol 2018; 234:6965-6975. [PMID: 30417422 DOI: 10.1002/jcp.27439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 08/28/2018] [Indexed: 12/19/2022]
Abstract
Lung cancer is the most frequently diagnosed cancer worldwide. Epigenetic regulation contributes to lung cancer pathogenesis. The ADAMTS18 tumor suppressor gene is inactivated in some cancers, but its involvement in lung cancer has not been shown. Immunohistochemistry, quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and methylation-specific PCR were used to assay ADAMTS18 expression and promoter methylation in lung tumor tissues and adjacent tissues. Cell viability, transwell, and wound-healing assays, as well as flow cytometry were used to characterize the biological activity of ADAMTS18. The influence of ADAMTS18 on protein expression was assayed using western blots analysis, and its effect on chemosensitivity was assayed by the response to cisplatin. We found that ADAMTS18 was silenced in lung cancer cells by promoter methylation. Demethylation by the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine, with or without the histone deacetylase inhibitor trichostatin A, restored ADAMTS18 expression. Compared with normal lung tissue, ADAMTS18 in lung tumors was frequently methylated. Overexpression of ADAMTS18 in lung cancer cells inhibited cell proliferation, migration, and invasiveness and induced G0/G1 cell cycle arrest. Furthermore, ADAMTS18 suppressed epidermal growth factor receptor/protein kinase B (EGFR/AKT) signaling, which sensitized lung cancer cells to cisplatin. Thus, our results demonstrated that the tumor suppressor gene ADAMTS18 was downregulated in lung cancer by promoter CpG methylation, and it promoted sensitivity to cisplatin via EGFR/AKT signaling. Our study suggests that ADAMTS18 promoter methylation is a potential epigenetic biomarker for early detection of lung cancer and warrants investigation as a therapeutic target for early-stage lung cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongying Xu
- Department of Respiratory Medicine, The Sixth People's Hospital of Chongqing, Chongqing, China
| | - Junhao Mu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuliang Guo
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Ye
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dairong Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqian He
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Kordowski F, Kolarova J, Schafmayer C, Buch S, Goldmann T, Marwitz S, Kugler C, Scheufele S, Gassling V, Németh CG, Brosch M, Hampe J, Lucius R, Röder C, Kalthoff H, Siebert R, Ammerpohl O, Reiss K. Aberrant DNA methylation of ADAMTS16 in colorectal and other epithelial cancers. BMC Cancer 2018; 18:796. [PMID: 30081852 PMCID: PMC6080380 DOI: 10.1186/s12885-018-4701-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ADAMs (a disintegrin and metalloproteinase) have long been associated with tumor progression. Recent findings indicate that members of the closely related ADAMTS (ADAMs with thrombospondin motifs) family are also critically involved in carcinogenesis. Gene silencing through DNA methylation at CpG loci around e.g. transcription start or enhancer sites is a major mechanism in cancer development. Here, we aimed at identifying genes of the ADAM and ADAMTS family showing altered DNA methylation in the development or colorectal cancer (CRC) and other epithelial tumors. METHODS We investigated potential changes of DNA methylation affecting ADAM and ADAMTS genes in 117 CRC, 40 lung cancer (LC) and 15 oral squamous-cell carcinoma (SCC) samples. Tumor tissue was analyzed in comparison to adjacent non-malignant tissue of the same patients. The methylation status of 1145 CpGs in 51 ADAM and ADAMTS genes was measured with the HumanMethylation450 BeadChip Array. ADAMTS16 protein expression was analyzed in CRC samples by immunohistochemistry. RESULTS In CRC, we identified 72 CpGs in 18 genes which were significantly affected by hyper- or hypomethylation in the tumor tissue compared to the adjacent non-malignant tissue. While notable/frequent alterations in methylation patterns within ADAM genes were not observed, conspicuous changes were found in ADAMTS16 and ADAMTS2. To figure out whether these differences would be CRC specific, additional LC and SCC tissue samples were analyzed. Overall, 78 differentially methylated CpGs were found in LC and 29 in SCC. Strikingly, 8 CpGs located in the ADAMTS16 gene were commonly differentially methylated in all three cancer entities. Six CpGs in the promoter region were hypermethylated, whereas 2 CpGs in the gene body were hypomethylated indicative of gene silencing. In line with these findings, ADAMTS16 protein was strongly expressed in globlet cells and colonocytes in control tissue but not in CRC samples. Functional in vitro studies using the colorectal carcinoma cell line HT29 revealed that ADAMTS16 expression restrained tumor cell proliferation. CONCLUSIONS We identified ADAMTS16 as novel gene with cancer-specific promoter hypermethylation in CRC, LC and SCC patients implicating ADAMTS16 as potential biomarker for these tumors. Moreover, our results provide evidence that ADAMTS16 may have tumor suppressor properties.
