1
|
Dey S, Ghosh A, Banerjee M. A mathematical modeling technique to understand the role of decoy receptors in ligand-receptor interaction. Sci Rep 2023; 13:6523. [PMID: 37085701 PMCID: PMC10121693 DOI: 10.1038/s41598-023-33596-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/15/2023] [Indexed: 04/23/2023] Open
Abstract
The ligand-receptor interaction is fundamental to many cellular processes in eukaryotic cells such as cell migration, proliferation, adhesion, signaling and so on. Cell migration is a central process in the development of organisms. Receptor induced chemo-tactic sensitivity plays an important role in cell migration. However, recently some receptors identified as decoy receptors, obstruct some mechanisms of certain regular receptors. DcR3 is one such important decoy receptor, generally found in glioma cell, RCC cell and many various malignant cells which obstruct some mechanism including apoptosis cell-signaling, cell inflammation, cell migration. The goal of our work is to mathematically formulate ligand-receptor interaction induced cell migration in the presence of decoy receptors. We develop here a novel mathematical model, consisting of four coupled partial differential equations which predict the movement of glioma cells due to the reaction-kinetic mechanism between regular receptors CD95, its ligand CD95L and decoy receptors DcR3 as obtained in experimental results. The aim is to measure the number of cells in the chamber's filter for different concentrations of ligand in presence of decoy receptors and compute the distance travelled by the cells inside filter due to cell migration. Using experimental results, we validate our model which suggests that the concentration of ligands plays an important role in cell migration.
Collapse
Affiliation(s)
- Subrata Dey
- Department of Mathematics and Statistics, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Aditi Ghosh
- Department of Mathematics, Texas A &M Commerce, Commerce, TX, USA.
| | - Malay Banerjee
- Department of Mathematics and Statistics, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| |
Collapse
|
2
|
Wang B, Xue Y, Zhai W. Integration of Tumor Microenvironment in Patient-Derived Organoid Models Help Define Precision Medicine of Renal Cell Carcinoma. Front Immunol 2022; 13:902060. [PMID: 35592336 PMCID: PMC9111175 DOI: 10.3389/fimmu.2022.902060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common urological tumor, with a poor prognosis, as the result of insensitivity to chemotherapy and radiotherapy. About 20%–30% of patients with RCC have metastasis at the first diagnosis, so only systemic treatment is possible. Due to the heterogeneity of renal tumors, responses to drugs differ from person to person. Consequently, patient-derived organoid, highly recapitulating tumor heterogeneity, becomes a promising model for high-throughput ex vivo drug screening and thus guides the drug choice of patients with RCC. Systemic treatment of RCC mainly targets the tumor microenvironment, including neovasculature and immune cells. We reviewed several methods with which patient-derived organoid models mimic the heterogeneity of not only tumor epithelium but also the tumor microenvironment. We further discuss some new aspects of the development of patient-derived organoids, preserving in vivo conditions in patients with RCC.
Collapse
Affiliation(s)
- Bingran Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yizheng Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Hua X, Ge S, Zhang J, Xiao H, Tai S, Yang C, Zhang L, Liang C. A costimulatory molecule-related signature in regard to evaluation of prognosis and immune features for clear cell renal cell carcinoma. Cell Death Discov 2021; 7:252. [PMID: 34537809 PMCID: PMC8449780 DOI: 10.1038/s41420-021-00646-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/17/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
Costimulatory molecules have been proven to enhance antitumor immune responses, but their roles in clear cell renal cell carcinoma (ccRCC) remain unexplored. In this study, we aimed to explore the gene expression profiles of costimulatory molecule genes in ccRCC and construct a prognostic signature to improve treatment decision-making and clinical outcomes. We performed the first comprehensive analysis of costimulatory molecules in patients with ccRCC and identified 13 costimulatory molecule genes with prognostic values and diagnostic values. Consensus clustering analysis based on these 13 costimulatory molecular genes showed different distribution patterns and prognostic differences for the two clusters identified. Then, a costimulatory molecule-related signature was constructed based on these 13 costimulatory molecular genes, and validated in an external dataset, showing good performance for predicting a patient’s prognosis. The signature was an independent risk factor for ccRCC patients and was significantly correlated with patients’ clinical factors, which could be used as a complement for clinical factors. In addition, the signature was associated with the tumor immune microenvironment and the response to immunotherapy. Patients identified as high-risk based on our signature exhibited a high mutation frequency, a high level of immune cell infiltration, and an immunosuppressive microenvironment. High-risk patients tended to have high cytolytic activity scores and immunophenoscore of CTLA4 and PD1/PD-L1/PD-L2 blocker than low-risk patients, suggesting these patients may be more suitable for immunotherapy. Therefore, our signature could provide clinicians with prognosis predictions and help guide treatment for ccRCC patients.
Collapse
Affiliation(s)
- Xiaoliang Hua
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,The Institute of Urology, Anhui Medical University, Hefei, China
| | - Shengdong Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,The Institute of Urology, Anhui Medical University, Hefei, China
| | - Jiong Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,The Institute of Urology, Anhui Medical University, Hefei, China
| | - Haibing Xiao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,The Institute of Urology, Anhui Medical University, Hefei, China
| | - Sheng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China.,The Institute of Urology, Anhui Medical University, Hefei, China
| | - Cheng Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China. .,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China. .,The Institute of Urology, Anhui Medical University, Hefei, China.
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China. .,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China. .,The Institute of Urology, Anhui Medical University, Hefei, China.
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China. .,Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China. .,The Institute of Urology, Anhui Medical University, Hefei, China. .,Anhui Institute of translational medicine, Hefei, China.
| |
Collapse
|
4
|
Nivolumab Reduces PD1 Expression and Alters Density and Proliferation of Tumor Infiltrating Immune Cells in a Tissue Slice Culture Model of Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13184511. [PMID: 34572738 PMCID: PMC8471479 DOI: 10.3390/cancers13184511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Immune checkpoint inhibitors (ICIs) have become a first-choice therapy option in the treatment of clear cell renal cell carcinoma (ccRCC). A predictive biomarker is urgently needed since not all patients respond and adverse events occur. Therefore, an ex vivo tissue slice culture (TSC) model was tested to investigate the effects of nivolumab on tumor infiltrating immune cells (TIIC). A decrease in programmed death receptor 1 expression, as well as effects on density and proliferation of TIIC, were observed. Thus, the TSC model could serve as a test platform for response prediction to ICIs. Abstract Background: In the treatment of clear cell renal cell carcinoma (ccRCC), nivolumab is an established component of the first-line therapy with a favorable impact on progression free survival and overall survival. However, treatment-related adverse effects occur and, to date, there is no approved predictive biomarker for patient stratification. Thus, the aim of this study was to establish an ex vivo tissue slice culture model of ccRCC and to elucidate the impact of nivolumab on tumor infiltrating immune cells. Methods: Fresh tumor tissue of ccRCC was treated with the immune checkpoint inhibitor nivolumab using ex vivo tissue slice culture (TSC). After cultivation, tissue slices were formalin-fixed, immunohistochemically stained and analyzed via digital image analysis. Results: The TSC model was shown to be suitable for ex vivo pharmacological experiments on intratumoral immune cells in ccRCC. PD1 expression on tumor infiltrating immune cells was dose-dependently reduced after nivolumab treatment (p < 0.01), whereas density and proliferation of tumor infiltrating T-cells and cytotoxic T-cells were inter-individually altered with a remarkable variability. Tumor cell proliferation was not affected by nivolumab. Conclusions: This study could demonstrate nivolumab-dependent effects on PD1 expression and tumor infiltrating T-cells in TSC of ccRCC. This is in line with results from other scientific studies about changes in immune cell proliferation in peripheral blood in response to nivolumab. Thus, TSC of ccRCC could be a further step to personalized medicine in terms of testing the response of individual patients to nivolumab.
