1
|
Zhu X, Li Z, Chen L, Li L, Ouyang M, Zhou H, Xiao K, Lin L, Chu PK, Zhou C, Xun C, Yang L, Huang W, Ding X. Exosomes delivering miR-129-5p combined with sorafenib ameliorate hepatocellular carcinoma progression via the KCTD1/HIF-1α/VEGF pathway. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01044-x. [PMID: 40227531 DOI: 10.1007/s13402-025-01044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Potassium channel tetramerization domain-containing 1 (KCTD1) plays a critical role in transcriptional regulation and adipogenesis, but its significance in hepatocellular cancer (HCC) has not been reported. METHODS Immunohistochemistry, Western blotting and quantitative real-time PCR analysis were performed to assess the expression of KCTD1 and related genes in HCC cells. MTT assays, colony formation, cell migration, invasion and the in-vivo mouse models were utilized to evaluate the function of KCTD1 in HCC progression. Co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assays were conducted to elucidate the molecular mechanisms of KCTD1 in HCC. RESULTS KCTD1 expression was increased in human HCC tissues and closely associated with advanced tumor stages. KCTD1 overexpression enhanced growth, migration, and invasion of Huh7 and HepG2 cells both in vitro and in vivo, while KCTD1 knockdown reversed these effects in MHCC97H cells. Mechanistically, KCTD1 interacted with hypoxia-inducible factor 1 alpha (HIF-1α) and enhanced HIF-1α protein stability with the inhibited prolyl-hydroxylases (PHD)/Von Hippel-Lindau (VHL) pathway, consequently activating the Vascular Endothelial Growth Factor (VEGF)/VEGFR2 pathway in HCC cells. Sorafenib and KCTD1 knockdown synergistically inhibited intrahepatic tumor growth following in situ injection of MHCC97H cells. miR-129-5p downregulated KCTD1 by binding to KCTD1 3'UTR. Finally, 45 µg exosomes from miR-129-5p-overexpressing MHCC97H cells combined with 25 mg/kg sorafenib to decrease HCC tumor size. CONCLUSIONS These results suggested that KCTD1 protects HIF-1α from degradation and activates the VEGF signaling cascade to enhance HCC progression. Therefore, KCTD1 may serve as a novel target of HCC and pave the way for an efficient combined therapy in advanced HCC.
Collapse
Affiliation(s)
- Xinyu Zhu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Li Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Taozihu Road No. 68, Changsha, 410081, China.
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Lin
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Chang Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Liu Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Wenhuan Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China.
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| |
Collapse
|
2
|
Sun D, Hu Y, Peng J, Wang S. Construction of T-Cell-Related Prognostic Risk Models and Prediction of Tumor Immune Microenvironment Regulation in Pancreatic Adenocarcinoma via Integrated Analysis of Single-Cell RNA-Seq and Bulk RNA-Seq. Int J Mol Sci 2025; 26:2384. [PMID: 40141028 PMCID: PMC11942068 DOI: 10.3390/ijms26062384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a fatal malignant tumor of the digestive system, and immunotherapy has currently emerged as a key therapeutic approach for treating PAAD, with its efficacy closely linked to T-cell subsets and the tumor immune microenvironment. However, reliable predictive markers to guide clinical immunotherapy for PAAD are not available. We analyzed the single-cell RNA sequencing (scRNA-seq) data focused on PAAD from the GeneExpressionOmnibus (GEO) database. Then, the information from the Cancer Genome Atlas (TCGA) database was integrated to develop and validate a prognostic risk model derived from T-cell marker genes. Subsequently, the correlation between these risk models and the effectiveness of immunotherapy was explored. Analysis of scRNA-seq data uncovered six T-cell subtypes and 1837 T-cell differentially expressed genes (DEGs). Combining these data with the TCGA dataset, we constructed a T-cell prognostic risk model containing 16 DEGs, which can effectively predict patient survival and immunotherapy outcomes. We have found that patients in the low-risk group had better prognostic outcomes, increased immune cell infiltration, and signs of immune activation compared to those in the high-risk group. Additionally, analysis of tumor mutation burden showed higher mutation rates in patients with PAAD in the high-risk group. Risk scores with immune checkpoint gene expression and drug sensitivity analysis provide patients with multiple therapeutic targets and drug options. Our study constructed a prognostic risk model for PAAD patients based on T-cell marker genes, providing valuable insights into predicting patient prognosis and the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Dingya Sun
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacology, Central South University, Changsha 410083, China;
| | - Yijie Hu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China;
| | - Jun Peng
- Xiangya School of Pharmaceutical Sciences, Department of Pharmacology, Central South University, Changsha 410083, China;
| | - Shan Wang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China;
| |
Collapse
|
3
|
Qiu Y, Xu B, Feng J, Wang C, Chen Y, He Y, Xie X, Li Y. Loss of EPS8 sensitizes non-small-cell lung carcinoma to chemotherapy-induced DNA damage. Cancer Gene Ther 2023:10.1038/s41417-023-00606-1. [PMID: 36932195 DOI: 10.1038/s41417-023-00606-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023]
Abstract
Epidermal growth factor receptor pathway substrate number 8 (EPS8) has been reported to be critical in mediating tumor progression. However, the molecular and biological consequences of EPS8 overexpression remain unclear. Here we evaluated whether EPS8 increased DNA damage repair in non-small-cell lung carcinoma (NSCLC) cells and the mechanism of EPS8-mediated DNA damage repair which influenced chemosensitivity. Serial studies of functional experiments revealed that EPS8 knockdown inhibited cell growth, induced cell-cycle arrest and increased cisplatin therapeutic effects on NSCLC. EPS8 was found to induce DNA damage repair via upregulation of phosphorylated-ATM and downregulation of the tumor suppressor p53 and G1 cell kinase inhibitor p21. Moreover, in conjunction with cisplatin, decreasing EPS8 protein levels further increased p53 protein level and inhibited ATM signaling. Transplanted tumor studies were also performed to demonstrate that EPS8 knockdown inhibited tumor growth and sensitized tumors to cisplatin treatment. In conclusion, we have described a novel molecular mechanism through which EPS8 is likely to be involved in cancer progression and chemoresistance via DNA damage repair, indicating that EPS8 expression may influence the response to chemotherapy. Therefore, targeting EPS8 may be a potential therapeutic approach for patients with NSCLC.
Collapse
Affiliation(s)
- Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Binyan Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Jianhua Feng
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Chunsheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China
| | - Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 Gongye Dadao Zhong, Guangzhou, Guangdong, 510285, P. R. China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, P. R. China.
| |
Collapse
|
4
|
Routh ED, Woodcock MG, Beckabir W, Vensko SP, Serody JS, Vincent BG. Evaluation of tumor antigen-specific antibody responses in patients with metastatic triple negative breast cancer treated with cyclophosphamide and pembrolizumab. J Immunother Cancer 2023; 11:jitc-2022-005848. [PMID: 36882226 PMCID: PMC10008414 DOI: 10.1136/jitc-2022-005848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 03/09/2023] Open
Abstract
The role of B cells in antitumor immunity is becoming increasingly appreciated, as B cell populations have been associated with response to immune checkpoint blockade (ICB) in patients with breast cancer and murine models of breast cancer. Deeper understanding of antibody responses to tumor antigens is needed to clarify the function of B cells in determining response to immunotherapy. We evaluated tumor antigen-specific antibody responses in patients with metastatic triple negative breast cancer treated with pembrolizumab following low-dose cyclophosphamide therapy using computational linear epitope prediction and custom peptide microarrays. We found that a minority of predicted linear epitopes were associated with antibody signal, and signal was associated with both neoepitopes and self-peptides. No association was observed between signal presence and subcellular localization or RNA expression of parent proteins. Patient-specific patterns of antibody signal boostability were observed that were independent of clinical response. Intriguingly, measures of cumulative antibody signal intensity relative to immunotherapy treatment showed that the one complete responder in the trial had the greatest increase in total antibody signal, which supports a potential association between ICB-dependent antibody boosting and clinical response. The antibody boost in the complete responder was largely driven by increased levels of IgG specific to a sequence of N-terminal residues in native Epidermal Growth Factor Receptor Pathway Substrate 8 (EPS8) protein, a known oncogene in several cancer types including breast cancer. Structural protein prediction showed that the targeted epitope of EPS8 was in a region of the protein with mixed linear/helical structure, and that this region was solvent-exposed and not predicted to bind to interacting macromolecules. This study highlights the potential importance of the humoral immune response targeting neoepitopes as well as self epitopes in shaping clinical response to immunotherapy.
