1
|
Liu J, Li Y, Lian X, Zhang C, Feng J, Tao H, Wang Z. Potential target within the tumor microenvironment - MT1-MMP. Front Immunol 2025; 16:1517519. [PMID: 40196128 PMCID: PMC11973285 DOI: 10.3389/fimmu.2025.1517519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Matrix metalloproteinases are integral to the modification of the tumor microenvironment and facilitate tumor progression by degrading the extracellular matrix, releasing cytokines, and influencing the recruitment of immune cells. Among the matrix metalloproteinases, membrane-type matrix metalloproteinase 1 (MT1-MMP/MMP14) is the first identified membrane-type MMP and acts as an essential proteolytic enzyme that enables tumor infiltration and metastatic progression. Given the pivotal role of MT1-MMP in tumor progression and the correlation between its overexpression in tumors and unfavorable prognoses across multiple cancer types, a comprehensive understanding of the potential functional mechanisms of MT1-MMP is essential. This knowledge will aid in the advancement of diverse anti-tumor therapies aimed at targeting MT1-MMP. Although contemporary research has highlighted the considerable potential of MT1-MMP in targeted cancer therapy, studies pertaining to its application in cell therapy remain relatively limited. In this review, we delineate the structural characteristics and regulatory mechanisms of MT1-MMP expression, as well as its biological significance in tumorigenesis. Finally, we discussed the current status and prospects of anti-tumor therapies targeting MT1-MMP.
Collapse
Affiliation(s)
- Jinlong Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yijing Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xueqi Lian
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chenglin Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianing Feng
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hongfei Tao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhimin Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Kirsch KE, Little ME, Cundari TR, El-Shaer E, Barone G, Lynch VM, Toledo SA. Direct O 2 mediated oxidation of a Ni(II)N 3O structural model complex for the active site of nickel acireductone dioxygenase (Ni-ARD): characterization, biomimetic reactivity, and enzymatic implications. Dalton Trans 2024; 53:17852-17863. [PMID: 39421893 DOI: 10.1039/d4dt02538e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
A new biomimetic model complex of the active site of acireductone dioxygenase (ARD) was synthesized and crystallographically characterized ([Ni(ii)(N-(ethyl-N'Me2)(Py)(2-t-ButPhOH))(OTf)]-1). 1 displays carbon-carbon oxidative cleavage activity in the presence of O2 towards the substrate 2-hydroxyacetophenone. This reactivity was monitored via UV-Visible and NMR spectroscopy. We postulate that the reactivity of 1 with O2 leads to the formation of a putative Ni(III)-superoxo transient species resulting from the direct activation of O2via the nickel center during the oxidative reaction. This proposed intermediate and reaction mechanism were studied in detail using DFT calculations. 1 and its substrate bound derivatives display reactivity toward mild outer sphere oxidants, suggesting ease of access to high valent Ni coordination complexes, consistent with our calculations. If confirmed, the direct activation of O2 at a nickel center could have implications for the mechanism of action of ARD and other nickel-based dioxygenases and their respective non-traditional, enzymatic moonlighting functions, as well as contribute to a general understanding of direct oxidation of nickel(II) coordination complexes by O2.
Collapse
Affiliation(s)
- Kelsey E Kirsch
- Department of Chemistry, American University, 4400 Massachusetts Ave NW, Washington, DC, 20016, USA.
| | - Mary E Little
- Department of Chemistry, St Edward's University, 3001 South Congress Ave, Austin, Texas 78704, USA
| | - Thomas R Cundari
- Department of Chemistry, University of North Texas, 1155 Union Cir, Denton, Texas 76203, USA
| | - Emily El-Shaer
- Department of Chemistry, St Edward's University, 3001 South Congress Ave, Austin, Texas 78704, USA
| | - Georgia Barone
- Department of Chemistry, St Edward's University, 3001 South Congress Ave, Austin, Texas 78704, USA
| | - Vincent M Lynch
- Department of Chemistry, The University of Texas at Austin, 120 Inner Campus Dr Stop G2500, Austin, Texas 78712, USA
| | - Santiago A Toledo
- Department of Chemistry, American University, 4400 Massachusetts Ave NW, Washington, DC, 20016, USA.
| |
Collapse
|
3
|
Grafinger OR, Hayward JJ, Meng Y, Geddes-McAlister J, Li Y, Mar S, Sheng M, Su B, Thillainadesan G, Lipsman N, Coppolino MG, Trant JF, Jerzak KJ, Leong HS. Cancer cell extravasation requires iplectin-mediated delivery of MT1-MMP at invadopodia. Br J Cancer 2024; 131:931-943. [PMID: 38969866 PMCID: PMC11369281 DOI: 10.1038/s41416-024-02782-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Invadopodia facilitate cancer cell extravasation, but the molecular mechanism whereby invadopodia-specific proteases such as MT1-MMP are called to invadopodia is unclear. METHODS Mass spectrometry and immunoprecipitation were used to identify interactors of MT1-MMP in metastatic breast cancer cells. After identification, siRNA and small molecule inhibitors were used to assess the effect these interactors had on cellular invasiveness. The chicken embryo chorioallantoic membrane (CAM) model was used to assess extravasation and invadopodia formation in vivo. RESULTS In metastatic breast cancer cells, MT1-MMP was found to associate with plectin, a cytolinker and scaffolding protein. Complex formation between plectin and MT1-MMP launches invadopodia formation, a subtype we termed iplectin (i = invadopodial). iPlectin delivers MT1-MMP to invadopodia and is indispensable for regulating cell surface levels of the enzyme. Genetic depletion of plectin with siRNA reduced invadopodia formation and cell invasion in vitro. In vivo extravasation efficiency assays and intravital imaging revealed iplectin to be a key contributor to invadopodia ultrastructure and essential for extravasation. Pharmacologic inhibition of plectin using the small molecule Plecstatin-1 (PST-1) abrogated MT1-MMP delivery to invadopodia and extravasation efficiency. CONCLUSIONS Anti-metastasis therapeutic approaches that target invadopodia are possible by disrupting interactions between MT1-MMP and iplectin. CLINICAL TRIAL REGISTRATION NUMBER NCT04608357.
