1
|
Darvishi B, Eisavand MR, Majidzadeh-A K, Farahmand L. Matrix stiffening and acquired resistance to chemotherapy: concepts and clinical significance. Br J Cancer 2022; 126:1253-1263. [PMID: 35124704 PMCID: PMC9043195 DOI: 10.1038/s41416-021-01680-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2020] [Revised: 10/10/2021] [Accepted: 12/16/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular matrix (ECM) refers to the non-cellular components of the tumour microenvironment, fundamentally providing a supportive scaffold for cellular anchorage and transducing signaling cues that orchestrate cellular behaviour and function. The ECM integrity is abrogated in several cases of cancer, ending in aberrant activation of a number of mechanotransduction pathways and induction of multiple tumorigenic events such as extended proliferation, cell death resistance, epithelial-mesenchymal transition and most importantly the development of chemoresistance. In this regard, the present study mainly aims to elucidate how the ECM-stiffening process may contribute to the development of chemoresistance during cancer progression and what pharmacological approaches are required for tackling this issue. Hence, the first section of this review explains the process of ECM stiffening and the ways it may affect biochemical pathways to induce chemoresistance in a clinic. In addition, the second part focuses on describing some of the most important pharmacological agents capable of targeting ECM components and underlying pathways for overcoming ECM-induced chemoresistance. Finally, the third part discusses the obtained results from the application of these agents in the clinic for overcoming chemoresistance.
Collapse
Affiliation(s)
- Behrad Darvishi
- grid.417689.5Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Reza Eisavand
- grid.417689.5Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- grid.417689.5Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- grid.417689.5Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
2
|
Conformation-Specific Inhibitory Anti-MMP-7 Monoclonal Antibody Sensitizes Pancreatic Ductal Adenocarcinoma Cells to Chemotherapeutic Cell Kill. Cancers (Basel) 2021; 13:cancers13071679. [PMID: 33918254 PMCID: PMC8038143 DOI: 10.3390/cancers13071679] [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] [Academic Contribution Register] [Received: 02/23/2021] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteases (MMPs) undergo post-translational modifications including pro-domain shedding. The activated forms of these enzymes are effective drug targets, but generating potent biological inhibitors against them remains challenging. We report the generation of anti-MMP-7 inhibitory monoclonal antibody (GSM-192), using an alternating immunization strategy with an active site mimicry antigen and the activated enzyme. Our protocol yielded highly selective anti-MMP-7 monoclonal antibody, which specifically inhibits MMP-7's enzyme activity with high affinity (IC50 = 132 ± 10 nM). The atomic model of the MMP-7-GSM-192 Fab complex exhibited antibody binding to unique epitopes at the rim of the enzyme active site, sterically preventing entry of substrates into the catalytic cleft. In human PDAC biopsies, tissue staining with GSM-192 showed characteristic spatial distribution of activated MMP-7. Treatment with GSM-192 in vitro induced apoptosis via stabilization of cell surface Fas ligand and retarded cell migration. Co-treatment with GSM-192 and chemotherapeutics, gemcitabine and oxaliplatin elicited a synergistic effect. Our data illustrate the advantage of precisely targeting catalytic MMP-7 mediated disease specific activity.
Collapse
|
3
|
Yang J, Liu X, Huang Y, He L, Zhang W, Ren J, Wang Y, Wu J, Wu X, Shan L, Yang X, Sun L, Liang J, Zhang Y, Shang Y. TRPS1 drives heterochromatic origin refiring and cancer genome evolution. Cell Rep 2021; 34:108814. [PMID: 33691114 DOI: 10.1016/j.celrep.2021.108814] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2020] [Revised: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Exploitation of naturally occurring genetic mutations could empower the discovery of novel aspects of established cancer genes. We report here that TRPS1, a gene linked to the tricho-rhino-phalangeal syndrome (TRPS) and recently identified as a potential breast cancer driver, promotes breast carcinogenesis through regulating replication. Epigenomic decomposition of TRPS1 landscape reveals nearly half of H3K9me3-marked heterochromatic origins are occupied by TRPS1, where it encourages the chromatin loading of APC/C, resulting in uncontrolled origin refiring. TRPS1 binds to the genome through its atypical H3K9me3 reading via GATA and IKAROS domains, while TRPS-related mutations affect its chromatin binding, replication boosting, and tumorigenicity. Concordantly, overexpression of wild-type but not TRPS-associated mutants of TRPS1 is sufficient to drive cancer genome amplifications, which experience an extrachromosomal route and dynamically evolve to confer therapeutic resistance. Together, these results uncover a critical function of TRPS1 in driving heterochromatin origin firing and breast cancer genome evolution.