Collapse
Affiliation(s)
- Felix Kordowski
- Department of Dermatology and Allergology, University Hospital Schleswig-Holstein, University of Kiel, Rosalind-Franklin-Straße 7, 24105 Kiel, Germany
| | - Julia Kolarova
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stephan Buch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Torsten Goldmann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
- Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel, Germany
| | - Sebastian Marwitz
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
- Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel, Germany
| | - Christian Kugler
- Thoracic Surgery, LungenClinic Grosshansdorf, Grosshansdorf, Germany
| | | | - Volker Gassling
- Department of Oral and Maxillofacial Surgery, University of Kiel, Kiel, Germany
| | | | - Mario Brosch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ralph Lucius
- Anatomical Institute, University of Kiel, Kiel, Germany
| | - Christian Röder
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Ole Ammerpohl
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Karina Reiss
- Department of Dermatology and Allergology, University Hospital Schleswig-Holstein, University of Kiel, Rosalind-Franklin-Straße 7, 24105 Kiel, Germany
| |
Collapse
|
17
|
Shao B, Feng Y, Zhang H, Yu F, Li Q, Tan C, Xu H, Ying J, Li L, Yang D, Peng W, Tang J, Li S, Ren G, Tao Q, Xiang T. The 3p14.2 tumour suppressor ADAMTS9 is inactivated by promoter CpG methylation and inhibits tumour cell growth in breast cancer. J Cell Mol Med 2017; 22:1257-1271. [PMID: 29193730 PMCID: PMC5783842 DOI: 10.1111/jcmm.13404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 08/29/2017] [Indexed: 12/14/2022] Open
Abstract
Chromosome region 3p12‐14 is an important tumour suppressor gene (TSG) locus for multiple cancers. ADAMTS9, a member of the metalloprotease large family, has been identified as a candidate 3p14.2 TSG inactivated by aberrant promoter CpG methylation in several carcinomas, but little known about its expression and function in breast cancer. In this report, ADAMTS9 expression and methylation was analysed in breast cancer cell lines and tissue samples. ADAMTS9 RNA was significantly down‐regulated in breast cancer cell lines (6/8). After treating the cells with demethylation agent Aza and TSA,ADAMTS9 expression was dramatically increased. Bisulphite genomic sequencing and methylation‐specific PCR detected promoter methylation, which was associated with decreased ADAMTS9 expression. Hypermethylation was also detected in 130/219 (59.4%) of primary tumours but only in 4.5% (2/44) of paired surgical margin tissues. Ectopic expression of ADAMTS9 in tumor cells induced significant growth suppression, cell cycle arrest at the G0/G1 phase, enhanced apoptosis and reduced cell migration and invasion. Conditioned culture medium from ADAMTS9‐transfected BT549 cells markedly disrupted tube formation ability of human umbilical vein endothelial cell (HUVEC) in Matrigel. Furthermore, ADAMTS9 inhibited AKT signaling and its downstream targets (MDM2, p53, p21, p27, E‐cadherin, VIM, SNAIL, VEGFA, NFκB‐p65 and MMP2). In addition, we demonstrated, for the first time, that ADAMTS9 inhibits AKT signaling, through suppressing its upstream activators EGFR and TGFβ1/TβR(I/II) in breast cancer cells. Our results suggest that ADAMTS9 is a TSG epigenetically inactivated in breast cancer, which functions through blocking EGFR‐ and TGFβ1/TβR(I/II)‐activated AKT signaling.
