1
|
Jiang H. Prostate Cancer Bone Metastasis: Molecular Mechanisms of Tumor and Bone Microenvironment. Cancer Manag Res 2025; 17:219-237. [PMID: 39912095 PMCID: PMC11796448 DOI: 10.2147/cmar.s495169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Prostate cancer is prevalent among men aged 65 and older. Bone metastasis occurs in up to 90% of advanced prostate cancer patients, metastatic prostate cancer is generally considered a non-curative condition which can impact quality of life. The tumor microenvironment, comprising diverse cellular and non-cellular elements, interacts with prostate cancer cells to affect tumor growth and bone metastasis. Within the bone microenvironment, different cell types, including osteoblasts, osteoclasts, adipocytes, endothelial cells, hematopoietic stem cells, and immune cells, engage with tumor cells. Some cells alter tumor behavior, while others are impacted or overpowered by tumor cells, leading to different phases of tumor cell movement, dormancy, latency, resistance to treatment, and advancement to visible bone metastasis. This review summarizes recent research on the tumor microenvironment and bone microenvironment in prostate cancer bone metastasis, exploring underlying mechanisms and the potential value of targeting these environments for treatment.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Urology, Fifth Affiliated Hospital of Zunyi Medical University (Zhuhai Sixth People’s Hospital), Zhuhai, People’s Republic of China
| |
Collapse
|
2
|
Samaržija I. The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery. Biomedicines 2023; 12:79. [PMID: 38255186 PMCID: PMC10813710 DOI: 10.3390/biomedicines12010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is among the top five cancer types according to incidence and mortality. One of the main obstacles in prostate cancer management is the inability to foresee its course, which ranges from slow growth throughout years that requires minimum or no intervention to highly aggressive disease that spreads quickly and resists treatment. Therefore, it is not surprising that numerous studies have attempted to find biomarkers of prostate cancer occurrence, risk stratification, therapy response, and patient outcome. However, only a few prostate cancer biomarkers are used in clinics, which shows how difficult it is to find a novel biomarker. Cell adhesion to the extracellular matrix (ECM) through integrins is among the essential processes that govern its fate. Upon activation and ligation, integrins form multi-protein intracellular structures called integrin adhesion complexes (IACs). In this review article, the focus is put on the biomarker potential of the ECM- and IAC-related molecules stemming from both body fluids and prostate cancer tissue. The processes that they are involved in, such as tumor stiffening, bone turnover, and communication via exosomes, and their biomarker potential are also reviewed.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
3
|
Ito K, Kitajima Y, Kai K, Matsufuji S, Yamada K, Egawa N, Kitagawa H, Okuyama K, Tanaka T, Noshiro H. Matrix metalloproteinase‑1 expression is regulated by HIF‑1‑dependent and epigenetic mechanisms and serves a tumor‑suppressive role in gastric cancer progression. Int J Oncol 2021; 59:102. [PMID: 34738626 PMCID: PMC8577796 DOI: 10.3892/ijo.2021.5282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/11/2021] [Indexed: 02/05/2023] Open
Abstract
The matrix metalloproteinase (MMP) family is associated with degradation of the extracellular matrix and is known to promote cancer invasion. The present study aimed to investigate the biological role of MMP‑1 in gastric cancer cells and analyze the association between MMP‑1 expression and the clinical outcomes of gastric cancer patients. In the present study, hypoxia accelerated invasion, accompanied by elevated MMP‑1 expression in the gastric cancer cell line 58As9. Additionally, hypoxia‑inducible factor‑1α (HIF‑1α) knockdown in 58As9 cells reduced MMP‑1 expression under hypoxic conditions. Treatment with 5‑aza‑2‑deoxycytidine and trichostatin A restored MMP‑1 expression in the MMP‑1‑deficient cell lines MKN45 and MKN74. These results indicated that MMP‑1 expression was controlled by both HIF‑1α‑dependent and epigenetic mechanisms in gastric cancer cell lines. In addition, MMP‑1 knockdown impaired the hypoxia‑induced invasiveness of 58As9 cells, implicating MMP‑1 in the elevated invasion. By contrast, knockdown enhanced the proliferative ability of 58As9 cells, whereby expression of cell cycle‑related genes was subsequently altered. In nude mouse models, the knockdown accelerated the growth of xenograft tumor and the development of peritoneal dissemination. In an immunohistochemical study using 161 surgically resected cancer tissues, the Ki67 score was significantly higher in the group with low MMP‑1 expression (P<0.001). Disease‑free survival (DFS) and disease‑specific survival (DSS) were both significantly reduced in patients with low MMP‑1 expression (log‑rank test; DFS: P=0.005; DSS: P=0.022). Multivariate analysis demonstrated that MMP‑1 expression was an independent prognostic factor for DFS and DSS [DFS: HR=2.11 (1.22‑3.92) P=0.005, DSS: HR=2.90 (1.23‑8.50) P=0.012]. In conclusion, the present study indicated that MMP‑1 may serve as a tumor‑suppressive factor that inhibits gastric cancer progression, although it promoted invasion in vitro.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Humans