Collapse
Affiliation(s)
- Olivia R Grafinger
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - John J Hayward
- Department of Chemistry, University of Windsor, Windsor, ON, Canada
| | - Ying Meng
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | | | - Yan Li
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sara Mar
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Minzhi Sheng
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Boyang Su
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gobi Thillainadesan
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - John F Trant
- Department of Chemistry, University of Windsor, Windsor, ON, Canada
| | - Katarzyna J Jerzak
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Hu S, Xia C, Zou H, Ren W, Liu L, Wang L, Kang Q, He K, Wang T, Zhang X. HS6ST1 overexpressed in cancer-associated fibroblast and inhibited cholangiocarcinoma progression. Dig Liver Dis 2023; 55:1114-1125. [PMID: 36586771 DOI: 10.1016/j.dld.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/11/2022] [Accepted: 12/07/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUD Fibroblasts turn into cancer associated fibroblasts (CAFs) in the tumor microenvironment, which play an important role in tumor progression. However, the mechanism is unclear. AIMS To investigate the role of CAFs with HS6ST1-overexpression in cell migration and invasion effects. METHODS Human primary CAFs were isolated and identified from intrahepatic cholangiocarcinoma. mRNA profiles differences between CAFs and NFs were examined by using transcriptome sequencing. Using Transwell® migration assays, ICCA cells (RBE and HUCCT1) with NF-CM, CAF-CM, CAFsNC-CM, and CAFsHS6ST1-CM were analyzed. Immunohistochemical staining were used to analyze the expression of HS6ST1 in CAF in 152 patients with ICCA. Overall survival (OS) was compared based on CAF HS6ST1 expression were analysed. The relationship between clinicopathological parameters and survival was also examined. RESULTS Successfully isolated CAFs is positive staining with αSMA, FSP-1, FAP, and PDGFR-β. Transcriptome sequencing showed that differently expressed genes were enriched in the function of the extracellular matrix and chemokine signaling pathway. HS6ST1 is differentially expressed between CAFs and NFs, and associated with the migration and invasion of ICCA cells. Moreover, HS6ST1 positive expression of CAFs predicted unfavorable prognosis in patients with intrahepatic cholangiocarcinoma and showed correlation with the presence of lymph node metastasis. CONCLUSION HS6ST1 is new possibilities for targeting the CAFs to reduce cholangiocarcinoma growth and metastasis.
Collapse
Affiliation(s)
- Sheng Hu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Chuqi Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hao Zou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Wenjun Ren
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lixin Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Lianmin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Qiang Kang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Kai He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Tao Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China.
| | - Xiaowen Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China.
| |
Collapse
|
5
|
Knapinska AM, Drotleff G, Chai C, Twohill D, Ernce A, Tokmina-Roszyk D, Grande I, Rodriguez M, Larson B, Fields GB. Screening MT1-MMP Activity and Inhibition in Three-Dimensional Tumor Spheroids. Biomedicines 2023; 11:biomedicines11020562. [PMID: 36831098 PMCID: PMC9953393 DOI: 10.3390/biomedicines11020562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Membrane type 1 matrix metalloproteinase (MT1-MMP) has been shown to be crucial for tumor angiogenesis, invasion, and metastasis, and thus MT1-MMP is a high priority target for potential cancer therapies. To properly evaluate MT1-MMP inhibitors, a screening protocol is desired by which enzyme activity can be quantified in a tumor microenvironment-like model system. In the present study, we applied a fluorogenic, collagen model triple-helical substrate to quantify MT1-MMP activity for tumor spheroids embedded in a collagen hydrogel. The substrate was designed to be MT1-MMP selective and to possess fluorescent properties compatible with cell-based assays. The proteolysis of the substrate correlated to glioma spheroid invasion. In turn, the application of either small molecule or protein-based MMP inhibitors reduced proteolytic activity and glioma spheroid invasion. The presence of MT1-MMP in glioma spheroids was confirmed by western blotting. Thus, spheroid invasion was dependent on MT1-MMP activity, and inhibitors of MT1-MMP and invasion could be conveniently screened in a high-throughput format. The combination of the fluorogenic, triple-helical substrate, the three-dimensional tumor spheroids embedded in collagen, and Hit-Pick software resulted in an easily adaptable in vivo-like tumor microenvironment for rapidly processing inhibitor potential for anti-cancer use.
Collapse
Affiliation(s)
- Anna M. Knapinska
- Alphazyme, Jupiter, FL 33458, USA
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Gary Drotleff
- Alphazyme, Jupiter, FL 33458, USA
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Cedric Chai
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Destiny Twohill
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Alexa Ernce
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Dorota Tokmina-Roszyk
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Isabella Grande
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Michelle Rodriguez
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
| | - Brad Larson
- Agilent Technologies, Raleigh, NC 27606, USA
| | - Gregg B. Fields
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, FL 33458, USA
- Correspondence:
| |
Collapse
|
6
|
Ohta T, Tanaka M, Taki S, Nakagawa H, Nagase S. Honeycomb-like Structured Film, a Novel Therapeutic Device, Suppresses Tumor Growth in an In Vivo Ovarian Cancer Model. Cancers (Basel) 2022; 15:cancers15010237. [PMID: 36612230 PMCID: PMC9818543 DOI: 10.3390/cancers15010237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Ovarian cancer cell dissemination can lead to the mortality of patients with advanced ovarian cancer. Complete surgery for no gross residual disease contributes to a more favorable prognosis than that of patients with residual disease. HCFs have highly regular porous structures and their 3D porous structures act as scaffolds for cell adhesion. HCFs are fabricated from biodegradable polymers and have been widely used in tissue engineering. This study aimed to show that HCFs suppress tumor growth in an in vivo ovarian cancer model. The HCF pore sizes had a significant influence on tumor growth inhibition, and HCFs induced morphological changes that rounded out ovarian cancer cells. Furthermore, we identified gene ontology (GO) terms and clusters of genes downregulated by HCFs. qPCR analysis demonstrated that a honeycomb structure downregulated the expression of CXCL2, FOXC1, MMP14, and SNAI2, which are involved in cell proliferation, migration, invasion, angiogenesis, focal adhesion, extracellular matrix (ECM), and epithelial-mesenchymal transition (EMT). Collectively, HCFs induced abnormal focal adhesion and cell morphological changes, subsequently inhibiting the differentiation, proliferation and motility of ovarian cancer cells. Our data suggest that HCFs could be a novel device for inhibiting residual tumor growth after surgery, and could reduce surgical invasiveness and improve the prognosis for patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Tsuyoshi Ohta
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
- Correspondence: ; Tel.: +81-23-628-5393
| | - Masaru Tanaka
- Institute for Materials Chemistry and Engineering, Kyushu University, CE41 744 Motooka, Nishi, Fukuoka 819-0395, Japan
| | - Seitaro Taki
- Toyoda Gosei Co., Ltd., 1-1, Higashitakasuka, Futatudera, Miwa-cho, Ama-gun, Aichi 490-1207, Japan
| | - Hiroyuki Nakagawa
- Toyoda Gosei Co., Ltd., 1-1, Higashitakasuka, Futatudera, Miwa-cho, Ama-gun, Aichi 490-1207, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
7
|
Pratt J, Haidara K, Annabi B. MT1-MMP Expression Levels and Catalytic Functions Dictate LDL Receptor-Related Protein-1 Ligand Internalization Capacity in U87 Glioblastoma Cells. Int J Mol Sci 2022; 23:14214. [PMID: 36430705 PMCID: PMC9692856 DOI: 10.3390/ijms232214214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Modulations in cell surface receptor ectodomain proteolytic shedding impact on receptor function and cancer biomarker expression. As such, heavily pursued therapeutic avenues have exploited LDL receptor-related protein-1 (LRP-1)-mediated capacity in internalizing Angiopep-2 (An2), a brain-penetrating peptide that allows An2-drug conjugates to cross the blood-brain tumor barrier (BBTB). Given that LRP-1 is proteolytically shed from the cell surface through matrix metalloproteinase (MMP) activity, the balance between MMP expression/function and LRP-1-mediated An2 internalization is unknown. In this study, we found that membrane type-1 (MT1)-MMP expression increased from grade 1 to 4 brain tumors, while that of LRP-1 decreased inversely. MMP pharmacological inhibitors such as Ilomastat, Doxycycline and Actinonin increased in vitro An2 internalization by up to 2.5 fold within a human grade IV-derived U87 glioblastoma cell model. Transient siRNA-mediated MT1-MMP gene silencing resulted in increased basal An2 cell surface binding and intracellular uptake, while recombinant MT1-MMP overexpression reduced both cell surface LRP-1 expression as well as An2 internalization. The addition of Ilomastat to cells overexpressing recombinant MT1-MMP restored LRP-1 expression at the cell surface and An2 uptake to levels comparable to those observed in control cells. Collectively, our data suggest that MT1-MMP expression status dictates An2-mediated internalization processes in part by regulating cell surface LRP-1 functions. Such evidence prompts preclinical evaluations of combined MMP inhibitors/An2-drug conjugate administration to potentially increase the treatment of high-MT1-MMP-expressing brain tumors.