Collapse
Affiliation(s)
- Bianfei Shao
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongbin Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Yu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,The Second people's hospital of JingDe Zhen, Jiangxi, China
| | - Qianqian Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cui Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongying Xu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,The Sixth people's hospital of Chongqing, Chongqing, China
| | - Jianming Ying
- Cancer Epigenetics Laboratory, State Key Laboratory of Oncology in South China, Department of Clinical Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong and CUHK Shenzhen Research Institute, Hong Kong, China.,Department of Pathology, Cancer Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Li
- Cancer Epigenetics Laboratory, State Key Laboratory of Oncology in South China, Department of Clinical Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong and CUHK Shenzhen Research Institute, Hong Kong, China
| | - Dejuan Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Tang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuman Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Tao
- Cancer Epigenetics Laboratory, State Key Laboratory of Oncology in South China, Department of Clinical Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong and CUHK Shenzhen Research Institute, Hong Kong, China
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Joosten SC, Deckers IA, Aarts MJ, Hoeben A, van Roermund JG, Smits KM, Melotte V, van Engeland M, Tjan-Heijnen VC. Prognostic DNA methylation markers for renal cell carcinoma: a systematic review. Epigenomics 2017; 9:1243-1257. [PMID: 28803494 DOI: 10.2217/epi-2017-0040] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM Despite numerous published prognostic methylation markers for renal cell carcinoma (RCC), none of these have yet changed patient management. Our aim is to systematically review and evaluate the literature on prognostic DNA methylation markers for RCC. MATERIALS & METHODS We conducted an exhaustive search of PubMed, EMBASE and MEDLINE up to April 2017 and identified 49 publications. Studies were reviewed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement, assessed for their reporting quality using the Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK) criteria, and were graded to determine the level of evidence (LOE) for each biomarker. RESULTS We identified promoter methylation of BNC1, SCUBE3, GATA5, SFRP1, GREM1, RASSF1A, PCDH8, LAD1 and NEFH as promising prognostic markers. Extensive methodological heterogeneity across the included studies was observed, which hampers comparability and reproducibility of results, providing a possible explanation why these biomarkers do not reach the clinic. CONCLUSION Potential prognostic methylation markers for RCC have been identified, but they require further validation in prospective studies to determine their true clinical value.
Collapse
Affiliation(s)
- Sophie C Joosten
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Ivette Ag Deckers
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Maureen J Aarts
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Ann Hoeben
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Joep G van Roermund
- Department of Urology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Kim M Smits
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Manon van Engeland
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Vivianne C Tjan-Heijnen
- Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
19
|
Xu H, Xiao Q, Fan Y, Xiang T, Li C, Li C, Li S, Hui T, Zhang L, Li H, Li L, Ren G. Epigenetic silencing of ADAMTS18 promotes cell migration and invasion of breast cancer through AKT and NF-κB signaling. Cancer Med 2017; 6:1399-1408. [PMID: 28503860 PMCID: PMC5463072 DOI: 10.1002/cam4.1076] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 12/16/2022] Open
Abstract
ADAMTS18 dysregulation plays an important role in many disease processes including cancer. We previously found ADAMTS18 as frequently methylated tumor suppressor gene (TSG) for multiple carcinomas, however, its biological functions and underlying molecular mechanisms in breast carcinogenesis remain unknown. Here, we found that ADAMTS18 was silenced or downregulated in breast cancer cell lines. ADAMTS18 was reduced in primary breast tumor tissues as compared with their adjacent noncancer tissues. ADAMTS18 promoter methylation was detected in 70.8% of tumor tissues by methylation-specific PCR, but none of the normal tissues. Demethylation treatment restored ADAMTS18 expression in silenced breast cell lines. Ectopic expression of ADAMTS18 in breast tumor cells resulted in inhibition of cell migration and invasion. Nude mouse model further confirmed that ADAMTS18 suppressed breast cancer metastasis in vivo. Further mechanistic studies showed that ADAMTS18 suppressed epithelial-mesenchymal transition (EMT), further inhibited migration and invasion of breast cancer cells. ADAMT18 deregulated AKT and NF-κB signaling, through inhibiting phosphorylation levels of AKT and p65. Thus, ADAMTS18 as an antimetastatic tumor suppressor antagonizes AKT and NF-κB signaling in breast tumorigenesis. Its methylation could be a potential tumor biomarker for breast cancer.