Collapse
Affiliation(s)
- Eric D Routh
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Division of Medical Oncology, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Wolfgang Beckabir
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Steven P Vensko
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan S Serody
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Department of Microbiology and Immunology, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA .,Department of Microbiology and Immunology, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Division of Hematology, Department of Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Computational Medicine Program, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Nasimian A, Ahmed M, Hedenfalk I, Kazi JU. A deep tabular data learning model predicting cisplatin sensitivity identifies BCL2L1 dependency in cancer. Comput Struct Biotechnol J 2023; 21:956-964. [PMID: 36733702 PMCID: PMC9876747 DOI: 10.1016/j.csbj.2023.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
Cisplatin, a platinum-based chemotherapeutic agent, is widely used as a front-line treatment for several malignancies. However, treatment outcomes vary widely due to intrinsic and acquired resistance. In this study, cisplatin-perturbed gene expression and pathway enrichment were used to define a gene signature, which was further utilized to develop a cisplatin sensitivity prediction model using the TabNet algorithm. The TabNet model performed better (>80 % accuracy) than all other machine learning models when compared to a wide range of machine learning algorithms. Moreover, by using feature importance and comparing predicted ovarian cancer patient samples, BCL2L1 was identified as an important gene contributing to cisplatin resistance. Furthermore, the pharmacological inhibition of BCL2L1 was found to synergistically increase cisplatin efficacy. Collectively, this study developed a tool to predict cisplatin sensitivity using cisplatin-perturbed gene expression and pathway enrichment knowledge and identified BCL2L1 as an important gene in this setting.
Collapse
Affiliation(s)
- Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden,Correspondence to: Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon village Building 404:C3, Scheelevägen 8, 22363 Lund, Sweden.
| |
Collapse
|
6
|
Tan M, Meng J, Sun X, Fu X, Wang R. EPS8 supports pancreatic cancer growth by inhibiting BMI1 mediated proteasomal degradation of ALDH7A1. Exp Cell Res 2021; 407:112782. [PMID: 34391775 DOI: 10.1016/j.yexcr.2021.112782] [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: 06/01/2021] [Revised: 07/11/2021] [Accepted: 07/17/2021] [Indexed: 01/29/2023]
Abstract
Aldehyde dehydrogenase 7 family member A1 (ALDH7A1) is an enzyme catalyzing lipid peroxidation of fatty aldehydes. It plays a critical role in sustaining high oxygen consumption rate (OCR) and ATP production in pancreatic ductal adenocarcinoma (PADC). However, why PADC cells maintain a relatively high level of ALDH7A1 concentration is still not well understood. In the current study, we explored the interplay between epidermal growth factor receptor kinase substrate 8 (EPS8) and ALDH7A1 in PADC cells. PADC cell lines MIA PaCa-2 and AsPANC-1 were used for in vitro and in vivo studies. The co-IP assay showed mutual interactions between Flag-EPS8 and Myc-ALDH7A1 in both MIA PaCa-2 and AsPANC-1 cells. EPS8 knockdown resulted in decreased ALDH7A1 protein levels and increased poly-ubiquitination. An interaction was observed between ALDH7A1 and BMI1 but not between BMI1 and EPS8. BMI1 knockdown reduced ALDH7A1 poly-ubiquitination and degradation caused by EPS8 knockdown. Dual EPS8 and ALDH7A1 knockdown had a synergistic effect on suppressing PADC cell proliferation in vitro and in vivo. In conclusion, this study revealed that EPS8 supports PADC growth by interacting with ALDH7A1 and inhibiting BMI1 mediated proteasomal degradation of ALDH7A1.
Collapse
Affiliation(s)
- Mingzhu Tan
- Internal Medicine, Weifang People's Hospital Brain Hospital, Weifang, Shandong, 261000, China
| | - Jun Meng
- Occupational Medicine, Weifang People's Hospital, Weifang, Shandong, 261000, China
| | - Xiaojuan Sun
- Occupational Medicine, Weifang People's Hospital, Weifang, Shandong, 261000, China
| | - Xiaowei Fu
- Department of Neonatology, Weifang People's Hospital, Weifang, Shandong, 261000, China
| | - Ruihao Wang
- Internal Medicine, Weifang People's Hospital Brain Hospital, Weifang, Shandong, 261000, China.
| |
Collapse
|
7
|
Barazetti JF, Jucoski TS, Carvalho TM, Veiga RN, Kohler AF, Baig J, Al Bizri H, Gradia DF, Mader S, Carvalho de Oliveira J. From Micro to Long: Non-Coding RNAs in Tamoxifen Resistance of Breast Cancer Cells. Cancers (Basel) 2021; 13:3688. [PMID: 34359587 PMCID: PMC8345104 DOI: 10.3390/cancers13153688] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/03/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer mortality among women. Two thirds of patients are classified as hormone receptor positive, based on expression of estrogen receptor alpha (ERα), the main driver of breast cancer cell proliferation, and/or progesterone receptor, which is regulated by ERα. Despite presenting the best prognosis, these tumors can recur when patients acquire resistance to treatment by aromatase inhibitors or antiestrogen such as tamoxifen (Tam). The mechanisms that are involved in Tam resistance are complex and involve multiple signaling pathways. Recently, roles for microRNAs and lncRNAs in controlling ER expression and/or tamoxifen action have been described, but the underlying mechanisms are still little explored. In this review, we will discuss the current state of knowledge on the roles of microRNAs and lncRNAs in the main mechanisms of tamoxifen resistance in hormone receptor positive breast cancer. In the future, this knowledge can be used to identify patients at a greater risk of relapse due to the expression patterns of ncRNAs that impact response to Tam, in order to guide their treatment more efficiently and possibly to design therapeutic strategies to bypass mechanisms of resistance.
Collapse
Affiliation(s)
- Jéssica Fernanda Barazetti
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Tayana Shultz Jucoski
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Tamyres Mingorance Carvalho
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Rafaela Nasser Veiga
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Ana Flávia Kohler
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Jumanah Baig
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; (J.B.); (H.A.B.)
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Hend Al Bizri
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; (J.B.); (H.A.B.)
| | - Daniela Fiori Gradia
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| | - Sylvie Mader
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; (J.B.); (H.A.B.)
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Jaqueline Carvalho de Oliveira
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-000, Parana, Brazil; (J.F.B.); (T.S.J.); (T.M.C.); (R.N.V.); (A.F.K.); (D.F.G.)
| |
Collapse
|
8
|
Dexmedetomidine suppresses the progression of esophageal cancer via miR-143-3p/epidermal growth factor receptor pathway substrate 8 axis. Anticancer Drugs 2021; 31:693-701. [PMID: 32701250 PMCID: PMC7365679 DOI: 10.1097/cad.0000000000000934] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Esophageal cancer is one of the fatal cancers around the world. Dexmedetomidine (DEX) is widely used during anesthesia of esophageal cancer surgery. Nevertheless, the role of DEX in the progression of esophageal cancer remains barely known. The proliferation, apoptosis and metastasis of esophageal cancer cells were detected by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, transwell migration and invasion assays and Western blot assay. The expression of miR-143-3p was measured by quantitative real-time PCR in esophageal cancer tissues and cells. The binding sites between miR-143-3p and epidermal growth factor receptor pathway substrate 8 (EPS8) were predicted by Starbase online software, and the combination was verified by dual-luciferase reporter assay. The murine xenograft model was established using KYSE150 cells to verify the function of DEX in vivo. DEX inhibited the proliferation and metastasis while accelerated the apoptosis of esophageal cancer cells. The abundance of miR-143-3p was lower in esophageal cancer tissues and cells than that in paring normal tissues and normal esophageal mucosal cells Het-1A. MiR-143-3p could be induced by DEX treatment in esophageal cancer cells, and miR-143-3p also suppressed the development of esophageal cancer. EPS8 was a functional target of miR-143-3p, and it played an oncogenic role in esophageal cancer. DEX inhibited the growth of tumor via miR-143-3p/EPS8 in vivo. DEX suppressed the growth and metastasis while facilitated the apoptosis of esophageal cancer cells through upregulating the abundance of miR-143-3p and reducing the level of EPS8 in vivo and in vitro, providing promising target for the treatment of esophageal cancer.
