1
|
Byrling J, Kristl T, Hu D, Pla I, Sanchez A, Sasor A, Andersson R, Marko-Varga G, Andersson B. Mass spectrometry-based analysis of formalin-fixed, paraffin-embedded distal cholangiocarcinoma identifies stromal thrombospondin-2 as a potential prognostic marker. J Transl Med 2020; 18:343. [PMID: 32887625 PMCID: PMC7487897 DOI: 10.1186/s12967-020-02498-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Distal cholangiocarcinoma is an aggressive malignancy with a dismal prognosis. Diagnostic and prognostic biomarkers for distal cholangiocarcinoma are lacking. The aim of the present study was to identify differentially expressed proteins between distal cholangiocarcinoma and normal bile duct samples. METHODS A workflow utilizing discovery mass spectrometry and verification by parallel reaction monitoring was used to analyze surgically resected formalin-fixed, paraffin-embedded samples from distal cholangiocarcinoma patients and normal bile duct samples. Bioinformatic analysis was used for functional annotation and pathway analysis. Immunohistochemistry was performed to validate the expression of thrombospondin-2 and investigate its association with survival. RESULTS In the discovery study, a total of 3057 proteins were identified. Eighty-seven proteins were found to be differentially expressed (q < 0.05 and fold change ≥ 2 or ≤ 0.5); 31 proteins were upregulated and 56 were downregulated in the distal cholangiocarcinoma samples compared to controls. Bioinformatic analysis revealed an abundance of differentially expressed proteins associated with the tumor reactive stroma. Parallel reaction monitoring verified 28 proteins as upregulated and 18 as downregulated in distal cholangiocarcinoma samples compared to controls. Immunohistochemical validation revealed thrombospondin-2 to be upregulated in distal cholangiocarcinoma epithelial and stromal compartments. In paired lymph node metastases samples, thrombospondin-2 expression was significantly lower; however, stromal thrombospondin-2 expression was still frequent (72%). Stromal thrombospondin-2 was an independent predictor of poor disease-free survival (HR 3.95, 95% CI 1.09-14.3; P = 0.037). CONCLUSION Several proteins without prior association with distal cholangiocarcinoma biology were identified and verified as differentially expressed between distal cholangiocarcinoma and normal bile duct samples. These proteins can be further evaluated to elucidate their biomarker potential and role in distal cholangiocarcinoma carcinogenesis. Stromal thrombospondin-2 is a potential prognostic marker in distal cholangiocarcinoma.
Collapse
Affiliation(s)
- Johannes Byrling
- Department of Clinical Sciences Lund, Surgery, Lund University, and Skåne University Hospital, Lund, Sweden
| | - Theresa Kristl
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Dingyuan Hu
- Department of Clinical Sciences Lund, Surgery, Lund University, and Skåne University Hospital, Lund, Sweden
| | - Indira Pla
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Aniel Sanchez
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Agata Sasor
- Department of Clinical Sciences Lund, Pathology, Lund University, and Skåne University Hospital, Lund, Sweden
| | - Roland Andersson
- Department of Clinical Sciences Lund, Surgery, Lund University, and Skåne University Hospital, Lund, Sweden
| | - György Marko-Varga
- Department of Biomedical Engineering, Clinical Protein Science and Imaging, Lund University, Lund, Sweden
| | - Bodil Andersson
- Department of Clinical Sciences Lund, Surgery, Lund University, and Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
2
|
Xiao D, Lu Y, Zhu L, Liang T, Wang Z, Ren J, He R, Wang K. Anti-osteosarcoma property of decorin-modified titanium surface: A novel strategy to inhibit oncogenic potential of osteosarcoma cells. Biomed Pharmacother 2020; 125:110034. [PMID: 32187963 DOI: 10.1016/j.biopha.2020.110034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 01/03/2023] Open
Abstract