- Hypoxia/physiopathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Matrix Metalloproteinase 1/genetics
- Matrix Metalloproteinase 1/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplasm Invasiveness
- Prognosis
- Stomach Neoplasms/genetics
- Stomach Neoplasms/pathology
- Survival Rate
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kotaro Ito
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Yoshihiko Kitajima
- Department of Surgery, National Hospital Organization Higashisaga Hospital, Miyaki, Saga 849-0101, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, Saga 849-8501, Japan
| | - Shohei Matsufuji
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Kohei Yamada
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Noriyuki Egawa
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Hiroshi Kitagawa
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Keiichiro Okuyama
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Tomokazu Tanaka
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | - Hirokazu Noshiro
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| |
Collapse
|
4
|
Tumor-associated macrophages (TAMs) depend on MMP1 for their cancer-promoting role. Cell Death Discov 2021; 7:343. [PMID: 34753916 PMCID: PMC8578434 DOI: 10.1038/s41420-021-00730-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/01/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022] Open
Abstract
The complex interaction between tumor-associated macrophages (TAMs) and tumor cells through several soluble factors and signaling is essential for colorectal cancer (CRC) progression. However, the molecular mechanism involved remains elusive. In this study, we demonstrated that MMP1 derived from TAMs markedly facilitated colon cancer cell proliferation via accelerating cell cycle transition from G0/G1 to S and G2/M phase. Moreover, exogenous MMP1 activated cdc25a/CDK4-cyclin D1 and p21/cdc2-cyclin B1 complexes through altering c-Myc and ETV4. Mechanistic studies indicated that inhibition of PAR1 or blockage of MAPK/Erk signaling eliminated the proliferation induced by exogenous MMP1 in vitro and in vivo. In addition, ETV4 could bind to the promoter of MMP1 and activate MMP1 transcription, which confirmed the MMP1/ETV4/MMP1 positive feedback. Altogether, our study identified a cytokine paracrine manner between colon cancer cells and TAMs. MMP1/PAR1/Erk1/2/ETV4 positive feedback loop may represent to be a therapeutic target and prognostic marker in CRC.
Collapse
|
5
|
Chen Y, Hou W, Zhong M, Wu B. Comprehensive Proteomic Analysis of Colon Cancer Tissue Revealed the Reason for the Worse Prognosis of Right-Sided Colon Cancer and Mucinous Colon Cancer at the Protein Level. Curr Oncol 2021; 28:3554-3572. [PMID: 34590603 PMCID: PMC8482240 DOI: 10.3390/curroncol28050305] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
To clarify the molecular mechanisms underlying the poor prognosis of right-sided and mucinous colon cancer at the proteomic level. A tandem mass tag-proteomics approach was used to identify differentially expressed proteins (DEPs) in colon carcinoma tissues from different locations and with different histological types to reveal the underlying mechanisms of these differences at the protein level. In additional, the DEPs were analyzed using bioinformatics methods. The proteomics profiles among colon cancers with different tumor locations and histological types were dramatically distinguished. In terms of tumor locations, the right-sided carcinoma specific DEPs may promote the tumor progression via activating inflammation, metastasis associated pathways. When referring to histological types, the mucinous colon cancers perhaps increased the invasion and metastasis through distinct mechanisms in different tumor locations. For mucinous cancer located in right-sided colon, the mucinous specific DEPs were mainly associated with ECM-related remodeling and the IL-17 signal pathway. For mucinous cancer located in left-sided colon, the mucinous specific DEPs showed a strong relationship with ACE2/Ang-(1–7)/MasR axis. The proteomics profiles of colon cancers showed distinct differences related to locations and histological types. These results suggested a distinct mechanism underlying the diverse subtypes of colon cancers.