Collapse
|
9
|
Kaniowski D, Ebenryter-Olbińska K, Kulik K, Suwara J, Cypryk W, Jakóbik-Kolon A, Leśnikowski Z, Nawrot B. Composites of Nucleic Acids and Boron Clusters (C 2B 10H 12) as Functional Nanoparticles for Downregulation of EGFR Oncogene in Cancer Cells. Int J Mol Sci 2021; 22:ijms22094863. [PMID: 34064412 PMCID: PMC8125477 DOI: 10.3390/ijms22094863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is one of the most promising molecular targets for anticancer therapy. We used boron clusters as a platform for generation of new materials. For this, functional DNA constructs conjugated with boron clusters (B-ASOs) were developed. These B-ASOs, built from 1,2-dicarba-closo-dodecaborane linked with two anti-EGFR antisense oligonucleotides (ASOs), form with their complementary congeners torus-like nanostructures, as previously shown by atomic force microscope (AFM) and transmission electron cryo-microscopy (cryo-TEM) imaging. In the present work, deepened studies were carried out on B-ASO's properties. In solution, B-ASOs formed four dominant complexes as confirmed by non-denaturing polyacrylamide gel electrophoresis (PAGE). These complexes exhibited increased stability in cell lysate comparing to the non-modified ASO. Fluorescently labeled B-ASOs localized mostly in the cytoplasm and decreased EGFR expression by activating RNase H. Moreover, the B-ASO complexes altered the cancer cell phenotype, decreased cell migration rate, and arrested the cells in the S phase of cell cycle. The 1,2-dicarba-closo-dodecaborane-containing nanostructures did not activate NLRP3 inflammasome in human macrophages. In addition, as shown by inductively coupled plasma mass spectrometry (ICP MS), these nanostructures effectively penetrated the human squamous carcinoma cells (A431), showing their potential applicability as anticancer agents.
Collapse
Affiliation(s)
- Damian Kaniowski
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
| | - Katarzyna Ebenryter-Olbińska
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
| | - Katarzyna Kulik
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
| | - Justyna Suwara
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
| | - Wojciech Cypryk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
| | - Agata Jakóbik-Kolon
- Department of Inorganic, Analytical Chemistry and Electrochemistry, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 6, 44-100 Gliwice, Poland;
| | - Zbigniew Leśnikowski
- Laboratory of Medicinal Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 92-232 Lodz, Poland;
| | - Barbara Nawrot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (D.K.); (K.E.-O.); (K.K.); (J.S.); (W.C.)
- Correspondence: ; Tel.: +48-42-6803248
| |
Collapse
|
10
|
Kalathil D, John S, Nair AS. FOXM1 and Cancer: Faulty Cellular Signaling Derails Homeostasis. Front Oncol 2021; 10:626836. [PMID: 33680951 PMCID: PMC7927600 DOI: 10.3389/fonc.2020.626836] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Forkhead box transcription factor, FOXM1 is implicated in several cellular processes such as proliferation, cell cycle progression, cell differentiation, DNA damage repair, tissue homeostasis, angiogenesis, apoptosis, and redox signaling. In addition to being a boon for the normal functioning of a cell, FOXM1 turns out to be a bane by manifesting in several disease scenarios including cancer. It has been given an oncogenic status based on several evidences indicating its role in tumor development and progression. FOXM1 is highly expressed in several cancers and has also been implicated in poor prognosis. A comprehensive understanding of various aspects of this molecule has revealed its role in angiogenesis, invasion, migration, self- renewal and drug resistance. In this review, we attempt to understand various mechanisms underlying FOXM1 gene and protein regulation in cancer including the different signaling pathways, post-transcriptional and post-translational modifications. Identifying crucial molecules associated with these processes can aid in the development of potential pharmacological approaches to curb FOXM1 mediated tumorigenesis.
Collapse
Affiliation(s)
- Dhanya Kalathil
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Samu John
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| | - Asha S Nair
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| |
Collapse
|
11
|
Luo K, Zhang L, Liao Y, Zhou H, Yang H, Luo M, Qing C. Effects and mechanisms of Eps8 on the biological behaviour of malignant tumours (Review). Oncol Rep 2021; 45:824-834. [PMID: 33432368 PMCID: PMC7859916 DOI: 10.3892/or.2021.7927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022] Open
Abstract
Epidermal growth factor receptor pathway substrate 8 (Eps8) was initially identified as the substrate for the kinase activity of EGFR, improving the responsiveness of EGF, which is involved in cell mitosis, differentiation and other physiological functions. Numerous studies over the last decade have demonstrated that Eps8 is overexpressed in most ubiquitous malignant tumours and subsequently binds with its receptor to activate multiple signalling pathways. Eps8 not only participates in the regulation of malignant phenotypes, such as tumour proliferation, invasion, metastasis and drug resistance, but is also related to the clinicopathological characteristics and prognosis of patients. Therefore, Eps8 is a potential tumour diagnosis and prognostic biomarker and even a therapeutic target. This review aimed to describe the structural characteristics, role and related molecular mechanism of Eps8 in malignant tumours. In addition, the prospect of Eps8 as a target for cancer therapy is examined.
Collapse
Affiliation(s)
- Kaili Luo
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Lei Zhang
- Department of Gynecology, Yunnan Tumor Hospital and The Third Affiliated Hospital of Kunming Medical University; Kunming, Yunnan 650118, P.R. China
| | - Yuan Liao
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Hongyu Zhou
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Hongying Yang
- Department of Gynecology, Yunnan Tumor Hospital and The Third Affiliated Hospital of Kunming Medical University; Kunming, Yunnan 650118, P.R. China
| | - Min Luo
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Chen Qing
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
12
|
Dong HC, Li PN, Chen CJ, Xu X, Zhang H, Liu G, Zheng LJ, Li P. Sinomenine Attenuates Cartilage Degeneration by Regulating miR-223-3p/NLRP3 Inflammasome Signaling. Inflammation 2020; 42:1265-1275. [PMID: 30847744 DOI: 10.1007/s10753-019-00986-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sinomenine (SIN) has been shown to protect against IL-1β-induced chondrocyte apoptosis in vitro. However, the role of SIN in the anterior cruciate ligament transection (ACLT)-induced osteoarthritis (OA) mouse model and its underlying molecular mechanisms remain unclear. In the present study, the protective effect of SIN on ACLT-induced articular cartilage degeneration and IL-1β-induced chondrocyte apoptosis miR-223-3p/NLRP3 signaling regulation was investigated. Safranin O staining was performed to evaluate the pathological changes of articular cartilage. Chondrocyte apoptosis was measured with Annexin V-fluorescein isothiocyanate/polyimide (annexin V-FITC/PI) staining using flow cytometry. Gene and protein expression were detected by RT-qPCR and Western blotting, respectively. SIN administration markedly improved articular cartilage degradation in mice undergoing ACLT surgery. In addition, SIN treatment downregulated the levels of inflammatory cytokines and the protein expression of NLRP3 inflammasome components and upregulated the expression of miR-223-3p in OA mice and IL-1β-stimulated chondrocytes. In vitro, we found that NLRP3 was a direct target of miR-223-3p, and overexpression of miR-223-3p blocked IL-1β-induced apoptosis and the inflammatory response in chondrocytes. These findings indicate that miR-223-3p/NLRP3 signaling could be used as a potential target of SIN for the treatment of OA.
Collapse
Affiliation(s)
- Hai-Chao Dong
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China.