Osteosarcoma is the most common bone sarcoma in adolescents. Decorin (DCN) has been proposed to be a new anti-osteosarcoma therapeutic strategy. Our previous study has loaded decorin on titanium (Ti) surface by polydopamine (DOPA) as an anchor to enhance osseointegration. In this study, we investigated the effect of decorin-coated Ti substrates (TI-DOPA-DCN) on the oncogenic potential of osteosarcoma cells SAOS-2. The substrates were placed in 24-well plates for cell culture. Cell viability was determined by Cell Counting Kit-8 (CCK8) assay. Apoptosis was evaluated by DAPI staining and Annexin V-FITC/PI double staining analysis. Cell cycle was analyzed by flow cytometry. Cell migration and invasion were evaluated by Transwell assay. For co-culture, the pre-osteogenic cells MEC3T3-E1 and osteosarcoma cells SAOS-2 were stained with cell membrane fluorescent dyes, and then mixed (1:1) for co-culture. The cells were observed under a fluorescence microscope at four time points of 24, 48, 72, and 96 h. The results showed that TI-DOPA-DCN substrate can selectively inhibit cell proliferation of osteosarcoma cells but not pre-osteoblasts. However, the cell cycle of SAOS-2 was not affected by TI-DOPA-DCN substrates. Both DAPI staining and Annexin V-FITC/PI double staining analysis revealed that TI-DOPA-DCN substrates induced apoptosis of osteosarcoma cells. Transwell assay showed that TI-DOPA-DCN substrates inhibited invasion and migration of osteosarcoma cells. Moreover, TI-DOPA-DCN substrates inhibited the growth of osteosarcoma cells but promoted that of pre-osteoblasts in the coculture system. Taken together, these findings suggested that decorin coating on Ti surface simultaneously inhibited the oncogenic potential of osteosarcoma cells but enhanced cell growth of pre-osteoblasts, which could be applied to surface modification of Ti orthopedic implant.
Collapse
Affiliation(s)
- Dahai Xiao
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunxiang Lu
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Zhu
- Department of Plastic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tangzhao Liang
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Wang
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianhua Ren
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ronghan He
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Kun Wang
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Yi Q, Liu Y, Cao M, Liu J, Xiang Q, Tan G, Zhang H, Lai G, Xie Y. Transcriptional analysis and differentially expressed gene screening of spontaneous liver tumors in CBA/CaJ mice. Gene 2020; 725:144159. [PMID: 31629818 DOI: 10.1016/j.gene.2019.144159] [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: 06/17/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide due to its frequent metastasis, tumor recurrence, and lack of curative treatment. However, the underlying molecular mechanisms involved in HCC progression remain unclear. Here, we analyzed the global gene expression of spontaneous liver tumor tissue from CBA/CaJ mice by RNA-Seq and identified 10,706 and 10,374 genes in the normal and liver tumor groups, respectively. Only 9793 genes were expressed in both, 913 genes were identified in only the liver tumor group, and 581 genes were found in normal liver tissues. There were 2054 differentially expressed genes (DEGs), with 975 down-regulated genes and 1079 up-regulated genes. Gene ontology (GO) term enrichment analysis showed that 43 up-regulated genes were significantly associated with cell cycle regulation and hundreds of up-regulated genes were related to cell migration, adhesion, or metabolic processes. KEGG pathway enrichment also demonstrated that some DEGs were tightly associated with the cell cycle, extracellular matrix (ECM)-receptor interactions, as well as protein digestion and absorption pathways, indicating that the activation of these oncogenic cascades was closely related to tumor liver progression in CBA/CaJ mice. Ninety-three genes with elevated expression levels preferentially localized in microtubules, kinetochores, and spindles play an important role during mitosis and meiosis and are associated with the reorganization of the cytoskeleton in cancer cells during migration and invasion. Some ECM-related genes were significantly different in the tumor group, including collagen types I, III, IV, V, and VI, non-collagenous glycoproteins, laminin, and fibronectin. We further validated the functions of upregulated genes, such as cyclin-dependent kinase 1 (CDK1) and polo-like kinase 1 (PLK1), with regards to cell cycle regulation, apoptosis, and proliferation in normal human liver or liver tumor-derived cell lines. Our results indicated that the cell cycle dysregulation, ECM-receptor interaction, and cytoskeleton-associated genes in mouse livers may promote HCC progression and deciphering the function of the genes will help investigators understand the underlying molecular mechanism of HCC.