Collapse
Affiliation(s)
- Yanyu Chen
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Wenyun Hou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (W.H.); (M.Z.)
| | - Miner Zhong
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (W.H.); (M.Z.)
| | - Bin Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (W.H.); (M.Z.)
- Correspondence: ; Tel.: +86-010-69156470
| |
Collapse
|
6
|
The Role of the Metzincin Superfamily in Prostate Cancer Progression: A Systematic-Like Review. Int J Mol Sci 2021; 22:ijms22073608. [PMID: 33808504 PMCID: PMC8036576 DOI: 10.3390/ijms22073608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer remains a leading cause of cancer-related morbidity in men. Potentially important regulators of prostate cancer progression are members of the metzincin superfamily of proteases, principally through their regulation of the extracellular matrix. It is therefore timely to review the role of the metzincin superfamily in prostate cancer and its progression to better understand their involvement in this disease. A systematic-like search strategy was conducted. Articles that investigated the roles of members of the metzincin superfamily and their key regulators in prostate cancer were included. The extracted articles were synthesized and data presented in tabular and narrative forms. Two hundred and five studies met the inclusion criteria. Of these, 138 investigated the role of the Matrix Metalloproteinase (MMP) subgroup, 34 the Membrane-Tethered Matrix Metalloproteinase (MT-MMP) subgroup, 22 the A Disintegrin and Metalloproteinase (ADAM) subgroup, 8 the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) subgroup and 53 the Tissue Inhibitor of Metalloproteinases (TIMP) family of regulators, noting that several studies investigated multiple family members. There was clear evidence that specific members of the metzincin superfamily are involved in prostate cancer progression, which can be either in a positive or negative manner. However, further understanding of their mechanisms of action and how they may be used as prognostic indicators or molecular targets is required.
Collapse
|
7
|
Gao G, Yang M, Wang F, Dang G, Zhang X, Zhao J, Wang X, Jin B. Coagulation factor 2 thrombin receptor promotes malignancy in glioma under SOX2 regulation. Aging (Albany NY) 2020; 12:10594—10613. [PMID: 32507767 PMCID: PMC7346046 DOI: 10.18632/aging.103281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/17/2020] [Indexed: 01/22/2023]
Abstract
Glioma is the most common human primary brain cancer with high mortality and unfavorable clinical outcome. Coagulation factor 2 thrombin receptor (F2R), is a key component in the thrombosis process and has been demonstrated upregulated in various cancers. However, the effect and molecular mechanisms of F2R in glioma remains unclear. In our study, we confirmed that the expression of F2R was upregulated in glioma and predicted poor prognosis. Gene Set Enrichment Analysis (GSEA) and function assays demonstrated that F2R overexpression promoted glioma cell proliferation, metastasis and epithelial-mesenchymal transition (EMT) in vitro and tumor growth in vivo. Then, we identified and validated F2R was the target gene of SRY-box 2 (SOX2) by dual luciferase reporter assay and chromatin immunoprecipitation assay. Besides, High expression of F2R in malignant glioma was associated with β-catenint signaling pathway activation. Our findings conclude that F2R promotes glioma cell proliferation and metastasis under SOX2 and actives WNT/β-catenin Signaling pathway, which provides novel insight to the therapeutic regimen in glioma.