Collapse
|
5
|
Contartese D, Salamanna F, Veronesi F, Fini M. Relevance of humanized three-dimensional tumor tissue models: a descriptive systematic literature review. Cell Mol Life Sci 2020; 77:3913-3944. [PMID: 32285137 PMCID: PMC11104864 DOI: 10.1007/s00018-020-03513-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
Despite numerous advances in tumor screening, diagnosis, and treatment, to date, tumors remain one of the leading causes of death, principally due to metastasis and the physiological damage produced by tumor growth. Among the main limits related to the study of tumor physiology there is the complex and heterogeneity nature of its environment and the absence of relevant, simple and inexpensive models able to mimic the biological processes occurring in patients allowing the correct clinical translation of results. To enhance the understanding of the mechanisms of tumors and to develop and evaluate new therapeutic approaches the set-up of advanced and alternative models is mandatory. One of the more translational approaches seems to be the use of humanized three-dimensional (3D) tissue culture. This model allows to accurately mimic tumor morphology and biology, maintaining the native microenvironment without any manipulation. However, little is still known on the real clinical relevance of these models for the study of tumor mechanisms and for the screening of new therapy. The aim of this descriptive systematic literature review was to evaluate and summarize the current knowledge on human 3D tumor tissue culture models. We reviewed the strategies employed by researchers to set-up these systems, also considering the different approaches and culture conditions used. All these aspects greatly contribute to the existing knowledge on tumors, providing a specific link to clinical scenarios and making the humanized 3D tumor tissue models a more attractive tool both for researchers and clinicians.
Collapse
Affiliation(s)
- D Contartese
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - Francesca Salamanna
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy.
| | - F Veronesi
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - M Fini
- Laboratory Preclinical and Surgical Studies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
6
|
Zhu HF, Liu YP, Liu DL, Ma YD, Hu ZY, Wang XY, Gu CS, Zhong Y, Long T, Kan HP, Li ZG. Role of TGFβ3-Smads-Sp1 axis in DcR3-mediated immune escape of hepatocellular carcinoma. Oncogenesis 2019; 8:43. [PMID: 31409774 PMCID: PMC6692328 DOI: 10.1038/s41389-019-0152-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/04/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of tumour-associated mortality worldwide, but no significant improvement in treating HCC has been reported with currently available systemic therapies. Immunotherapy represents a new frontier in tumour therapy. Therefore, the immunobiology of hepatocarcinoma has been under intensive investigation. Decoy receptor 3 (DcR3), a member of the tumour necrosis factor receptor (TNFR) superfamily, is an immune suppressor associated with tumourigenesis and cancer metastasis. However, little is known about the role of DcR3 in the immunobiology of hepatocarcinoma. In this study, we found that overexpression of DcR3 in HCC is mediated by the TGFβ3-Smad-Sp1 signalling pathway, which directly targets DcR3 promoter regions. Moreover, overexpression of DcR3 in HCC tissues is associated with tumour invasion and metastasis and significantly promotes the differentiation and secretion of Th2 and Treg cells while inhibiting the differentiation and secretion of Th1 cells. Conversely, knockdown of DcR3 expression in HCC significantly restored the immunity of CD4+ T cells. Inhibition of DcR3 expression may provide a novel immunotherapeutic approach to restoring immunity in HCC patients.
Collapse
Affiliation(s)
- Hui-Fang Zhu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 601 Jinsui Road, 453003, Xinxiang, Henan, China
| | - Yan-Ping Liu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Ding-Li Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1023 South Shatai Road, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Yi-Dan Ma
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Zhi-Yan Hu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Xiao-Yan Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Chuan-Sha Gu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, 601 Jinsui Road, 453003, Xinxiang, Henan, China
| | - Yan Zhong
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - Ting Long
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China
| | - He-Ping Kan
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China.
| | - Zu-Guo Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 1023 South Shatai Rd, Baiyun District, 510515, Guangzhou, Guangdong, China. .,Department of Pathology, Shenzhen Hospital, Southern Medical University, 1333 Xin-hu Road, Bao'an District, 518100, Shenzhen, Guangdong, China. .,Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, 1333 Xin-hu Road, Bao'an District, 518100, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Chang WC, Yeh YC, Ho HL, Hsieh SL, Chou TY. Decoy Receptor 3 Expression Is Associated With Wild-Type EGFR Status, Poor Differentiation of Tumor, and Unfavorable Patient Outcome. Am J Clin Pathol 2019; 152:207-216. [PMID: 31077284 DOI: 10.1093/ajcp/aqz035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES The role of decoy receptor 3 (DcR3) in lung cancer, particularly adenocarcinoma, has not been well studied. In this study, we aim to investigate the expression profile and the clinicopathologic implications of DcR3 expression in lung adenocarcinoma. METHODS Immunohistochemistry was used to examine DcR3 expression in 461 lung adenocarcinomas. The differences in DcR3 expression among the various histopathologic patterns were analyzed. The relationship between DcR3 expression and clinicopathologic parameters, including epidermal growth factor receptor (EGFR) mutation, was also investigated. RESULTS DcR3 expression was more frequently expressed in solid, micropapillary, and acinar patterns (P < .0001) and in tumors with wild-type EGFR status (P = .018). In addition, DcR3 expression portends a less favorable disease-free survival in stage I patients (P = .012). CONCLUSIONS The expression of DcR3 might be involved in the differentiation and progression of lung adenocarcinoma. Therefore, DcR3 may be applied clinically for prediction of tumor progression in stage I lung adenocarcinoma.