Collapse
Affiliation(s)
- Qiying Yi
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China
| | - Yang Liu
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China
| | - Min Cao
- The M.O.E. Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing 400014, China
| | - Jianing Liu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qin Xiang
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China
| | - Guo Tan
- Department of Foreign Language, Minzu University of China, Beijing 100081, China
| | - Huatang Zhang
- Chongqing Academy of Science and Technology, Chongqing 400016, China
| | - Guoqi Lai
- Laboratory Animal Center, Chongqing Medical University, Chongqing 400016, China.
| | - Yajun Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
4
|
Deng M, Xue Y, Xu L, Wang Q, Wei J, Ke X, Wang J, Chen X. Chrysophanol exhibits inhibitory activities against colorectal cancer by targeting decorin. Cell Biochem Funct 2019; 38:47-57. [PMID: 31710116 DOI: 10.1002/cbf.3445] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/10/2019] [Accepted: 09/23/2019] [Indexed: 12/30/2022]
Abstract
Colorectal cancer (CRC) is a common human malignancy that accounts for 600,000 deaths annually worldwide. Chrysophanol, a naturally occurring anthraquinone compound, exhibits anti-neoplastic effects in various cancer cells. The aim of this study was to explore the biological effects of chrysophanol on CRC cells, and determine the underlying mechanism. Chrysophanol inhibited proliferation of and promoted apoptosis in CRC cells by activating the intrinsic mitochondrial apoptotic pathway. In addition, chrysophanol also suppressed tumor growth in vivo and increased the percentage of apoptotic cells in tumor xenografts, without general toxicity. Proteomic iTRAQ analysis revealed decorin (DCN) as the major target of chrysophanol. DCN was upregulated in the tumor tissues following chrysophanol treatment, and ectopic DCN expression markedly augmented the pro-apoptotic effects of chrysophanol in CRC cells. In contrast, DCN knockdown significantly abrogated chrysophanol-induced apoptosis in CRC cells. Taken together, chrysophanol exerts anti-neoplastic effects in vitro and in vivo in CRC cells by modulating DCN, there by highlighting its therapeutic potential in CRC.
Collapse
Affiliation(s)
- Min Deng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Yongju Xue
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Lerong Xu
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Qiangwu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Jun Wei
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Xiquan Ke
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Jianchao Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| | - Xiaodong Chen
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, P.R. China
| |
Collapse
|
5
|
Wang G, Liu G, Ye Y, Fu Y, Zhang X. Bufothionine exerts anti-cancer activities in gastric cancer through Pim3. Life Sci 2019; 232:116615. [PMID: 31260686 DOI: 10.1016/j.lfs.2019.116615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/19/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
AIM Gastric cancer (GC) is the fourth most common cancer globally. Bufothionine is a major active constituent of Cinobufacini (Huachansu), which is extracted from the skin and parotid venom gland of the toad Bufo bufo gargarizans Cantor. It exhibits anti-cancer activities in vitro. However, whether bufothionine exerts anti-cancer activities against GC is unknown. This study was designed to evaluate the efficacy of bufothionine in vitro and in vivo. MATERIAL AND METHODS MKN28 and AGS cells were chosen as cell models to study the anti-cancer effect of bufothionine. Cell viability was determined by CCK-8 assay, while the effect of bufothionine on cell membrane integrity was examined by LDH assay. Cell apoptosis was detected by Hoechst/PI staining and Annexin V-FITC/PI staining followed by flow cytometry analysis. The expression levels of proteins involved were examined using western blotting. I-Traq analysis was conducted to identify the differentially expressed genes in AGS cells following bufothionine treatment. The anti-growth effect of bufothionine was validated in vivo using a GC xenograft model. KEY FINDINGS The results revealed that bufothionine prevented the growth, destroyed cell membrane and promoted apoptotic cell death of GC cells. iTRAQ analysis revealed thatPIM3 might be a molecular target responsible for the anti-cancer effects of bufothionine. It was also found that PIM3 knockdown significantly augmented the anti-growth and pro-apoptotic effects of bufothionine in GC cells. In contrast, ectopic PIM3 expression markedly dampened the anti-neoplastic activities of bufothionine. The expression of PIM3 was also suppressed by bufothionine treatment in xenograft tumor tissue. SIGNIFICANCE Bufothionine exhibited anti-cancer activities in vitro and in vivo in GC via downregulating PIM3.