| | - Pei-Nan Li
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Chang-Jian Chen
- Dalian love cubic Health Management Co., Ltd, Dalian, 116000, China
| | - Xin Xu
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Hong Zhang
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Gang Liu
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Lian-Jie Zheng
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| | - Peng Li
- Department of Orthopedic Surgery, Second Clinical College, Dalian Medical University, No. 222 Zhongshan Road, Dalian, 116011, China
| |
Collapse
|
13
|
Yang G, Lu YB, Guan QL. EPS8 is a Potential Oncogene in Glioblastoma. Onco Targets Ther 2019; 12:10523-10534. [PMID: 31819533 PMCID: PMC6898995 DOI: 10.2147/ott.s227739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose In this study, we investigated the expression and function of Epidermal growth factor receptor kinase substrate 8 (EPS8) in glioblastoma (GBM), and further explored the underlying mechanisms that regulate it. Patients and methods The expression and potential mechanisms of EPS8 in GBM were evaluated through multiple online public databases. The expression level EPS8 in GBM tissues and cell lines were detected by immunohistochemical staining and Western blot. Then, the prognosis of EPS8 and GBM patients were analyzed. Loss-of-function experiments were conducted to determine the role of EPS8 for the biological behavior of GBM cells. In addition, the tumorigenic ability of nude mice was tested in vivo. Results EPS8 is highly expressed in GBM tissues and indicates poor patient prognosis. In cell experiments, EPS8 can promote the proliferation, migration and invasion of GBM cells. In vivo, EPS8 promotes tumor formation in nude mice. EPS8 can activate the PI3K/Akt signaling pathway to function. Conclusion EP8S plays a role in the development of GBM and may be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Gang Yang
- Department of Neurosurgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.,The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Yong-Bin Lu
- Department of Technology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Quan-Lin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.,Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| |
Collapse
|
14
|
Huang W, Zhong Z, Luo C, Xiao Y, Li L, Zhang X, Yang L, Xiao K, Ning Y, Chen L, Liu Q, Hu X, Zhang J, Ding X, Xiang S. The miR-26a/AP-2α/Nanog signaling axis mediates stem cell self-renewal and temozolomide resistance in glioma. Am J Cancer Res 2019; 9:5497-5516. [PMID: 31534499 PMCID: PMC6735392 DOI: 10.7150/thno.33800] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
Aberrant expression of transcription factor AP-2α has been functionally associated with various cancers, but its clinical significance and molecular mechanisms in human glioma are largely elusive. Methods: AP-2α expression was analyzed in human glioma tissues by immunohistochemistry (IHC) and in glioma cell lines by Western blot. The effects of AP-2α on glioma cell proliferation, migration, invasion and tumor formation were evaluated by the 3-(4,5-dimethyNCthiazol-2-yl)-25-diphenyltetrazolium bromide (MTT) and transwell assays in vitro and in nude mouse models in vivo. The influence of AP-2α on glioma cell stemness was analyzed by sphere-formation, self-renewal and limiting dilution assays in vitro and in intracranial mouse models in vivo. The effects of AP-2α on temozolomide (TMZ) resistance were detected by the MTT assay, cell apoptosis, real-time PCR analysis, western blotting and mouse experiments. The correlation between AP-2α expression and the expression of miR-26a, Nanog was determined by luciferase reporter assays, electrophoretic mobility shift assay (EMSA) and expression analysis. Results: AP-2α expression was downregulated in 58.5% of glioma tissues and in 4 glioma cell lines. AP-2α overexpression not only reduced the proliferation, migration and invasion of glioma cell lines but also suppressed the sphere-formation and self-renewal abilities of glioma stem cells in vitro. Moreover, AP-2α overexpression inhibited subcutaneous and intracranial xenograft tumor growth in vivo. Furthermore, AP-2α enhanced the sensitivity of glioma cells to TMZ. Finally, AP-2α directly bound to the regulatory region of the Nanog gene, reduced Nanog, Sox2 and CD133 expression. Meanwhile, AP-2α indirectly downregulated Nanog expression by inhibiting the interleukin 6/janus kinase 2/signal transducer and activator of transcription 3 (IL6/JAK2/STAT3) signaling pathway, consequently decreasing O6-methylguanine methyltransferase (MGMT) and programmed death-ligand 1 (PD-L1) expression. In addition, miR-26a decreased AP-2α expression by binding to the 3' untranslated region (UTR) of AP-2α and reversed the tumor suppressive role of AP-2α in glioma, which was rescued by a miR-26a inhibitor. TMZ and the miR-26a inhibitor synergistically suppressed intracranial GSC growth. Conclusion: These results suggest that AP-2α reduces the stemness and TMZ resistance of glioma by inhibiting the Nanog/Sox2/CD133 axis and IL6/STAT3 signaling pathways. Therefore, AP-2α and miR-26a inhibition might represent a new target for developing new therapeutic strategies in TMZ resistance and recurrent glioma patients.
Collapse
|
15
|
Li P, Hu T, Wang H, Tang Y, Ma Y, Wang X, Xu Y, Chen G. Upregulation of EPS8L3 is associated with tumorigenesis and poor prognosis in patients with liver cancer. Mol Med Rep 2019; 20:2493-2499. [PMID: 31322213 DOI: 10.3892/mmr.2019.10471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 05/23/2019] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor kinase substrate 8 (EPS8) plays critical roles in a variety of solid tumors. However, the biologic functions and clinical significance of EPS8‑like 3 (EPS8L3), an EPS8‑related protein, in liver cancer remain unclear. To measure EPS8L3 expression in liver cancer cell lines, reverse transcription‑quantitative PCR and western blot analyses were performed. The correlation between 338 patients with liver cancer and various clinicopathological factors obtained from the Oncomine database were evaluated using the χ2 test. Survival of patients with different expression of EPS8L3 was determined using Kaplan‑Meier survival analysis with a log rank test, and Cox regression analysis was performed to estimate the prognostic significance of EPS8L3 expression. Additionally, cell proliferation and migration were determined using Cell Counting Kit‑8 and wound healing assays. The results revealed that EPS8L3 expression was significantly upregulated in liver cancer tissues and cell lines (P<0.01), and that the expression of EPS8L3 was closely associated with grade (P=0.024) and mortality (P=0.011). Furthermore, survival analysis suggested patients with high EPS8L3 expression exhibited shorter survival compared with those with low EPS8L3 expression. Cox regression analysis indicated that EPS8L3 could be regarded as a prognostic biomarker in patients with liver cancer (hazard ratio, 1.58; 95% confidence interval, 1.085‑2.301; P=0.017). Additionally, in vitro assays revealed that EPS8L3 depletion significantly inhibited liver cancer cell proliferation and migration, and reduced the levels of phosphorylated PI3K and AKT in the PI3K/AKT signaling pathway. Collectively, the results of the present study, for the first time to the best of our knowledge, demonstrated that EPS8L3 serves as an oncogene in liver cancer development; therefore, EPS8L3 may be a valuable prognostic predictor for patients with liver cancer.
Collapse
Affiliation(s)
- Peng Li
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Ting Hu
- Department of Oncology, The First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Hongsheng Wang
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Ying Tang
- Department of Nursing, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Yue Ma
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Xiaodong Wang
- Medical Department, Huailai County Hospital of Traditional Chinese Medicine, Zhangjiakou, Hebei 075400, P.R. China
| | - Yansong Xu
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Guangyu Chen
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| |
Collapse
|
16
|
Molecular prognosticators in clinically and pathologically distinct cohorts of head and neck squamous cell carcinoma-A meta-analysis approach. PLoS One 2019; 14:e0218989. [PMID: 31310629 PMCID: PMC6634788 DOI: 10.1371/journal.pone.0218989] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) includes multiple subsites that exhibit differential treatment outcome, which is in turn reflective of tumor stage/histopathology and molecular profile. This study hypothesized that the molecular profile is an accurate prognostic adjunct in patients triaged based on clinico-pathological characteristics. Towards this effect, publically available micro-array datasets (n = 8), were downloaded, classified based on HPV association (n = 83) and site (tongue n = 88; laryngopharynx n = 53; oropharynx n = 51) and re-analyzed (Genespring; v13.1). The significant genes were validated in respective cohorts in The Cancer Genome Atlas (TCGA) for correlation with clinico-pathological parameters/survival. The gene entities (n = 3258) identified from HPV based analysis, when validated in TCGA identified the subset specifically altered in HPV+ HNSCC (n = 63), with three genes showing survival impact (RPP25, NUDCD2, NOVA1). Site-specific meta-analysis identified respective differentials (tongue: 3508, laryngopharynx: 4893, oropharynx: 2386); validation in TCGA revealed markers with high incidence (altered in >10% of patients) in tongue (n = 331), laryngopharynx (n = 701) and oropharynx (n = 404). Assessment of these genes in clinical sub-cohorts of TCGA indicated that early stage tongue (MTFR1, C8ORF33, OTUD6B) and laryngeal cancers (TWISTNB, KLHL13 and UBE2Q1) were defined by distinct prognosticators. Similarly, correlation with perineural/angiolymophatic invasion, identified discrete marker panels with survival impact (tongue: NUDCD1, PRKC1; laryngopharynx: SLC4A1AP, PIK3CA, AP2M1). Alterations in ANO1, NUDCD1, PIK3CA defined survival in tongue cancer patients with nodal metastasis (node+ECS-), while EPS8 is a significant differential in node+ECS- laryngopharyngeal cancers. In oropharynx, wherein HPV is a major etiological factor, distinct prognosticators were identified in HPV+ (ECHDC2, HERC5, GGT6) and HPV- (GRB10, EMILIN1, FNDC1). Meta-analysis in combination with TCGA validation carried out in this study emphasized on the molecular heterogeneity inherent within HNSCC; the feasibility of leveraging this information for improving prognostic efficacy is also established. Subject to large scale clinical validation, the marker panel identified in this study can prove to be valuable prognostic adjuncts.