Collapse
Affiliation(s)
- Jianguo Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Xiaoping Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Yunchao Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Lin He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Wenting Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jiajing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaodi Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Lin Shan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaohan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Luyang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Jing Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China.
| | - Yongfeng Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Health Science Center, Beijing 100191, China; Department of Biochemistry and Molecular Biology, School of Medicine, Hangzhou Normal University, Hangzhou 311121, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
Szarvas T, Hoffmann MJ, Olah C, Szekely E, Kiss A, Hess J, Tschirdewahn S, Hadaschik B, Grotheer V, Nyirady P, Csizmarik A, Varadi M, Reis H. MMP-7 Serum and Tissue Levels Are Associated with Poor Survival in Platinum-Treated Bladder Cancer Patients. Diagnostics (Basel) 2020; 11:diagnostics11010048. [PMID: 33396213 PMCID: PMC7824149 DOI: 10.3390/diagnostics11010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/17/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 01/09/2023] Open
Abstract
Chemotherapy resistance is a main cause of therapeutic failure and death in bladder cancer. With the approval of immune checkpoint inhibitors, prediction of platinum treatment became of great clinical importance. Matrix metalloproteinase-7 (MMP-7) was shown to be involved in cisplatin resistance. Therefore, tissue and circulating MMP-7 levels were evaluated in 124 bladder cancer patients who received postoperative platinum-based chemotherapy. Tissue MMP-7 levels were analyzed by immunohistochemistry in 72 formalin-fixed, paraffin-embedded chemo-naïve tumor samples, while MMP-7 serum concentrations were determined in 132 serum samples of an independent cohort of 52 patients. MMP-7 tissue and serum levels were correlated with clinicopathological and follow-up data. MMP-7 gene expression was determined by RT-qPCR in 20 urothelial cancer cell lines and two non-malignant urothelial cell lines. MMP-7 was overexpressed in RT-112 and T-24 cells by stable transfection, to assess its functional involvement in platinum sensitivity. High MMP-7 tissue expression and pretreatment serum concentrations were independently associated with poor overall survival (tissue HR = 2.296, 95%CI = 1.235–4.268 and p = 0.009; serum HR = 2.743, 95%CI = 1.258–5.984 and p = 0.011). Therefore, MMP-7 tissue and serum analysis may help to optimize therapeutic decisions. Stable overexpression in RT-112 and T-24 cells did not affect platinum sensitivity.
Collapse
Affiliation(s)
- Tibor Szarvas
- Department of Urology, University of Duisburg-Essen, 45147 Essen, Germany; (C.O.); (J.H.); (S.T.); (B.H.)
- Department of Urology, Semmelweis University, 1089 Budapest, Hungary; (P.N.); (A.C.); (M.V.)
- Correspondence: ; Tel.: +49-201-7238-4967
| | - Michèle J. Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Csilla Olah
- Department of Urology, University of Duisburg-Essen, 45147 Essen, Germany; (C.O.); (J.H.); (S.T.); (B.H.)
| | - Eszter Szekely
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.S.); (A.K.)
| | - Andras Kiss
- 2nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary; (E.S.); (A.K.)
| | - Jochen Hess
- Department of Urology, University of Duisburg-Essen, 45147 Essen, Germany; (C.O.); (J.H.); (S.T.); (B.H.)
| | - Stephan Tschirdewahn
- Department of Urology, University of Duisburg-Essen, 45147 Essen, Germany; (C.O.); (J.H.); (S.T.); (B.H.)
| | - Boris Hadaschik
- Department of Urology, University of Duisburg-Essen, 45147 Essen, Germany; (C.O.); (J.H.); (S.T.); (B.H.)
| | - Vera Grotheer
- Department of Orthopedics and Trauma Surgery, Medical Faculty, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Peter Nyirady
- Department of Urology, Semmelweis University, 1089 Budapest, Hungary; (P.N.); (A.C.); (M.V.)