Collapse
Affiliation(s)
| | | | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Centre de Recherche CERMO-FC, Département de Chimie, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
| |
Collapse
|
8
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
9
|
Mitkin NA, Ustiugova AS, Uvarova AN, Rumyantsev KA, Korneev KV, Pavshintsev VV. Serum of Mice Immunized with MT1-MMP Metalloproteinase Reduces Migration Potential of Pancreatic Cancer Cells. Mol Biol 2021. [DOI: 10.1134/s0026893321050095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
10
|
Pezeshkian Z, Nobili S, Peyravian N, Shojaee B, Nazari H, Soleimani H, Asadzadeh-Aghdaei H, Ashrafian Bonab M, Nazemalhosseini-Mojarad E, Mini E. Insights into the Role of Matrix Metalloproteinases in Precancerous Conditions and in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13246226. [PMID: 34944846 PMCID: PMC8699154 DOI: 10.3390/cancers13246226] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Colorectal cancer (CRC) is one of the most common cancer worldwide. CRC is derived from polyps and many factors, such as Matrix Metalloproteinases (MMPs) can gain the progression of colorectal carcinogenesis. Many investigations have indicated the role of MMPs in CRC development while there is not enough knowledge about the function of MMPs in precancerous conditions. This review summarizes the current information about the role of MMPs in polyps and CRC progression. Abstract Colorectal cancer (CRC) is the third and second cancer for incidence and mortality worldwide, respectively, and is becoming prevalent in developing countries. Most CRCs derive from polyps, especially adenomatous polyps, which can gradually transform into CRC. The family of Matrix Metalloproteinases (MMPs) plays a critical role in the initiation and progression of CRC. Prominent MMPs, including MMP-1, MMP-2, MMP-7, MMP-8, MMP-9, MMP-12, MMP-13, MMP-14, and MMP-21, have been detected in CRC patients, and the expression of most of them correlates with a poor prognosis. Moreover, many studies have explored the inhibition of MMPs and targeted therapy for CRC, but there is not enough information about the role of MMPs in polyp malignancy. In this review, we discuss the role of MMPs in colorectal cancer and its pathogenesis
Collapse
Affiliation(s)
- Zahra Pezeshkian
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19835-178, Iran; (Z.P.); (N.P.); (B.S.); (H.A.-A.)
| | - Stefania Nobili
- Department of Neurosciences, Imaging and Clinical Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Center for Advanced Studies and Technology (CAST), University “G. D’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Noshad Peyravian
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19835-178, Iran; (Z.P.); (N.P.); (B.S.); (H.A.-A.)
| | - Bahador Shojaee
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19835-178, Iran; (Z.P.); (N.P.); (B.S.); (H.A.-A.)
| | - Haniye Nazari
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran 19395-1495, Iran;
| | - Hiva Soleimani
- Department of General Biology, Faculty of Fundamental Science, Islamic Azad University of Shahr-E-Qods, Tehran 37515-374, Iran;
| | - Hamid Asadzadeh-Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19835-178, Iran; (Z.P.); (N.P.); (B.S.); (H.A.-A.)
| | - Maziar Ashrafian Bonab
- School of Medicine, University of Sunderland, City Campus, Chester Road, Sunderland SR1 3SD, UK;
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 19835-178, Iran
- Correspondence: (E.N.-M.); (E.M.)
| | - Enrico Mini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
- DENOTHE Excellence Center, University of Florence, 50139 Florence, Italy
- Correspondence: (E.N.-M.); (E.M.)
| |
Collapse
|
11
|
Li M, Li S, Zhou L, Yang L, Wu X, Tang B, Xie S, Fang L, Zheng S, Hong T. Immune Infiltration of MMP14 in Pan Cancer and Its Prognostic Effect on Tumors. Front Oncol 2021; 11:717606. [PMID: 34604053 PMCID: PMC8484967 DOI: 10.3389/fonc.2021.717606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background Matrix metalloproteinase 14 (MMP14) is a member of the MMP family, which interacts with tissue inhibitors of metalloproteinase (TIMPs), and is involved in normal physiological functions such as cell migration, invasion, metastasis, angiogenesis, and proliferation, as well as tumor genesis and progression. However, there has been a lack of relevant reports on the effect of MMP14 across cancers. This study aims to explore the correlation between MMP14 and pan-cancer prognosis, immune infiltration, and the effects of pan-cancer gene mismatch repair (MMR), microsatellite instability (MSI), tumor mutational burden (TMB), DNA methylation, and immune checkpoint genes. Methods In this study, we used bioinformatics to analyze data from multiple databases, including The Cancer Genome Atlas (TCGA), ONCOMINE, and Kaplan–Meier plotter. We investigated the relationship between the expression of MMP14 in tumors and tumor prognosis, the relationship between MMP14 expression and tumor cell immune infiltration, and the relationship between MMR gene MMR, MSI, TMB, DNA methylation, and immune checkpoint genes. Results MMP14 expression is highly associated with the prognosis of a variety of cancers and tumor immune invasion and has important effects on pan oncologic MMR, MSI, TMB, DNA methylation, and immune checkpoint genes. Conclusion MMP14 is highly correlated with tumor prognosis and immune invasion and affects the occurrence and progression of many tumors. All of these results fully indicate that MMP14 may be a biomarker for the prognosis, diagnosis, and treatment of many tumors and provide new ideas and direction for subsequent tumor immune research and treatment strategies.
