1
|
Li P, Xie W, Wei H, Yang F, Chen Y, Li Y. Transcriptome Analyses of Liver Sinusoidal Endothelial Cells Reveal a Consistent List of Candidate Genes Associated with Endothelial Dysfunction and the Fibrosis Progression. Curr Issues Mol Biol 2024; 46:7997-8014. [PMID: 39194690 DOI: 10.3390/cimb46080473] [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] [Received: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Liver fibrosis is an important step in the transformation of chronic liver disease into cirrhosis and liver cancer, and structural changes and functional disorders of liver sinusoidal endothelial cells (LSECs) are early events in the occurrence of liver fibrosis. Therefore, it is necessary to identify the key regulatory genes of endothelial dysfunction in the process of liver fibrosis to provide a reference for the diagnosis and treatment of liver fibrosis. In this study, we identified 230 common differentially expressed genes (Co-DEGs) by analyzing transcriptomic data of primary LSECs from three different liver fibrosis mouse models (carbon tetrachloride; choline-deficient, l-amino acid-defined diet; and nonalcoholic steatohepatitis). Enrichment analysis revealed that the Co-DEGs were mainly involved in regulating the inflammatory response, immune response, angiogenesis, formation and degradation of the extracellular matrix, and mediating chemokine-related pathways. A Venn diagram analysis was used to identify 17 key genes related to the progression of liver cirrhosis. Regression analysis using the Lasso-Cox method identified genes related to prognosis among these key genes: SOX4, LGALS3, SERPINE2, CD52, and LPXN. In mouse models of liver fibrosis (bile duct ligation and carbon tetrachloride), all five key genes were upregulated in fibrotic livers. This study identified key regulatory genes for endothelial dysfunction in liver fibrosis, namely SOX4, LGALS3, SERPINE2, CD52, and LPXN, which will provide new targets for the development of therapeutic strategies targeting endothelial dysfunction in LSECs and liver fibrosis.
Collapse
Affiliation(s)
- Penghui Li
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenjie Xie
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Hongjin Wei
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Fan Yang
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yan Chen
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yinxiong Li
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangzhou 510530, China
| |
Collapse
|
2
|
Wu S, Yang Y, Zhang M, Khan AU, Dai J, Ouyang J. Serpin peptidase inhibitor, clade E, member 2 in physiology and pathology: recent advancements. Front Mol Biosci 2024; 11:1334931. [PMID: 38469181 PMCID: PMC10927012 DOI: 10.3389/fmolb.2024.1334931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/01/2024] [Indexed: 03/13/2024] Open
Abstract
Serine protease inhibitors (serpins) are the most numerous and widespread multifunctional protease inhibitor superfamily and are expressed by all eukaryotes. Serpin E2 (serpin peptidase inhibitor, clade E, member 2), a member of the serine protease inhibitor superfamily is a potent endogenous thrombin inhibitor, mainly found in the extracellular matrix and platelets, and expressed in numerous organs and secreted by many cell types. The multiple functions of serpin E2 are mainly mediated through regulating urokinase-type plasminogen activator (uPA, also known as PLAU), tissue-type plasminogen activator (tPA, also known as PLAT), and matrix metalloproteinase activity, and include hemostasis, cell adhesion, and promotion of tumor metastasis. The importance serpin E2 is clear from its involvement in numerous physiological and pathological processes. In this review, we summarize the structural characteristics of the Serpin E2 gene and protein, as well as its roles physiology and disease.
Collapse
Affiliation(s)
- Shutong Wu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University), National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Xinjin Branch of Chengdu Municipal Public Security Bureau, Chengdu, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University), National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Yue Bei People’s Hospital Postdoctoral Innovation Practice Base, Southern Medical University, Guangzhou, China
| | - Meiling Zhang
- Chengdu Municipal Public Security Bureau Wenjiang Branch, Chengdu, China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University), National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University), National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University), National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Liu Y, Li X, Chen S, Zhu C, Shi Y, Dang S, Zhang W, Li W. Pan-cancer analysis of SERPINE family genes as biomarkers of cancer prognosis and response to therapy. Front Mol Biosci 2024; 10:1277508. [PMID: 38274096 PMCID: PMC10808646 DOI: 10.3389/fmolb.2023.1277508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Background: Serine protease inhibitor E (SERPINE) family genes participate in the tumor growth, cancer cell survival and metastasis. However, the SERPINE family members role in the prognosis and their clinical therapeutic potentials in various human cancer types have not been elaborately explored. Methods: We preliminarily analyzed expression levels and prognostic values of SERPINE family genes, and investigated the correlation between SERPINEs expression and tumor microenvironment (TME), Stemness score, clinical characteristic, immune infiltration, tumor mutational burden (TMB), immune subtype, and drug sensitivity in pan-cancer, which based on updated public databases and integrated some bioinformatics analysis methods. In addition, we conducted the enrichment analysis of SERPINEs from DAVID and KOBAS databases. Results: SERPINE1, SERPINE2, and SERPINE3 expression were upregulated in nine cancers, twelve cancers, and six cancers, respectively. The expression of SERPINE family genes was associated with the prognosis in several cancers from The Cancer Genome Atlas (TCGA). Furthermore, SERPINE family genes expression also had a significant relation to stromal and immune scores, and RNA stemness score and DNA stemness score in pan-cancer. SERPINE1 and SERPINE2 expression significantly increased in tumor advanced stage in colon adenocarcinoma (COAD). Results showed that SERPINE1 and SERPINE2 expression were negatively related with B cells and Monocytes, respectively. SERPINE2 expression had a significantly positive relation with B cells and Macrophages. In terms of TMB, SERPINE1, SERPINE2, and SERPINE3 were found to associated with TMB in seven cancers, fourteen cancers, and four cancers, respectively. Moreover, all SERPINE gene family members were significantly correlated with immune subtypes. SERPINE1 expression had a significantly positive or negative correlation with drug sensitivity. Conclusion: The study indicated the great potential of SERPINE family genes as biomarkers for prognosis and provided valuable strategies for further investigation of SERPINE family genes as potential targets in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Li
- Department of Cancer Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Chuang HW, Lin LH, Ji DD, Fu TY, Lee HS, Yang YF, Tseng HC, Hsia KT. Serpin peptidase inhibitor, clade E, member 2 is associated with malignant progression and clinical prognosis in oral squamous cell carcinoma. J Dent Sci 2024; 19:70-78. [PMID: 38303830 PMCID: PMC10829680 DOI: 10.1016/j.jds.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/19/2023] [Indexed: 02/03/2024] Open
Abstract
Background/purpose The serpin peptidase inhibitor, clade E, member 2 (SERPINE2), is upregulated in breast cancer, prostate cancer, and urothelial carcinoma; however, limited information exists regarding its expression in oral cancer. Therefore, this study aimed to analyze the association between SERPINE2 expression and oral squamous cell carcinoma (OSCC) outcomes. Materials and methods SERPINE2 mRNA and protein expression in patients with head and neck squamous cell carcinoma and OSCC were investigated using online databases and tissue-array analysis. Its relationship with clinicopathological characteristics, OSCC prognosis and its biological function in OSCC cells were explored. Results Analysis using online databases revealed higher SERPINE2 expression in tumor tissues and its role as a prognostic factor. High SERPINE2 protein levels were significantly correlated with adverse pathological parameters, including advanced clinical stage and tumor status (P < 0.001), lymph nodes (P = 0.014), and distant metastases (P = 0.013). High SERPINE2 expression was associated with worse overall survival (P < 0.001) and was identified as an independent prognostic factor for OSCC. In vitro studies revealed that SERPINE2 knockdown significantly reduced cell proliferation, migration, and invasion in OSCC cell lines. Conclusion This study suggests that SERPINE2 may serve as a prognostic biomarker and potential therapeutic target for oral cancer.
Collapse
Affiliation(s)
- Hao-Wen Chuang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Han Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Dar-Der Ji
- Department of Tropical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Ying Fu
- Department of Pathology, Yuan's General Hospital, Kaohsiung, Taiwan
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hsing-Cheng Tseng
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Kan-Tai Hsia
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
5
|
Zhang D, Sun R, Di C, Li L, Zhao F, Han Y, Zhang W. Microdissection of cancer-associated fibroblast infiltration subtypes unveils the secreted SERPINE2 contributing to immunosuppressive microenvironment and immuotherapeutic resistance in gastric cancer: A large-scale study integrating bulk and single-cell transcriptome profiling. Comput Biol Med 2023; 166:107406. [PMID: 37729702 DOI: 10.1016/j.compbiomed.2023.107406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/23/2023] [Accepted: 08/26/2023] [Indexed: 09/22/2023]
Abstract
In the era of immunotherapy, the suboptimal response rate and the development of acquired resistance among the initial beneficiaries continue to present significant challenges across multiple malignancies, including gastric cancer (GC). Considering that the interactions of tumor stroma, especially the cancer-associated fibroblasts (CAFs), with immune and tumor cells, play indispensable roles in tumor progression, tumor microenvironment remodeling and therapeutic responsiveness, in-depth exploration on the roles of CAFs and pivotal mediators of their functions may provide novel clues to increase the effectiveness of current immunotherapeutic drugs and further achieve synergistic antitumor response. Herein, through the consensus clustering of canonical biomarkers, three GC subclasses with different abundance of CAFs were virtually microdissected in four integrated bulk cohorts encompassing 2148 GC patients from 11 independent datasets. An extensive immunogenomic analysis revealed that tumors with high CAFs infiltration were characterized with unfavorable outcomes, aggressive phenotypes, decreased tumor immunogenicity, high risk of immune evasion and thus immunotherapeutic resistance. By leveraging large-scale single-cell transcriptomic profiling, a series of CAF-secreted proteins were identified, among which the SERPINE2 was confirmed to be restrictively enriched in stromal fibroblasts of GC tissues and contribute to promoting a protumor milieu and fostering an immunosuppressive microenvironment via bioinformatics computations and tissue microarray analysis. Moreover, pan-cancer investigations generalized the immunological roles of SERPINE2, especially in pan-gastrointestinal malignancies, with multiple real-world immunotherapy cohorts further confirming its implications on predicting immunotherapeutic efficacy. In conclusion, these findings suggest that the CAF-derived SERPINE2 is a promising immune-oncology target with therapeutic implications to further synergize the immunotherapeutic combinations.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Breast and Thyroid Surgery, General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Breast and Thyroid Surgery, General Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China; Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China; Department of Clinical Medicine, The First Clinical College, Shandong University, Jinan, Shandong, 250012, China.
| | - Rui Sun
- Department of Clinical Medicine, The First Clinical College, Shandong University, Jinan, Shandong, 250012, China; Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Chenyu Di
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China; Department of Clinical Medicine, The First Clinical College, Shandong University, Jinan, Shandong, 250012, China
| | - Lin Li
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250000, China
| | - Faming Zhao
- Key Laboratory of Environmental Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Han
- Department of Pathology, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Wenjie Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250011, China; Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250011, China.