Collapse
Affiliation(s)
- Guojun Gao
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Ming Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Fan Wang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Ge Dang
- Department of Operating Theatre, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Xiaoling Zhang
- Department of Operating Theatre, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Jing Zhao
- Department of Operating Theatre, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Xiangyang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| | - Baozhe Jin
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, PR. China
| |
Collapse
|
8
|
Wang K, Zheng J, Yu J, Wu Y, Guo J, Xu Z, Sun X. Knockdown of MMP‑1 inhibits the progression of colorectal cancer by suppressing the PI3K/Akt/c‑myc signaling pathway and EMT. Oncol Rep 2020; 43:1103-1112. [PMID: 32323782 PMCID: PMC7057971 DOI: 10.3892/or.2020.7490] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to investigate the role of matrix metalloproteinase-1 (MMP-1) in the development of colorectal cancer and reveal the mechanism underlying this progression. Bioinformatics methods and a public dataset were first used to analyze MMP-1 gene expression in a public dataset. MMP-1 expression in colorectal cancer patients was assessed by immunohistochemistry; its association with clinicopathological parameters and its significance for prognosis were analyzed. Then proliferation, scratch and Transwell assays and a xenograft model were used to assess the change in malignant behavior in cells transfected with an MMP-1 shRNA. Changes involved in epithelial-mesenchymal transition (EMT) and the Akt signaling pathway were detected by western blotting. According to the results, MMP-1 expression was higher in colorectal cancer tissues than it was in matched adjacent noncancerous tissues, and its high expression was significantly related to lymphatic metastasis as well as TNM stage (P<0.05). Downregulation of MMP-1 expression inhibited the progression of colorectal cancer in vitro and in vivo. Furthermore, after the cells were stably transfected with MMP-1 shRNA, the expression of N-cadherin, vimentin and Twist1 decreased while that of E-cadherin increased. The expression of p-Akt and c-Myc also decreased. In conclusion, MMP-1 may promote malignant behavior in colorectal cancer via EMT and the Akt signaling pathway.
Collapse
Affiliation(s)
- Kai Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianbao Zheng
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Junhui Yu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yunhua Wu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Guo
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zhengshui Xu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xuejun Sun
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
9
|
Adams GN, Sharma BK, Rosenfeldt L, Frederick M, Flick MJ, Witte DP, Mosnier LO, Harmel-Laws E, Steinbrecher KA, Palumbo JS. Protease-activated receptor-1 impedes prostate and intestinal tumor progression in mice. J Thromb Haemost 2018; 16:2258-2269. [PMID: 30152921 PMCID: PMC6214773 DOI: 10.1111/jth.14277] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Indexed: 12/28/2022]
Abstract
Essentials Protease activated receptor-1 (PAR-1) has been proposed to drive cancer progression. Surprisingly, PAR-1 deletion accelerated tumor progression in two distinct experimental settings. PAR-1 deletion was shown to limit the apoptosis of transformed epithelial cells. Thrombin- and activated protein C-mediated PAR-1 activation have unique effects on tumor cell biology. SUMMARY: Background Multiple studies have implicated protease-activated receptor-1 (PAR-1), a G-protein-coupled receptor activated by proteolytic cleavage of its N-terminus, as one target coupling thrombin-mediated proteolysis to tumor progression. Objective To analyze the role of PAR-1 in the setting of two distinct spontaneously developing tumor models in mice. Methods We interbred PAR-1-deficient mice with Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice, which spontaneously develop prostate tumors, and adenomatous polyposis coli Min (APCMin/+ ) mice, which spontaneously develop intestinal adenomas. Results Analyses of TRAMP mice with advanced disease (30 weeks) revealed that PAR-1 deficiency resulted in significantly larger and more aggressive prostate tumors. Prostates collected at an earlier time point (12 weeks of age) revealed that PAR-1 promotes apoptosis in transformed epithelia. In vitro analyses of TRAMP-derived cells revealed that activated protein C-mediated PAR-1 cleavage can induce tumor cell apoptosis, suggesting that tumor cell-intrinsic PAR-1 functions can limit tumor progression. Paralleling results in TRAMP mice, PAR-1-deficient APCMin/+ mice developed three-fold more adenomas than PAR-1-expressing mice, and the adenomas that formed were significantly larger. Moreover, loss of PAR-1 expression was shown to limit apoptosis in transformed intestinal epithelial cells. Conclusions Together, these results demonstrate a previously unrecognized role for PAR-1 in impeding tumor progression in vivo. These results also offer a cautionary note suggesting that long-term PAR-1 inhibition could increase malignancy risk in some contexts.