Collapse
Affiliation(s)
- Wei-Chin Chang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology, MacKay Memorial Hospital and MacKay Medical College, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Liang DY, Sha S, Yi Q, Shi J, Chen Y, Hou Y, Chang Q. Hepatitis B X protein upregulates decoy receptor 3 expression via the PI3K/NF-κB pathway. Cell Signal 2019; 62:109346. [PMID: 31229617 DOI: 10.1016/j.cellsig.2019.109346] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
Chronic hepatitis B (CHB) is associated with the development of hepatocellular carcinoma (HCC). Decoy receptor 3 (DcR3) is a tumor necrosis factor receptor that promotes tumor cell survival by inhibiting apoptosis and interfering with immune surveillance. Previous studies showed that DcR3 was overexpressed in HCC cells and that short hairpin RNA (shDcR3) sensitizes TRAIL-resistant HCC cells. However, the expression of DcR3 during hepatitis B virus (HBV) infection has not been investigated. Here, we demonstrated that DcR3 was overexpressed in CHB patients and that DcR3 upregulation was positively correlated with the HBV DNA load and liver injury (determined by histological activity index, serum alanine aminotransferase level, and aspartate aminotransferase level). We found that hepatitis B virus X protein (HBx) upregulated DcR3 expression in a dose-dependent manner, but this increase was blocked by NF-κB inhibitors. HBx also induced the activation of NF-κB, and the NF-κB subunits p65 and p50 upregulated DcR3 by directly binding to the DcR3 promoters. Inhibition of PI3K significantly downregulated DcR3 and inhibited the binding of NF-κB to the DcR3 promoters. Our results demonstrate that the HBx induced DcR3 expression via the PI3K/NF-κB pathway; this process may contribute to the development of HBV-mediated HCC.
Collapse
MESH Headings
- Binding Sites/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/virology
- Gene Expression Regulation, Neoplastic/genetics
- Hep G2 Cells
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/pathology
- Hepatitis B, Chronic/virology
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/virology
- NF-kappa B p50 Subunit/genetics
- Phosphatidylinositol 3-Kinases/genetics
- Promoter Regions, Genetic/genetics
- Protein Binding/genetics
- RNA, Small Interfering/genetics
- Receptors, Tumor Necrosis Factor, Member 6b/genetics
- Signal Transduction/genetics
- Trans-Activators/genetics
- Transcription Factor RelA/genetics
- Viral Regulatory and Accessory Proteins
Collapse
Affiliation(s)
- Dong-Yu Liang
- Shanghai General Practice Medical Education and Research Center, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, China; College of medical technology, Shanghai University of Medicine & Health Sciences, China
| | - Shuang Sha
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Qingqing Yi
- Shanghai General Practice Medical Education and Research Center, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, China
| | - Junfeng Shi
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yingmin Chen
- Shanghai General Practice Medical Education and Research Center, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, China
| | - Yanqiang Hou
- Department of Central Laboratory, Shanghai Songjiang District Central Hospital, Shanghai 201600, China.
| | - Qing Chang
- Shanghai General Practice Medical Education and Research Center, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, China.
| |
Collapse
|
9
|
DcR3 induces epithelial-mesenchymal transition through activation of the TGF-β3/SMAD signaling pathway in CRC. Oncotarget 2018; 7:77306-77318. [PMID: 27764793 PMCID: PMC5363587 DOI: 10.18632/oncotarget.12639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/28/2016] [Indexed: 12/11/2022] Open
Abstract
Decoy receptor 3 (DcR3), a novel member of the tumor necrosis factor receptor (TNFR) family, was recently reported to be associated with tumorigenesis and metastasis. However, the role of DcR3 in human colorectal cancer (CRC) has not been fully elucidated. In this study, we found that DcR3 expression was significantly higher in human colorectal cancer tissues than in paired normal tissues, and that DcR3 expression was strongly correlated with tumor invasion, lymph node metastases and poor prognoses. Moreover, DcR3 overexpression significantly enhanced CRC cell proliferation and migration in vitro and tumorigenesis in vivo. Conversely, DcR3 knockdown significantly repressed CRC cell proliferation and migration in vitro, and DcR3 deficiency also attenuated CRC tumorigenesis and metastasis in vivo. Functionally, DcR3 was essential for TGF-β3/SMAD-mediated epithelial-mesenchymal transition (EMT) of CRC cells. Importantly, cooperation between DcR3 and TGF-β3/SMAD-EMT signaling-related protein expression was correlated with survival and survival time in CRC patients. In conclusion, our results demonstrate that DcR3 may be a prognostic biomarker for CRC and that this receptor facilitates CRC development and metastasis by participating in TGF-β3/SMAD-mediated EMT of CRC cells.
Collapse
|
10
|
Xu YC, Cui J, Zhang LJ, Zhang DX, Xing BC, Huang XWY, Wu JX, Liang CJ, Li GM. Anti-apoptosis Effect of Decoy Receptor 3 in Cholangiocarcinoma Cell Line TFK-1. Chin Med J (Engl) 2018; 131:82-87. [PMID: 29271385 PMCID: PMC5754963 DOI: 10.4103/0366-6999.221271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: Decoy receptor 3 (DcR3) is a protein with anti-apoptotic effect that belongs to the tumor necrosis factor receptor superfamily. DcR3 is highly expressed in a variety of malignant tumors including cholangiocarcinoma and its expression was found to be related to the clinical stage, the invasion, and the metastasis of the tumor. This in vitro study aimed to investigate the effect of downregulated expression of DcR3 on cell viability, cell apoptosis, and cell cycle in cholangiocarcinoma cell line TFK-1. Methods: Three different cell lines were cultured: human cholangiocarcinoma TFK-1, human biliary epithelial carcinoma HuCCT-1, and human cholangiocarcinoma RBE. The cholangiocarcinoma cell line with the highest expression of DcR3 was selected for further investigation. The expression of DcR3 was silenced/knocked down by transfection with DcR3-siRNA in the selected cell line. Various biological phenotype parameters such as cell viability, apoptosis, and cell cycle were observed. Results: The mRNA and protein levels of DcR3 were measured in the three cell lines, and TFK-1 was selected. After the treatment with DcR3-siRNA for 48 h, DcR3 mRNA and protein expression in the treatment group were 38.45% (P < 0.01) and 48.03% (P < 0.05) of that of the control, respectively. It was found that the cell viability decreased to 61.87% of the control group (P < 0.01) after the downregulation of DcR3 in cholangiocarcinoma cell line TFK-1 by transfection with DcR3-siRNA, while the percentage of apoptotic cells was 2.98 times as compared with the control group (P < 0.05). Compared with the control group the ratio of G0/G1 increased, and the ratio of G2/M decreased in the treatment group. However, the differences were not statistically significant. Conclusions: The effect of DcR3 on the growth and apoptosis of cholangiocarcinoma has been demonstrated. DcR3 is not only a predictive marker for malignant tumor but it is also likely to be a potential target for cancer gene therapy. Further studies should focus on exploring the binding ligand of DcR3, the signaling pathway involved, and the molecular mechanism for the regulation of DcR3 expression in cholangiocarcinoma.