Collapse
Affiliation(s)
- Minde Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shaoyang Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Le Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shenhao Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Linchun Fang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Suyue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Abooshahab R, Al-Salami H, Dass CR. The increasing role of pigment epithelium-derived factor in metastasis: from biological importance to a promising target. Biochem Pharmacol 2021; 193:114787. [PMID: 34571004 DOI: 10.1016/j.bcp.2021.114787] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a non-inhibitory member of the serpin (serine protease inhibitor) family and is a well-known potent anti-tumor factor in a variety of cancers. It has been ascertained that PEDF regulates multiple metastatic processes through various plausible mechanisms, including inhibiting angiogenesis, inducing apoptosis, stimulating extracellular matrix (ECM) degradation, and suppressing the epithelial-to-mesenchymal transition (EMT) process. Although PEDF has been recognized as an anti-metastatic marker in most studies, its role remains controversial with conflicting reports of PEDF as a metastatic marker. The emerging insights into the mechanism(s) of PEDF in tumor progression and its therapeutic effects are discussed systematically in this review, aiming to improve our understanding in the context of metastasis and drug development.
Collapse
Affiliation(s)
- Raziyeh Abooshahab
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hani Al-Salami
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia.
| |
Collapse
|
13
|
Dynamic Expression of Membrane Type 1-Matrix Metalloproteinase (Mt1-mmp/Mmp14) in the Mouse Embryo. Cells 2021; 10:cells10092448. [PMID: 34572097 PMCID: PMC8465375 DOI: 10.3390/cells10092448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 01/13/2023] Open
Abstract
MT1-MMP/MMP14 belongs to a subgroup of the matrix metalloproteinases family that presents a transmembrane domain, with a cytosolic tail and the catalytic site exposed to the extracellular space. Deficient mice for this enzyme result in early postnatal death and display severe defects in skeletal, muscle and lung development. By using a transgenic line expressing the LacZ reporter under the control of the endogenous Mt1-mmp promoter, we reported a dynamic spatiotemporal expression pattern for Mt1-mmp from early embryonic to perinatal stages during cardiovascular development and brain formation. Thus, Mt1-mmp shows expression in the endocardium of the heart and the truncus arteriosus by E8.5, and is also strongly detected during vascular system development as well as in endothelial cells. In the brain, LacZ reporter expression was detected in the olfactory bulb, the rostral cerebral cortex and the caudal mesencephalic tectum. LacZ-positive cells were observed in neural progenitors of the spinal cord, neural crest cells and the intersomitic region. In the limb, Mt1-mmp expression was restricted to blood vessels, cartilage primordium and muscles. Detection of the enzyme was confirmed by Western blot and immunohistochemical analysis. We suggest novel functions for this metalloproteinase in angiogenesis, endocardial formation and vascularization during organogenesis. Moreover, Mt1-mmp expression revealed that the enzyme may contribute to heart, muscle and brain throughout development.
Collapse
|
14
|
Wang X, Meng Q, Wang Y, Gao Y. Overexpression of MMP14 predicts the poor prognosis in gastric cancer: Meta-analysis and database validation. Medicine (Baltimore) 2021; 100:e26545. [PMID: 34397871 PMCID: PMC8360427 DOI: 10.1097/md.0000000000026545] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 06/14/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Plenty of studies have showed matrix metalloproteinase 14 (MMP14) expression might be associated with the prognosis of gastric cancer (GC). However, no definite conclusion has been obtained for the contradictory results. METHODS We searched PubMed, Web of science, Embase, and Cochrane library for eligible studies. The association between MMP14 expression and prognostic outcomes of GC was evaluated. Hazard ratio (HR) and 95% confidence interval (CI) were integrated to show the effect of MMP14 expression on the overall survival (OS) or recurrence-free survival (RFS). Data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) was used to validate the association of MMP14 expression with OS or RFS in GC. A brief bioinformatics analysis was also performed to determine the prognostic role of MMP14 expression in GC. RESULTS High MMP14 expression was associated with shorter OS compared to low MMP14 expression in GC (HR = 1.95, P < .01). Patients with high MMP14 expression tended to have worse differentiation (P = .03), deeper tumor invasion (P < .01), earlier lymph node metastasis (P < .01), earlier distant metastasis (P < .01) and more advanced clinical stage (P < .01) compared to those with low MMP14 expression. The data from TCGA and GEO showed MMP14 was overexpressed in tumor tissues compared to normal tissues (P < .05), and high MMP14 expression was significantly related to shorter OS (HR = 1.70, 95% CI = 1.32-2.20, P < .01) and RFS (HR = 1.45, 95% CI = 1.15-1.83, P < .01) compared to low MMP14 expression in GC. Expression of MMP14 was linked to functional networks involving the biological process, metabolic process, response to stimulus, cell communication and so on. Functional network analysis suggested that MMP14 regulated the protein digestion and absorption, extracellular matrix receptor interaction, focal adhesion, ribosome, spliceosome, and so on. CONCLUSION High MMP14 expression was associated with worse prognosis of GC compared to low MMP14 expression. MMP14 expression could serve as a prognostic factor and potential therapeutic target of GC.
Collapse
Affiliation(s)
- Xikai Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qinghe Meng
- School of Public Health, Peking University, Beijing, China
| | - Yuanyuan Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Yanlu Gao
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
15
|
Oberska P, Jedrzejczak-Silicka M, Michałek K, Grabowska M. Initial assessment of suitability of MCF-7 and HepG2 cancer cell lines for AQP3 research in cancer biology. Acta Histochem 2021; 123:151716. [PMID: 33933702 DOI: 10.1016/j.acthis.2021.151716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022]
Abstract
Cancer cell lines are widely used as in vitro models to elucidate biological processes in cancer, and as a tool to evaluate anticancer agents. In fact, the use of an appropriate cancer cell line in cancer research is crucial for investigating new, potential factors involved in carcinogenesis. One of them is aquaporin-3 (AQP3), which is a small, hydrophobic, integral membrane protein with a predominant role in water and glycerol transport. Recently, altered expression of AQP3 has been reported in many types of cancer. Increasing evidence strongly suggests that AQP3 plays a key role in cancer cell proliferation, migration and invasion. In this study, we performed an insightful characteristic of AQP3 location and its protein expression in MCF-7 human breast adenocarcinoma and HepG2 hepatocellular carcinoma cell lines in the context of cancer biology using immunocytochemistry, immunofluorescence and Western blot analyses. AQP3 was found to be located in the cell membrane and cytoplasm of MCF-7 cells, and in the cytoplasm and nuclear membrane of HepG2 cells. Immunoblotting of proteins derived from both cell lines revealed a clear band with a molecular weight of approx. 30 kDa representing an unglycosylated form of AQP3. However, the expression of this protein was higher in MCF-7 than in HepG2. Concluding, our results clearly indicated variability in both the expression levels and subcellular location of the AQP3 protein in MCF-7 and HepG2 cell lines. This leads to the possibility that the expression patterns and subcellular location of AQP3 in the tested cancer cell lines are tissue-of-origin specific, and may be related to the aggressiveness of cancer cells and their mobility.