Collapse
Affiliation(s)
- Gregory N. Adams
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Bal Krishan Sharma
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Leah Rosenfeldt
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Malinda Frederick
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Matthew J. Flick
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - David P. Witte
- Department of Pathology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | | | - Eleana Harmel-Laws
- Division of Gastroenterology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Kris A. Steinbrecher
- Division of Gastroenterology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| | - Joseph S. Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine
| |
Collapse
|
10
|
Staal J, Beyaert R. Inflammation and NF-κB Signaling in Prostate Cancer: Mechanisms and Clinical Implications. Cells 2018; 7:E122. [PMID: 30158439 PMCID: PMC6162478 DOI: 10.3390/cells7090122] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is a highly prevalent form of cancer that is usually slow-developing and benign. Due to its high prevalence, it is, however, still the second most common cause of death by cancer in men in the West. The higher prevalence of prostate cancer in the West might be due to elevated inflammation from metabolic syndrome or associated comorbidities. NF-κB activation and many other signals associated with inflammation are known to contribute to prostate cancer malignancy. Inflammatory signals have also been associated with the development of castration resistance and resistance against other androgen depletion strategies, which is a major therapeutic challenge. Here, we review the role of inflammation and its link with androgen signaling in prostate cancer. We further describe the role of NF-κB in prostate cancer cell survival and proliferation, major NF-κB signaling pathways in prostate cancer, and the crosstalk between NF-κB and androgen receptor signaling. Several NF-κB-induced risk factors in prostate cancer and their potential for therapeutic targeting in the clinic are described. A better understanding of the inflammatory mechanisms that control the development of prostate cancer and resistance to androgen-deprivation therapy will eventually lead to novel treatment options for patients.
Collapse
Affiliation(s)
- Jens Staal
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
11
|
Wang T, Jiao J, Zhang H, Zhou W, Li Z, Han S, Wang J, Yang X, Huang Q, Wu Z, Yan W, Xiao J. TGF-β induced PAR-1 expression promotes tumor progression and osteoclast differentiation in giant cell tumor of bone. Int J Cancer 2017; 141:1630-1642. [PMID: 28670703 DOI: 10.1002/ijc.30862] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 06/11/2017] [Accepted: 06/22/2017] [Indexed: 11/07/2022]
Abstract
Although protease activated receptor-1 (PAR-1) has been confirmed as an oncogene in many cancers, the role of PAR-1 in giant cell tumor (GCT) of bone has been rarely reported. The mechanism of PAR-1 in tumor-induced osteoclastogenesis still remains unclear. In the present study, we detected that PAR-1 was significantly upregulated in GCT of bone compared to normal tissues, while TGF-β was also overexpressed in GCT tissues and could promote the expression of PAR-1 in a dose and time dependent manner. Using the luciferase reporter assay, we found that two downstreams of TGF-β, Smad3 and Smad4, could activate the promoter of PAR-1, which might explain the mechanism of TGF-β induced PAR-1 expression. Meanwhile, PAR-1 was also overexpressed in microvesicles from stromal cells of GCT (GCTSCs), and might be transported from GCTSCs to monocytes through microvesicles. In addition, knockout of PAR-1 by TALENs in GCTSCs inhibited tumor growth, angiogenesis and osteoclastogenesis in GCT in vitro. Using the chick CAM models, we further showed that inhibition of PAR-1 suppressed tumor growth and giant cell formation in vivo. Using microarray assay, we detected a number of genes involved in osteoclastogenesis as the possible downstreams of PAR-1, which may partly explain the mechanism of PAR-1 in GCT. In brief, for the first time, these results reveal an upstream regulatory role of TGF-β in PAR-1 expression, and PAR-1 expression promotes tumor growth, angiogenesis and osteoclast differentiation in GCT of bone. Hence, PAR-1 represents a novel potential therapeutic target for GCT of bone.