| |
Collapse
|
6
|
Zhou B, Mo Z, Lai G, Chen X, Li R, Wu R, Zhu J, Zheng F. Targeting tumor exosomal circular RNA cSERPINE2 suppresses breast cancer progression by modulating MALT1-NF-𝜅B-IL-6 axis of tumor-associated macrophages. J Exp Clin Cancer Res 2023; 42:48. [PMID: 36797769 PMCID: PMC9936722 DOI: 10.1186/s13046-023-02620-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have important regulatory functions in cancer, but the role of circRNAs in the tumor microenvironment (TME) remains unclear. Moreover, we also explore the effects of si-circRNAs loaded in nanoparticles as therapeutic agent for anti-tumor in vivo. METHODS We conducted bioinformatics analysis, qRT-PCR, EdU assays, Transwell assays, co-culture system and multiple orthotopic xenograft models to investigate the expression and function of circRNAs. Additionally, PLGA-based nanoparticles loaded with si-circRNAs were used to evaluate the potential of nanotherapeutic strategy in anti-tumor response. RESULTS We identified oncogene SERPINE2 derived circRNA, named as cSERPINE2, which was notably elevated in breast cancer and was closely related to poor clinical outcome. Functionally, tumor exosomal cSERPINE2 was shuttled to tumor associated macrophages (TAMs) and enhanced the secretion of Interleukin-6 (IL-6), leading to increased proliferation and invasion of breast cancer cells. Furthermore, IL-6 in turn increased the EIF4A3 and CCL2 levels within tumor cells in a positive feedback mechanism, further enhancing tumor cSERPINE2 biogenesis and promoting the recruitment of TAMs. More importantly, we developed a PLGA-based nanoparticle loaded with si-cSERPINE2, which effectively attenuated breast cancer progression in vivo. CONCLUSIONS Our study illustrates a novel mechanism that tumor exosomal cSERPINE2 mediates a positive feedback loop between tumor cells and TAMs to promote cancer progression, which may serve as a promising nanotherapeutic strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Boxuan Zhou
- grid.452437.3Department of Breast Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000 China ,grid.412536.70000 0004 1791 7851Medical Research Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Zhaohong Mo
- grid.412558.f0000 0004 1762 1794Department of Hepatobiliary Surgery, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630 China
| | - Guie Lai
- grid.452437.3Department of Breast Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000 China
| | - Xiaohong Chen
- grid.452437.3Department of Laboratory, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000 China
| | - Ruixi Li
- grid.12981.330000 0001 2360 039XDepartment of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033 China
| | - Runxin Wu
- grid.12981.330000 0001 2360 039XZhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Jia Zhu
- Department of Breast Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
| | - Fang Zheng
- Medical Research Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
7
|
Chen WJ, Dong KQ, Pan XW, Gan SS, Xu D, Chen JX, Chen WJ, Li WY, Wang YQ, Zhou W, Rini B, Cui XG. Single-cell RNA-seq integrated with multi-omics reveals SERPINE2 as a target for metastasis in advanced renal cell carcinoma. Cell Death Dis 2023; 14:30. [PMID: 36646679 PMCID: PMC9842647 DOI: 10.1038/s41419-023-05566-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
Tumor growth, metastasis and therapeutic response are believed to be regulated by the tumor and its microenvironment (TME) in advanced renal cell carcinoma (RCC). However, the mechanisms underlying genomic, transcriptomic and epigenetic alternations in RCC progression have not been completely defined. In this study, single-cell RNA-sequencing (scRNA-seq) data were obtained from eight tissue samples of RCC patients, including two matched pairs of primary and metastatic sites (lymph nodes), along with Hi-C, transposable accessible chromatin by high-throughput (ATAC-seq) and RNA-sequencing (RNA-seq) between RCC (Caki-1) and human renal tubular epithelial cell line (HK-2). The identified target was verified in clinical tissue samples (microarray of 407 RCC patients, TMA-30 and TMA-2020), whose function was further validated by in vitro and in vivo experiments through knockdown or overexpression. We profiled transcriptomes of 30514 malignant cells, and 14762 non-malignant cells. Comprehensive multi-omics analysis revealed that malignant cells and TME played a key role in RCC. The expression programs of stromal cells and immune cells were consistent among the samples, whereas malignant cells expressed distinct programs associated with hypoxia, cell cycle, epithelial differentiation, and two different metastasis patterns. Comparison of the hierarchical structure showed that SERPINE2 was related to these NNMF expression programs, and at the same time targeted the switched compartment. SERPINE2 was highly expressed in RCC tissues and lowly expressed in para-tumor tissues or HK-2 cell line. SERPINE2 knockdown markedly suppressed RCC cell growth and invasion, while SERPINE2 overexpression dramatically promoted RCC cell metastasis both in vitro and in vivo. In addition, SERPINE2 could activate the epithelial-mesenchymal transition pathway. The above findings demonstrated that the role of distinct expression patterns of malignant cells and TME played a distinct role in RCC progression. SERPINE2 was identified as a potential therapeutic target for inhibiting metastasis in advanced RCC.
Collapse
Affiliation(s)
- Wen-Jin Chen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Ke-Qin Dong
- Department of Urology, General Hospital of Central Theater Command of PLA, Wuhan, 430070, China
| | - Xiu-Wu Pan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Si-Shun Gan
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Da Xu
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Jia-Xin Chen
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Wei-Jie Chen
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Wen-Yan Li
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yu-Qi Wang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wang Zhou
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Brian Rini
- Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Xin-Gang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China.
| |
Collapse
|
8
|
Mondal P, Patel NS, Bailey K, Aravind S, Cartwright SB, Hollingsworth MA, Lazenby AJ, Carlson MA. Induction of pancreatic neoplasia in the KRAS/TP53 Oncopig. Dis Model Mech 2023; 16:286617. [PMID: 36579622 PMCID: PMC9884120 DOI: 10.1242/dmm.049699] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022] Open
Abstract
The 5-year survival of pancreatic cancer (PC) remains low. Murine models may not adequately mimic human PC and can be too small for medical device development. A large-animal PC model could address these issues. We induced and characterized pancreatic tumors in Oncopigs (transgenic swine containing KRASG12D and TP53R167H). The oncopigs underwent injection of adenovirus expressing Cre recombinase (AdCre) into one of the main pancreatic ducts. Resultant tumors were characterized by histology, cytokine expression, exome sequencing and transcriptome analysis. Ten of 14 Oncopigs (71%) had gross tumor within 3 weeks. At necropsy, all of these subjects had gastric outlet obstruction secondary to pancreatic tumor and phlegmon. Oncopigs with injections without Cre recombinase and wild-type pigs with AdCre injection did not show notable effect. Exome and transcriptome analysis of the porcine pancreatic tumors revealed similarity to the molecular signatures and pathways of human PC. Although further optimization and validation of this porcine PC model would be beneficial, it is anticipated that this model will be useful for focused research and development of diagnostic and therapeutic technologies for PC. This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Neesha S. Patel
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Katie Bailey
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Shruthishree Aravind
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Sara B. Cartwright
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Audrey J. Lazenby
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA,Department of Surgery and VA Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA,Author for correspondence ()
| |
Collapse
|
9
|
LHX2 Enhances the Malignant Phenotype of Esophageal Squamous Cell Carcinoma by Upregulating the Expression of SERPINE2. Genes (Basel) 2022; 13:genes13081457. [PMID: 36011368 PMCID: PMC9408536 DOI: 10.3390/genes13081457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 12/01/2022] Open
Abstract
LHX2 dysregulations have been found to present in cancers, but the function of LHX2 in esophageal squamous cell carcinoma (ESCC) remains unknown. Here, we report that LHX2 was upregulated in ESCC tissues in comparison to the LHX2 levels in adjacent normal tissues. Loss- and gain-of-function experiments demonstrated that the knockdown of LHX2 markedly inhibited ESCC cells’ proliferation, migration, invasion, tumor growth and metastasis, whereas the overexpression of LHX2 had the opposite effects. A mechanistic investigation revealed that LHX2 bound to the promoter of SERPINE2 gene and transcriptionally regulated the expression of SERPINE2. Collectively, LHX2 facilitates ESCC tumor progression, and it could be a potential therapeutic target for ESCC.
Collapse
|
10
|
Li X, Shi X, Mesalam NM, Liu L, Chen Z, Yang B. Mechanism of Lysoforte in Improving Jejuna Morphology and Health in Broiler Chickens. Front Vet Sci 2022; 9:946148. [PMID: 35928108 PMCID: PMC9343761 DOI: 10.3389/fvets.2022.946148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Lysoforte (LFT) plays a vital role in maintaining broilers' health and intestinal morphology. However, the mechanism behind the effects of LFT improving intestinal morphology and health is still unclear. Therefore, this study was implemented to explore the central genes linked to the regulatory effect of LFT. Seventy-five newly hatched Cobb 500 male broilers were randomly divided into three groups: control, LFT500, and LFT1000 groups, with 25 chicks per group. The control chicks were provided with the basal diet, and the birds in LFT500 and LFT1000 groups were offered the same basal diet with 500 g/ton and 1,000 g/ton LFT, respectively. GSE94622 dataset consisted of the control and two LFT-treated groups (LFT500 and LFT1000). Jejuna samples were obtained from Gene Expression Omnibus (GEO). Totally 106–344 DEGs were obtained by comparing LFT500 and LFT1000 vs. control and LFT1000 vs. LFT500. Gene ontology (GO) enrichment suggested that the DEGs are mainly related to the phosphatidylethanolamine biosynthetic process and neuron projection extension. KEGG analysis suggested the DEGs were enriched in AGE-RAGE, fatty acid elongation, ECM-receptor interaction (ECMRI), glycerophospholipid metabolism, focal adhesion, unsaturated fatty acids biosynthesis, and ABC transporters. Moreover, 29 genes, such as REG4, GJB1, KAT2A, APOA5, SERPINE2, ELOVL1, ABCC2, ANKRD9, CYP4V2, and PISD, might be closely related to promoting jejuna morphology in broilers. Taken together, our observation enhances the understanding of LFT in maintaining intestinal architecture and the general health of broiler chickens.
Collapse
Affiliation(s)
- Xiaofeng Li
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| | - Xiaoli Shi
- College of Animal Science, Guizhou University, Guiyang, China
| | - Noura M. Mesalam
- Biological Applications Department, Nuclear Research Center, Egyptian Atomic Energy Authority, Abu-Zaabal, Egypt
| | - Lei Liu
- Center of Reproductive Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhihao Chen
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| | - Bing Yang
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
- *Correspondence: Bing Yang
| |
Collapse
|
11
|
Zhang J, Wu Q, Zhu L, Xie S, Tu L, Yang Y, Wu K, Zhao Y, Wang Y, Xu Y, Chen X, Ma S, Zhang S. SERPINE2/PN-1 regulates the DNA damage response and radioresistance by activating ATM in lung cancer. Cancer Lett 2022; 524:268-283. [PMID: 34648881 DOI: 10.1016/j.canlet.2021.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/20/2021] [Accepted: 10/03/2021] [Indexed: 12/12/2022]
Abstract
Although the DNA damage response (DDR) is associated with the radioresistance characteristics of lung cancer cells, the specific regulators and underlying mechanisms of the DDR are unclear. Here, we identified the serine proteinase inhibitor clade E member 2 (SERPINE2) as a modulator of radiosensitivity and the DDR in lung cancer. Cells exhibiting radioresistance after ionizing radiation show upregulation of SERPINE2, and SERPINE2 knockdown improves tumor radiosensitivity in vitro and in vivo. Functionally, SERPINE2 deficiency causes a reduction in homologous recombination repair, rapid recovery of cell cycle checkpoints, and suppression of migration and invasion. Mechanistically, SERPINE2 knockdown inhibits the accumulation of p-ATM and the downstream repair protein RAD51 during DNA repair, and RAD51 can restore DNA damage and radioresistance phenotypes in lung cancer cells. Furthermore, SERPINE2 can directly interact with MRE11 and ATM to facilitate its phosphorylation in HR-mediated DSB repair. In addition, high SERPINE2 expression correlates with dismal prognosis in lung adenocarcinoma patients, and a high serum SERPINE2 concentration predicts a poor response to radiotherapy in non-small cell lung cancer patients. In summary, these findings indicate a novel regulatory mechanism by which SERPINE2 modulates the DDR and radioresistance in lung cancer.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Qiong Wu
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Lucheng Zhu
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shujun Xie
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Linglan Tu
- Center for Molecular Medicine, Hangzhou Medical College, Hangzhou, 310013, China
| | - Yuhong Yang
- Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Kan Wu
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yanyan Zhao
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Yuqing Wang
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Yasi Xu
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Xueqin Chen
- Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China
| | - Shenglin Ma
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Department of Cancer Medical Center, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311201, China.