Collapse
Affiliation(s)
- Ying-Chen Xu
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Jing Cui
- Department of Pathology, Capital Medical University, Beijing 100069, China
| | - Li-Jun Zhang
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Dong-Xin Zhang
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Bing-Chen Xing
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiong-Wei-Ye Huang
- Department of Pathology, Capital Medical University, Beijing 100069, China
| | - Ji-Xiang Wu
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Chao-Jie Liang
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| | - Guang-Ming Li
- Department of Hepatobiliary Surgery, Surgical Laboratory, Beijing Tong Ren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
11
|
Tsai HW, Huang MT, Wang PH, Huang BS, Chen YJ, Hsieh SL. Decoy receptor 3 promotes cell adhesion and enhances endometriosis development. J Pathol 2017; 244:189-202. [PMID: 29057478 DOI: 10.1002/path.5000] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/07/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023]
Abstract
Endometriosis is a multifactorial inflammatory disease with persistent activation of the nuclear factor-κB (NF-κB) signalling pathway. Aberrant adhesion of endometrium is the essential step in the progression of endometriosis, but the molecular mechanism of ectopic growth of endometrium is still unclear. Decoy receptor 3 (DcR3)/TNFRSF6B, a pleiotropic immunomodulator regulated by oestrogen, is able to activate focal adhesion kinase to promote cell adhesion. We found that DcR3 is upregulated in human ectopic endometrial cells via activation of the Akt-NF-κB signalling pathway, and its expression level correlates positively with that of the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) and homing cell adhesion molecule (HCAM; CD44). In a multivariate regression model, DcR3 expression level was the most significant parameter associated with endometriosis severity. Knockdown of DcR3 not only downregulated the expression of ICAM-1 and HCAM, but also reduced cell adhesion and migration. In vivo investigation further showed that DcR3 promoted the growth and spread of endometrium, whereas knockdown of DcR3 by lentivirus-delivered short hairpin RNA inhibited ectopic adhesion of endometrium and abrogated endometriosis progression. These observations are in support of DcR3 playing a critical role in the pathogenesis of endometriosis, and the inhibition of DcR3 expression being a promising approach for the treatment of endometriosis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hsiao-Wen Tsai
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | - Peng-Hui Wang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Obstetrics and Gyneacology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ben-Shian Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Obstetrics and Gyneacology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Obstetrics and Gyneacology, Taipei Veterans General Hospital, Taipei, Taiwan.,Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Genomics Research Centre, Academia Sinica, Taipei, Taiwan.,Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Abstract
Decoy receptor 3 (DcR3), also known as tumor necrosis factor receptor (TNFR) superfamily member 6b (TNFRSF6B), is a soluble decoy receptor which can neutralize the biological functions of three members of tumor necrosis factor superfamily (TNFSF): Fas ligand (FasL), LIGHT, and TL1A. In addition to ‘decoy’ function, recombinant DcR3.Fc is able to modulate the activation and differentiation of dendritic cells (DCs) and macrophages via ‘non-decoy’ action. DcR3-treated DCs skew T cell differentiation into Th2 phenotype, while DcR3-treated macrophages behave M2 phenotype. DcR3 is upregulated in various cancer cells and several inflammatory tissues, and is regarded as a potential biomarker to predict inflammatory disease progression and cancer metastasis. However, whether DcR3 is a pathogenic factor or a suppressor to attenuate inflammatory reactions, has not been discussed comprehensively yet. Because mouse genome does not have DcR3, it is not feasible to investigate its physiological functions by gene-knockout approach. However, DcR3-mediated effects in vitro are determined via overexpressing DcR3 or addition of recombinant DcR3.Fc fusion protein. Moreover, CD68-driven DcR3 transgenic mice are used to investigate DcR3-mediated systemic effects in vivo. Upregulation of DcR3 during inflammatory reactions exerts negative-feedback to suppress inflammation, while tumor cells hijack DcR3 to prevent apoptosis and promote tumor growth and invasion. Thus, ‘switch-on’ of DcR3 expression may be feasible for the treatment of inflammatory diseases and enhance tissue repairing, while ‘switch-off’ of DcR3 expression can enhance tumor apoptosis and suppress tumor growth in vivo.
Collapse
Affiliation(s)
- Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan. .,Institute of Clinical Medicine & Immunology Research Center, National Yang-Ming University, Taipei, Taiwan. .,Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan. .,Institute of Immunology, College of Medicine, National Taiwan University Taipei, Taipei, Taiwan. .,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, No. 1 Section 1, Jen Ai Road, Taipei, 10001, Taiwan.
| |
Collapse
|
13
|
Zhang H, Chen X, Li D, Cui L, Li X, Ye X, Wan X. DcR3 promotes hepatoma cell migration by downregulating E-cadherin expression. Oncol Rep 2017; 38:377-383. [PMID: 28560426 DOI: 10.3892/or.2017.5685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/27/2016] [Indexed: 11/06/2022] Open
Abstract
Decoy receptor 3 (DcR3), a decoy molecule belonging to the tumor necrosis factor receptor superfamily (TNFRSF), is a soluble receptor that can neutralize the biological effects of three other TNFSF members, namely, Fas ligand (FasL/TNFSF6/CD95L), LIGHT (TNFSF14) and TNF-like molecule 1A (TL1A/TNFSF15). DcR3 expression is increased in tumor cells. As such, DcR3 has been considered a potential biomarker to predict cancer invasion and progression of inflammation. However, the molecular mechanisms of DcR3 in tumor progression and metastasis remain poorly described. In the present study, DcR3 induced cytoskeleton remodeling, inhibited E-cadherin expression, and promoted cancer cell migration. Immunofluorescence and flow cytometry demonstrated that DcR3 expression was increased in hepatoma cells, whereas E-cadherin expression was significantly downregulated. Immunohistochemistry revealed that DcR3 and E-cadherin exhibited an opposite expression pattern between normal and cancerous liver tissues. Moreover, DcR3 treatment promoted IκBα degradation and p65 nuclear translocation. Therefore, the present study uncovered the mechanism underlying the function of DcR3 in cancer cell migration and provides evidence that DcR3 may be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Hongling Zhang
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xuhong Chen
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, P.R. China
| | - Dongming Li
- Basic Medicine College, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lulu Cui
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xin Li
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| | - Xiufeng Ye
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, P.R. China
| | - Xiaochun Wan
- Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P.R. China
| |
Collapse
|
14
|
Chen J, Guo XZ, Li HY, Zhao JJ, Xu WD. Dendritic cells engineered to secrete anti-DcR3 antibody augment cytotoxic T lymphocyte response against pancreatic cancer in vitro. World J Gastroenterol 2017; 23:817-829. [PMID: 28223726 PMCID: PMC5296198 DOI: 10.3748/wjg.v23.i5.817] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/04/2016] [Accepted: 12/21/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the enhanced cytotoxic T lymphocyte responses against pancreatic cancer (PC) in vitro induced by dendritic cells (DCs) engineered to secrete anti-DcR3 monoclonal antibody (mAb).