Collapse
Affiliation(s)
- Guojun Wang
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Guanghui Liu
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanwei Ye
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yang Fu
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiefu Zhang
- The Department of Gastrointestinal surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
6
|
Sainio AO, Järveläinen HT. Decorin-mediated oncosuppression - a potential future adjuvant therapy for human epithelial cancers. Br J Pharmacol 2018; 176:5-15. [PMID: 29488209 PMCID: PMC6284329 DOI: 10.1111/bph.14180] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/24/2022] Open
Abstract
Currently, the multifaceted role of the extracellular matrix (ECM) in tumourigenesis has been realized. One ECM macromolecule exhibiting potent oncosuppressive actions in tumourigenesis is decorin, the prototype of the small leucine-rich proteoglycan gene family. The actions of decorin include its ability to function as an endogenous pan-receptor tyrosine kinase inhibitor, a regulator of both autophagy and mitophagy, as well as a modulator of the immune system. In this review, we will discuss these topics in more detail. We also provide a summary of preclinical studies exploring the value of decorin-mediated oncosuppression, as a potential future adjuvant therapy for epithelial cancers. LINKED ARTICLES: This article is part of a themed section on Translating the Matrix. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.1/issuetoc.
Collapse
Affiliation(s)
- Annele Orvokki Sainio
- Institute of Biomedicine, Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Hannu Tapio Järveläinen
- Institute of Biomedicine, Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.,Department of Internal Medicine, Satakunta Central Hospital, Sairaalantie 3, 28500, Pori, Finland
| |
Collapse
|
7
|
Yang Y, Xu W, Neill T, Hu Z, Wang CH, Xiao X, Stock SR, Guise T, Yun CO, Brendler CB, Iozzo RV, Seth P. Systemic Delivery of an Oncolytic Adenovirus Expressing Decorin for the Treatment of Breast Cancer Bone Metastases. Hum Gene Ther 2015; 26:813-25. [PMID: 26467629 DOI: 10.1089/hum.2015.098] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The development of novel therapies for breast cancer bone metastasis is a major unmet medical need. Toward that end, we have constructed an oncolytic adenovirus, Ad.dcn, and a nonreplicating adenovirus, Ad(E1-).dcn, both containing the human decorin gene. Our in vitro studies showed that Ad.dcn produced high levels of viral replication and the decorin protein in the breast tumor cells. Ad(E1-).dcn-mediated decorin expression in MDA-MB-231 cells downregulated the expression of Met, β-catenin, and vascular endothelial growth factor A, all of which are recognized decorin targets and play pivotal roles in the progression of breast tumor growth and metastasis. Adenoviral-mediated decorin expression inhibited cell migration and induced mitochondrial autophagy in MDA-MB-231 cells. Mice bearing MDA-MB-231-luc skeletal metastases were systemically administered with the viral vectors, and skeletal tumor growth was monitored over time. The results of bioluminescence imaging and X-ray radiography indicated that Ad.dcn and Ad(E1-).dcn significantly inhibited the progression of bone metastases. At the terminal time point, histomorphometric analysis, micro-computed tomography, and bone destruction biomarkers showed that Ad.dcn and Ad(E1-).dcn reduced tumor burden and inhibited bone destruction. A nonreplicating adenovirus Ad(E1-).luc expressing the luciferase 2 gene had no significant effect on inhibiting bone metastases, and in several assays, Ad.dcn and Ad(E1-).dcn were better than Ad.luc, a replicating virus expressing the luciferase 2 gene. Our data suggest that adenoviral replication coupled with decorin expression could produce effective antitumor responses in a MDA-MB-231 bone metastasis model of breast cancer. Thus, Ad.dcn could potentially be developed as a candidate gene therapy vector for treating breast cancer bone metastases.