Collapse
|
17
|
Li M, Yang J, Zhang L, Tu S, Zhou X, Tan Z, Zhou W, He Y, Li Y. A low-molecular-weight compound exerts anticancer activity against breast and lung cancers by disrupting EGFR/Eps8 complex formation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:211. [PMID: 31118055 PMCID: PMC6532146 DOI: 10.1186/s13046-019-1207-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 05/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and epidermal growth factor receptor pathway substrate 8 (Eps8) have been widely reported to be expressed in various tumors. Eps8 is an important active kinase substrate of EGFR that directly binds to the juxtamembrane (JXM) domain of EGFR to form an EGFR/Eps8 complex. The EGFR/Eps8 complex is involved in regulating cancer progression and might be an ideal target for antitumor therapy. This study focused on the screening of small-molecule inhibitors that target the EGFR/Eps8 complex in breast cancer and non-small cell lung cancer (NSCLC). METHODS In silico virtual screening was used to identify small-molecule EGFR/Eps8 complex inhibitors. These compounds were screened for the inhibition of A549 and BT549 cell viability. The direct interaction between EGFR and Eps8 was measured using coimmunoprecipitation (CoIP) and JXM domain replacement assays. The antitumor effects of the inhibitors were analyzed in cancer cells and xenograft models. An acute toxicity study of EE02 was performed in a mouse model. In addition, the effect of the EE02 inhibitor on the protein expression of elements downstream of the EGFR/Eps8 complex was determined by western blotting and protein chip assays. RESULTS In this study of nearly 390,000 compounds screened by virtual database screening, the top 29 compounds were identified as candidate small-molecule EGFR/Eps8 complex inhibitors and evaluated by using cell-based assays. The compound EE02 was identified as the best match to our selection criteria. Further investigation demonstrated that EE02 directly bound to the JXM domain of EGFR and disrupted EGFR/Eps8 complex formation. EE02 selectively suppressed growth and induced apoptosis in EGFR-positive and Eps8-positive breast cancer and NSCLC cells. More importantly, the PI3K/Akt/mTOR and MAPK/Erk pathways downstream of the EGFR/Eps8 complex were suppressed by EE02. In addition, the suppressive effect of EE02 on tumor growth in vivo was comparable to that of erlotinib at the same dose. CONCLUSIONS We identified EE02 as an EGFR/Eps8 complex inhibitor that demonstrated promising antitumor effects in breast cancer and NSCLC. Our data suggest that the EGFR/Eps8 complex offers a novel cancer drug target.
Collapse
Affiliation(s)
- Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Jilong Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Lenghe Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Xuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Ze Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Weijun Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China.
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
18
|
Wu M, Zhang L, Zhang H, Ning J, Tu S, He Y, Li Y. CD19 chimeric antigen receptor-redirected T cells combined with epidermal growth factor receptor pathway substrate 8 peptide-derived dendritic cell vaccine in leukemia. Cytotherapy 2019; 21:659-670. [PMID: 31031152 DOI: 10.1016/j.jcyt.2019.03.313] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy opens a new era for cancer treatment. However, in prolonged follow-up, relapse has emerged as one of the major obstacles. Dendritic cell (DC) vaccination is a promising treatment to eradicate tumor cells and prevent relapse. The epidermal growth factor receptor (EGFR) pathway substrate 8 (Eps8) gene is involved in regulating cancer progression and is considered an attractive target for specific cancer immunotherapy. The purpose of this study was to explore a combinatorial therapy using CAR-T cells and a DC vaccine such as Eps8-DCs to increase leukemia treatment efficacy. METHODS We pulsed DCs with Eps8-derived peptides to generate Eps8-DCs, engineered T cells to express a second-generation CAR specific for CD19, and analyzed the effects of the Eps8-DCs on the in vitro expansion, phenotype and effector functions of the CD19 CAR-T cells. RESULTS The Eps8-DCs significantly reduced the activation-induced cell death and enhanced the proliferative potential of CAR-T cells during in vitro expansion. In addition, the expanded T cells co-cultured with the Eps8-DCs exhibited an increased percentage of central memory T cells (Tcms) and a decreased percentage of effector memory T cells (Tems). The Eps8-DCs enhanced CD19 CAR-T cell immune functions, including cytokine production, CD107a degranulation activity and cytotoxicity. DISCUSSION This study demonstrates that Eps8-DCs exert synergistic effect on CD19 targeting CAR-T cells and paves the way for clinical trials using the combination of DC vaccination and engineered T cells in relapsed leukemia.
Collapse
Affiliation(s)
- Meirong Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Litian Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hanzhen Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingxuan Ning
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Hematology, Yuncheng Central Hospital, Shanxi Medical University, Yuncheng, China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
19
|
Fukuhisa H, Seki N, Idichi T, Kurahara H, Yamada Y, Toda H, Kita Y, Kawasaki Y, Tanoue K, Mataki Y, Maemura K, Natsugoe S. Gene regulation by antitumor miR-130b-5p in pancreatic ductal adenocarcinoma: the clinical significance of oncogenic EPS8. J Hum Genet 2019; 64:521-534. [DOI: 10.1038/s10038-019-0584-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022]
|
20
|
Beltrán-Anaya FO, Romero-Córdoba S, Rebollar-Vega R, Arrieta O, Bautista-Piña V, Dominguez-Reyes C, Villegas-Carlos F, Tenorio-Torres A, Alfaro-Riuz L, Jiménez-Morales S, Cedro-Tanda A, Ríos-Romero M, Reyes-Grajeda JP, Tagliabue E, Iorio MV, Hidalgo-Miranda A. Expression of long non-coding RNA ENSG00000226738 (LncKLHDC7B) is enriched in the immunomodulatory triple-negative breast cancer subtype and its alteration promotes cell migration, invasion, and resistance to cell death. Mol Oncol 2019; 13:909-927. [PMID: 30648789 PMCID: PMC6441920 DOI: 10.1002/1878-0261.12446] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/29/2018] [Accepted: 12/30/2018] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) represents an aggressive phenotype with poor prognosis compared with ER, PR, and HER2‐positive tumors. TNBC is a heterogeneous disease, and gene expression analysis has identified seven molecular subtypes. Accumulating evidence demonstrates that long non‐coding RNA (lncRNA) are involved in regulation of gene expression and cancer biology, contributing to essential cancer cell functions. In this study, we analyzed the expression profile of lncRNA in TNBC subtypes from 156 TNBC samples, and then characterized the functional role of LncKLHDC7B (ENSG00000226738). A total of 710 lncRNA were found to be differentially expressed between TNBC subtypes, and a subset of these altered lncRNA were independently validated. We discovered that LncKLHDC7B (ENSG00000226738) acts as a transcriptional modulator of its neighboring coding gene KLHDC7B in the immunomodulatory subtype. Furthermore, LncKLHDC7B knockdown enhanced migration and invasion, and promoted resistance to cellular death. Our findings confirmed the contribution of LncKLHDC7B to induction of apoptosis and inhibition of cell migration and invasion, suggesting that TNBC tumors with enrichment of LncKLHDC7B may exhibit distinct regulatory activity, or that this may be a generalized process in breast cancer. Additionally, in silico analysis confirmed for the first time that the low expression of KLHDC7B and LncKLHDC7B is associated with poor prognosis in patients with breast cancer.