| | - Shirong Zhang
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, 310006, China; Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Cancer Center, Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
12
|
Chuang HW, Hsia KT, Liao JB, Yeh CC, Kuo WT, Yang YF. SERPINE2 Overexpression Is Associated with Poor Prognosis of Urothelial Carcinoma. Diagnostics (Basel) 2021; 11:diagnostics11101928. [PMID: 34679626 PMCID: PMC8535068 DOI: 10.3390/diagnostics11101928] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 01/17/2023] Open
Abstract
Recent studies have reported that SERPINE2 contributes to the development of various cancers. However, its association with urothelial carcinoma (UC) remains unclear. In this study, data on urinary bladder UC (UBUC) cases from The Cancer Genome Atlas (TCGA) database were used to investigate the prognostic value of SERPINE2 mRNA expression. Then, SERPINE2 expression was analyzed with tissue microarrays constructed from 117 upper tract UC (UTUC) and 84 UBUC tissue specimens using immunohistochemical staining. Results were compared to clinicopathologic data by multivariate analysis. In the TCGA database, high SERPINE2 mRNA expression indicated a poor prognosis in patients with UBUC. Furthermore, Mann-Whitney U test showed that high SERPINE2 immunoexpression was significantly associated with adverse pathologic parameters including invasion, high grade, coexistence of UC in situ, and advanced pT stage (all p < 0.05, except for a marginal association with high-grade UBUC, p = 0.066). Kaplan-Meier analysis revealed that high SERPINE2 expression was associated with worse overall survival (OS; UTUC, p = 0.003; UBUC, p = 0.014) and disease-free survival (UTUC, p = 0.031; UBUC, p = 0.033). Moreover, multivariate analysis identified high SERPINE2 expression as an independent prognostic factor for OS (UTUC, p = 0.002; UBUC, p = 0.024). Taken together, our findings demonstrated that increased SERPINE2 expression is associated with adverse pathologic features and may serve as a prognostic biomarker for UC.
Collapse
Affiliation(s)
- Hao-Wen Chuang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
- Institute of Oral Biology, School of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Kan-Tai Hsia
- Institute of Oral Biology, School of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
| | - Chih-Ching Yeh
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (H.-W.C.); (J.-B.L.); (C.-C.Y.)
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung 82144, Taiwan
| | - Wei-Ting Kuo
- Division of Urology, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: (W.-T.K.); (Y.-F.Y.)
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Correspondence: (W.-T.K.); (Y.-F.Y.)
| |
Collapse
|
13
|
NCOR1 Sustains Colorectal Cancer Cell Growth and Protects against Cellular Senescence. Cancers (Basel) 2021; 13:cancers13174414. [PMID: 34503224 PMCID: PMC8430780 DOI: 10.3390/cancers13174414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary NCOR1 is a scaffold protein that interacts with multiple partners to repress gene transcription. NCOR1 controls immunometabolic functions in several tissues and has been recently shown to protect against experimental colitis in mice. Our laboratory has observed a pro-proliferative role of NCOR1 in normal intestinal epithelial cells. However, it is unclear whether NCOR1 is functionally involved in colon cancer. This study demonstrated that NCOR1 is required for colorectal cancer cell growth. Depletion of NCOR1 caused these cells to become senescent. Transcriptomic signatures confirmed these observations but also predicted the potential for these cells to become pro-invasive. Thus, NCOR1 plays a novel role in preventing cancer-associated senescence and could represent a target for controlling colon cancer progression. Abstract NCOR1 is a corepressor that mediates transcriptional repression through its association with nuclear receptors and specific transcription factors. Some evidence supports a role for NCOR1 in neonatal intestinal epithelium maturation and the maintenance of epithelial integrity during experimental colitis in mice. We hypothesized that NCOR1 could control colorectal cancer cell proliferation and tumorigenicity. Conditional intestinal epithelial deletion of Ncor1 in ApcMin/+ mice resulted in a significant reduction in polyposis. RNAi targeting of NCOR1 in Caco-2/15 and HT-29 cell lines led to a reduction in cell growth, characterized by cellular senescence associated with a secretory phenotype. Tumor growth of HT-29 cells was reduced in the absence of NCOR1 in the mouse xenografts. RNA-seq transcriptome profiling of colon cancer cells confirmed the senescence phenotype in the absence of NCOR1 and predicted the occurrence of a pro-migration cellular signature in this context. SOX2, a transcription factor essential for pluripotency of embryonic stem cells, was induced under these conditions. In conclusion, depletion of NCOR1 reduced intestinal polyposis in mice and caused growth arrest, leading to senescence in human colorectal cell lines. The acquisition of a pro-metastasis signature in the absence of NCOR1 could indicate long-term potential adverse consequences of colon-cancer-induced senescence.
Collapse
|
14
|
Unveiling the Roles of Low-Density Lipoprotein Receptor-Related Protein 6 in Intestinal Homeostasis, Regeneration and Oncogenesis. Cells 2021; 10:cells10071792. [PMID: 34359960 PMCID: PMC8307932 DOI: 10.3390/cells10071792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022] Open
Abstract
Intestinal epithelial self-renewal is tightly regulated by signaling pathways controlling stem cell proliferation, determination and differentiation. In particular, Wnt/β-catenin signaling controls intestinal crypt cell division, survival and maintenance of the stem cell niche. Most colorectal cancers are initiated by mutations activating the Wnt/β-catenin pathway. Wnt signals are transduced through Frizzled receptors and LRP5/LRP6 coreceptors to downregulate GSK3β activity, resulting in increased nuclear β-catenin. Herein, we explored if LRP6 expression is required for maintenance of intestinal homeostasis, regeneration and oncogenesis. Mice with an intestinal epithelial cell-specific deletion of Lrp6 (Lrp6IEC-KO) were generated and their phenotype analyzed. No difference in intestinal architecture nor in proliferative and stem cell numbers was found in Lrp6IEC-KO mice in comparison to controls. Nevertheless, using ex vivo intestinal organoid cultures, we found that LRP6 expression was critical for crypt cell proliferation and stem cell maintenance. When exposed to dextran sodium sulfate, Lrp6IEC-KO mice developed more severe colitis than control mice. However, loss of LRP6 did not affect tumorigenesis in ApcMin/+ mice nor growth of human colorectal cancer cells. By contrast, Lrp6 silencing diminished anchorage-independent growth of BRafV600E-transformed intestinal epithelial cells (IEC). Thus, LRP6 controls intestinal stem cell functionality and is necessary for BRAF-induced IEC oncogenesis.
Collapse
|
15
|
Sohail M, Shkreta L, Toutant J, Rabea S, Babeu JP, Huard C, Coulombe-Huntington J, Delannoy A, Placet M, Geha S, Gendron FP, Boudreau F, Tyers M, Grierson DS, Chabot B. A novel class of inhibitors that target SRSF10 and promote p53-mediated cytotoxicity on human colorectal cancer cells. NAR Cancer 2021; 3:zcab019. [PMID: 34316707 PMCID: PMC8210162 DOI: 10.1093/narcan/zcab019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/22/2021] [Accepted: 05/04/2021] [Indexed: 01/07/2023] Open
Abstract
The elevated expression of the splicing regulator SRSF10 in metastatic colorectal cancer (CRC) stimulates the production of the pro-tumorigenic BCLAF1-L splice variant. We discovered a group of small molecules with an aminothiazole carboxamide core (GPS167, GPS192 and others) that decrease production of BCLAF1-L. While additional alternative splicing events regulated by SRSF10 are affected by GPS167/192 in HCT116 cells (e.g. in MDM4, WTAP, SLK1 and CLK1), other events are shifted in a SRSF10-independent manner (e.g. in MDM2, NAB2 and TRA2A). GPS167/192 increased the interaction of SRSF10 with the CLK1 and CLK4 kinases, leading us to show that GPS167/192 can inhibit CLK kinases preferentially impacting the activity of SRSF10. Notably, GPS167 impairs the growth of CRC cell lines and organoids, inhibits anchorage-independent colony formation, cell migration, and promotes cytoxicity in a manner that requires SRSF10 and p53. In contrast, GPS167 only minimally affects normal colonocytes and normal colorectal organoids. Thus, GPS167 reprograms the tumorigenic activity of SRSF10 in CRC cells to elicit p53-dependent apoptosis.
Collapse
Affiliation(s)
- Muhammad Sohail
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Lulzim Shkreta
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Johanne Toutant
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Safwat Rabea
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jean-Philippe Babeu
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Caroline Huard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | | | - Aurélie Delannoy
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Morgane Placet
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
| | - Sameh Geha
- Department of Pathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche Clinique du CHUS, CIUSSS de l’Estrie, Sherbrooke, QC, Canada
| | - Fernand-Pierre Gendron
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
- Centre de Recherche Clinique du CHUS, CIUSSS de l’Estrie, Sherbrooke, QC, Canada
| | - François Boudreau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
- Centre de Recherche Clinique du CHUS, CIUSSS de l’Estrie, Sherbrooke, QC, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - David S Grierson
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke. Sherbrooke, Quebec, Canada
- Centre de Recherche Clinique du CHUS, CIUSSS de l’Estrie, Sherbrooke, QC, Canada
| |
Collapse
|
16
|
Sivasudhan E, Blake N, Lu ZL, Meng J, Rong R. Dynamics of m6A RNA Methylome on the Hallmarks of Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:642443. [PMID: 33869193 PMCID: PMC8047153 DOI: 10.3389/fcell.2021.642443] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Epidemiological data consistently rank hepatocellular carcinoma (HCC) as one of the leading causes of cancer-related deaths worldwide, often posing severe economic burden on health care. While the molecular etiopathogenesis associated with genetic and epigenetic modifications has been extensively explored, the biological influence of the emerging field of epitranscriptomics and its associated aberrant RNA modifications on tumorigenesis is a largely unexplored territory with immense potential for discovering new therapeutic approaches. In particular, the underlying cellular mechanisms of different hallmarks of hepatocarcinogenesis that are governed by the complex dynamics of m6A RNA methylation demand further investigation. In this review, we reveal the up-to-date knowledge on the mechanistic and functional link between m6A RNA methylation and pathogenesis of HCC.
Collapse
Affiliation(s)
- Enakshi Sivasudhan
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Neil Blake
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Zhi-Liang Lu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Rong Rong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
17
|
High expression level of serpin peptidase inhibitor clade E member 2 is associated with poor prognosis in lung adenocarcinoma. Respir Res 2020; 21:331. [PMID: 33317533 PMCID: PMC7737331 DOI: 10.1186/s12931-020-01597-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/06/2020] [Indexed: 12/27/2022] Open
Abstract
Background Recent studies have revealed that serpin peptidase inhibitor clade E member 2 (SERPINE2) is associated with tumorigenesis. However, SERPINE2 expression and its role in lung adenocarcinomas are still unknown. Methods The expression levels of SERPINE2 in 74 consecutively resected lung adenocarcinomas were analyzed by using immunostaining. Inhibition of SERPINE2 expression by small interfering RNA (siRNA) was detected by quantitative PCR. Cell number assays and cell apoptosis assays were performed to clarify the cell-autonomous function of SERPINE2 in A549 and PC9 lung cancer cells. Results The overall survival of patients with high SERPINE2 expression was significantly worse than that of patients with low SERPINE2 expression (P = 0.0172). Multivariate analysis revealed that SERPINE2 expression was an independent factor associated with poor prognosis (P = 0.03237). The interference of SERPINE2 decreased cell number and increased apoptosis in A549 and PC9 cells Conclusion These results suggest that SERPINE2 can be used as a novel prognostic marker of lung adenocarcinoma.