METHODS DCs, T lymphocytes and primary PC cells were obtained from PC patients. DCs were transfected with a designed humanized anti-DcR3 monoclonal antibody heavy and light chain mRNA and/or total tumor RNA (DC-tumor-anti-DcR3 RNA or DC-total tumor RNA) by using electroporation technology. The identification, concentration and function of anti-DcR3 mAb secreted by DC-tumor-anti-DcR3 RNA were determined by western blotting and enzyme-linked immunosorbent assay. After co-culturing of autologous isolated PC cells with target DCs, the effects of secreting anti-DcR3 mAb on RNA-DCs’ viability and apoptosis were assessed by MTT assay and flow cytometry. Analysis of enhanced antigen-specific immune response against PC induced by anti-DcR3 mAb secreting DCs was performed using a 51Cr releasing test. T cell responses induced by RNA-loaded DCs were analyzed by measuring cytokine levels, including IFN-γ, IL-10, IL4, TNF-α and IL-12.
RESULTS The anti-DcR3 mAb secreted by DCs reacted with recombinant human DcR3 protein and generated a band with 35 kDa molecular weight. The secreting mAb was transient, peaking at 24 h and becoming undetectable after 72 h. After co-incubation with DC-tumor-anti-DcR3 RNA for designated times, the DcR3 level in the supernatant of autologous PC cells was significantly down-regulated (P < 0.05). DCs secreting anti-DcR3 mAb could improve cell viability and slow down the apoptosis of RNA-loaded DCs, compared with DC-total tumor RNA (P < 0.01). The anti-DcR3 mAb secreted by DC-tumor-anti-DcR3 RNA could enhance the induction of cytotoxic T lymphocytes (CTLs) activity toward RNA-transfected DCs, primary tumor cells, and PC cell lines, compared with CTLs stimulated by DC-total tumor RNA or control group (P < 0.05). Meanwhile, the antigen-specific CTL responses were MHC class I-restricted. The CD4+ T cells and CD8+ T cells incubated with anti-DcR3 mAb secreting DCs could produce extremely higher level IFN-γ and lower level IL4 than those incubated with DC-total tumor RNA or controls (P < 0.01).
CONCLUSION DCs engineered to secrete anti-DcR3 antibody can augment CTL responses against PC in vitro, and the immune-enhancing effects may be partly due to their capability of down-regulating DC apoptosis and adjusting the Th1/Th2 cytokine network.
Collapse
|
15
|
Liang C, Xu Y, Li G, Zhao T, Xia F, Li G, Zhang D, Wu J. Downregulation of DcR3 sensitizes hepatocellular carcinoma cells to TRAIL-induced apoptosis. Onco Targets Ther 2017; 10:417-428. [PMID: 28176915 PMCID: PMC5261847 DOI: 10.2147/ott.s127202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Decoy receptor 3 (DcR3) has been recently described as an antiapoptosis and prometastasis factor since it can competitively bind to FasL, TL1A, and LIGHT, and it is highly expressed in many malignant tumors. Downregulation of DcR3 can promote tumor cell apoptosis and inhibit metastasis. A previous study demonstrated that reduction of DcR3 could induce tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis in pancreatic cancer cells. However, whether such an effect is seen in hepatocellular carcinoma (HCC) remains to be explored. This study was designed to investigate the sensitivity of HCC cells to TRAIL after silencing DcR3, and this was done by evaluating the expression of DcR3 in HCC cells and the effect on TRAIL-mediated apoptosis after downregulation of DcR3. Our data showed that DcR3 was highly expressed in HepG2, BEL-7402, Hep3B, Huh-7, MHCC97H, and SMCC7721 cell lines compared with normal liver cell line LO-2. Both HepG2 and BEL-7402 were tolerant to TRAIL-mediated apoptosis, and the tolerance was negatively correlated to the expression of DcR3. Silencing of DcR3 with shRNA and treatment with TRAIL induced obvious apoptosis in HepG2 and BEL-7402, with more cancer cells found in the G1 phase. SiDcR3 combined with TRAIL could induce activation of caspases-3, -8, and -9, raise the expression of the apoptotic protein Bax, and reduce the expression of antiapoptotic proteins (Bcl-2, Mcl-1, Bcl-XL, IAP-2, and survivin). Caspase-8 inhibitor Ac-IETD-CHO significantly decreased the activation of caspase cascade, indicating that the extrinsic pathway may have a vital role in the apoptotic events induced by SiDcR3/TRAIL. Furthermore, our results showed that the TRAIL death receptor 5 (DR5) was upregulated and that DR5 neutralizing antibody abrogated the effect of SiDcR3. Our results demonstrated that downregulation of DcR3 could enhance TRAIL-mediated apoptosis in HCC through the death receptor pathway. In the future, this might be useful as a clinical treatment method of liver cancer.
Collapse
Affiliation(s)
- Chaojie Liang
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yingchen Xu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guangming Li
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Tuanjie Zhao
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Feng Xia
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guanqun Li
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dongxin Zhang
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jixiang Wu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
16
|
Roth W. [Cell death in malignant tumors. Relevance of cell death regulation for metastasis]. DER PATHOLOGE 2016; 36 Suppl 2:181-4. [PMID: 26400565 DOI: 10.1007/s00292-015-0080-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Defects in the regulation of cell death are important causes for both the development and therapy resistance of malignant tumors. Several distinct, molecularly defined types of cell death are known, such as apoptosis, anoikis, and necroptosis. Moreover, the specific triggering of cell death plays an important role in the prevention of metastasis. The results of recent studies have shown that various types of cell death are pivotal at different steps of the metastasis cascade, in order to prevent cellular detachment, migration, invasion, intravasation, extravasation and the establishment of micrometastasis and macrometastasis. At the subcellular level, numerous links exist between cell death regulation and metastasis, specifically regarding signaling pathways and individual proteins with dual or multiple functions. As an example, the decoy receptor 3 protein (DcR3) functions both as an anti-apoptotic protein and as a direct promotor of invasion and migration of tumor cells. In summary, the specific triggering of cell death plays a pivotal role for the prevention of metastasis. On the other hand, the stepwise process of metastasis represents a mechanism of selection resulting in established metastases with a multiresistant phenotype which corresponds to the clinical observation that many metastasized cancers are therapy resistant. In the future, innovative diagnostic tests to individually predict the resistance pattern and possibilities to overcome resistance are urgently needed.