| | - Anita Csizmarik
- Department of Urology, Semmelweis University, 1089 Budapest, Hungary; (P.N.); (A.C.); (M.V.)
| | - Melinda Varadi
- Department of Urology, Semmelweis University, 1089 Budapest, Hungary; (P.N.); (A.C.); (M.V.)
| | - Henning Reis
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| |
Collapse
|
5
|
Yuan S, Lin LS, Gan RH, Huang L, Wu XT, Zhao Y, Su BH, Zheng D, Lu YG. Elevated matrix metalloproteinase 7 expression promotes the proliferation, motility and metastasis of tongue squamous cell carcinoma. BMC Cancer 2020; 20:33. [PMID: 31937294 PMCID: PMC6958600 DOI: 10.1186/s12885-020-6521-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2019] [Accepted: 01/07/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase 7 (MMP7), as the smallest member of the matrix metalloproteinase family, has been verified to be implicated in cancer progression, especially metastasis. However, its expression pattern and function in tongue cancer is not clear. METHODS The expression of MMP7 in human tongue squamous cell carcinoma (TSCC) specimens compared with their respective paired nontumour tissues by real-time PCR and immunohistochemical staining. The effect of MMP7 on the proliferation, apoptosis, migration, invasion of tongue cancer cells was tested in appropriate ways after MMP7 siRNA knockdown or overexpression. The effect of MMP7 on lymph node metastasis in vivo was analyzed using a high-metastasis orthotopic nude mouse tongue transplanted tumour model. RESULTS We found markedly elevated expression of MMP7 in human TSCC specimens compared with their respective paired nontumour tissues, and this high expression was correlated with the patients' lymph node metastasis. Furthermore, the results of molecular functional assays confirmed that MMP7 promotes cell proliferation, migration and invasion of TSCC cells. Knockdown of MMP7 inhibited lymph nodes metastasis in vivo. CONCLUSIONS MMP7 plays an oncogenic role in carcinogenesis and metastasis of tongue cancer, and may serve as a potential therapeutic target for tongue cancer.
Collapse
Affiliation(s)
- Shuo Yuan
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou, 350000, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China
| | - Li-Song Lin
- Department of Oral and Maxillofacial Surgery, Affiliated First Hospital of Fujian Medical University, 20 Cha Zhong Road, Fuzhou, 350005, China
| | - Rui-Huan Gan
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou, 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China
| | - Li Huang
- Department of Oral and Maxillofacial Surgery, Affiliated First Hospital of Fujian Medical University, 20 Cha Zhong Road, Fuzhou, 350005, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China
| | - Xiao-Ting Wu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou, 350000, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xue Yuan Road, University Town, Fuzhou, 350122, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China
| | - Yong Zhao
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China
| | - Bo-Hua Su
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou, 350000, China
| | - Dali Zheng
- Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China.
| | - You-Guang Lu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Middle Road, Fuzhou, 350000, China.,Key laboratory of Stomatology of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Rd, Fuzhou, 350004, China
| |
Collapse
|
6
|
Muraki M, Hirota K. Confirmation of covalently-linked structure and cell-death inducing activity in site-specific chemical conjugates of human Fas ligand extracellular domain. BMC Res Notes 2018; 11:395. [PMID: 29907131 PMCID: PMC6003068 DOI: 10.1186/s13104-018-3501-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/04/2018] [Accepted: 06/07/2018] [Indexed: 12/03/2022] Open
Abstract
Objective In this study, we aimed to identify the structural components and to clarify the biological activity in the site-specific conjugates of human Fas ligand extracellular domain (hFasLECD) with either fluorescein moiety (FL) or chicken egg-white avidin (Avi). The conjugates were characterized by molecular-weight measurement using MALDI-TOF mass-spectrometric analysis and by cell-death inducing activity measurement against a human colorectal cancer cell line, HT-29, using MTT cell-viability assay. Pretreatment effect with human interferon-γ (IFN-γ) on the cell-death inducing activity was evaluated. Results The mass-spectrometric analysis of the hFasLECD-Avi conjugate showed that it was possible to detect the signal peak of molecular-weight to electric charge (m/z) derived from the component involved in the covalent linking as the sum of the molecular-weight of unconjugated hFasLECD- and Avi-derivative subunits, in addition to the signals from each corresponding subunit component irrelevant to the covalent linking. The cell-viability assay revealed that both conjugates possessed a remarkable death-inducing activity against HT-29 cells in synergy with the pretreatment using human IFN-γ. Following 24 h pretreatment with 100 IU/ml of human IFN-γ, almost no viable cells existed after 72 h treatment with either 100 or 1000 ng/ml of FL-hFasLECD and hFasLECD-Avi conjugates. Electronic supplementary material The online version of this article (10.1186/s13104-018-3501-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michiro Muraki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| | - Kiyonori Hirota