Collapse
Affiliation(s)
- Ting Wang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Jian Jiao
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Hao Zhang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Wang Zhou
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Zhenxi Li
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Shuai Han
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Jing Wang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Xinghai Yang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Quan Huang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Zhipeng Wu
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| | - Wangjun Yan
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
- Department of Bone and Soft Tissue Tumor, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Jianru Xiao
- Department of Bone Tumor Surgery, Changzheng Hospital, Second MilitaryMedical University, Shanghai, China
| |
Collapse
|
12
|
Liu X, Yu J, Song S, Yue X, Li Q. Protease-activated receptor-1 (PAR-1): a promising molecular target for cancer. Oncotarget 2017; 8:107334-107345. [PMID: 29291033 PMCID: PMC5739818 DOI: 10.18632/oncotarget.21015] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 08/27/2017] [Indexed: 12/18/2022] Open
Abstract
PAR-1 is expressed not only in epithelium, neurons, astrocytes, immune cells, but also in cancer-associated fibroblasts, ECs (epithelial cells), myocytes of blood vessels, mast cells, and macrophages in tumor microenvironment, whereas PAR-1 stimulates macrophages to synthesize and secrete thrombin as well as other growth factors, resulting in enhanced cell proliferation, tumor growth and metastasis. Therefore, considerable effort has been devoted to the development of inhibitors targeting PAR-1. Here, we provide a comprehensive review of PAR-1’s role in cancer invasiveness and dissemination, as well as potential therapeutic strategies targeting PAR-1 signaling.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jiahui Yu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Shangjin Song
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xiaoqiang Yue
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
13
|
Wyatt RA, Trieu NPV, Crawford BD. Zebrafish Xenograft: An Evolutionary Experiment in Tumour Biology. Genes (Basel) 2017; 8:E220. [PMID: 28872594 PMCID: PMC5615353 DOI: 10.3390/genes8090220] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/27/2017] [Accepted: 08/29/2017] [Indexed: 12/17/2022] Open
Abstract
Though the cancer research community has used mouse xenografts for decades more than zebrafish xenografts, zebrafish have much to offer: they are cheap, easy to work with, and the embryonic model is relatively easy to use in high-throughput assays. Zebrafish can be imaged live, allowing us to observe cellular and molecular processes in vivo in real time. Opponents dismiss the zebrafish model due to the evolutionary distance between zebrafish and humans, as compared to mice, but proponents argue for the zebrafish xenograft's superiority to cell culture systems and its advantages in imaging. This review places the zebrafish xenograft in the context of current views on cancer and gives an overview of how several aspects of this evolutionary disease can be addressed in the zebrafish model. Zebrafish are missing homologs of some human proteins and (of particular interest) several members of the matrix metalloproteinase (MMP) family of proteases, which are known for their importance in tumour biology. This review draws attention to the implicit evolutionary experiment taking place when the molecular ecology of the xenograft host is significantly different than that of the donor.
Collapse
Affiliation(s)
- Rachael A Wyatt
- Department of Biology, University of New Brunswick, Fredericton, NB E3B 5A3, Canada.
| | - Nhu P V Trieu
- Department of Biology, University of New Brunswick, Fredericton, NB E3B 5A3, Canada.
| | - Bryan D Crawford
- Department of Biology, University of New Brunswick, Fredericton, NB E3B 5A3, Canada.