Collapse
Affiliation(s)
- W Roth
- Pathologisches Institut, Deutsches Krebsforschungszentrum Heidelberg, Universität Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland.
| |
Collapse
|
17
|
Zhang X, Takata K, Cui W, Miyata-Takata T, Sato Y, Noujima-Harada M, Yoshino T. Protocadherin γ A3 is expressed in follicular lymphoma irrespective of BCL2 status and is associated with tumor cell growth. Mol Med Rep 2016; 14:4622-4628. [PMID: 27748813 PMCID: PMC5102029 DOI: 10.3892/mmr.2016.5808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/18/2016] [Indexed: 01/24/2023] Open
Abstract
Protocadherin genes (PCDHs) have been suggested to act as tumor suppressor genes in various tumor types. Previous studies have demonstrated the upregulation of certain PCDH-γ subfamily genes in nodal and duodenal follicular lymphoma (FL) using gene expression analyses. However, the mechanisms and associated molecular function of PCDH-γ subfamily gene upregulation in FL remain to be elucidated. The present study examined the expression of PCDHGA3, an upregulated PCDH-γ gene subfamily member, in B-cell lymphoma 2 (BCL2)-positive and -negative FL, and evaluated its association with tumor cell proliferation in an FL-derived cell line. Immunohistochemical analysis demonstrated that the majority of FL grade 1–2 samples (19/20; 95%) and over half of grade 3A FL samples (5/9; 56%) were PCDHGA3-positive, whereas only 1/17 reactive lymphoid hyperplasia samples was positive. Notably, this positivity was widely observed in samples of BCL2-negative FL (13/15; 87%) and FL with diffuse area (10/10; 100%). The FL-derived cell line FL18 exhibited strong PCDHGA3 expression, similar to the patient samples, and its proliferation was suppressed by PCDHGA3 gene knockdown. Genes expressed concomitantly with PCDHGA3 were selected from gene expression data, and TNFRSF6B, a member of the tumor necrosis factor receptor superfamily, was among the top five most strongly correlated genes. Coexpression of TNFRSF6B and PCDHGA3 was observed immunohistochemically in FL18 cells, suggesting potential cooperation in tumor cell maintenance. In conclusion, the results of the present study indicated that PCDHGA3 was expressed in FL irrespective of BCL2 status and grading and was associated with cell proliferation. Further studies involving molecular genetic analyses are required to elucidate the mechanisms underlying the activity of PCDHGA3 in FL.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Katsuyoshi Takata
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Wei Cui
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Tomoko Miyata-Takata
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Yasuharu Sato
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Mai Noujima-Harada
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700‑8558, Japan
| |
Collapse
|
18
|
Zhang Y, Huang S, Leng Y, Chen X, Liu T, Wang H, Wei F, Luo D, Chen G, Wei Z. Effect of DcR3-specific siRNA on cell growth suppression and apoptosis induction in glioma cells via affecting ERK and AKT. Onco Targets Ther 2016; 9:5195-202. [PMID: 27621648 PMCID: PMC5010168 DOI: 10.2147/ott.s108395] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previously, we found that the expression of decoy receptor 3 (DcR3) in gliomas was significantly upregulated compared to normal brain tissues. However, the effect of DcR3-specific small interfering RNA (siRNA) on cell biological function of glioma cells remains incompletely understood. OBJECTIVE The aim of this study was to explore the effect of DcR3 siRNA on cell growth and apoptosis of glioma cells and to investigate the potential downstream pathways affected by DcR3. METHODS DcR3-specific siRNA was transfected into three glioma cell lines (U251MG, LN-308, and U87MG) using combiMAGnetofection method. MTS tetrazolium assay and fluorimetric resorufin viability assay were used to assess the growth of glioma cells. Then, apoptosis was examined using the Hoechst 33342/propidium iodide double-staining assay and fluorescent caspase-3/7 assay. Meanwhile, Western blot was performed to explore the probable pathway by which DcR3-specific siRNA acts in glioma cells. Also, microarray dataset analysis was applied to analyze the potential function of DcR3 in glioma. RESULTS The DcR3-specific siRNA had a potent effect on cell growth and apoptosis of all three glioma cells tested, and the effects were time dependent. Among these three glioma cell lines, U251MG had the most significant effect with regard to growth inhibition and apoptosis induction. MTS assay showed that the proliferation rate at 72 and 96 hours after the transfection was 76.333%±5.131% (t=7.611, P=0.002) and 64.333%±5.859% (t=10.983, P<0.001), respectively. The viability rate of U251MG cells was 80.667%±2.309% (t=12.302, P<0.001) and 62.333%±2.082% (t=21.213, P<0.001) at 72 and 96 hours posttreatment, respectively. The caspase-3/7 activity of U251MG cells was 2.76 (t=-6.601, P=0.003) and 4.75 (t=-9.189, P=0.001) folds that of the mock control at 72 and 96 hours, respectively. The apoptosis rate was increased to 1.85 (t=-2.496, P=0.067) and 3.93 (t=-12.587, P<0.001) folds at 72 and 96 hours after transfection, respectively. Furthermore, the levels of phospho-ERK1/2 and phospho-AKT were significantly downregulated after DcR3 silencing. CONCLUSION The DcR3-specific siRNA could efficiently inhibit growth and induce apoptosis of cells via affecting ERK and AKT. Hence, DcR3-specific siRNA treatment could act as a supplementary targeted therapy strategy for gliomas.
Collapse
Affiliation(s)
| | - Suning Huang
- Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | | | | | | | | | | | | | | | - Zhuxin Wei
- Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
19
|
Scherr AL, Gdynia G, Salou M, Radhakrishnan P, Duglova K, Heller A, Keim S, Kautz N, Jassowicz A, Elssner C, He YW, Jaeger D, Heikenwalder M, Schneider M, Weber A, Roth W, Schulze-Bergkamen H, Koehler BC. Bcl-xL is an oncogenic driver in colorectal cancer. Cell Death Dis 2016; 7:e2342. [PMID: 27537525 PMCID: PMC5108319 DOI: 10.1038/cddis.2016.233] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/22/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the second most common malignant neoplasia in women and men worldwide. The B-cell lymphoma 2 (Bcl-2) protein family is mainly known for its pivotal role in the regulation of the mitochondrial death pathway. Anti-apoptotic Bcl-2 proteins may provide survival benefits and induce therapy resistance in cancer cells. Among anti-apoptotic Bcl-2 proteins, we found solely Bcl-xL strongly upregulated in human CRC specimens. In order to study protein function in the context of tumor initiation and progression in vivo, we generated a mouse model lacking Bcl-xL in intestinal epithelial cells (Bcl-xLIEC-KO). If challenged in an inflammation-driven tumor model, Bcl-xLIEC-KO mice showed a significantly reduced tumor burden with lower tumor numbers per animal and decreased tumor sizes. Analysis of cell death events by immunohistochemistry and immunoblotting revealed a striking increase of apoptosis in Bcl-xL-negative tumors. qRT-PCR and immunohistochemistry excluded changes in proliferative capacity and immune cell infiltration as reasons for the reduced tumor load and thereby identify apoptosis as key mechanism. Human CRC tissue was cultured ex vivo and treated with the small molecule compound ABT-737, which inhibits Bcl-xL and Bcl-2. Under ABT-737 treatment, the amount of apoptotic tumor cells significantly increased compared with controls, whereas proliferation levels remained unaltered. In summary, our findings identify Bcl-xL as a driver in colorectal tumorigenesis and cancer progression, making it a valuable target for clinical application.