Collapse
Affiliation(s)
- Fredy Omar Beltrán-Anaya
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Sandra Romero-Córdoba
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Department of Experimental Oncology and Molecular Medicine, Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosa Rebollar-Vega
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | | | | | | | | | - Luis Alfaro-Riuz
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Alberto Cedro-Tanda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Magdalena Ríos-Romero
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | | - Elda Tagliabue
- Department of Experimental Oncology and Molecular Medicine, Istituto Nazionale dei Tumori, Milan, Italy
| | - Marilena V Iorio
- Department of Experimental Oncology and Molecular Medicine, Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
21
|
MiR-590-3p Attenuates Acute Kidney Injury by Inhibiting Tumor Necrosis Factor Receptor-Associated Factor 6 in Septic Mice. Inflammation 2018; 42:637-649. [DOI: 10.1007/s10753-018-0921-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Ma Y, Bu D, Long J, Chai W, Dong J. LncRNA DSCAM‐AS1 acts as a sponge of miR‐137 to enhance Tamoxifen resistance in breast cancer. J Cell Physiol 2018; 234:2880-2894. [PMID: 30203615 DOI: 10.1002/jcp.27105] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/29/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Yun Ma
- Department of Breast SurgeryThe First Affiliated Hospital of Kunming Medical UniversityKunming Yunnan China
| | - Deyong Bu
- Department of Geriatric General SurgeryThe First Affiliated Hospital of Kunming Medical UniversityKunming Yunnan China
| | - Jiang Long
- Department of NeurosurgeryThe First Affiliated Hospital of Kunming Medical UniversityKunming Yunnan China
| | - Wenying Chai
- Department of Breast SurgeryThe First Affiliated Hospital of Kunming Medical UniversityKunming Yunnan China
| | - Jian Dong
- Yunnan Cancer HospitalThe Third Affiliated Hospital of Kunming Medical UniversityKunming Yunnan China
| |
Collapse
|
23
|
Yang L, Qiu J, Xiao Y, Hu X, Liu Q, Chen L, Huang W, Li X, Li L, Zhang J, Ding X, Xiang S. AP-2β inhibits hepatocellular carcinoma invasion and metastasis through Slug and Snail to suppress epithelial-mesenchymal transition. Theranostics 2018; 8:3707-3721. [PMID: 30026878 PMCID: PMC6037033 DOI: 10.7150/thno.25166] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/18/2018] [Indexed: 12/14/2022] Open
Abstract
Transcription factor AP-2β plays an important role in human cancer, but its clinical significance in hepatocellular carcinogenesis is largely unknown. Methods: AP-2β expression was detected in human hepatocellular cancer (HCC) tissues and cell lines. The effects of AP-2β on HCC proliferation, migration, invasion, tumor formation and metastasis were evaluated by MTT, colony formation and transwell assays in vitro and mouse experiments in vivo. The association between AP-2β and miR-27a/EMT markers in HCC cell lines and tissues was analyzed. Results: AP-2β expression was decreased in HCC tissues and cell lines. Reduced expression of AP-2β was significantly associated with more advanced tumor stages and larger tumor sizes. The overexpression of AP-2β reduced HCC proliferation, migration, invasion, tumor formation and metastasis in vitro and in vivo. Additionally, AP-2β overexpression increased the sensitivity of HCC cells to cisplatin. Moreover, AP-2β modulates the levels of EMT markers through Slug and Snail in HCC cell lines and tissues. Furthermore, oncogenic miR-27a inhibits AP-2β expression by binding to the AP-2β 3′ untranslated region (UTR) and reverses the tumor suppressive role of AP-2β. Conclusion: These results suggested that AP-2β is lowly expressed in HCC by inhibiting EMT signaling to regulate HCC cell growth and migration. Therefore, AP-2β in the novel miR-27a/AP-2β/Slug/EMT regulatory axis enhances the chemotherapeutic drug sensitivity of HCC and might represent a potential target for evaluating the treatment and prognosis of human HCC.
Collapse
|
24
|
A dual-function epidermal growth factor receptor pathway substrate 8 (Eps8)-derived peptide exhibits a potent cytotoxic T lymphocyte-activating effect and a specific inhibitory activity. Cell Death Dis 2018. [PMID: 29515106 PMCID: PMC5841361 DOI: 10.1038/s41419-018-0420-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The identification and characterization of tumor-associated antigens (TAAs) that generate specific cytotoxic T lymphocytes (CTLs) are vital to the development of cancer immunotherapy. The epidermal growth factor receptor (EGFR) pathway substrate 8 gene (Eps8) is involved in regulating cancer progression and might be an ideal antigen. In this study, we searched for novel human leukocyte antigen (HLA)-A*2402-restricted epitopes derived from the Eps8 protein via the HLA-binding prediction algorithm. Among four candidates, peptides 327 (EFLDCFQKF), 534 (KYAKSKYDF) and 755 (LFSLNKDEL) induced peptide-specific CTLs to secrete higher levels of interferon-gamma (IFN-γ) and showed enhanced cytotoxic activity against malignant cancer cells. Our results demonstrated that peptide-specific CTLs showed effective antitumor responses, including upregulation of interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF-α), granzyme B and perforin. Treatment with peptide-sensitized peripheral blood mononuclear cells (PBMCs) significantly reduced the tumor growth in vivo compared with the non-peptide-sensitized PBMC treatment. Importantly, our results indicated that peptide 327 may interfere with EGFR signaling by mechanistically disrupting Eps8/EGFR complex formation. We extended this observation that peptide 327 also suppressed the viability of cancer cells, blocked EGFR signal pathway and reduced the expression of downstream targets. Notably, conjugation of peptide 327 to the TAT sequence (TAT-327) resulted in potent antitumor activity and selective insertion into cancer cell membranes, where it adopted a punctate distribution. Furthermore, peptide 327 and TAT-327 displayed anticancer properties in xenograft models. Our results indicated that 327, 534 and 755 were novel HLA-A*2402-restricted epitopes from Eps8. By inhibiting the Eps8/EGFR interaction, peptide 327 and TAT-327 may serve as novel peptide inhibitors, which could provide an innovative approach for treating various cancers.
Collapse
|
25
|
Sun P, Zhou X, He Y, Liu H, Wang Y, Chen Y, Li M, He Y, Li G, Li Y. Effect of trichostatin A on Burkitt's lymphoma cells: Inhibition of EPS8 activity through Phospho-Erk1/2 pathway. Biochem Biophys Res Commun 2018; 497:990-996. [PMID: 29462617 DOI: 10.1016/j.bbrc.2018.02.128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/14/2018] [Indexed: 01/24/2023]
Abstract
Histone deacetylase inhibitors (HDACi) manifest great potential for treatment of Burkitt's lymphoma (BL), an aggressive B-cell lymphoma. Epidermal growth factor receptor pathway substrate 8 (EPS8) is confirmed overexpressed and associated with poor prognosis in solid tumors and leukemia. However, EPS8 expression and the relationship between EPS8 and HDACi on BL remains obscure. Here, we hypothesized that trichostatin A (TSA), a pan-HDACi, could inhibit BL cells by downregulating EPS8. We demonstrated that TSA reduced cell viability, induced apoptosis and cell arrest at G0/G1. Mechanismly, TSA attenuated EPS8 and downstream Phospho-Erk1/2 pathway. Knockdown of EPS8 resulted in a significant reduction in cellular proliferation and suppressed Phospho-Erk1/2 pathway activity, particularly when combined with TSA. Conversely, overexpression of EPS8 rescued this phenomenon. Then we showed that the combination of TSA and Epirubicin had a more significant effect when compared with TSA or Epirubicin alone. Finally, knockdown of EPS8 and TSA had a synergistic suppression effect on BALB/c nude mice. In conclusion, this study reveals that TSA affects BL cells by suppressing Phospho-Erk1/2 pathway through downregulating EPS8.
Collapse
Affiliation(s)
- Peipei Sun
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Xin Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Yingzhi He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Huimin Liu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Yuxin Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Guowei Li
- Department of Hematology, Huizhou Municipal Central Hospital, 41 Eling Road North, 516001, Huizhou, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| |
Collapse
|
26
|
Msaki A, Pastò A, Curtarello M, Arigoni M, Barutello G, Calogero RA, Macagno M, Cavallo F, Amadori A, Indraccolo S. A hypoxic signature marks tumors formed by disseminated tumor cells in the BALB-neuT mammary cancer model. Oncotarget 2018; 7:33081-95. [PMID: 27105499 PMCID: PMC5078077 DOI: 10.18632/oncotarget.8859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 03/31/2016] [Indexed: 12/29/2022] Open
Abstract
Metastasis is the final stage of cancer progression. Some evidence indicates that tumor cell dissemination occurs early in the natural history of cancer progression. Disseminated tumor cells (DTC) have been described in the bone marrow (BM) of cancer patients as well as in experimental models, where they correlate with later development of metastasis. However, little is known about the tumorigenic features of DTC obtained at different time points along tumor progression. Here, we found that early DTC isolated from BM of 15-17 week-old Her2/neu transgenic (BALB-neuT) mice were not tumorigenic in immunodeficient mice. In contrast, DTC-derived tumors were easily detectable when late DTC obtained from 19-22 week-old BALB-neuT mice were injected. Angiogenesis, which contributes to regulate tumor dormancy, appeared dispensable to reactivate late DTC, although it accelerated growth of secondary DTC tumors. Compared with parental mammary tumors, gene expression profiling disclosed a distinctive transcriptional signature of late DTC tumors which was enriched for hypoxia-related transcripts and was maintained in ex-vivo cell culture. Altogether, these findings highlight a different tumorigenic potential of early and late DTC in the BALB-neuT model and describe a HIF-1α-related transcriptional signature in DTC tumors, which may render DTC angiogenesis-competent, when placed in a favourable environment.