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| |
Collapse
|
7
|
Mitochondrial UQCRB as a new molecular prognostic biomarker of human colorectal cancer. Exp Mol Med 2017; 49:e391. [PMID: 29147009 PMCID: PMC5704184 DOI: 10.1038/emm.2017.152] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 12/17/2022] Open
Abstract
Ubiquinol cytochrome c reductase binding protein (UQCRB) is important for mitochondrial complex III stability, electron transport, cellular oxygen sensing and angiogenesis. However, its potential as a prognostic marker in colorectal cancer (CRC) remains unclear. The aim of this study was to determine whether UQCRB can be used as a diagnostic molecular marker for CRC. The correlation between the expression of three genes (UQCRB, UQCRFS1 and MT-CYB) in the mitochondrial respiratory chain complex III and clinico-pathological features was determined. Compared to non-tumor tissues, UQCRB gene expression was upregulated in CRC tissues. Gene and protein expression of the genes were positively correlated. Copy number variation (CNV) differences in UQCRB were observed in CRC tissues (1.32-fold) compared to non-tumor tissues. The CNV of UQCRB in CRC tissues increased proportionally with gene expression and clinical stage. Single-nucleotide polymorphisms in the 3′-untranslated region of UQCRB (rs7836698 and rs10504961) were investigated, and the rs7836698 polymorphism was associated with CRC clinical stage. DNA methylation of the UQCRB promoter revealed that most CRC patients had high methylation levels (12/15 patients) in CRC tissues compared to non-tumor tissues. UQCRB overexpression and CNV gain were correlated with specific CRC clinico-pathological features, indicating clinical significance as a prognostic predictor in CRC. Gene structural factors may be more important than gene transcription repression factors with respect to DNA methylation in UQCRB overexpression. Our results provide novel insights into the critical role of UQCRB in regulating CRC, supporting UQCRB as a new candidate for the development of diagnostics for CRC patients.
Collapse
|
8
|
Down-regulation of miRNA-148a and miRNA-625-3p in colorectal cancer is associated with tumor budding. BMC Cancer 2017; 17:607. [PMID: 28863773 PMCID: PMC5580437 DOI: 10.1186/s12885-017-3575-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2016] [Accepted: 08/22/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND MiRNAs are often deregulated in colorectal cancer and might function as tumor suppressors or as oncogenes. They participate in controlling key signaling pathways involved in proliferation, invasion and apoptosis and may serve as prognostic and predictive markers. In this study we aimed to evaluate the role of miRNA-148a and miRNA-625-3p in metastatic colorectal cancer. METHODS Fifty-four patients with a first-time diagnosed CRC receiving FOLFOX ± Bevacizumab were involved in the study. Tumor samples underwent routine pathology examination including evaluation for tumor budding and KRAS. MiRNA-148a and miRNA-625-3p expression analysis was done by RT-PCR. Associations between expression of both miRNAs and clinico-pathological factors, treatment outcomes and survival were analyzed. RESULTS Both miRNA-148a and miRNA-625-3p were down-regulated in the tumors compared to normal colonic mucosa. Significantly lower expression of both miRNAs was noticed in tumors with budding phenomenon compared to tumors without it (median values of miRNA-148a were 0.314 and 0.753 respectively, p = 0.011, and 0.404 and 0.620 respectively for miRNA-625-3p, p = 0.036). Significantly lower expression of miRNA-625-3p was detected in rectal tumors, compared to tumors in the colon (median 0.390 and 0.665 respectively, p = 0.037). Progression free survival was significantly lower in patients with high miRNA-148a expression (6 and 9 months respectively, p = 0.033), but there were no significant differences in PFS for miRNA-625-3p and in overall survival for both miRNAs. CONCLUSIONS There was a significant relationship between low miRNA-148a and miRNA-625-3p expression and tumor budding, which is thought to represent epithelial-mesenchymal transition. Both studied miRNAs may be associated with a more aggressive phenotype and could be the potential prognostic and predictive biomarkers in CRC. Further investigation is needed to confirm miRNAs involvement in EMT, and their prognostic and predictive value.