Collapse
|
18
|
Zhang X, Jiang D, Yang S, Sun Y, Liu Y, Shi J, Hu C, Pan J, Liu T, Jin B, Yang K. BAP31 Promotes Tumor Cell Proliferation by Stabilizing SERPINE2 in Hepatocellular Carcinoma. Front Cell Dev Biol 2020; 8:607906. [PMID: 33363167 PMCID: PMC7759511 DOI: 10.3389/fcell.2020.607906] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) patients are mostly diagnosed at an advanced stage, resulting in systemic therapy and poor prognosis. Therefore, the identification of a novel treatment target for HCC is important. B-cell receptor-associated protein 31 (BAP31) has been identified as a cancer/testis antigen; however, BAP31 function and mechanism of action in HCC remain unclear. In this study, BAP31 was demonstrated to be upregulated in HCC and correlated with the clinical stage. BAP31 overexpression promoted HCC cell proliferation and colony formation in vitro and tumor growth in vivo. RNA-sequence (RNA-seq) analysis demonstrated that serpin family E member 2 (SERPINE2) was downregulated in BAP31-knockdown HCC cells. Coimmunoprecipitation and immunofluorescence assays demonstrated that BAP31 directly binds to SERPINE2. The inhibition of SERPINE2 significantly decreased the BAP31-induced cell proliferation and colony formation of HCC cells and phosphorylation of Erk1/2 and p38. Moreover, multiplex immunohistochemistry staining of the HCC tissue microarray showed positive associations between the expression levels of BAP31, SERPINE2, its downstream gene LRP1, and a tumor proliferation marker, Ki-67. The administration of anti-BAP31 antibody significantly inhibited HCC cell xenograft tumor growth in vivo. Thus, these findings suggest that BAP31 promotes tumor cell proliferation by stabilizing SERPINE2 and can serve as a promising candidate therapeutic target for HCC.
Collapse
Affiliation(s)
- Xiyang Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Dongbo Jiang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Shuya Yang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yuanjie Sun
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Jingqi Shi
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Chenchen Hu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Jingyu Pan
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Tianyue Liu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Kun Yang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
19
|
Lin J, Yang J, Xu X, Wang Y, Yu M, Zhu Y. A robust 11-genes prognostic model can predict overall survival in bladder cancer patients based on five cohorts. Cancer Cell Int 2020; 20:402. [PMID: 32843852 PMCID: PMC7441568 DOI: 10.1186/s12935-020-01491-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022] Open
Abstract
Background Bladder cancer is the tenth most common cancer globally, but existing biomarkers and prognostic models are limited. Method In this study, we used four bladder cancer cohorts from The Cancer Genome Atlas and Gene Expression Omnibus databases to perform univariate Cox regression analysis to identify common prognostic genes. We used the least absolute shrinkage and selection operator regression to construct a prognostic Cox model. Kaplan-Meier analysis, receiver operating characteristic curve, and univariate/multivariate Cox analysis were used to evaluate the prognostic model. Finally, a co-expression network, CIBERSORT, and ESTIMATE algorithm were used to explore the mechanism related to the model. Results A total of 11 genes were identified from the four cohorts to construct the prognostic model, including eight risk genes (SERPINE2, PRR11, DSEL, DNM1, COMP, ELOVL4, RTKN, and MAPK12) and three protective genes (FABP6, C16orf74, and TNK1). The 11-genes model could stratify the risk of patients in all five cohorts, and the prognosis was worse in the group with a high-risk score. The area under the curve values of the five cohorts in the first year are all greater than 0.65. Furthermore, this model's predictive ability is stronger than that of age, gender, grade, and T stage. Through the weighted co-expression network analysis, the gene module related to the model was found, and the key genes in this module were mainly enriched in the tumor microenvironment. B cell memory showed low infiltration in high-risk patients. Furthermore, in the case of low B cell memory infiltration and high-risk score, the prognosis of the patients was the worst. Conclusion The proposed 11-genes model is a promising biomarker for estimating overall survival in bladder cancer. This model can be used to stratify the risk of bladder cancer patients, which is beneficial to the realization of individualized treatment.
Collapse
Affiliation(s)
- Jiaxing Lin
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Jieping Yang
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Xiao Xu
- Department of Pediatric Intensive Care Unit, The Shengjing Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Yutao Wang
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Meng Yu
- Department of Reproductive Biology and Transgenic Animal, China Medical University, Shenyang, 110001 Liaoning China
| | - Yuyan Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning China
| |
Collapse
|
20
|
Zhang J, Luo A, Huang F, Gong T, Liu Z. SERPINE2 promotes esophageal squamous cell carcinoma metastasis by activating BMP4. Cancer Lett 2019; 469:390-398. [PMID: 31730904 DOI: 10.1016/j.canlet.2019.11.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022]
Abstract
Metastasis is a major lethal cause of esophageal squamous cell carcinoma (ESCC) and confers a poor prognosis. Previous studies demonstrated that serpin family E member 2 (SERPINE2) is involved in tumor metastasis. However, the function and mechanism of SERPINE2 in ESCC metastasis remains unclear. In this study, we found that SERPINE2 was increased in ESCC and associated with tumor metastasis. SERPINE2 knockdown inhibited tumor cell invasion and lymph node and lung metastasis by inducing epithelial-mesenchymal transition (EMT). We identified a total of 410 differentially expressed genes in SERPINE2-knockdown cells by RNA-Seq analysis. Among them, bone morphogenetic protein 4 (BMP4) was significantly downregulated. Conversely, BMP4 was increased in SERPINE2-overexpressing cells. Inhibiting BMP4 could attenuate SERPINE2-induced migration and invasion. Moreover, SERPINE2 was positively correlated with clinical stage, tumor invasion depth and lymph node metastasis in ESCC patients. These findings suggest that SERPINE2 promotes tumor metastasis by activating BMP4 and could serve as a potential therapeutic target for clinical intervention in ESCC.
Collapse
Affiliation(s)
- Jianglan Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Aiping Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Furong Huang
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tongyang Gong
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
21
|
Protease Nexin I is a feedback regulator of EGF/PKC/MAPK/EGR1 signaling in breast cancer cells metastasis and stemness. Cell Death Dis 2019; 10:649. [PMID: 31501409 PMCID: PMC6733841 DOI: 10.1038/s41419-019-1882-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 01/18/2023]
Abstract
Breast cancer is the most prevalent cancer in women worldwide, which remains incurable once metastatic. Breast cancer stem cells (BCSCs) are a small subset of breast cancer cells, which are the radical cause of drug resistance, tumor relapse, and metastasis in breast cancer. The extracellular serine protease inhibitor serpinE2, also named protease nexin-1 (PN-1), contributes to enhanced metastasis of cancer cells mainly by remodeling the tumor matrix. In this study, we found that PN-1 was up-regulated in breast cancer, which promoted cell invasion, migration and stemness. Furthermore, by using specific inhibitors, we discovered that epidermal growth factor (EGF) up-regulated PN-1 in breast cancer cells through cascade activation of epidermal growth factor receptor (EGFR) to the activation of protein kinase Cδ (PKCδ), mitogen-activated protein kinase (MEK) and extracellular signal-related kinase (ERK), which finally led to the up-regulation of early growth response protein 1 (EGR1). Moreover, EGF signaling was further activated as a feedback of PN-1 up-regulation through PN-1 blocking HtrA1. Taken together, our findings revealed a novel signaling axis that up-regulated PN-1 expression in breast cancer cells, and the new mechanism of PN-1-promoted breast cancer metastasis, which may provide new insights into identifying novel therapeutic targets for breast cancer.
Collapse
|
22
|
Zeng C, Chen Y. HTR1D, TIMP1, SERPINE1, MMP3 and CNR2 affect the survival of patients with colon adenocarcinoma. Oncol Lett 2019; 18:2448-2454. [PMID: 31452735 PMCID: PMC6676656 DOI: 10.3892/ol.2019.10545] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 05/09/2019] [Indexed: 01/30/2023] Open
Abstract
Colorectal cancer (CRC) is a tumor that derives from the rectum or colon, and colon adenocarcinoma (COAD) is the most common type of CRC. The present study was performed to identify genes that serve critical roles in the survival of patients with COAD. RNA-sequencing data of COAD was extracted from The Cancer Genome Atlas database, which included 480 tumor samples and 41 normal samples. Using the limma package, differential expression analysis was performed to identify the differentially expressed genes (DEGs). In addition, the potential functions and pathways for the identified DEGs were analyzed using the clusterProfiler package. After the samples were divided into high and low expression groups, survival analysis for the two groups was performed using the Kaplan-Meier model. Using Cytoscape software, a protein-protein interaction network was generated for the survival-associated genes. A total of 1,519 DEGs, including 568 upregulated genes and 951 downregulated genes, were identified in the COAD samples. Enrichment analysis suggested that the DEGs were implicated in numerous functional terms and pathways. Furthermore, 109 DEGs were identified to be survival-associated genes in COAD. According to the degrees of the network nodes, 5-hydroxytryptamine receptor 1D (HTR1D), TIMP metallopeptidase inhibitor 1 (TIMP1), serpin family E member 1 (SERPINE1), matrix metallopeptidase 3 (MMP3) and cannabinoid receptor 2 (CNR2) were key nodes, and the expression levels of these genes were analyzed in clinical samples of CRC. Therefore, the results of the present study suggest HTR1D, TIMP1, SERPINE1, MMP3 and CNR2 may affect the prognosis of patients with COAD.
Collapse
Affiliation(s)
- Chunyan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Youxiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
23
|
Ward Rashidi MR, Mehta P, Bregenzer M, Raghavan S, Fleck EM, Horst EN, Harissa Z, Ravikumar V, Brady S, Bild A, Rao A, Buckanovich RJ, Mehta G. Engineered 3D Model of Cancer Stem Cell Enrichment and Chemoresistance. Neoplasia 2019; 21:822-836. [PMID: 31299607 PMCID: PMC6624324 DOI: 10.1016/j.neo.2019.06.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 06/03/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
Intraperitoneal dissemination of ovarian cancers is preceded by the development of chemoresistant tumors with malignant ascites. Despite the high levels of chemoresistance and relapse observed in ovarian cancers, there are no in vitro models to understand the development of chemoresistance in situ. Method: We describe a highly integrated approach to establish an in vitro model of chemoresistance and stemness in ovarian cancer, using the 3D hanging drop spheroid platform. The model was established by serially passaging non-adherent spheroids. At each passage, the effectiveness of the model was evaluated via measures of proliferation, response to treatment with cisplatin and a novel ALDH1A inhibitor. Concomitantly, the expression and tumor initiating capacity of cancer stem-like cells (CSCs) was analyzed. RNA-seq was used to establish gene signatures associated with the evolution of tumorigenicity, and chemoresistance. Lastly, a mathematical model was developed to predict the emergence of CSCs during serial passaging of ovarian cancer spheroids. Results: Our serial passage model demonstrated increased cellular proliferation, enriched CSCs, and emergence of a platinum resistant phenotype. In vivo tumor xenograft assays indicated that later passage spheroids were significantly more tumorigenic with higher CSCs, compared to early passage spheroids. RNA-seq revealed several gene signatures supporting the emergence of CSCs, chemoresistance, and malignant phenotypes, with links to poor clinical prognosis. Our mathematical model predicted the emergence of CSC populations within serially passaged spheroids, concurring with experimentally observed data. Conclusion: Our integrated approach illustrates the utility of the serial passage spheroid model for examining the emergence and development of chemoresistance in ovarian cancer in a controllable and reproducible format.