| |
Collapse
|
14
|
Saus E, Brunet-Vega A, Iraola-Guzmán S, Pegueroles C, Gabaldón T, Pericay C. Long Non-Coding RNAs As Potential Novel Prognostic Biomarkers in Colorectal Cancer. Front Genet 2016; 7:54. [PMID: 27148353 PMCID: PMC4828582 DOI: 10.3389/fgene.2016.00054] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/21/2016] [Indexed: 01/27/2023] Open
Abstract
Colorectal cancer (CRC) is the fourth most common cause of death worldwide. Surgery is usually the first line of treatment for patients with CRC but many tumors with similar histopathological features show significantly different clinical outcomes. The discovery of robust prognostic biomarkers in patients with CRC is imperative to achieve more effective treatment strategies and improve patient's care. Recent progress in next generation sequencing methods and transcriptome analysis has revealed that a much larger part of the genome is transcribed into RNA than previously assumed. Collectively referred to as non-coding RNAs (ncRNAs), some of these RNA molecules such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have been shown to be altered and to play critical roles in tumor biology. This discovery leads to exciting possibilities for personalized cancer diagnosis, and therapy. Many lncRNAs are tissue and cancer-type specific and have already revealed to be useful as prognostic markers. In this review, we focus on recent findings concerning aberrant expression of lncRNAs in CRC tumors and emphasize their prognostic potential in CRC. Further studies focused on the mechanisms of action of lncRNAs will contribute to the development of novel biomarkers for diagnosis and disease progression.
Collapse
Affiliation(s)
- Ester Saus
- Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Anna Brunet-Vega
- Department of Oncology Research, Parc Taulí Foundation, Corporació Sanitària Parc Taulí - University Institute - UAB Barcelona Sabadell, Spain
| | - Susana Iraola-Guzmán
- Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Cinta Pegueroles
- Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Toni Gabaldón
- Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Institució Catalana de Recerca i Estudis AvançatsBarcelona, Spain
| | - Carles Pericay
- Department of Oncology Research, Parc Taulí Foundation, Corporació Sanitària Parc Taulí - University Institute - UAB BarcelonaSabadell, Spain; Oncology Service, Hospital de Sabadell, Corporació Sanitària Parc Taulí - University Institute - UAB BarcelonaSabadell, Spain
| |
Collapse
|
15
|
Fan HX, Chen Y, Ni BX, Wang S, Sun M, Chen D, Zheng JH. Expression of MMP-1/PAR-1 and patterns of invasion in oral squamous cell carcinoma as potential prognostic markers. Onco Targets Ther 2015; 8:1619-26. [PMID: 26170698 PMCID: PMC4498722 DOI: 10.2147/ott.s84561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Matrix metalloproteinase (MMP)-1 degrades type I collagen of the extracellular matrix and also activates protease activated receptor (PAR)-1 to induce angiogenesis. The aims of this study were to evaluate microvessel density (MVD) and the expression of PAR-1 and MMP-1 in oral squamous cell carcinoma (SCC) specimens with different patterns of invasion (POI) and to evaluate their association with clinical outcomes. Methods Seventy-four surgically obtained oral SCC samples were classified by POI according to hematoxylin-eosin staining. MVD and the localization and intensity of PAR-1 and MMP-1 expression were detected by immunohistochemistry. Results Of the 74 oral SCC samples, 18, 5, 34, and 17 showed type I, II, III, and IV POI, respectively. MVD and expression levels of MMP-1 and PAR-1 differed between POI types I–II and POI types III–IV. Patients with low tumor expression of MMP-1 and PAR-1 and low MVD had a longer survival time than those with high tumor expression of MMP-1 and PAR-1. Moreover, the survival time of patients with POI types III–IV was shorter than that of patients with POI types I–II. Conclusion POI combined with expression levels of MMP-1 and PAR-1 may be a valuable tool for assessing the clinical prognosis of patients with oral SCC.
Collapse
Affiliation(s)
- Hai-Xia Fan
- Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, People's Republic of China
| | - Yan Chen
- Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, People's Republic of China
| | - Bo-Xiong Ni
- Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, People's Republic of China
| | - Shan Wang
- Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, People's Republic of China
| | - Miao Sun
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Dong Chen
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Jin-Hua Zheng
- Department of Anatomy, Basic Medical Science College, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|