Collapse
|
9
|
Liu H, Liu X, Zhang C, Zhu H, Xu Q, Bu Y, Lei Y. Redox Imbalance in the Development of Colorectal Cancer. J Cancer 2017; 8:1586-1597. [PMID: 28775778 PMCID: PMC5535714 DOI: 10.7150/jca.18735] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2016] [Accepted: 02/27/2017] [Indexed: 01/10/2023] Open
Abstract
Redox imbalance is resulted from the destruction of balance between oxidants and antioxidants. The dominant oxidants are reactive oxygen species (ROS), which are involved in multiple cellular processes by physiologically transporting signal as a second messenger or pathologically oxidizing DNA, lipids, and proteins. Generally speaking, low concentration of ROS is indispensable for cell survival and proliferation. However, high concentration of ROS is cytotoxic. Additionally, ROS are now known to induce the oxidative modification of macromolecules especially proteins. The redox modification of proteins is involved in numerous biological processes related to diseases including CRC. Herein, we attempt to afford an overview that highlights the crosstalk between redox imbalance and CRC.
Collapse
Affiliation(s)
- Hao Liu
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Xin Liu
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Chundong Zhang
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Huifang Zhu
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Qian Xu
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Youquan Bu
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, P. R. China
| |
Collapse
|
10
|
Isaacson KJ, Martin Jensen M, Subrahmanyam NB, Ghandehari H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J Control Release 2017; 259:62-75. [PMID: 28153760 DOI: 10.1016/j.jconrel.2017.01.034] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/16/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
While commonly known for degradation of the extracellular matrix, matrix metalloproteinases (MMPs) exhibit broad potential for use in targeting of bioactive and imaging agents in cancer treatment. MMPs are upregulated at all stages of expression in cancers. A comprehensive analysis of published literature on expression of all MMP subtypes at the genetic, protein, and activity levels in normal and diseased tissues indicate targeting applicability in a variety of cancers. This expression significantly increases at advanced cancer stages, providing an improved opportunity for controlled release in higher-stage patients. Since MMPs are integral at every stage of metastasis, MMP roles in cancer are discussed with a focus on MMP distribution and mobility within cells and tumors for cancer targeting applications. Several strategies for MMP utilization in targeting - such as matrix degradation, MMP cleavage, MMP binding, and MMP-induced environmental changes - are addressed.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Nithya B Subrahmanyam
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
11
|
Choi S, Choi Y, Jun E, Kim IS, Kim SE, Jung SA, Oh ES. Shed syndecan-2 enhances tumorigenic activities of colon cancer cells. Oncotarget 2016; 6:3874-86. [PMID: 25686828 PMCID: PMC4414160 DOI: 10.18632/oncotarget.2885] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2014] [Accepted: 12/09/2014] [Indexed: 12/14/2022] Open
Abstract
Because earlier studies showed the cell surface heparan sulfate proteoglycan, syndecan-2, sheds from colon cancer cells in culture, the functional roles of shed syndecan-2 were assessed. A non-cleavable mutant of syndecan-2 in which the Asn148-Leu149 residues were replaced with Asn148-Ile149, had decreased shedding, less cancer-associated activities of syndecan-2 in vitro, and less syndecan-2-mediated metastasis of mouse melanoma cells in vivo, suggesting the importance of shedding on syndecan-2-mediated pro-tumorigenic functions. Indeed, shed syndecan-2 from cancer-conditioned media and recombinant shed syndecan-2 enhanced cancer-associated activities, and depletion of shed syndecan-2 abolished these effects. Similarly, shed syndecan-2 was detected from sera of patients from advanced carcinoma (625.9 ng/ml) and promoted cancer-associated activities. Furthermore, a series of syndecan-2 deletion mutants showed that the tumorigenic activity of shed syndecan-2 resided in the C-terminus of the extracellular domain and a shed syndecan-2 synthetic peptide (16 residues) was sufficient to establish subcutaneous primary growth of HT29 colon cancer cells, pulmonary metastases (B16F10 cells), and primary intrasplenic tumor growth and liver metastases (4T1 cells). Taken together, these results demonstrate that shed syndecan-2 directly enhances colon cancer progression and may be a promising therapeutic target for controlling colon cancer development.