Collapse
Affiliation(s)
- Yuefeng Yang
- 1 Gene Therapy Program, Department of Medicine, NorthShore Research Institute, An Affiliate of the University of Chicago , Evanston, Illinois
| | - Weidong Xu
- 1 Gene Therapy Program, Department of Medicine, NorthShore Research Institute, An Affiliate of the University of Chicago , Evanston, Illinois
| | - Thomas Neill
- 2 Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Zebin Hu
- 3 1st Division of In Vitro Diagnostic Reagents, National Institutes for Food and Drug Control , Beijing, China
| | - Chi-Hsiung Wang
- 4 Department of Surgery, NorthShore Research Institute , Evanston, Illinois
| | - Xianghui Xiao
- 5 Advanced Photon Source, Argonne National Lab. , Argonne, Illinois
| | - Stuart R Stock
- 6 Department of Cell and Molecular Biology, Northwestern University , Chicago, Illinois
| | - Theresa Guise
- 7 Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Chae-Ok Yun
- 8 Department of Bioengineering, Hanyang University , Seoul, Korea
| | - Charles B Brendler
- 4 Department of Surgery, NorthShore Research Institute , Evanston, Illinois
| | - Renato V Iozzo
- 2 Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Prem Seth
- 1 Gene Therapy Program, Department of Medicine, NorthShore Research Institute, An Affiliate of the University of Chicago , Evanston, Illinois
| |
Collapse
|
8
|
Nyman MC, Sainio AO, Pennanen MM, Lund RJ, Vuorikoski S, Sundström JTT, Järveläinen HT. Decorin in Human Colon Cancer: Localization In Vivo and Effect on Cancer Cell Behavior In Vitro. J Histochem Cytochem 2015; 63:710-20. [PMID: 26001829 DOI: 10.1369/0022155415590830] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/17/2015] [Indexed: 12/15/2022] Open
Abstract
Decorin is generally recognized as a tumor suppressing molecule. Nevertheless, although decorin has been shown to be differentially expressed in malignant tissues, it has often remained unclear whether, in addition to non-malignant stromal cells, cancer cells also express it. Here, we first used two publicly available databases to analyze the current information about decorin expression and immunoreactivity in normal and malignant human colorectal tissue samples. The analyses demonstrated that decorin expression and immunoreactivity may vary in cancer cells of human colorectal tissues. Therefore, we next examined decorin expression in normal, premalignant and malignant human colorectal tissues in more detail using both in situ hybridization and immunohistochemistry for decorin. Our results invariably demonstrate that malignant cells within human colorectal cancer tissues are devoid of both decorin mRNA and immunoreactivity. Identical results were obtained for cells of neuroendocrine tumors of human colon. Using RT-qPCR, we showed that human colon cancer cell lines are also decorin negative, in accordance with the above in vivo results. Finally, we demonstrate that decorin transduction of human colon cancer cell lines causes a significant reduction in their colony forming capability. Thus, strategies to develop decorin-based adjuvant therapies for human colorectal malignancies are highly rational.