Collapse
Affiliation(s)
- Maria R Ward Rashidi
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michael Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Elyse M Fleck
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Eric N Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Zainab Harissa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Visweswaran Ravikumar
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel Brady
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Andrea Bild
- Division of Molecular Pharmacology, Department of Medical Oncology and Therapeutics, City of Hope Cancer Institute, Duarte, CA, USA
| | - Arvind Rao
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Radiation Oncology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ronald J Buckanovich
- Director of Ovarian Cancer Research, Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Geeta Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, USA..
| |
Collapse
|
24
|
Desjobert C, Carrier A, Delmas A, Marzese DM, Daunay A, Busato F, Pillon A, Tost J, Riond J, Favre G, Etievant C, Arimondo PB. Demethylation by low-dose 5-aza-2'-deoxycytidine impairs 3D melanoma invasion partially through miR-199a-3p expression revealing the role of this miR in melanoma. Clin Epigenetics 2019; 11:9. [PMID: 30651148 PMCID: PMC6335767 DOI: 10.1186/s13148-018-0600-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/17/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Efficient treatments against metastatic melanoma dissemination are still lacking. Here, we report that low-cytotoxic concentrations of 5-aza-2'-deoxycytidine, a DNA demethylating agent, prevent in vitro 3D invasiveness of metastatic melanoma cells and reduce lung metastasis formation in vivo. RESULTS We unravelled that this beneficial effect is in part due to MIR-199A2 re-expression by promoter demethylation. Alone, this miR showed an anti-invasive and anti-metastatic effect. Throughout integration of micro-RNA target prediction databases with transcriptomic analysis after 5-aza-2'-deoxycytidine treatments, we found that miR-199a-3p downregulates set of genes significantly involved in invasion/migration processes. In addition, analysis of data from melanoma patients showed a stage- and tissue type-dependent modulation of MIR-199A2 expression by DNA methylation. CONCLUSIONS Thus, our data suggest that epigenetic- and/or miR-based therapeutic strategies can be relevant to limit metastatic dissemination of melanoma.
Collapse
Affiliation(s)
- Cécile Desjobert
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France
| | - Arnaud Carrier
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France
| | - Audrey Delmas
- Cancer Research Center of Toulouse, CRCT, Toulouse, France
| | - Diego M Marzese
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Antoine Daunay
- Laboratory for Functional Genomics, Fondation Jean Dausset - CEPH, Paris, France
| | - Florence Busato
- Laboratory for Epigenetics and Environment, Centre National de la Recherche en Génomique Humaine, CEA, Evry, France
| | - Arnaud Pillon
- Institut de Recherche Pierre Fabre, CRDPF, Toulouse, France
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de la Recherche en Génomique Humaine, CEA, Evry, France
| | - Joëlle Riond
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France.,UMR 1037 INSERM/Université Toulouse III, CRCT, Toulouse, France
| | - Gilles Favre
- Cancer Research Center of Toulouse, CRCT, Toulouse, France
| | | | - Paola B Arimondo
- FRE no. 3600 CNRS, Epigenetic Targeting of Cancer (ETaC), Toulouse, France. .,Institut Pasteur CNRS UMR3523, Epigenetic Chemical Biology, Paris, France.
| |
Collapse
|
25
|
Shen Y, Wang X, Xu J, Lu L. SerpinE2, a poor biomarker of endometrial cancer, promotes the proliferation and mobility of EC cells. Cancer Biomark 2018; 19:271-278. [PMID: 28453461 DOI: 10.3233/cbm-160442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The SerpinE2 pathway is evolutionarily conserved and plays an important role in tumorigenesis. SerpinE2 (a small ubiquitin-related modifier), like ubiquitin, conjugates SerpinE2 proteins onto lysine residues of target proteins. SerpinE2 over-expression has been found in several tumors. Here, we detected the level of SerpinE2 in 72 samples of EC tissue using immunohistochemistry to assess the role of SerpinE2 in EC prognosis. Meanwhile, we knocked down SerpinE2 by siRNA in the HTB-111 and Ishikawa EC cell lines and analyzed the viability and mobility change using an MTT assay, an annexin V/PI apoptosis assay, a wound scratch test and a transwell assay. A Kaplan-Meier analysis indicated a negative correlation between the level of SerpinE2 and the EC prognosis. Silencing SerpinE2 induced cell apoptosis and reduced the migration ability. Our data suggest SerpinE2 works as an oncogene in EC.
Collapse
Affiliation(s)
- Yuan Shen
- Gynecology and Obstetrics Department of the First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Xiaoyu Wang
- Gynecology and Obstetrics Department of the First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Jianping Xu
- Gynecology and Obstetrics Department of the First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Lin Lu
- Gynecology and Obstetrics Department, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
26
|
Smirnova T, Bonapace L, MacDonald G, Kondo S, Wyckoff J, Ebersbach H, Fayard B, Doelemeyer A, Coissieux MM, Heideman MR, Bentires-Alj M, Hynes NE. Serpin E2 promotes breast cancer metastasis by remodeling the tumor matrix and polarizing tumor associated macrophages. Oncotarget 2018; 7:82289-82304. [PMID: 27793045 PMCID: PMC5347692 DOI: 10.18632/oncotarget.12927] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/19/2016] [Indexed: 12/26/2022] Open
Abstract
The extracellular serine protease inhibitor serpinE2 is overexpressed in breast cancer and has been shown to foster metastatic spread. Here, we investigated the hypothesis that serpinE2 creates tumor-promoting conditions in the tumor microenvironment (TME) by affecting extracellular matrix remodeling. Using two different breast cancer models, we show that blocking serpinE2, either by knock-down (KD) in tumor cells or in response to a serpinE2 binding antibody, decreases metastatic dissemination from primary tumors to the lungs. We demonstrate that in response to serpinE2 KD or antibody treatment there are dramatic changes in the TME. Multiphoton intravital imaging revealed deposition of a dense extracellular collagen I matrix encapsulating serpinE2 KD or antibody-treated tumors. This is accompanied by a reduction in the population of tumor-promoting macrophages, as well as a decrease in chemokine ligand 2, which is known to affect macrophage abundance and polarization. In addition, TIMP-1 secretion is increased, which may directly inhibit matrix metalloproteases critical for collagen degradation in the tumor. In summary, our findings suggest that serpinE2 is required in the extracellular milieu of tumors where it acts in multiple ways to regulate tumor matrix deposition, thereby controlling tumor cell dissemination.
Collapse
Affiliation(s)
- Tatiana Smirnova
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Laura Bonapace
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Gwen MacDonald
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Shunya Kondo
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jeffrey Wyckoff
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Bérengère Fayard
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Arno Doelemeyer
- Novartis Institute for Biomedical Research, Basel, Switzerland
| | | | - Marinus R Heideman
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | | | - Nancy E Hynes
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
27
|
Yang Y, Xin X, Fu X, Xu D. Expression pattern of human SERPINE2 in a variety of human tumors. Oncol Lett 2018; 15:4523-4530. [PMID: 29556291 DOI: 10.3892/ol.2018.7819] [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: 03/12/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022] Open
Abstract
Serine proteinase inhibitor, clade E member 2 (SERPINE2), also known as protease nexin-1 (PN-1), is a member of the serpin family. Despite several reported roles of SERPINE2 in tumor development the histological distribution of SERPINE2 and its expression levels in a large variety of tumors remains unclear. Through expressed sequence tag database analysis, immunohistochemical staining of tissue microarrays and a literature review, it was revealed that SERPINE2 expression varied according to growth stages and tissue types. SERPINE2 is differentially expressed in a number of tumors and their normal tissue counterparts. SERPINE2 is identified most abundantly in adenocarcinomas. SERPINE2 serves diverse roles in a variety of tumors and therefore may serve as a promising biomarker for tumor diagnosis and prognosis.
Collapse
Affiliation(s)
- Ying Yang
- Department of Haematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangke Xin
- Department of Haematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xing Fu
- Department of Haematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Danmei Xu
- Department of Haematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China.,Department of Haematology, Imperial College London, Hammersmith Hospital, London, W12 0HS, UK
| |
Collapse
|
28
|
Perego M, Maurer M, Wang JX, Shaffer S, Müller AC, Parapatics K, Li L, Hristova D, Shin S, Keeney F, Liu S, Xu X, Raj A, Jensen JK, Bennett KL, Wagner SN, Somasundaram R, Herlyn M. A slow-cycling subpopulation of melanoma cells with highly invasive properties. Oncogene 2018; 37:302-312. [PMID: 28925403 PMCID: PMC5799768 DOI: 10.1038/onc.2017.341] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/02/2017] [Accepted: 08/12/2017] [Indexed: 12/16/2022]
Abstract
Melanoma is a heterogeneous tumor with different subpopulations showing different proliferation rates. Slow-cycling cells were previously identified in melanoma, but not fully biologically characterized. Using the label-retention method, we identified a subpopulation of slow-cycling cells, defined as label-retaining cells (LRC), with strong invasive properties. We demonstrate through live imaging that LRC are leaving the primary tumor mass at a very early stage and disseminate to peripheral organs. Through global proteome analyses, we identified the secreted protein SerpinE2/protease nexin-1 as causative for the highly invasive potential of LRC in melanomas.
Collapse
Affiliation(s)
- M Perego
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - M Maurer
- Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - J X Wang
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - S Shaffer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - A C Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - K Parapatics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - L Li
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - D Hristova
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - S Shin
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - F Keeney
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - S Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - X Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - A Raj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - J K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - K L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - S N Wagner
- Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - R Somasundaram
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - M Herlyn
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| |
Collapse
|
29
|
Metri R, Mohan A, Nsengimana J, Pozniak J, Molina-Paris C, Newton-Bishop J, Bishop D, Chandra N. Identification of a gene signature for discriminating metastatic from primary melanoma using a molecular interaction network approach. Sci Rep 2017; 7:17314. [PMID: 29229936 PMCID: PMC5725601 DOI: 10.1038/s41598-017-17330-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/10/2017] [Indexed: 01/15/2023] Open
Abstract
Understanding the biological factors that are characteristic of metastasis in melanoma remains a key approach to improving treatment. In this study, we seek to identify a gene signature of metastatic melanoma. We configured a new network-based computational pipeline, combined with a machine learning method, to mine publicly available transcriptomic data from melanoma patient samples. Our method is unbiased and scans a genome-wide protein-protein interaction network using a novel formulation for network scoring. Using this, we identify the most influential, differentially expressed nodes in metastatic as compared to primary melanoma. We evaluated the shortlisted genes by a machine learning method to rank them by their discriminatory capacities. From this, we identified a panel of 6 genes, ALDH1A1, HSP90AB1, KIT, KRT16, SPRR3 and TMEM45B whose expression values discriminated metastatic from primary melanoma (87% classification accuracy). In an independent transcriptomic data set derived from 703 primary melanomas, we showed that all six genes were significant in predicting melanoma specific survival (MSS) in a univariate analysis, which was also consistent with AJCC staging. Further, 3 of these genes, HSP90AB1, SPRR3 and KRT16 remained significant predictors of MSS in a joint analysis (HR = 2.3, P = 0.03) although, HSP90AB1 (HR = 1.9, P = 2 × 10-4) alone remained predictive after adjusting for clinical predictors.
Collapse
Affiliation(s)
- Rahul Metri
- IISc Mathematics Initiative (IMI), Indian Institute of Science, Bangalore, Karnataka, India
| | - Abhilash Mohan
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Jérémie Nsengimana
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Joanna Pozniak
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Carmen Molina-Paris
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| | - Julia Newton-Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - David Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Nagasuma Chandra
- IISc Mathematics Initiative (IMI), Indian Institute of Science, Bangalore, Karnataka, India.