Collapse
Affiliation(s)
- Sojoong Choi
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea
| | - Youngsil Choi
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Eunsung Jun
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea.,Department of Biochemistry and Cell Biology, School of Medicine and Cell & Matrix Research Institute, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea
| | - Sung-Ae Jung
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, Republic of Korea
| |
Collapse
|
12
|
Hibino Y, Sakamoto N, Naito Y, Goto K, Oo HZ, Sentani K, Hinoi T, Ohdan H, Oue N, Yasui W. Significance of miR-148a in Colorectal Neoplasia: Downregulation of miR-148a Contributes to the Carcinogenesis and Cell Invasion of Colorectal Cancer. Pathobiology 2015; 82:233-41. [PMID: 26389729 DOI: 10.1159/000438826] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/20/2015] [Accepted: 07/20/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Colorectal cancer (CRC) develops through the deregulation of gene expression and the accumulation of epigenetic abnormalities, leading to tumor cell acquisition of malignant features. MicroRNAs (miRNAs) play a critical role in cancer development, where they can act as oncogenes or oncosuppressors. METHODS miR-148a expression was measured by qRT-PCR in patients with colorectal adenoma (n = 21) and CRC (stage I-IV, n = 159) using formalin-fixed paraffin-embedded tissue samples. In situ hybridization (ISH) using an miR-148a-specific probe was also performed. To further confirm the direct effect of miR-148a on matrix metalloproteinase (MMP)7 expression in CRC, MTT and cell invasion assays using HT29 and WiDr cells were performed. RESULTS miR-148a expression was found to be clearly downregulated in high-grade adenoma compared to low-grade adenoma on both qRT-PCR and ISH analysis. Downregulation of miR-148a expression was significantly correlated with advanced clinicopathological features and was an independent prognostic classifier in patients with stage III CRC. In CRC cells and tissues, miR-148a expression was inversely correlated with the expression of MMP7. CONCLUSION We showed the collaborative participation of miR-148a and MMP7 in CRC cell invasion. These results also demonstrate that the downregulation of miR-148a expression promotes CRC progression, especially carcinogenesis and cancer cell invasion.
Collapse
Affiliation(s)
- Yumi Hibino
- Department of Molecular Pathology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Deng W, Jiao N, Li N, Wan X, Luo S, Zhang Y. Decreased expression of PinX1 protein predicts poor prognosis of colorectal cancer patients receiving 5-FU adjuvant chemotherapy. Biomed Pharmacother 2015. [DOI: 10.1016/j.biopha.2015.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022] Open
|
14
|
Sun DW, Zhang YY, Qi Y, Zhou XT, Lv GY. Prognostic significance of MMP-7 expression in colorectal cancer: a meta-analysis. Cancer Epidemiol 2015; 39:135-42. [PMID: 25677090 DOI: 10.1016/j.canep.2015.01.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2014] [Revised: 01/07/2015] [Accepted: 01/11/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Matrix metalloproteinase-7 (MMP-7) is a member of the family of matrix metalloproteinases (MMPs); it is associated with invasive tumor growth and distant metastasis in colorectal cancer (CRC). However, the prognostic value of MMP-7 in CRC remains controversial. Thus, we conducted a meta-analysis to investigate the prognostic value of MMP-7 expression in CRC. METHODS We systematically searched for studies evaluating the relationship between MMP-7 expression and the outcome of CRC patients in PubMed, the Cochrane library, Embase, Google Scholar, and Wanfang databases updated to August 2014. Studies were assessed for quality using REMARK (REporting recommendations for tumor MARKer prognostic studies). Hazard ratios (HRs) with 95% confidence intervals (95%CIs) were pooled to estimate the hazard for overall survival (OS) and disease-free survival (DFS), and odds ratios (ORs) with 95%CIs were pooled to estimate the impact of MMP-7 on the 5-year survival rate. RESULTS In total, 2985 patients from 17 studies were included in this meta-analysis. Our results showed that MMP-7 over-expression predicted poor OS (HR=3.57, 95%CI 2.21-5.75, P=0.000, random-effect model), poor DFS (HR=2.49, 95%CI 1.73-3.57, P=0.000, fixed-effect model), and decreased 5-year survival rate (OR=0.26, 95%CI 0.19-0.37, P=0.000, fixed-effect model). Therefore, MMP-7 is an indicator of poor survival and high recurrence rate in CRC patients. CONCLUSION MMP-7 can serve as a prognostic indicator and a potential novel target for treatment in CRC patients. More well-designed prospective studies with better methodology for MMP-7 assessment are required to clarify the prognostic significance of MMP-7 expression in CRC patients.