Collapse
Affiliation(s)
- Hongying Xu
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Qian Xiao
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yu Fan
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tingxiu Xiang
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chen Li
- Cancer Epigenetics LaboratoryDepartment of Clinical OncologyState Key Laboratory of Oncology in South China, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health SciencesCUHK Shenzhen Research InstituteThe Chinese University of Hong KongHong Kong
| | - Chunhong Li
- Department of OncologySuining Center HospitalSuiningChina
| | - Shuman Li
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tianli Hui
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lu Zhang
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lili Li
- Cancer Epigenetics LaboratoryDepartment of Clinical OncologyState Key Laboratory of Oncology in South China, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health SciencesCUHK Shenzhen Research InstituteThe Chinese University of Hong KongHong Kong
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
20
|
Detection of piRNAs in whitespotted bamboo shark liver. Gene 2016; 590:51-6. [PMID: 27267405 DOI: 10.1016/j.gene.2016.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/20/2016] [Accepted: 06/03/2016] [Indexed: 01/12/2023]
Abstract
Piwi-interacting RNAs (piRNAs) are 26 to 31-nt small non-coding RNAs that have been reported mostly in germ-line cells and cancer cells. However, the presence of piRNAs in the whitespotted bamboo shark liver has not yet been reported. In a previous study of microRNAs in shark liver, some piRNAs were detected from small RNAs sequenced by Solexa technology. A total of 4857 piRNAs were predicted and found in shark liver. We further selected 17 piRNAs with high and significantly differential expression between normal and regenerative liver tissues for subsequent verification by Northern blotting. Ten piRNAs were further identified, and six of these were matched to known piRNAs in piRNABank. The actual expression of six known and four novel piRNAs was validated by qRT-PCR. In addition, a total of 401 target genes of the 10 piRNAs were predicted by miRanda. Through GO and pathway function analyses, only five piRNAs could be annotated with eighteen GO annotations. The results indicated that the identified piRNAs are involved in many important biological responses, including immune inflammation, cell-specific differentiation and development, and angiogenesis. This manuscript provides the first identification of piRNAs in the liver of whitespotted bamboo shark using Solexa technology as well as further elucidation of the regulatory role of piRNAs in whitespotted bamboo shark liver. These findings may provide a useful resource and may facilitate the development of therapeutic strategies against liver damage.
Collapse
|
21
|
The Silencing of CCND2 by Promoter Aberrant Methylation in Renal Cell Cancer and Analysis of the Correlation between CCND2 Methylation Status and Clinical Features. PLoS One 2016; 11:e0161859. [PMID: 27583477 PMCID: PMC5008725 DOI: 10.1371/journal.pone.0161859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/13/2016] [Indexed: 12/21/2022] Open
Abstract
Cyclin D2 (CCND2) is a member of the D-type cyclins, which plays a pivotal role in cell cycle regulation, differentiation and malignant transformation. However, its expression status and relative regulation mechanism remains unclear in renal cell cancer (RCC). In our study, the mRNA expression level of CCND2 is down-regulated in 22/23 paired RCC tissues (p<0.05). In addition, its protein expression level is also decreased in 43/43 RCC tumor tissues compared with its corresponding non-malignant tissues (p<0.001). We further detected that CCND2 was down-regulated or silenced in 6/7 RCC cell lines, but expressed in “normal” human proximal tubular (HK-2) cell line. Subsequently, MSP and BGS results showed that the methylation status in CCND2 promoter region is closely associated with its expression level in RCC cell lines. Treatment with 5-Aza with or without TSA restored CCND2 expression in several methylated RCC cell lines. Among the 102 RCC tumors, methylation of CCND2 was detected in 29/102 (28%) cases. Only 2/23 (8.7%) adjacent non-malignant tissues showed methylation. We then analyzed the correlation of clinical features and its promoter methylation. Collectively, our data suggested that loss of CCND2 expression is closely associated with the promoter aberrant methylation.
Collapse
|