Collapse
Affiliation(s)
- Anna-Lena Scherr
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - Georg Gdynia
- Clinical Cooperation Unit (CCU) Molecular Tumor Pathology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Institute of Pathology, Department of Surgical Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Mariam Salou
- Division of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Katarina Duglova
- Institute of Pathology, Department of Surgical Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Anette Heller
- Institute of Pathology, Department of Surgical Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Sophia Keim
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - Nicole Kautz
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - Adam Jassowicz
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - Christin Elssner
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dirk Jaeger
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München and Helmholtz Zentrum München, Munich 81675, Germany.,Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Achim Weber
- Institute of Surgical Pathology, University and University Hospital Zurich, Zurich 8091, Switzerland
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz 55131, Germany
| | | | - Bruno Christian Koehler
- National Center for Tumor Diseases, Department of Medical Oncology and Heidelberg University Hospital, Internal Medicine VI, 69120 Heidelberg, Germany
| |
Collapse
|
20
|
Zhang Y, Luo J, He R, Huang W, Li Z, Li P, Dang Y, Chen G, Li S. Expression and clinicopathological implication of DcR3 in lung cancer tissues: a tissue microarray study with 365 cases. Onco Targets Ther 2016; 9:4959-68. [PMID: 27570459 PMCID: PMC4986681 DOI: 10.2147/ott.s105225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Decoy receptor 3 (DcR3) has been reported to be involved in different cancers. However, few related researches have been accomplished on the role of DcR3 in lung cancer. OBJECTIVE To explore the expression level and clinicopathological implication of DcR3 protein in lung cancer tissues. MATERIALS AND METHODS Immunohistochemistry was used to examine DcR3 protein expression in lung cancer (n=365) and normal lung tissues (n=26). The relationships between DcR3 expression and clinical parameters were further investigated. Furthermore, the diagnostic and clinicopathological value of DcR3 mRNA was analyzed based on The Cancer Genome Atlas database in lung cancer patients. RESULTS Compared to normal lung tissues, DcR3 expression was significantly higher in lung cancer (P=0.007) tissues, including small-cell lung cancer (P=0.001) and non-small-cell lung cancer (P=0.008). In addition, DcR3 expression was related to tumor-node-metastasis (TNM) stage (P<0.001), tumor diameter (P=0.007), distant metastasis (P<0.001), and lymph node metastasis (P<0.001) in lung cancers. When concerning non-small-cell lung cancer, consistent correlations between DcR3 expression and TNM stage (P<0.001), tumor diameter (P=0.019), distant metastasis (P<0.001), and lymph node metastasis (P<0.001) were found. Simultaneously, in small-cell lung cancer, TNM stage (P=0.004) and lymph node metastasis (P=0.005) were also associated with DcR3 expression. Additionally, receiver operator characteristic curve revealed that the area under curve (AUC) of DcR3 was 0.637 (95% confidence interval [CI] 0.531-0.742) for lung cancer. Furthermore, DcR3 was overexpressed in both adenocarcinoma and squamous cell carcinoma tissues than in noncancerous lung tissues (all P<0.0001) based on the data from The Cancer Genome Atlas. AUC of DcR3 was 0.726 (95% CI 0.644-0.788) for lung adenocarcinoma patients and 0.647 (95% CI 0.566-0.728) for squamous cell carcinoma patients. DcR3 expression was also related to the overall survival (P<0.001) and disease-free survival (P<0.001) of lung adenocarcinoma according to the data from The Cancer Genome Atlas. CONCLUSION Our study confirms that DcR3 might be involved in the tumorigenesis and deterioration of lung cancer. Therefore, the detection of DcR3 gains the potential to be applied in the clinic for screening and progression prediction of lung cancer.
Collapse
Affiliation(s)
| | - Jie Luo
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University
| | - Rongquan He
- Center for Genomic and Personalized Medicine, Guangxi Medical University
| | | | | | | | | | | | - Shikang Li
- Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
21
|
Im J, Kim K, Hergert P, Nho RS. Idiopathic pulmonary fibrosis fibroblasts become resistant to Fas ligand-dependent apoptosis via the alteration of decoy receptor 3. J Pathol 2016; 240:25-37. [PMID: 27218286 DOI: 10.1002/path.4749] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 11/06/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible lethal lung disease with an unknown etiology. IPF patients' lung fibroblasts express inappropriately high Akt activity, protecting them in response to an apoptosis-inducing type I collagen matrix. FasL, a ligand for Fas, is known to be increased in the lung tissues of patients with IPF, implicated with the progression of IPF. Expression of Decoy Receptor3 (DcR3), which binds to FasL, thereby subsequently suppressing the FasL-Fas-dependent apoptotic pathway, is frequently altered in various human disease. However, the role of DcR3 in IPF fibroblasts in regulating their viability has not been examined. We found that enhanced DcR3 expression exists in the majority of IPF fibroblasts on collagen matrices, resulting in the protection of IPF fibroblasts from FasL-induced apoptosis. Abnormally high Akt activity suppresses GSK-3β function, thereby accumulating the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) in the nucleus, increasing DcR3 expression in IPF fibroblasts. This alteration protects IPF cells from FasL-induced apoptosis on collagen. However, the inhibition of Akt or NFATc1 decreases DcR3 mRNA and protein levels, which sensitizes IPF fibroblasts to FasL-mediated apoptosis. Furthermore, enhanced DcR3 and NFATc1 expression is mainly present in myofibroblasts in the fibroblastic foci of lung tissues derived from IPF patients. Our results showed that when IPF cells interact with collagen matrix, aberrantly activated Akt increases DcR3 expression via GSK-3β-NFATc1 and protects IPF cells from the FasL-dependent apoptotic pathway. These findings suggest that the inhibition of DcR3 function may be an effective approach for sensitizing IPF fibroblasts in response to FasL, limiting the progression of lung fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kyutae Kim
- College of Biological Science, University of Minnesota, St. Paul, MN, USA
| | - Polla Hergert
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.,Lung Morphology Research Core Department, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
22
|
Jia HY, Liu JL, Yuan MZ, Zhou CJ, Sun WD, Zhao JJ, Wang J, Liu L, Luan Y. Regulation Roles of MICA and NKG2D in Human Renal Cancer Cells. Asian Pac J Cancer Prev 2016; 16:3901-5. [PMID: 25987057 DOI: 10.7314/apjcp.2015.16.9.3901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Our aim was to investigation the roles of MHC class I chain-related gene A(MICA) and natural killer cell group 2D(NKG2D) in human renal cancer cells. MATERIALS AND METHODS The expression of membrane MICA (mMICA) on renal cells and NKG2D on NK cells were detected by flow cytometry (FCM); the content of sMICA were detected by enzyme linked immunosorbent assay (ELISA) and the distribution of mMICA on renal tumor tissues by immunohistochemistry; the interaction between MICA and NKG2D was observed by antibody closed method. RESULTS Our results showed that the expression of mMICA in renal cancer tissues was significantly higher than in controls, where the soluble MICA was not expressed. Cytotoxic activity of NK cells was significantly reduced after exposure to NKG2D and MICA antibodies (P<0.05), and serum containing sMICA can obviously lower the function of NKG2D (P<0.05). CONCLUSIONS The interaction of mMICA and NKG2D play important roles in mediation of cytotoxicity of NK cells in RCC. On the other hand, sMICA may mediate tumor immune escape through down- regulated NKG2D expression.