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
30
|
Hankey W, McIlhatton MA, Ebede K, Kennedy B, Hancioglu B, Zhang J, Brock GN, Huang K, Groden J. Mutational Mechanisms That Activate Wnt Signaling and Predict Outcomes in Colorectal Cancer Patients. Cancer Res 2017; 78:617-630. [PMID: 29212857 DOI: 10.1158/0008-5472.can-17-1357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/14/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022]
Abstract
APC biallelic loss-of-function mutations are the most prevalent genetic changes in colorectal tumors, but it is unknown whether these mutations phenocopy gain-of-function mutations in the CTNNB1 gene encoding β-catenin that also activate canonical WNT signaling. Here we demonstrate that these two mutational mechanisms are not equivalent. Furthermore, we show how differences in gene expression produced by these different mechanisms can stratify outcomes in more advanced human colorectal cancers. Gene expression profiling in Apc-mutant and Ctnnb1-mutant mouse colon adenomas identified candidate genes for subsequent evaluation of human TCGA (The Cancer Genome Atlas) data for colorectal cancer outcomes. Transcriptional patterns exhibited evidence of activated canonical Wnt signaling in both types of adenomas, with Apc-mutant adenomas also exhibiting unique changes in pathways related to proliferation, cytoskeletal organization, and apoptosis. Apc-mutant adenomas were characterized by increased expression of the glial nexin Serpine2, the human ortholog, which was increased in advanced human colorectal tumors. Our results support the hypothesis that APC-mutant colorectal tumors are transcriptionally distinct from APC-wild-type colorectal tumors with canonical WNT signaling activated by other mechanisms, with possible implications for stratification and prognosis.Significance: These findings suggest that colon adenomas driven by APC mutations are distinct from those driven by WNT gain-of-function mutations, with implications for identifying at-risk patients with advanced disease based on gene expression patterns. Cancer Res; 78(3); 617-30. ©2017 AACR.
Collapse
Affiliation(s)
- William Hankey
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Michael A McIlhatton
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Kenechi Ebede
- Department of Anesthesiology, University of Florida, Gainesville, Florida
| | - Brian Kennedy
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Baris Hancioglu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, Indiana
| | - Guy N Brock
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Kun Huang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, Indiana
| | - Joanna Groden
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
31
|
Elevated Concentrations of SERPINE2/Protease Nexin-1 and Secretory Leukocyte Protease Inhibitor in the Serum of Patients with Papillary Thyroid Cancer. DISEASE MARKERS 2017; 2017:4962137. [PMID: 28255192 PMCID: PMC5306996 DOI: 10.1155/2017/4962137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/02/2017] [Accepted: 01/16/2017] [Indexed: 01/02/2023]
Abstract
Introduction. SERPINE2 and secretory leukocyte protease inhibitor (SLPI) are proteins with anticoagulant properties which could promote solid tumor growth. However, their role in the pathogenesis of thyroid cancer has not been determined. Materials and Methods. The aim of this study was to assess serum SERPINE2 and SLPI concentrations in a group of 36 patients with papillary thyroid cancer (PTC) and a group of 19 subjects with multinodular nontoxic goiter (MNG). The control group (CG) consisted of 20 healthy volunteers. Blood samples were collected one day before surgery. Serum SERPINE2 and SLPI concentrations were measured using specific ELISA methods. Results. Significantly higher concentrations of SERPINE2 and SLPI were found in patients with PTC as compared with MNG and controls. Positive correlation was found between SERPINE2 and SLPI concentrations in PTC patients. The levels of SERPINE2 and SLPI did not differ significantly between MNG and healthy controls. Conclusions. Our results indicate that SERPINE2 and SLPI play a significant role in the development of papillary thyroid cancer and imply that the evaluation of serum concentrations of both anticoagulant molecules may be considered as additional marker for the differentiation of malignancies during the preoperative diagnosis of patients with thyroid gland tumors.
Collapse
|
32
|
Kim BA, Jee HG, Yi JW, Kim SJ, Chai YJ, Choi JY, Lee KE. Expression Profiling of a Human Thyroid Cell Line Stably Expressing the BRAFV600E Mutation. Cancer Genomics Proteomics 2017; 14:53-67. [PMID: 28031237 DOI: 10.21873/cgp.20018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/AIM The BRAFV600E mutation acts as an initiator of cancer development in papillary thyroid carcinoma (PTC). Gene expression changes caused by the BRAFV600E mutation may have an important role in thyroid cancer development. MATERIALS AND METHODS To study genomic alterations caused by the BRAFV600E mutation, we made human thyroid cell lines that harbor the wild-type BRAF gene (Nthy/WT) and the V600E mutant-type BRAF gene (Nthy/V600E). RESULTS Flow cytometry and western blotting showed stable transfection of the BRAF gene. In functional experiments, Nthy/V600E showed increased anchorage-independent growth and invasion through Matrigel, compared to Nthy/WT. Microarray analysis revealed that 2,441 genes were up-regulated in Nthy/V600E compared to Nthy/WT. Gene ontology analysis showed that the up-regulated genes were associated with cell adhesion, migration, and the ERK and MAPK cascade, and pathway analysis showed enrichment in cancer-related pathways. CONCLUSION Our Nthy/WT and Nthy/V600E cell line pair could be a suitable model to study the molecular characteristics of BRAFV600E PTC.
Collapse
Affiliation(s)
- Byoung-Ae Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyeon-Gun Jee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Wook Yi
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jun Chai
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - June Young Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kyu Eun Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea .,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
SERPINE2/Protease Nexin-1 in vivo multiple functions: Does the puzzle make sense? Semin Cell Dev Biol 2016; 62:160-169. [PMID: 27545616 DOI: 10.1016/j.semcdb.2016.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/17/2016] [Accepted: 08/17/2016] [Indexed: 11/21/2022]
Abstract
Cultures of glial cells and fibroblasts allowed and lead to the identification SERPINE2/Protease Nexin-1 (SERPINE2/PN-1). Cellular, biochemical, immunological and molecular characterization substantiated its variable expression in many organs as a function of development, adult stages, pathological situations or following injury. It is not a circulating serpin, but as other members of the family, its target specificity is influenced by components of the extracellular matrix. The challenges are to identify where and when SERPINE2/PN-1 modulatory action becomes crucial or even possibly specific in a mosaic of feasible in vivo impacts. Data providing correlations are not sufficient to satisfy this aim. Genetically modified mice, or tissue derived thereof, provide interesting in vivo models to identify and study the relevance of this serpin. This review will highlight sometimes-intriguing results indicating a crucial impact of SERPINE2/PN-1, especially in the vasculature, the nervous system or the behavior of cancer cells in vivo. Data presently available will be discussed in an attempt to define general trends in the diversity of SERPINE2/PN-1 modes of action in vivo.
Collapse
|
34
|
MAO MINZHI, WANG WANCHUN. SerpinE2 promotes multiple cell proliferation and drug resistance in osteosarcoma. Mol Med Rep 2016; 14:881-7. [DOI: 10.3892/mmr.2016.5316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 04/15/2016] [Indexed: 11/06/2022] Open
|
35
|
Wu QW. Serpine2, a potential novel target for combating melanoma metastasis. Am J Transl Res 2016; 8:1985-1997. [PMID: 27347308 PMCID: PMC4891413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 11/17/2015] [Indexed: 06/06/2023]
Abstract
Early stages of melanoma can be treated by surgical resection of tumor, but there is still no effective treatment once it is progressed to metastatic phases. Although growing family of both metastasis promoting and metastasis suppressor genes have been reported, the molecular mechanisms governing melanoma metastatic cascade are still not completely understood. Therefore, defining the molecules that govern melanoma metastasis may aid the development of more effective therapeutic strategies for combating cancer. In the present study, we found that Serpin Peptidase Inhibitor 2, Serpine2 was involved in the metastasis of melanoma cells. The requirement of Serpine2 in the migration of melanoma cells was confirmed by gene silencing and over-expression in vitro. Moreover, down-regulation of Serpine2 expression strikingly inhibited melanoma cellular metastasis in vivo. Finally, we found that Serpine2 promotes melanoma metastasis through the glycogen synthesis kinase 3β, GSK-3β signaling pathway. To conclude, our findings suggested a novel mechanism underlying the metastasis of melanoma cells which might serve as a new intervention target for the treatment of melanoma.
Collapse
Affiliation(s)
- Qi Wei Wu
- Department of Dermatology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine 1111 Xianxia Road, Shanghai 200336, China
| |
Collapse
|
36
|
Ohta N, Ishiguro S, Kawabata A, Uppalapati D, Pyle M, Troyer D, De S, Zhang Y, Becker KG, Tamura M. Human umbilical cord matrix mesenchymal stem cells suppress the growth of breast cancer by expression of tumor suppressor genes. PLoS One 2015; 10:e0123756. [PMID: 25942583 PMCID: PMC4420498 DOI: 10.1371/journal.pone.0123756] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/24/2015] [Indexed: 12/25/2022] Open
Abstract
Human and rat umbilical cord matrix mesenchymal stem cells (UCMSC) possess the ability to control the growth of breast carcinoma cells. Comparative analyses of two types of UCMSC suggest that rat UCMSC-dependent growth regulation is significantly stronger than that of human UCMSC. Their different tumoricidal abilities were clarified by analyzing gene expression profiles in the two types of UCMSC. Microarray analysis revealed differential gene expression between untreated naïve UCMSC and those co-cultured with species-matched breast carcinoma cells. The analyses screened 17 differentially expressed genes that are commonly detected in both human and rat UCMSC. The comparison between the two sets of gene expression profiles identified two tumor suppressor genes, adipose-differentiation related protein (ADRP) and follistatin (FST), that were specifically up-regulated in rat UCMSC, but down-regulated in human UCMSC when they were co-cultured with the corresponding species' breast carcinoma cells. Over-expression of FST, but not ADRP, in human UCMSC enhanced their ability to suppress the growth of MDA-231 cells. The growth of MDA-231 cells was also significantly lower when they were cultured in medium conditioned with FST, but not ADRP over-expressing human UCMSC. In the breast carcinoma lung metastasis model generated with MDA-231 cells, systemic treatment with FST-over-expressing human UCMSC significantly attenuated the tumor burden. These results suggest that FST may play an important role in exhibiting stronger tumoricidal ability in rat UCMSC than human UCMSC and also implies that human UCMSC can be transformed into stronger tumoricidal cells by enhancing tumor suppressor gene expression.