Collapse
Affiliation(s)
- Aichi Msaki
- Istituto Oncologico Veneto - IRCCS, Padova, Italy
| | - Anna Pastò
- Istituto Oncologico Veneto - IRCCS, Padova, Italy
| | | | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Raffaele Adolfo Calogero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alberto Amadori
- Istituto Oncologico Veneto - IRCCS, Padova, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | |
Collapse
|
27
|
A synthetic cell-penetrating peptide derived from nuclear localization signal of EPS8 exerts anticancer activity against acute myeloid leukemia. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:12. [PMID: 29357910 PMCID: PMC5778704 DOI: 10.1186/s13046-018-0682-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023]
Abstract
Background Oncogenic roles of epidermal growth factor receptor pathway substrate no.8 (EPS8) have been widely reported in various tumors, making targeting of EPS8 an appealing prospect. Here, we describe the role of EPS8 in acute myeloid leukemia (AML) and consider the potential of EPS8 as an anti-AML target. Nuclear localization signal (NLS) residues of tumor-associated proteins are crucial for cell cycle progression, and specific inhibitors derived from the NLS have inhibitory effect on cancer cells. The NLS in EPS8 has potential as a specific anti-AML target. Methods Gene Expression Omnibus expression profiles of AML patients were used to test associations between EPS8 expression and AML patient outcome. The biological characteristics of AML cells after EPS8 knockdown were analyzed in vitro and in vivo. A specific peptide (CP-EPS8-NLS) derived from the NLS of EPS8 (amino acids 298–310) was synthesized, and the anti-AML effects of CP-EPS8-NLS were analyzed in cancer cells and in xenograft models. Mutated CP-EPS8-NLS and penetratin served as controls. Results We observed that elevated EPS8 expression in AML patients is associated with poor outcome. Knockdown of EPS8 significantly suppressed the survival of AML cells in vitro and in vivo. CP-EPS8-NLS interfered with EPS8-associated signaling and consequently exerted anti-AML activity. Importantly, CP-EPS8-NLS displayed anti-AML activity in various AML cell types, with diminished activity in PBMCs. CP-ESP8-NLS suppressed U937 cell proliferation, and injection of CP-EPS8-NLS exerted potent antitumor activity in the xenograft tumor models. A synergistic effect of CP-EPS8-NLS and chemotherapeutic agents was also observed in vitro and in vivo. Mechanistically, treatment of various AML cells with CP-EPS8-NLS downregulated the expression of EPS8 and its downstream pathways. Conclusions The function of CP-EPS8-NLS is explained by the presence of a NLS in EPS8, which has been shown to induce nuclear translocation, consequently resulting in EPS8 overexpression. These results indicate that EPS8 is a potential target for AML treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0682-x) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Huang R, Liu H, Chen Y, He Y, Kang Q, Tu S, He Y, Zhou X, Wang L, Yang J, Wu A, Li Y. EPS8 regulates proliferation, apoptosis and chemosensitivity in BCR-ABL positive cells via the BCR-ABL/PI3K/AKT/mTOR pathway. Oncol Rep 2017; 39:119-128. [PMID: 29192326 PMCID: PMC5783592 DOI: 10.3892/or.2017.6102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023] Open
Abstract
Although the introduction of tyrosine kinase inhibitors greatly improved the survival of patients with chronic myeloid leukemia (CML), drug resistance remains a problem. Thus, mechanism-based novel therapeutic targets warrant exploration. Recently, epidermal growth factor receptor kinase substrate 8 (EPS8), which has been identified as an oncogene and plays an important role in a broad spectrum of solid tumours, was reported to be related to poor prognosis or chemoresistance in acute leukemia patients. However, its role in CML remains unclear. In the present study, using q-RT-PCR, we demonstrated that CML patients expressed a higher level of EPS8 mRNA in bone marrow mononuclear cells than healthy controls. Then, to determine the effect of EPS8 on the biological functions of CML cells, EPS8 expression was knocked down in the human CML cell line K562. Reduced proliferation, increased apoptosis, impaired adhesion and migration were observed in K562 cells after EPS8 silencing. Notably, attenuation of EPS8 increased chemosensitivity both in imatinib-sensitive K562 cells and in the imatinib-resistant murine BCR-ABL+ 32D-p210BCR/ABL-T315I cells. Mechanistically, knockdown of EPS8 downregulated p-BCR/ABL and its downstream AKT/mTOR signalling pathway. Finally, knockdown of EPS8 attenuated K562 cell proliferation in BALB/c nude mice. These data indicated that EPS8 regulated the proliferation, apoptosis and chemosensitivity in BCR-ABL positive cells via the BCR-ABL/PI3K/AKT/mTOR pathway. Targeting EPS8 alone or combined with a tyrosine kinase inhibitor may be a promising alternative therapeutic strategy.
Collapse
Affiliation(s)
- Rui Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Huimin Liu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Qian Kang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yingzhi He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Xuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Lei Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jilong Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Anqin Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
29
|
Shin WS, Shim HJ, Lee YH, Pyo M, Park JS, Ahn SY, Lee ST. PTK6 Localized at the Plasma Membrane Promotes Cell Proliferation and MigratiOn Through Phosphorylation of Eps8. J Cell Biochem 2017; 118:2887-2895. [PMID: 28214294 DOI: 10.1002/jcb.25939] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 02/16/2017] [Indexed: 12/26/2022]
Abstract
Protein tyrosine kinase 6 (PTK6; also known as Brk) is closely related to the Src family kinases, but lacks a membrane-targeting myristoylation signal. Sublocalization of PTK6 at the plasma membrane enhances its oncogenic potential. To understand the mechanism(s) underlying the oncogenic property of plasma---membrane-associated PTK6, proteins phosphorylated by membrane-targeted myristoylated PTK6 (Myr-PTK6) were enriched and analyzed using a proteomics approach. Eps8 which was identified by this method is phosphorylated by Myr-PTK6 in HEK293 cells. Mouse Eps8 expressed in HEK293 cells is phosphorylated by Myr-PTK6 at residues Tyr497, Tyr524, and Tyr534. Compared to wild-type Eps8 (Eps8 WT), the phosphorylation-defective 3YF mutant (Eps8 3YF) reverts the increased proliferation, migration, and phosphorylation of ERK and FAK mediated by Eps8 WT in HEK293 cells overexpressing PTK6. PTK6 knockdown in T-47D breast cancer cells decreased EGF-induced phosphorylation of Eps8. Endogenous PTK6 phosphorylates ectopically expressed Eps8 WT, but not Eps8 3YF mutant, in EGF-stimulated T-47D cells. The EGF-induced Eps8 phosphorylation enhances activation of ERK and FAK, cell adhesion, and anchorage-independent colony formation in T-47D cells, but not in the PTK6-knokdown T-47D cells. These results indicate that plasma-membrane-associated PTK6 phosphorylates Eps8, which promotes cell proliferation, adhesion, and migration and, thus, tumorigenesis. J. Cell. Biochem. 118: 2887-2895, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Won-Sik Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hyun Jae Shim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young Hun Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Minju Pyo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jun Sang Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - So Yun Ahn
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Wang F, Huang W, Hu X, Chen C, Li X, Qiu J, Liang Z, Zhang J, Li L, Wang X, Ding X, Xiang S, Zhang J. Transcription factor AP-2β suppresses cervical cancer cell proliferation by promoting the degradation of its interaction partner β-catenin. Mol Carcinog 2017; 56:1909-1923. [PMID: 28277615 DOI: 10.1002/mc.22646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Transcription factor AP-2β mediates the transcription of a number of genes implicated in mammalian development, cell proliferation, and carcinogenesis. Although the expression pattern of AP-2β has been analyzed in cervical cancer cell lines, the functions and molecular mechanism of AP-2β are unknown. Here, we found that AP-2β significantly inhibits TCF/LEF reporter activity. Moreover, AP-2β and β-catenin interact both in vitro through GST pull-down assays and in vivo by co-immunoprecipitation. We further identified the interaction regions to the DNA-binding domain of AP-2β and the 1-9 Armadillo repeats of β-catenin. Moreover, AP-2β binds with β-TrCP and promotes the degradation of endogenous β-catenin via the proteasomal degradation pathway. Immunohistochemistry analysis revealed a negative correlation between the two proteins in cervical cancer tissues and cell lines. Finally, functional analysis showed that AP-2β suppresses cervical cancer cell growth in vitro and in vivo by inhibiting the expression of Wnt downstream genes. Taken together, these findings demonstrated that AP-2β functions as a novel inhibitor of the Wnt/β-catenin signaling pathway in cervical cancer.