Collapse
Affiliation(s)
- Hong-Ying Jia
- Clinical Molecular Biology Laboratory, the Second Hospital of Shandong University, Jinan, Shandong Province, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
NFATC1 promotes cell growth and tumorigenesis in ovarian cancer up-regulating c-Myc through ERK1/2/p38 MAPK signal pathway. Tumour Biol 2015; 37:4493-500. [DOI: 10.1007/s13277-015-4245-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 10/12/2015] [Indexed: 12/11/2022] Open
|
24
|
Macher-Goeppinger S, Keith M, Tagscherer KE, Singer S, Winkler J, Hofmann TG, Pahernik S, Duensing S, Hohenfellner M, Kopitz J, Schirmacher P, Roth W. PBRM1 (BAF180) protein is functionally regulated by p53-induced protein degradation in renal cell carcinomas. J Pathol 2015; 237:460-71. [PMID: 26178300 DOI: 10.1002/path.4592] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/23/2015] [Accepted: 07/13/2015] [Indexed: 01/28/2023]
Abstract
About 40% of clear-cell renal cell carcinomas (ccRCC) harbour mutations in Polybromo-1 (PBRM1), encoding the BAF180 subunit of a SWI/SNF chromatin remodelling complex. This qualifies PBRM1 as a major cancer gene in ccRCC. The PBRM1 protein alters chromatin structure and its known functions include transcriptional regulation by controlling the accessibility of DNA and influencing p53 transcriptional activity. Since little is known about the regulation of PBRM1, we studied possible mechanisms and interaction partners involved in the regulation of PBRM1 expression. Activation of p53 in RCC cells resulted in a marked decrease of PBRM1 protein levels. This effect was abolished by siRNA-mediated down-regulation of p53, and transcriptional activity was not crucial for p53-dependent PBRM1 regulation. Pulse-chase experiments determined post-translational protein degradation to be the underlying mechanism for p53-dependent PBRM1 regulation, which was accordingly inhibited by proteasome inhibitors. The effects of p53 activation on PBRM1 expression were confirmed in RCC tissue ex vivo. Our results demonstrate that PBRM1 is a target of p53-induced proteasomal protein degradation and provide further evidence for the influence of PBRM1 on p53 function in RCC tumour cells. Considering the paramount role of p53 in carcinogenesis and the presumptive impact of PBRM1 in RCC development, this novel regulation mechanism might be therapeutically exploited in the future.
Collapse
Affiliation(s)
- Stephan Macher-Goeppinger
- Institute of Pathology, University Hospital Heidelberg, Germany.,Molecular Tumour Pathology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Martina Keith
- Institute of Pathology, University Hospital Heidelberg, Germany.,Molecular Tumour Pathology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Katrin E Tagscherer
- Institute of Pathology, University Hospital Heidelberg, Germany.,Molecular Tumour Pathology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Stephan Singer
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Juliane Winkler
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Thomas G Hofmann
- Cellular Senescence, German Cancer Research Centre (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sascha Pahernik
- Department of Urology, University Hospital Heidelberg, Germany
| | - Stefan Duensing
- Molecular Uro-oncology, Department of Urology, University Hospital Heidelberg, Germany
| | | | - Juergen Kopitz
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Germany
| | | | - Wilfried Roth
- Institute of Pathology, University Hospital Heidelberg, Germany.,Molecular Tumour Pathology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Hickman JA, Graeser R, de Hoogt R, Vidic S, Brito C, Gutekunst M, van der Kuip H. Three-dimensional models of cancer for pharmacology and cancer cell biology: capturing tumor complexity in vitro/ex vivo. Biotechnol J 2015; 9:1115-28. [PMID: 25174503 DOI: 10.1002/biot.201300492] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/11/2014] [Accepted: 08/05/2014] [Indexed: 12/12/2022]
Abstract
Cancers are complex and heterogeneous pathological "organs" in a dynamic interplay with their host. Models of human cancer in vitro, used in cancer biology and drug discovery, are generally highly reductionist. These cancer models do not incorporate complexity or heterogeneity. This raises the question as to whether the cancer models' biochemical circuitry (not their genome) represents, with sufficient fidelity, a tumor in situ. Around 95% of new anticancer drugs eventually fail in clinical trial, despite robust indications of activity in existing in vitro pre-clinical models. Innovative models are required that better capture tumor biology. An important feature of all tissues, and tumors, is that cells grow in three dimensions. Advances in generating and characterizing simple and complex (with added stromal components) three-dimensional in vitro models (3D models) are reviewed in this article. The application of stirred bioreactors to permit both scale-up/scale-down of these cancer models and, importantly, methods to permit controlled changes in environment (pH, nutrients, and oxygen) are also described. The challenges of generating thin tumor slices, their utility, and potential advantages and disadvantages are discussed. These in vitro/ex vivo models represent a distinct move to capture the realities of tumor biology in situ, but significant characterization work still remains to be done in order to show that their biochemical circuitry accurately reflects that of a tumor.
Collapse
|
26
|
Levin-Gromiko U, Koshelev V, Kushnir P, Fedida-Metula S, Voronov E, Fishman D. Amplified lipid rafts of malignant cells constitute a target for inhibition of aberrantly active NFAT and melanoma tumor growth by the aminobisphosphonate zoledronic acid. Carcinogenesis 2014; 35:2555-66. [DOI: 10.1093/carcin/bgu178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
27
|
Jia HY, Liu JL, Zhou CJ, Kong F, Yuan MZ, Sun WD, Wang J, Liu L, Zhao JJ, Luan Y. High Expression of MICA in Human Kidney Cancer Tissue and Renal Cell Carcinoma Lines. Asian Pac J Cancer Prev 2014; 15:1715-7. [DOI: 10.7314/apjcp.2014.15.4.1715] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|