Collapse
Affiliation(s)
- Patrycja Oberska
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270, Szczecin, Poland
| | - Magdalena Jedrzejczak-Silicka
- Laboratory of Cytogenetics, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270, Szczecin, Poland.
| | - Katarzyna Michałek
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270, Szczecin, Poland
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Pomeranian Medical University, Żołnierska 48, 71-210, Szczecin, Poland
| |
Collapse
|
16
|
Cyclooxygenase Inhibition Alters Proliferative, Migratory, and Invasive Properties of Human Glioblastoma Cells In Vitro. Int J Mol Sci 2021; 22:ijms22094297. [PMID: 33919029 PMCID: PMC8122446 DOI: 10.3390/ijms22094297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Prostaglandin E2 (PGE2) is known to increase glioblastoma (GBM) cell proliferation and migration while cyclooxygenase (COX) inhibition decreases proliferation and migration. The present study investigated the effects of COX inhibitors and PGE2 receptor antagonists on GBM cell biology. Cells were grown with inhibitors and dose response, viable cell counting, flow cytometry, cell migration, gene expression, Western blotting, and gelatin zymography studies were performed. The stimulatory effects of PGE2 and the inhibitory effects of ibuprofen (IBP) were confirmed in GBM cells. The EP2 and EP4 receptors were identified as important mediators of the actions of PGE2 in GBM cells. The concomitant inhibition of EP2 and EP4 caused a significant decrease in cell migration which was not reverted by exogenous PGE2. In T98G cells exogenous PGE2 increased latent MMP2 gelatinolytic activity. The inhibition of COX1 or COX2 caused significant alterations in MMP2 expression and gelatinolytic activity in GBM cells. These findings provide further evidence for the importance of PGE2 signalling through the EP2 and the EP4 receptor in the control of GBM cell biology. They also support the hypothesis that a relationship exists between COX1 and MMP2 in GBM cells which merits further investigation as a novel therapeutic target for drug development.
Collapse
|
17
|
Development of a LC-MS/MS method for the quantification of toxic payload DM1 cleaved from BT1718 in a Phase I study. Bioanalysis 2021; 13:101-113. [PMID: 33496610 DOI: 10.4155/bio-2020-0256] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: BT1718 is a novel bicyclic peptide anticancer drug targeting membrane type I matrix metalloproteinase to release its toxic payload DM1. A LC-MS/MS method was validated to quantify DM1 generated from BT1718 in a Phase I/IIa clinical trial. Materials & methods: Plasma samples underwent a reduction reaction to artificially cleave BT1718 into DM1 and its bicycle components. An alkylation step was carried out to stabilize the reaction products, and plasma proteins extracted using acetonitrile. LC-MS/MS analysis utilized a C18 column and Agilent 6460 triple quadrupole mass spectrometer (Agilent, Cheshire, UK). Results: The method was fully validated over a linear range of 200-50,000 ng/ml BT1718, with overall precision ≤10% and accuracy 89-102%. Conclusion: A novel method for quantifying DM1 yielded from BT1718 has been validated and is now being utilized clinically.
Collapse
|
18
|
Deng F, Mu J, Qu C, Yang F, Liu X, Zeng X, Peng X. A Novel Prognostic Model of Endometrial Carcinoma Based on Clinical Variables and Oncogenomic Gene Signature. Front Mol Biosci 2021; 7:587822. [PMID: 33490103 PMCID: PMC7817972 DOI: 10.3389/fmolb.2020.587822] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Due to the difficulty in predicting the prognosis of endometrial carcinoma (EC) patients by clinical variables alone, this study aims to build a new EC prognosis model integrating clinical and molecular information, so as to improve the accuracy of predicting the prognosis of EC. The clinical and gene expression data of 496 EC patients in the TCGA database were used to establish and validate this model. General Cox regression was applied to analyze clinical variables and RNAs. Elastic net-penalized Cox proportional hazard regression was employed to select the best EC prognosis-related RNAs, and ridge regression was used to construct the EC prognostic model. The predictive ability of the prognostic model was evaluated by the Kaplan-Meier curve and the area under the receiver operating characteristic curve (AUC-ROC). A clinical-RNA prognostic model integrating two clinical variables and 28 RNAs was established. The 5-year AUC of the clinical-RNA prognostic model was 0.932, which is higher than that of the clinical-alone (0.897) or RNA-alone prognostic model (0.836). This clinical-RNA prognostic model can better classify the prognosis risk of EC patients. In the training group (396 patients), the overall survival of EC patients was lower in the high-risk group than in the low-risk group [HR = 32.263, (95% CI, 7.707-135.058), P = 8e-14]. The same comparison result was also observed for the validation group. A novel EC prognosis model integrating clinical variables and RNAs was established, which can better predict the prognosis and help to improve the clinical management of EC patients.
Collapse
Affiliation(s)
- Fang Deng
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jing Mu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Chiwen Qu
- School of Mathematics and Statistics, Hunan Normal University, Changsha, China
| | - Fang Yang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xing Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiaomin Zeng
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiaoning Peng
- School of Mathematics and Statistics, Hunan Normal University, Changsha, China.,Department of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Department of Pathophysiology, Jishou University School of Medicine, Jishou, China
| |
Collapse
|
19
|
Niland S, Eble JA. Hold on or Cut? Integrin- and MMP-Mediated Cell-Matrix Interactions in the Tumor Microenvironment. Int J Mol Sci 2020; 22:ijms22010238. [PMID: 33379400 PMCID: PMC7794804 DOI: 10.3390/ijms22010238] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) has become the focus of interest in cancer research and treatment. It includes the extracellular matrix (ECM) and ECM-modifying enzymes that are secreted by cancer and neighboring cells. The ECM serves both to anchor the tumor cells embedded in it and as a means of communication between the various cellular and non-cellular components of the TME. The cells of the TME modify their surrounding cancer-characteristic ECM. This in turn provides feedback to them via cellular receptors, thereby regulating, together with cytokines and exosomes, differentiation processes as well as tumor progression and spread. Matrix remodeling is accomplished by altering the repertoire of ECM components and by biophysical changes in stiffness and tension caused by ECM-crosslinking and ECM-degrading enzymes, in particular matrix metalloproteinases (MMPs). These can degrade ECM barriers or, by partial proteolysis, release soluble ECM fragments called matrikines, which influence cells inside and outside the TME. This review examines the changes in the ECM of the TME and the interaction between cells and the ECM, with a particular focus on MMPs.