Collapse
Affiliation(s)
- Naomi Ohta
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Susumu Ishiguro
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Atsushi Kawabata
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Deepthi Uppalapati
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Marla Pyle
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Deryl Troyer
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
| | - Supriyo De
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, NIH Biomedical Research Center, National Institute on Aging, NIH, Baltimore, MD, 21224, United States of America
| | - Masaaki Tamura
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66506, United States of America
- * E-mail:
| |
Collapse
|
37
|
Serpine2/PN-1 Is Required for Proliferative Expansion of Pre-Neoplastic Lesions and Malignant Progression to Medulloblastoma. PLoS One 2015; 10:e0124870. [PMID: 25901736 PMCID: PMC4406471 DOI: 10.1371/journal.pone.0124870] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/18/2015] [Indexed: 12/13/2022] Open
Abstract
Background Medulloblastomas are malignant childhood brain tumors that arise due to the aberrant activity of developmental pathways during postnatal cerebellar development and in adult humans. Transcriptome analysis has identified four major medulloblastoma subgroups. One of them, the Sonic hedgehog (SHH) subgroup, is caused by aberrant Hedgehog signal transduction due to mutations in the Patched1 (PTCH1) receptor or downstream effectors. Mice carrying a Patched-1 null allele (Ptch1∆/+) are a good model to study the alterations underlying medulloblastoma development as a consequence of aberrant Hedgehog pathway activity. Results Transcriptome analysis of human medulloblastomas shows that SERPINE2, also called Protease Nexin-1 (PN-1) is overexpressed in most medulloblastomas, in particular in the SHH and WNT subgroups. As siRNA-mediated lowering of SERPINE2/PN-1 in human medulloblastoma DAOY cells reduces cell proliferation, we analyzed its potential involvement in medulloblastoma development using the Ptch1∆/+ mouse model. In Ptch1∆/+ mice, medulloblastomas arise as a consequence of aberrant Hedgehog pathway activity. Genetic reduction of Serpine2/Pn-1 interferes with medulloblastoma development in Ptch1∆/+ mice, as ~60% of the pre-neoplastic lesions (PNLs) fail to develop into medulloblastomas and remain as small cerebellar nodules. In particular the transcription factor Atoh1, whose expression is essential for development of SHH subgroup medulloblastomas is lost. Comparative molecular analysis reveals the distinct nature of the PNLs in young Ptch1∆/+Pn-1Δ/+ mice. The remaining wild-type Ptch1 allele escapes transcriptional silencing in most cases and the aberrant Hedgehog pathway activity is normalized. Furthermore, cell proliferation and the expression of the cell-cycle regulators Mycn and Cdk6 are significantly reduced in PNLs of Ptch1∆/+Pn-1Δ/+ mice. Conclusions Our analysis provides genetic evidence that aberrant Serpine2/Pn-1 is required for proliferation of human and mouse medulloblastoma cells. In summary, our analysis shows that Serpine2/PN-1 boosts malignant progression of PNLs to medulloblastomas, in which the Hedgehog pathway is activated in a SHH ligand-independent manner.
Collapse
|
38
|
Oncogenic KRAS signalling promotes the Wnt/β-catenin pathway through LRP6 in colorectal cancer. Oncogene 2014; 34:4914-27. [PMID: 25500543 PMCID: PMC4687460 DOI: 10.1038/onc.2014.416] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 11/04/2014] [Accepted: 11/08/2014] [Indexed: 12/14/2022]
Abstract
Aberrant regulation of the Wnt/β-catenin signaling pathway is one of the major causes of colorectal cancer (CRC). Loss-of-function mutations in APC are commonly found in CRC, leading to inappropriate activation of canonical Wnt signaling. Conversely, gain-of-function mutations in KRAS and BRAF genes are detected in up to 60% of CRCs. Whereas KRAS/mitogen-activated protein kinase (MAPK) and canonical Wnt/β-catenin pathways are critical for intestinal tumorigenesis, mechanisms integrating these two important signaling pathways during CRC development are unknown. Results herein demonstrate that transformation of normal intestinal epithelial cells (IECs) by oncogenic forms of KRAS, BRAF or MEK1 was associated with a marked increase in β-catenin/TCF4 and c-MYC promoter transcriptional activities and mRNA levels of c-Myc, Axin2 and Lef1. Notably, expression of a dominant-negative mutant of T-Cell Factor 4 (ΔNTCF4) severely attenuated IEC transformation induced by oncogenic MEK1 and markedly reduced their tumorigenic and metastatic potential in immunocompromised mice. Interestingly, the Frizzled co-receptor LRP6 was phosphorylated in a MEK-dependent manner in transformed IECs and in human CRC cell lines. Expression of LRP6 mutant in which serine/threonine residues in each particular ProlineProlineProlineSerine/ThreonineProline motif were mutated to alanines (LRP6-5A) significantly reduced β-catenin/TCF4 transcriptional activity. Accordingly, MEK inhibition in human CRC cells significantly diminished β-catenin/TCF4 transcriptional activity and c-MYC mRNA and protein levels without affecting β-catenin expression or stability. Lastly, LRP6 phosphorylation was also increased in human colorectal tumors, including adenomas, in comparison with healthy adjacent normal tissues. Our data indicate that oncogenic activation of KRAS/BRAF/MEK signaling stimulates the canonical Wnt/β-catenin pathway, which in turn promotes intestinal tumor growth and invasion. Moreover, LRP6 phosphorylation by ERK1/2 may provide a unique point of convergence between KRAS/MAPK and Wnt/β-catenin signalings during oncogenesis.
Collapse
|
39
|
Protease Nexin-1 affects the migration and invasion of C6 glioma cells through the regulation of urokinase Plasminogen Activator and Matrix Metalloproteinase-9/2. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2631-44. [DOI: 10.1016/j.bbamcr.2014.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/26/2014] [Accepted: 07/17/2014] [Indexed: 01/23/2023]
|
40
|
Prognostic significance of SERPINE2 in gastric cancer and its biological function in SGC7901 cells. J Cancer Res Clin Oncol 2014; 141:805-12. [DOI: 10.1007/s00432-014-1858-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 10/16/2014] [Indexed: 01/09/2023]
|
41
|
Song Y, Gong K, Yan H, Hong W, Wang L, Wu Y, Li W, Li W, Cao Z. Sj7170, a unique dual-function peptide with a specific α-chymotrypsin inhibitory activity and a potent tumor-activating effect from scorpion venom. J Biol Chem 2014; 289:11667-11680. [PMID: 24584937 DOI: 10.1074/jbc.m113.540419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
A new peptide precursor, termed Sj7170, was characterized from the venomous gland cDNA library of the scorpion Scorpiops jendeki. Sj7170 was deduced to be a 62-amino acid peptide cross-linked by five disulfide bridges. The recombinant Sj7170 peptide (rSj7170) with chromatographic purity was produced by a prokaryotic expression system. Enzyme inhibition assay in vitro and in vivo showed that rSj7170 specifically inhibited the activity of α-chymotrypsin at micromole concentrations. In addition, Sj7170 not only promoted cell proliferation and colony formation by up-regulating the expression of cyclin D1 in vitro but also enhanced tumor growth in nude mice. Finally, Sj7170 accelerated cellular migration and invasion by increasing the expression of the transcription factor Snail and then inducing the epithelial-mesenchymal transition. Moreover, Sj7170 changed cell morphology and cytoskeleton of U87 cells by the GTPase pathway. Taken together, Sj7170 is a unique dual-function peptide, i.e. a specific α-chymotrypsin inhibitor and a potent tumorigenesis/metastasis activator. Our work not only opens an avenue of developing new modulators of tumorigenesis/metastasis from serine protease inhibitors but also strengthens the functional link between protease inhibitors and tumor activators.
Collapse
Affiliation(s)
- Yu Song
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ke Gong
- College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Hong Yan
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Wei Hong
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Le Wang
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yingliang Wu
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Wenhua Li
- College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| | - Wenxin Li
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| | - Zhijian Cao
- State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei 430072, China; College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| |
Collapse
|
42
|
Zhao BZ, Cao J, Shao JC, Sun YB, Fan LM, Wu CY, Liang S, Guo BF, Yang G, Xie WH, Yang QC, Yang SF. Novel esophageal squamous cell carcinoma bone metastatic clone isolated by scintigraphy, X ray and micro PET/CT. World J Gastroenterol 2014; 20:1030-1037. [PMID: 24574775 PMCID: PMC3921526 DOI: 10.3748/wjg.v20.i4.1030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a Chinese esophageal squamous cell carcinoma (ESCC) cell line with high bone metastasis potency using 99mTc-methylene diphosphonate (99mTc-MDP) micro-pinhole scintigraphy, X ray and micro-positron emission tomography/computed tomography (PET/CT) for exploring the mechanism of occurrence and development in esophageal cancer.
METHODS: The cells came from a BALB/c nu/nu immunodeficient mouse, and oncogenic tumor tissue was from a surgical specimen from a 61-year-old male patient with ESCC. The cell growth curve was mapped and analysis of chromosome karyotype was performed. Approximately 1 × 106 oncogenic cells were injected into the left cardiac ventricle of immunodeficient mice. The bone metastatic lesions of tumor-bearing mice were detected by 99mTc-MDP scintigraphy, micro-PET/CT and X-ray, and were resected from the mice under deep anesthesia. The bone metastatic cells in the lesions were used for culture and for repeated intracardiac inoculation. This in vivo/in vitro experimental metastasis study was repeated for four cycles. All of the suspicious bone sites were confirmed by pathology. Real-time polymerase chain reaction was used to compare the gene expression in the parental cells and in the bone metastatic clone.
RESULTS: The surgical specimen was implanted subcutaneously in immunodeficient mice and the tumorigenesis rate was 100%. First-passage oncogenic cells were named CEK-Sq-1. The chromosome karyotype analysis of the cell line was hypotriploid. The bone metastasis rate went from 20% with the first-passage oncogenic cells via intracardiac inoculation to 90% after four cycles. The established bone metastasis clone named CEK-Sq-1BM had a high potential to metastasize in bone, including mandible, humerus, thoracic and lumbar vertebrae, scapula and femur. The bone metastasis lesions were successfully detected by micro-pinhole bone scintigraphy, micro-PET/CT, and X-ray. The sensitivity, specificity and accuracy of the micro-pinhole scintigraphy, X-ray, and micro-PET/CT imaging examinations were: 89.66%/32%/80%, 88.2%/100%/89.2%, and 88.75%/77.5%/87.5%, respectively. Some gene expression difference was found between parental and bone metastasis cells.
CONCLUSION: This newly established Chinese ESCC cell line and animal model may provide a useful tool for the study of the pathogenesis and development of esophageal carcinoma.
Collapse
|
43
|
Jones C, St-Jean S, Fréchette I, Bergeron D, Rivard N, Boudreau F. Identification of a novel promyelocytic leukemia zinc-finger isoform required for colorectal cancer cell growth and survival. Int J Cancer 2013; 133:58-66. [PMID: 23280881 DOI: 10.1002/ijc.28008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 10/28/2012] [Accepted: 12/17/2012] [Indexed: 11/06/2022]
Abstract
Promyelocytic leukemia zinc-finger (PLZF) is a transcriptional repressor that regulates proliferation, differentiation and apoptosis among various cellular origins. PLZF expression is upregulated in colorectal cancer cell lines but its putative functional role in this context is unknown. Here, we report the identification of a novel p65 PLZF isoform that results from the usage of an evolutionarily conserved alternative translational initiation site. This isoform is devoid of the classical BTB/POZ domain required for nuclear localization and transcriptional repression. Depletion of p65 PLZF expression in colorectal cancer cell lines results in reduction of cell growth, loss of cell anchorage and increase in cell apoptosis. Overall, these results indicate that p65 PLZF is crucial to maintain colorectal cancer cell adhesion as well as survival and must occur independently of the traditionally viewed transcriptional role of PLZF in the course of these biological processes.
Collapse
Affiliation(s)
- Christine Jones
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Muff R, Ram Kumar RM, Botter SM, Born W, Fuchs B. Genes regulated in metastatic osteosarcoma: evaluation by microarray analysis in four human and two mouse cell line systems. Sarcoma 2012; 2012:937506. [PMID: 23213280 PMCID: PMC3504467 DOI: 10.1155/2012/937506] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/07/2012] [Indexed: 12/27/2022] Open
Abstract
Osteosarcoma (OS) is a rare bone neoplasm that affects mainly adolescents. It is associated with poor prognosis in case of metastases formation. The search for metastasis predicting markers is therefore imperative to optimize treatment strategies for patients at risk and important for the search of new drugs for the treatment of this devastating disease. Here, we have analyzed by microarray the differential gene expression in four human and two mouse OS cell line systems consisting of parental cell lines with low metastatic potential and derivatives thereof with increased metastatic potential. Using two osteoblastic cell line systems, the most common OS phenotype, we have identified forty-eight common genes that are differentially expressed in metastatic cell lines compared to parental cells. The identified subset of metastasis relevant genes in osteoblastic OS overlapped only minimally with differentially expressed genes in the other four preosteoblast or nonosteoblastic cell line systems. The results imply an OS phenotype specific expression pattern of metastasis regulating proteins and form a basis for further investigation of gene expression profiles in patients' samples combined with survival analysis with the aim to optimize treatment strategies to develop new drugs and to consequently improve the survival of patients with the most common form of osteoblastic OS.