Collapse
Affiliation(s)
- Da-wei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Ying-yi Zhang
- Department of Chemotherapy, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Yue Qi
- Department of Hepatobiliary and Pancreatic Medicine, The First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Xing-tong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Guo-yue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Bethune Hospital, Jilin University, Changchun 130021, China.
| |
Collapse
|
15
|
Shay G, Lynch CC, Fingleton B. Moving targets: Emerging roles for MMPs in cancer progression and metastasis. Matrix Biol 2015; 44-46:200-6. [PMID: 25652204 DOI: 10.1016/j.matbio.2015.01.019] [Citation(s) in RCA: 319] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/03/2014] [Revised: 01/24/2015] [Accepted: 01/24/2015] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases have long been associated with cancer. Clinical trials of small molecule inhibitors for this family of enzymes however, were spectacularly unsuccessful in a variety of tumor types. Here, we discuss some of the newer roles that have been uncovered for MMPs in cancer that would not have been targeted with those initial inhibitors or in the patient populations analyzed. We also consider novel ways of using cancer-associated MMP activity for clinical benefit.
Collapse
Affiliation(s)
- Gemma Shay
- Dept. of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Conor C Lynch
- Dept. of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Barbara Fingleton
- Dept of Cancer Biology, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232, USA.
| |
Collapse
|
16
|
Sizemore ST, Sizemore GM, Booth CN, Thompson CL, Silverman P, Bebek G, Abdul-Karim FW, Avril S, Keri RA. Hypomethylation of the MMP7 promoter and increased expression of MMP7 distinguishes the basal-like breast cancer subtype from other triple-negative tumors. Breast Cancer Res Treat 2014; 146:25-40. [PMID: 24847890 DOI: 10.1007/s10549-014-2989-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2013] [Accepted: 04/30/2014] [Indexed: 12/30/2022]
Abstract
Identification of novel targets for the treatment of basal-like breast cancer is essential for improved outcomes in patients with this disease. This study investigates the association of MMP7 expression and MMP7 promoter methylation with subtype and outcome in breast cancer patient cohorts. Immunohistochemical analysis was performed on a breast cancer tissue microarray and validated in independent histological samples. MMP7 expression significantly correlated with patient age, tumor size, triple-negative (TN) status, and recurrence. Analysis of publically available datasets confirmed MMP7 gene expression as a prognostic marker of breast cancer metastasis, particularly metastasis to the brain and lungs. Methylation of the MMP7 promoter was assessed by methylation-specific PCR in a panel of breast cancer cell lines and patient tumor samples. Hypomethylation of the MMP7 promoter significantly correlated with TN status in DNA from patient tumor samples, and this association was confirmed using The Cancer Genome Atlas (TCGA) dataset. Evaluation of a panel of breast cancer cell lines and data from the Curtis and TCGA breast carcinoma datasets revealed that elevated MMP7 expression and MMP7 promoter hypomethylation are specific biomarkers of the basal-like molecular subtype which shares considerable, but not complete, overlap with the clinical TN subtype. Importantly, MMP7 expression was identified as an independent predictor of pathological complete response in a large breast cancer patient cohort. Combined, these data suggest that MMP7 expression and MMP7 promoter methylation may be useful as prognostic biomarkers. Furthermore, MMP7 expression and promoter methylation analysis may be effective mechanisms to distinguish basal-like breast cancers from other triple-negative subtypes. Finally, these data implicate MMP7 as a potential therapeutic target for the treatment of basal-like breast cancers.
Collapse
Affiliation(s)
- Steven T Sizemore
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH, 44106-4965, USA
| | | | | | | | | | | | | | | | | |
Collapse
|