Collapse
|
20
|
Raeeszadeh-Sarmazdeh M, Do LD, Hritz BG. Metalloproteinases and Their Inhibitors: Potential for the Development of New Therapeutics. Cells 2020; 9:E1313. [PMID: 32466129 PMCID: PMC7290391 DOI: 10.3390/cells9051313] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
The metalloproteinase (MP) family of zinc-dependent proteases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) plays a crucial role in the extracellular matrix (ECM) remodeling and degradation activities. A wide range of substrates of the MP family includes ECM components, chemokines, cell receptors, and growth factors. Metalloproteinases activities are tightly regulated by proteolytic activation and inhibition via their natural inhibitors, tissue inhibitors of metalloproteinases (TIMPs), and the imbalance of the activation and inhibition is responsible in progression or inhibition of several diseases, e.g., cancer, neurological disorders, and cardiovascular diseases. We provide an overview of the structure, function, and the multifaceted role of MMPs, ADAMs, and TIMPs in several diseases via their cellular functions such as proteolysis of other cell signaling factors, degradation and remodeling of the ECM, and other essential protease-independent interactions in the ECM. The significance of MP inhibitors targeting specific MMP or ADAMs with high selectivity is also discussed. Recent advances and techniques used in developing novel MP inhibitors and MP responsive drug delivery tools are also reviewed.
Collapse
Affiliation(s)
- Maryam Raeeszadeh-Sarmazdeh
- Chemical and Materials Engineering Department, University of Nevada, Reno, NV 89557, USA; (L.D.D.); (B.G.H.)
| | | | | |
Collapse
|
21
|
Ji X, Xie S, Jiao Y, Zhang X, Sun D, Yang VC, Wang M, He H, Sun L. MT1-MMP activatable fluorogenic probes with enhanced specificity via high-affinity peptide conjugation for tumor imaging. Biomater Sci 2020; 8:2308-2317. [PMID: 32186291 DOI: 10.1039/c9bm02007a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Overlapping substrate specificities within the family of matrix metalloproteinases (MMPs), usually caused by their highly conserved structural topology, increase the potential for a substrate to be cleaved by multiple enzymes within this family, which leads to the decrease in the selectivity of MMP substrate-based probes. To resolve this issue, MT1-MMP activatable fluorogenic probes for tumor detection with enhanced specificity were developed by combining a fluorescence resonance energy transfer (FRET) peptide substrate and its specific binding peptide with different lengths of linkers. The specificity of the probes increased profiting from the high affinity of the MT1-MMP specific binding peptide while keeping the ability to amplify the output imaging signals in response to MMP activity with the FRET substrate. Enzyme kinetics analysis clearly demonstrated that the conjugation of P-1 and MT1-AF7p enhanced both the specificity and selectivity of the fluorogenic probes for MT1-MMP, and introducing a linker composed of 12 PEG subunits into these two fragments led to optimized specificity and selectivity of the fluorogenic probe for MT1-MMP. Both in vitro and in vivo results revealed that the imaging probe with the linker composed of 12 PEG subunits based on our designed strategy could be effectively applied for MT1-MMP positive tumor imaging. Since this strategy for enhancing the specificity of protease sensing probes can be applied to other proteases and is not just limited to MT1-MMP, it is an appealing platform to achieve selective tumor imaging.
Collapse
Affiliation(s)
- Xiuru Ji
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
The Expanding Role of MT1-MMP in Cancer Progression. Pharmaceuticals (Basel) 2019; 12:ph12020077. [PMID: 31137480 PMCID: PMC6630478 DOI: 10.3390/ph12020077] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
For over 20 years, membrane type 1 matrix metalloproteinase (MT1-MMP) has been recognized as a key component in cancer progression. Initially, the primary roles assigned to MT1-MMP were the activation of proMMP-2 and degradation of fibrillar collagen. Proteomics has revealed a great array of MT1-MMP substrates, and MT1-MMP selective inhibitors have allowed for a more complete mapping of MT1-MMP biological functions. MT1-MMP has extensive sheddase activities, is both a positive and negative regulator of angiogenesis, can act intracellularly and as a transcription factor, and modulates immune responses. We presently examine the multi-faceted role of MT1-MMP in cancer, with a consideration of how the diversity of MT1-MMP behaviors impacts the application of MT1-MMP inhibitors.
Collapse
|
23
|
Han KY, Chang JH, Azar DT. MMP14-Containing Exosomes Cleave VEGFR1 and Promote VEGFA-Induced Migration and Proliferation of Vascular Endothelial Cells. Invest Ophthalmol Vis Sci 2019; 60:2321-2329. [PMID: 31117124 PMCID: PMC6532701 DOI: 10.1167/iovs.18-26277] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose Investigate the impact matrix metalloproteinase 14 (MMP14) delivered via exosomes produced by corneal fibroblasts on vascular endothelial growth factor receptor 1 (VEGFR1) cleavage on endothelial cells, and other key processes of angiogenesis. Methods Proteolysis of VEGFR1 and R2 by the catalytic domain of MMP14 was investigated via immunocytochemistry with anti-VEGFR1, anti-VEGFR2, and anti-MMP14 antibodies. Exosomes were isolated via precipitation and serial ultracentrifugation from wild-type (WT) and MMP14 exon4-deficient corneal fibroblasts. Transmission electron microscopy and nanotracking analysis were used to characterize the isolated exosomes. The presence of MMP14 in exosomes from WT fibroblasts was confirmed by Western blotting. VEGFR1 cleavage upon treatment with WT-derived exosomes, Δexon4-derived exosomes, or the pan-MMP inhibitor GM60001 was examined via in vitro proteolysis analysis using recombinant mouse (rm) VEGFR1/R2. Endothelial cell migration and proliferation were investigated using a Boyden chamber assay and BrdU incorporation, respectively. Results WT-derived exosomes specifically cleaved rmVEGFR1 in vitro, whereas Δexon4-derived exosomes did not. Treatment with the pan-MMP inhibitor GM6001 effectively inhibited VEGFR1 cleavage by WT-derived exosomes, confirming the role of MMP14 in this cleavage. WT-derived exosomes induced greater endothelial cell migration (P < 0.01) and proliferation (P < 0.5) compared to Δexon4-derived exosomes. Conclusions MMP14-containing exosomes may be involved in the regulation of corneal neovascularization through degradation of VEGFR1 and VEGFA-induced endothelial cell proliferation and migration.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States
| |
Collapse
|
24
|
Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis 2019; 36:171-198. [PMID: 30972526 DOI: 10.1007/s10585-019-09966-1] [Citation(s) in RCA: 367] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) constitutes the scaffold of tissues and organs. It is a complex network of extracellular proteins, proteoglycans and glycoproteins, which form supramolecular aggregates, such as fibrils and sheet-like networks. In addition to its biochemical composition, including the covalent intermolecular cross-linkages, the ECM is also characterized by its biophysical parameters, such as topography, molecular density, stiffness/rigidity and tension. Taking these biochemical and biophysical parameters into consideration, the ECM is very versatile and undergoes constant remodeling. This review focusses on this remodeling of the ECM under the influence of a primary solid tumor mass. Within this tumor stroma, not only the cancer cells but also the resident fibroblasts, which differentiate into cancer-associated fibroblasts (CAFs), modify the ECM. Growth factors and chemokines, which are tethered to and released from the ECM, as well as metabolic changes of the cells within the tumor bulk, add to the tumor-supporting tumor microenvironment. Metastasizing cancer cells from a primary tumor mass infiltrate into the ECM, which variably may facilitate cancer cell migration or act as barrier, which has to be proteolytically breached by the infiltrating tumor cell. The biochemical and biophysical properties therefore determine the rates and routes of metastatic dissemination. Moreover, primed by soluble factors of the primary tumor, the ECM of distant organs may be remodeled in a way to facilitate the engraftment of metastasizing cancer cells. Such premetastatic niches are responsible for the organotropic preference of certain cancer entities to colonize at certain sites in distant organs and to establish a metastasis. Translational application of our knowledge about the cancer-primed ECM is sparse with respect to therapeutic approaches, whereas tumor-induced ECM alterations such as increased tissue stiffness and desmoplasia, as well as breaching the basement membrane are hallmark of malignancy and diagnostically and histologically harnessed.