Collapse
Affiliation(s)
- Roman Muff
- Laboratory for Orthopedic Research, Balgrist University Hospital, Forchstrasse 340, 8008 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
45
|
Zsippai A, Szabó DR, Tömböl Z, Szabó PM, Éder K, Pállinger É, Gaillard RC, Patócs A, Tóth S, Falus A, Rácz K, Igaz P. Effects of mitotane on gene expression in the adrenocortical cell line NCI-H295R: a microarray study. Pharmacogenomics 2012; 13:1351-61. [DOI: 10.2217/pgs.12.116] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The adrenolytic agent mitotane is widely used in the treatment of adrenocortical cancer; however, its mechanism of action is poorly elucidated. We have studied mitotane-induced mRNA expression changes in the NCI-H295R adrenocortical cancer cell line. Materials & methods: Cell viability and hormone assays were used to select the optimal mitotane concentration effectively inhibiting hormone secretion without affecting cell viability. RNA isolated from cultures treated for 48 and 72 h was subjected to Agilent 4×44K microarray platforms. Microarray results were validated by quantitative reverse-transcription PCR. Results: Altogether, 117 significantly differentially expressed genes were detected at 48 h and 72 h (p < 0.05) in mitotane-treated samples relative to controls. Three significantly underexpressed genes involved in steroid hormone biosynthesis (HSD3B1, HSD3B2 and CYP21A2) and four significantly overexpressed genes (GDF15, ALDH1L2, TRIB3 and SERPINE2) have been validated. Conclusion: Gene-expression changes might be involved in the adrenal action of mitotane and in the inhibition of hormone secretion. Original submitted 20 January 2012; Revision submitted 17 May 2012
Collapse
Affiliation(s)
- Adrienn Zsippai
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Diana Rita Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Zsófia Tömböl
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Peter M Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Katalin Éder
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Rolf C Gaillard
- Division of Endocrinology, Diabetology & Metabolism, University Hospital Lausanne, Rue du Bugnon 46., CH-1011 Lausanne, Switzerland
| | - Attila Patócs
- Molecular Medicine Research Group, Hungarian Academy of Sciences & Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Sára Tóth
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - András Falus
- Department of Genetics, Cell & Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad square 4, H-1089 Budapest, Hungary
| | - Károly Rácz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| | - Peter Igaz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi street 46, H-1088 Budapest, Hungary
| |
Collapse
|
46
|
Xu HY, Chen ZW, Pan YM, Fan L, Guan J, Lu YY. Transfection of PDCD5 effect on the biological behavior of tumor cells and sensitized gastric cancer cells to cisplatin-induced apoptosis. Dig Dis Sci 2012; 57:1847-56. [PMID: 22359193 DOI: 10.1007/s10620-012-2090-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 02/06/2012] [Indexed: 12/09/2022]
Abstract
BACKGROUND Programmed cell death 5 (PDCD5) expression is reduced in various human tumor cells, and the protein concentration and nuclear translocation of PDCD5 is also observed during tumor cell apoptosis. AIMS The purpose of this study was to investigate the differential expression of PDCD5 in six gastric cell lines, and to explore the changes of biological behavior mechanism underlying enhanced apoptosis-inducing effects of cisplatin by PDCD5 over-expression on gastric cancer BGC823 cells. METHODS RT-PCR and real-time PCR were used to determine PDCD5 expression. BGC823/PDCD5 cells were assessed the cellular proliferating ability by MTT assay, soft agar cloning experiments and tumorigenicity in nude mice experiments in vivo. The effects of cisplatin in combination with PDCD5 on the proliferation and apoptosis were measured by MTT, Annexin-V-FITC/PI dual labeling and cell cycle analysis, respectively. Immunofluorescence was used to detect co-localization of p53 and PDCD5 protein to explore the mechanism underlying the synergistic therapeutic effect of PDCD5 with cisplatin (5 μg/ml for 24 h). RESULTS PDCD5 had the highest expression level in the GES1 cell among other cell lines. The growths of BGC823 cells transfected with PDCD5 for six (6th) or 17 (17th) days were both slower than that of BGC823 and BGC823/Neo (P < 0.01). The stable transfection of PDCD5 demonstrated G2/M cell cycle arrest, increased apoptosis and nuclear translocation of PDCD5 and p53 after cisplatin treatment. CONCLUSIONS Stable transfection of the PDCD5 gene can inhibit the growth of the BGC823 cell line and notably improve apoptosis-inducing effects of cisplatin, indicating a novel strategy for better chemotherapeutic effects on gastric cancer.
Collapse
Affiliation(s)
- Hui-Yu Xu
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang, People's Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Yao L, Lao W, Zhang Y, Tang X, Hu X, He C, Hu X, Xu LX. Identification of EFEMP2 as a Serum Biomarker for the Early Detection of Colorectal Cancer with Lectin Affinity Capture Assisted Secretome Analysis of Cultured Fresh Tissues. J Proteome Res 2012; 11:3281-94. [PMID: 22506683 DOI: 10.1021/pr300020p] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
| | - Weifeng Lao
- Biomedical Research Center and
Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw
Hospital, Zhejiang University, Hangzhou,
China
| | | | | | - Xiaotong Hu
- Biomedical Research Center and
Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw
Hospital, Zhejiang University, Hangzhou,
China
| | - Chao He
- Biomedical Research Center and
Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw
Hospital, Zhejiang University, Hangzhou,
China
| | | | | |
Collapse
|
48
|
Oncogenic KRAS and BRAF activation of the MEK/ERK signaling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition. Oncogene 2012; 32:564-76. [PMID: 22430215 DOI: 10.1038/onc.2012.88] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gain-of-function mutations in KRAS and BRAF genes are found in up to 50% of colorectal cancers. These mutations result in the activation of the BRAF/MEK signaling pathway culminating in the stimulation of ERK1/2 mitogen-activated protein kinases. Upon activation, ERK1/2 translocate from the cytoplasm to the nucleus. This process has been shown to be required for the induction of many cellular responses, although the molecular mechanisms regulating ERK nuclear function, especially under oncogenic stimulation, remain to be explored. Herein, we examined the spatiotemporal regulation of ERK1/2 activity upon oncogenic activation of KRAS(G12V) and BRAF(V600E) in normal intestinal epithelial crypt cells (IECs). Results demonstrate that expression of these oncogenes markedly stimulated ERK1/2 activities and morphologically transformed IECs. Importantly however, ERK phosphorylation was not observed in the nucleus, but restricted to the cytoplasm of KRAS(G12V)- and BRAF(V600E)-transformed IECs. The absence of nuclear ERK phosphorylation was due to a vanadate-sensitive phosphatase activity. Nuclear ERK dephosphorylation was found to be tightly correlated with the rapid expression of DUSP4 phosphatase induced in an MEK-dependent manner. In addition, MEK-dependent phosphorylation of T361, T363, S390 and S395 residues highly stabilized DUSP4 protein. Finally, in human colorectal cancer cells, ERK1/2 activities were also confined to the cytoplasm and treatment with pervanadate reactivated ERK1/2 in the nucleus. Accordingly, DUSP4 mRNAs were found to be highly expressed, in an MEK-dependent manner, in all colorectal cancer cells analyzed. These findings indicate that DUSP4 functions as part of a negative feedback mechanism in the control of the duration and magnitude of nuclear ERK activation during intestinal tumorigenesis.
Collapse
|
49
|
de Paula CAA, Coulson-Thomas VJ, Ferreira JG, Maza PK, Suzuki E, Nakahata AM, Nader HB, Sampaio MU, Oliva MLV. Enterolobium contortisiliquum trypsin inhibitor (EcTI), a plant proteinase inhibitor, decreases in vitro cell adhesion and invasion by inhibition of Src protein-focal adhesion kinase (FAK) signaling pathways. J Biol Chem 2011; 287:170-182. [PMID: 22039045 DOI: 10.1074/jbc.m111.263996] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tumor cell invasion is vital for cancer progression and metastasis. Adhesion, migration, and degradation of the extracellular matrix are important events involved in the establishment of cancer cells at a new site, and therefore molecular targets are sought to inhibit such processes. The effect of a plant proteinase inhibitor, Enterolobium contortisiliquum trypsin inhibitor (EcTI), on the adhesion, migration, and invasion of gastric cancer cells was the focus of this study. EcTI showed no effect on the proliferation of gastric cancer cells or fibroblasts but inhibited the adhesion, migration, and cell invasion of gastric cancer cells; however, EcTI had no effect upon the adhesion of fibroblasts. EcTI was shown to decrease the expression and disrupt the cellular organization of molecules involved in the formation and maturation of invadopodia, such as integrin β1, cortactin, neuronal Wiskott-Aldrich syndrome protein, membrane type 1 metalloprotease, and metalloproteinase-2. Moreover, gastric cancer cells treated with EcTI presented a significant decrease in intracellular phosphorylated Src and focal adhesion kinase, integrin-dependent cell signaling components. Together, these results indicate that EcTI inhibits the invasion of gastric cancer cells through alterations in integrin-dependent cell signaling pathways.
Collapse
Affiliation(s)
- Cláudia Alessandra Andrade de Paula
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Vivien Jane Coulson-Thomas
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Joana Gasperazzo Ferreira
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Paloma Korehisa Maza
- Department of Microbiology, Immunology, and Parasitology, Universidade Federal de São Paulo-Escola Paulista de Medicina, 04044-020 São Paulo, Brazil
| | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Universidade Federal de São Paulo-Escola Paulista de Medicina, 04044-020 São Paulo, Brazil
| | - Adriana Miti Nakahata
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Helena Bonciani Nader
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Misako Uemura Sampaio
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Maria Luiza V Oliva
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil.
| |
Collapse
|
50
|
Wiśniewski JR, Ostasiewicz P, Mann M. High recovery FASP applied to the proteomic analysis of microdissected formalin fixed paraffin embedded cancer tissues retrieves known colon cancer markers. J Proteome Res 2011; 10:3040-9. [PMID: 21526778 DOI: 10.1021/pr200019m] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteomic analysis of samples isolated by laser capture microdissection from clinical specimens requires sample preparation and fractionation methods suitable for small amounts of protein. Here we describe a streamlined filter-aided sample preparation (FASP) workflow that allows efficient analysis of lysates from low numbers of cells. Addition of carrier substances such as polyethylene glycol or dextran to the processed samples improves the peptide yields in the low to submicrogram range. In a single LC-MS/MS run, analyses of 500, 1000, and 3000 cells allowed identification of 905, 1536, and 2055 proteins, respectively. Incorporation of an additional SAX fractionation step at somewhat higher amounts enabled the analysis of formalin fixed and paraffin embedded human tissues prepared by LCM to a depth of 3600-4400 proteins per single experiment. We applied this workflow to compare archival neoplastic and matched normal colonic mucosa cancer specimens for three patients. Label-free quantification of more than 6000 proteins verified this technology through the differential expression of 30 known colon cancer markers. These included Carcino-Embryonic Antigen (CEA), the most widely used colon cancer marker, complement decay accelerating factor (DAF, CD55) and Metastasis-associated in colon cancer protein 1 (MACC1). Concordant with literature knowledge, mucin 1 was overexpressed and mucin 2 underexpressed in all three patients. These results show that FASP is suitable for the low level analysis of microdissected tissue and that it has the potential for exploration of clinical samples for biomarker and drug target discovery.
Collapse
Affiliation(s)
- Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, D-82152 Martinsried, Germany.
| | | | | |
Collapse
|