Collapse
Affiliation(s)
- Fangmei Wang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Wenhuan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Cheng Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Xinxin Li
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Junlu Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Zhongheng Liang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Jianmei Zhang
- Reproductive Medicine Center, Changsha Hospital for Maternal & Child Health Care, Changsha, Hunan, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoqing Wang
- Xiangya Second Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| |
Collapse
|
31
|
Huang S, Feng C, Chen L, Huang Z, Zhou X, Li B, Wang LL, Chen W, Lv FQ, Li TS. Identification of Potential Key Long Non-Coding RNAs and Target Genes Associated with Pneumonia Using Long Non-Coding RNA Sequencing (lncRNA-Seq): A Preliminary Study. Med Sci Monit 2016; 22:3394-3408. [PMID: 27663962 PMCID: PMC5040222 DOI: 10.12659/msm.900783] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background This study aimed to identify the potential key long non-coding RNAs (lncRNAs) and target genes associated with pneumonia using lncRNA sequencing (lncRNA-seq). Material/Methods A total of 9 peripheral blood samples from patients with mild pneumonia (n=3) and severe pneumonia (n=3), as well as volunteers without pneumonia (n=3), were received for lncRNA-seq. Based on the sequencing data, differentially expressed lncRNAs (DE-lncRNAs) were identified by the limma package. After the functional enrichment analysis, target genes of DE-lncRNAs were predicted, and the regulatory network was constructed. Results In total, 99 DE-lncRNAs (14 upregulated and 85 downregulated ones) were identified in the mild pneumonia group and 85 (72 upregulated and 13 downregulated ones) in the severe pneumonia group, compared with the control group. Among these DE-lncRNAs, 9 lncRNAs were upregulated in both the mild and severe pneumonia groups. A set of 868 genes were predicted to be targeted by these 9 DE-lncRNAs. In the network, RP11-248E9.5 and RP11-456D7.1 targeted the majority of genes. RP11-248E9.5 regulated several genes together with CTD-2300H10.2, such as QRFP and EPS8. Both upregulated RP11-456D7.1 and RP11-96C23.9 regulated several genes, such as PDK2. RP11-456D7.1 also positively regulated CCL21. Conclusions These novel lncRNAs and their target genes may be closely associated with the progression of pneumonia.
Collapse
Affiliation(s)
- Sai Huang
- Department of Hematology, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Cong Feng
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Li Chen
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Zhi Huang
- Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN, USA
| | - Xuan Zhou
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Bei Li
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Li-Li Wang
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Wei Chen
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| | - Fa-Qin Lv
- Department of Ultrasound, General Hospital of the PLA, Beijing, China (mainland)
| | - Tan-Shi Li
- Department of Emergency, General Hospital of the PLA, Beijing, China (mainland)
| |
Collapse
|
32
|
Hetmanski JHR, Zindy E, Schwartz JM, Caswell PT. A MAPK-Driven Feedback Loop Suppresses Rac Activity to Promote RhoA-Driven Cancer Cell Invasion. PLoS Comput Biol 2016; 12:e1004909. [PMID: 27138333 PMCID: PMC4854413 DOI: 10.1371/journal.pcbi.1004909] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/08/2016] [Indexed: 12/22/2022] Open
Abstract
Cell migration in 3D microenvironments is fundamental to development, homeostasis and the pathobiology of diseases such as cancer. Rab-coupling protein (RCP) dependent co-trafficking of α5β1 and EGFR1 promotes cancer cell invasion into fibronectin (FN) containing extracellular matrix (ECM), by potentiating EGFR1 signalling at the front of invasive cells. This promotes a switch in RhoGTPase signalling to inhibit Rac1 and activate a RhoA-ROCK-Formin homology domain-containing 3 (FHOD3) pathway and generate filopodial actin-spike protrusions which drive invasion. To further understand the signalling network that drives RCP-driven invasive migration, we generated a Boolean logical model based on existing network pathways/models, where each node can be interrogated by computational simulation. The model predicted an unanticipated feedback loop, whereby Raf/MEK/ERK signalling maintains suppression of Rac1 by inhibiting the Rac-activating Sos1-Eps8-Abi1 complex, allowing RhoA activity to predominate in invasive protrusions. MEK inhibition was sufficient to promote lamellipodia formation and oppose filopodial actin-spike formation, and led to activation of Rac and inactivation of RhoA at the leading edge of cells moving in 3D matrix. Furthermore, MEK inhibition abrogated RCP/α5β1/EGFR1-driven invasive migration. However, upon knockdown of Eps8 (to suppress the Sos1-Abi1-Eps8 complex), MEK inhibition had no effect on RhoGTPase activity and did not oppose invasive migration, suggesting that MEK-ERK signalling suppresses the Rac-activating Sos1-Abi1-Eps8 complex to maintain RhoA activity and promote filopodial actin-spike formation and invasive migration. Our study highlights the predictive potential of mathematical modelling approaches, and demonstrates that a simple intervention (MEK-inhibition) could be of therapeutic benefit in preventing invasive migration and metastasis. The majority of cancer-related fatalities are caused by the movement of cancer cells away from the primary site to form metastases, making understanding the signalling mechanisms which underpin cell migration and invasion through their local environment of paramount importance. Much has been discovered about key events leading to invasive cell migration. Here, we have taken this prior knowledge to build a powerful predictive model based on simple ON/OFF relations and logic to determine potential intervention targets to reduce harmful invasive migration. Interrogating our model, we have identified a negative feedback loop important to the signalling that determines invasive migration, the breaking of which reverts cells to a slower, less invasive phenotype. We have supported this feedback loop prediction using an array of in vitro experiments performed in cells within 2-D and physiologically relevant 3-D environments. Our findings demonstrate the predictive power of such modelling techniques, and could form the basis for clinical intervention to prevent metastasis in certain cancers.
Collapse
Affiliation(s)
- Joseph H. R. Hetmanski
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Egor Zindy
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Jean-Marc Schwartz
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Patrick T. Caswell
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Huang W, Chen C, Liang Z, Qiu J, Li X, Hu X, Xiang S, Ding X, Zhang J. AP-2α inhibits hepatocellular carcinoma cell growth and migration. Int J Oncol 2016; 48:1125-34. [PMID: 26780928 DOI: 10.3892/ijo.2016.3318] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/09/2015] [Indexed: 11/06/2022] Open
Abstract
Transcription factor AP-2α is involved in many types of human cancers, but its role in hepatocellular carcinogenesis is largely unknown. In this study, we found that expression of AP-2α was low in 40% of human hepatocellular cancers compared with adjacent normal tissues by immunohistochemical analysis. Moreover, AP-2α expression was low or absent in hepatocellular cancer cell lines (HepG2, Hep3B, SMMC-7721 and MHHC 97-H). Human liver cancer cell lines SMMC-7721 and Hep3B stably overexpressing AP-2α were established by lentiviral infection and puromycin screening, and the ectopic expression of AP-2α was able to inhibit hepatocellular cancer cell growth and proliferation by cell viability, MTT assay and liquid colony formation in vitro and in vivo. Furthermore, AP-2α overexpression decreased liver cancer cell migration and invasion as assessed by wound healing and Transwell assays, increasing the sensitivity of liver cancer cells to cisplatin analyzed by MTT assays. Also AP-2α overexpression suppressed the sphere formation and renewed the ability of cancer stem cells. Finally, we found that AP-2α is epigenetically modified and modulates the levels of phosphorylated extracellular signal-regulated protein kinase (ERK), β-catenin, p53, EMT, and CD133 expression in liver cancer cell lines. These results suggested that AP-2α expression is low in human hepatocellular cancers by regulating multiple signaling to affect hepatocellular cancer cell growth and migration. Therefore, AP-2α might represent a novel potential target in human hepatocellular cancer therapy.
Collapse
Affiliation(s)
- Wenhuan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Cheng Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhongheng Liang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Junlu Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xinxin Li
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
34
|
Silencing of Eps8 inhibits in vitro angiogenesis. Life Sci 2015; 131:30-6. [DOI: 10.1016/j.lfs.2015.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 02/25/2015] [Accepted: 03/19/2015] [Indexed: 11/21/2022]
|