Collapse
Affiliation(s)
- Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| |
Collapse
|
25
|
Li B, Lou G, Zhou J. MT1‑MMP promotes the proliferation and invasion of gastric carcinoma cells via regulating vimentin and E‑cadherin. Mol Med Rep 2019; 19:2519-2526. [PMID: 30720114 PMCID: PMC6423635 DOI: 10.3892/mmr.2019.9918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to explore the possible effects of membrane‑type 1 matrix metalloproteinase (MT1‑MMP) on gastric carcinoma cells proliferation and invasion. Immunohistochemistry analysis was conducted to measure MT1‑MMP expression level in 15 patients with gastric carcinoma. Subsequently, recombinant short hairpin RNA (shRNA) vectors targeting MT1‑MMP were constructed to silence the expression of MT1‑MMP in gastric carcinoma cells. Then, the inhibitive efficiency was verified via reverse transcription quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis. The effects of MT1‑MMP silencing on cell proliferation and invasion were detected through Cell Counting Kit‑8 test and Transwell assays. The expression levels of vimentin and epithelial cadherin (E‑cadherin) were detected by RT‑qPCR. The immunohistochemistry analysis results revealed that MT1‑MMP expression in gastric carcinoma tissues was markedly overexpressed compared with non‑cancerous adjacent tissues. The MT1‑MMP expression level in cancer‑derived cell line AGS cells was also significantly increased compared with that in non‑cancer‑derived GES‑1 cells. In addition, the MT1‑MMP expression level in AGS cells was significantly decreased via shRNA transfection. Cell proliferation and invasion were markedly inhibited following knockdown of MT1‑MMP level in AGS cells. These inhibitory effects were associated with the decreased expression of vimentin and increased expression of E‑cadherin. MT1‑MMP was overexpressed in gastric carcinoma cells, and silencing of MT1‑MMP inhibited the proliferation and invasion of cells via regulating the expression of vimentin and E‑cadherin.
Collapse
Affiliation(s)
- Bo Li
- Department of Radiotherapy, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Guochun Lou
- Department of Gastroenterology, Zhejiang Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Juying Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
26
|
MT4-MMP: The GPI-Anchored Membrane-Type Matrix Metalloprotease with Multiple Functions in Diseases. Int J Mol Sci 2019; 20:ijms20020354. [PMID: 30654475 PMCID: PMC6359745 DOI: 10.3390/ijms20020354] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 01/02/2023] Open
Abstract
MT4-MMP (or MMP17) belongs to the Membrane-Type Matrix Metalloproteinase (MT-MMP) family. This family of proteases contributes to extracellular matrix remodeling during several physiological processes, including embryogenesis, organogenesis, tissue regeneration, angiogenesis, wound healing, and inflammation. MT4-MMP (MMP17) presents unique characteristics compared to other members of the family in terms of sequence homology, substrate specificity, and internalization mode, suggesting distinct physiological and pathological functions. While the physiological functions of MT4-MMP are poorly understood, it has been involved in different pathological processes such as arthritis, cardiovascular disease, and cancer progression. The mt4-mmp transcript has been detected in a large diversity of cancers. The contribution of MT4-MMP to tumor development has been further investigated in gastric cancer, colon cancer, head and neck cancer, and more deeply in breast cancer. Given its contribution to different pathologies, particularly cancers, MT4-MMP represents an interesting therapeutic target. In this review, we examine its biological and structural properties, and we propose an overview of its physiological and pathological functions.
Collapse
|
27
|
Byeon HK, Ku M, Yang J. Beyond EGFR inhibition: multilateral combat strategies to stop the progression of head and neck cancer. Exp Mol Med 2019; 51:1-14. [PMID: 30700700 PMCID: PMC6353966 DOI: 10.1038/s12276-018-0202-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/03/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) overexpression is common in head and neck squamous cell carcinoma. Targeted therapy specifically directed towards EGFR has been an area of keen interest in head and neck cancer research, as EGFR is potentially an integration point for convergent signaling. Despite the latest advancements in cancer diagnostics and therapeutics against EGFR, the survival rates of patients with advanced head and neck cancer remain disappointing due to anti-EGFR resistance. This review article will discuss recent multilateral efforts to discover and validate actionable strategies that involve signaling pathways in heterogenous head and neck cancer and to overcome anti-EGFR resistance in the era of precision medicine. Particularly, this review will discuss in detail the issue of cancer metabolism, which has recently emerged as a novel mechanism by which head and neck cancer may be successfully controlled according to different perspectives. South Korean researchers propose novel combination strategies for overcoming drug resistance and halting the progression of head and neck cancer (HNC). Although high levels of epidermal growth factor receptor (EGFR) protein in HNC correlate with reduced survival, patients’ response to the EGFR inhibitor cetuximab often declines rapidly after a short period of effectiveness. Hyung Kwon Byeon at Korea University College of Medicine in Seoul and colleagues review current knowledge of the mechanisms underlying cetuximab resistance. They suggest that evaluating a patient’s genetic profile and combining cetuximab with drugs that enhance the effects of inhibiting EGFR signaling pathways (with inhibitors of other EGFR family members or proteins that mediate EGFR entry to the cell nucleus, for example) as well as with agents that inhibit cancer cell metabolism could be a more effective approach for treating HNC.