Collapse
Affiliation(s)
- Marie C Nyman
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland (MCN, AOS, MMP, HTJ)
| | - Annele O Sainio
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland (MCN, AOS, MMP, HTJ)
| | - Mirka M Pennanen
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland (MCN, AOS, MMP, HTJ)
| | - Riikka J Lund
- Turku Centre for Biotechnology , University of Turku, Turku, Finland(RJL, SV)
| | - Sanna Vuorikoski
- Turku Centre for Biotechnology , University of Turku, Turku, Finland(RJL, SV)
| | | | - Hannu T Järveläinen
- Division of Medicine, Department of Endocrinology, Turku University Hospital, Turku, Finland (HTJ)
| |
Collapse
|
9
|
Järveläinen H, Sainio A, Wight TN. Pivotal role for decorin in angiogenesis. Matrix Biol 2015; 43:15-26. [PMID: 25661523 DOI: 10.1016/j.matbio.2015.01.023] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 01/29/2015] [Accepted: 01/29/2015] [Indexed: 01/05/2023]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting vessels, is a highly complex process. It is regulated in a finely-tuned manner by numerous molecules including not only soluble growth factors such as vascular endothelial growth factor and several other growth factors, but also a diverse set of insoluble molecules, particularly collagenous and non-collagenous matrix constituents. In this review we have focused on the role and potential mechanisms of a multifunctional small leucine-rich proteoglycan decorin in angiogenesis. Depending on the cellular and molecular microenvironment where angiogenesis occurs, decorin can exhibit either a proangiogenic or an antiangiogenic activity. Nevertheless, in tumorigenesis-associated angiogenesis and in various inflammatory processes, particularly foreign body reactions and scarring, decorin exhibits an antiangiogenic activity, thus providing a potential basis for the development of decorin-based therapies in these pathological situations.
Collapse
Affiliation(s)
- Hannu Järveläinen
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland; Department of Medicine, Division of Endocrinology, Turku University Hospital, Turku, Finland.
| | - Annele Sainio
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
10
|
Shi X, Liang W, Yang W, Xia R, Song Y. Decorin is responsible for progression of non-small-cell lung cancer by promoting cell proliferation and metastasis. Tumour Biol 2014; 36:3345-54. [PMID: 25524578 DOI: 10.1007/s13277-014-2968-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/09/2014] [Indexed: 12/30/2022] Open
Abstract
Decorin, a member of the small leucine-rich proteoglycans family, exists and plays multifunctional roles in stromal and epithelial cells. Emerging evidences showed that decorin is dysregulated expression in a wide variety of human tumors and affects a broad biology process of cancer cells, including growth, metastasis, and angiogenesis. Recent studies demonstrated that decorin could affect A549 proliferation though decreasing TGF-β1, cycling D1 expression and increasing P53 and P21 expression. However, limited data are available on the effect of decorin on metastasis of non-small-cell lung cancer (NSCLC) cell lines and how decorin impacts metastasis is still unknown. In this study, we identified decorin mRNA expression through Oncomine database and verified the expression of decorin mRNA and protein in 50 patients who underwent primary surgical resection of a NSCLC in the Department of Thoracic Surgery, Jinling Hospital, Nanjing University School of Medicine, China, between September 2013 and March 2014 by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot. Also, the correlationship between decorin and the NSCLC patients' clinical characteristics or survival ( www.kmplot.com ) was analyzed. Via ectopic expression analyses and Western blot, the roles of decorin in proliferation, metastasis, and the underline mechanism for decorin expression were further explored. We found that decorin was downregulated in NSCLC tissues compared with the adjacent normal lung tissues or normal tissues. Additionally, the expression of decorin was correlated with tumor size, lymph node metastasis, tumor stage, and prognosis. We also showed that overexpression of decorin could inhibit NSCLC cell lines proliferation and metastasis. Through Western blot analysis, we identified that E-cadherin and vascular endothelial growth factor (VEGF) are two key factors responsible for the growth arrest and metastasis inhibition induced by decorin in NSCLC. Our results indicated that decorin plays crucial roles in NSCLC against carcinogenesis and progression. Decorin might be a predictive factor and an attractive therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Xuefei Shi
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China,
| | | | | | | | | |
Collapse
|