Collapse
Affiliation(s)
- Hyung Kwon Byeon
- Department of Otorhinolaryngology-Head and Neck Surgery, Soonchunhyang University College of Medicine, Seoul, Republic of Korea. .,Systems Molecular Oncology for Head and Neck Cancer, Seoul, Republic of Korea. .,Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea.
| | - Minhee Ku
- Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea.,Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Research Institute of Radiological Science, Yonsei University, Seoul, Republic of Korea
| | - Jaemoon Yang
- Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea. .,Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Research Institute of Radiological Science, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Pahwa S, Bhowmick M, Amar S, Cao J, Strongin AY, Fridman R, Weiss SJ, Fields GB. Characterization and regulation of MT1-MMP cell surface-associated activity. Chem Biol Drug Des 2018; 93:1251-1264. [PMID: 30480376 DOI: 10.1111/cbdd.13450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/18/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022]
Abstract
Quantitative assessment of MT1-MMP cell surface-associated proteolytic activity remains undefined. Presently, MT1-MMP was stably expressed and a cell-based FRET assay developed to quantify activity toward synthetic collagen-model triple-helices. To estimate the importance of cell surface localization and specific structural domains on MT1-MMP proteolysis, activity measurements were performed using a series of membrane-anchored MT1-MMP mutants and compared directly with those of soluble MT1-MMP. MT1-MMP activity (kcat /KM ) on the cell surface was 4.8-fold lower compared with soluble MT1-MMP, with the effect largely manifested in kcat . Deletion of the MT1-MMP cytoplasmic tail enhanced cell surface activity, with both kcat and KM values affected, while deletion of the hemopexin-like domain negatively impacted KM and increased kcat . Overall, cell surface localization of MT1-MMP restricts substrate binding and protein-coupled motions (based on changes in both kcat and KM ) for catalysis. Comparison of soluble and cell surface-bound MT2-MMP revealed 12.9-fold lower activity on the cell surface. The cell-based assay was utilized for small molecule and triple-helical transition state analog MMP inhibitors, which were found to function similarly in solution and at the cell surface. These studies provide the first quantitative assessments of MT1-MMP activity and inhibition in the native cellular environment of the enzyme.
Collapse
Affiliation(s)
- Sonia Pahwa
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Manishabrata Bhowmick
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Sabrina Amar
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida
| | - Jian Cao
- Departments of Medicine/Cancer Prevention and Pathology, Stony Brook University, Stony Brook, New York
| | - Alex Y Strongin
- Cancer Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, California
| | - Rafael Fridman
- Department of Pathology and the Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Stephen J Weiss
- Division of Molecular Medicine & Genetics, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Gregg B Fields
- Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, Florida.,The Scripps Research Institute/Scripps Florida, Jupiter, Florida
| |
Collapse
|
29
|
Qiang L, Cao H, Chen J, Weller SG, Krueger EW, Zhang L, Razidlo GL, McNiven MA. Pancreatic tumor cell metastasis is restricted by MT1-MMP binding protein MTCBP-1. J Cell Biol 2018; 218:317-332. [PMID: 30487181 PMCID: PMC6314558 DOI: 10.1083/jcb.201802032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 09/28/2018] [Accepted: 10/29/2018] [Indexed: 12/17/2022] Open
Abstract
Tumor cells utilize invadopodia to remodel the surrounding stroma during metastatic invasion. Qiang et al. demonstrate that MTCBP-1 significantly attenuates invadopodia formation and function by binding MT1-MMP and preventing the interaction of MT1-MMP with the actin cytoskeleton. The process by which tumor cells mechanically invade through surrounding stroma into peripheral tissues is an essential component of metastatic dissemination. The directed recruitment of the metalloproteinase MT1-MMP to invadopodia plays a critical role in this invasive process. Here, we provide mechanistic insight into MT1-MMP cytoplasmic tail binding protein 1 (MTCBP-1) with respect to invadopodia formation, matrix remodeling, and invasion by pancreatic tumor cells. MTCBP-1 localizes to invadopodia and interacts with MT1-MMP. We find that this interaction displaces MT1-MMP from invadopodia, thereby attenuating their number and function and reducing the capacity of tumor cells to degrade matrix. Further, we observe an inverse correlation between MTCBP-1 and MT1-MMP expression both in cultured cell lines and human pancreatic tumors. Consistently, MTCBP-1–expressing cells show decreased ability to invade in vitro and metastasize in vivo. These findings implicate MTCBP-1 as an inhibitor of the metastatic process.
Collapse
Affiliation(s)
- Li Qiang
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
| | - Hong Cao
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Jing Chen
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Shaun G Weller
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Eugene W Krueger
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Lizhi Zhang
- Department of Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Gina L Razidlo
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN.,Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Mark A McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN .,Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
30
|
Zhang X, Liu R, Yuan Q, Gao F, Li J, Zhang Y, Zhao Y, Chai Z, Gao L, Gao X. The Precise Diagnosis of Cancer Invasion/Metastasis via 2D Laser Ablation Mass Mapping of Metalloproteinase in Primary Cancer Tissue. ACS NANO 2018; 12:11139-11151. [PMID: 30359513 DOI: 10.1021/acsnano.8b05584] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cancer invasion and metastasis remain the major causes of over 90% of patient deaths. Molecular imaging methods such as computed tomography (CT)/magnetic resonance imaging (MRI) can precisely assess primary regional lymph node invasion and distant organ metastasis via body scanning; however, such diagnostic methods are often utilized too late for cancer therapy. To date, pathologic methods mainly provide information on differentiation/proliferation and potential drug therapy biomarkers of primary tumors rather than precisely reveal tumor regional invasion and distant metastasis in the body. We hypothesized that quantification of membrane type-1 matrix metalloproteinase (MT1-MMP) levels in primary tumor tissue will provide a precise assessment of tumor regional lymph node invasion and remote organ metastasis. In this work, we developed peptide-coated Au clusters with intrinsic red fluorescence and a specific mass signal. When these clusters labeled MT1-MMP in tumor tissue sections derived from the xenograft lung carcinoma model, human lung carcinoma and human renal carcinoma, we could directly observe MT1-MMP via optical fluorescence microscopy and quantitatively detect the MT1-MMP expression level via laser ablation inductively coupled plasma mass spectrometry 2D mapping (2D-LA-Mass Mapping). By observing and quantifying the MT1-MMP expression level in primary human lung carcinoma and human renal carcinoma tissue sections, we precisely assessed the risk of primary tumor invasion/metastasis. Importantly, the accuracy of this pathologic method was verified by CT/MRI molecular imaging of cancer patients and traditional hematoxylin and eosin (H&E) staining/immunohistochemistry (IHC)/immunofluorescence (IF) pathologic studies of primary tumor tissues.
Collapse
Affiliation(s)
- Xiangchun Zhang
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Ru Liu
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Qing Yuan
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Fuping Gao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Jiaojiao Li
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Ya Zhang
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Yuliang Zhao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Zhifang Chai
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Liang Gao
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| | - Xueyun Gao
- Department of Chemistry and Chemical Engineering , Beijing University of Technology , Beijing 100124 , China
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
31
|
MT1-MMP as a PET Imaging Biomarker for Pancreas Cancer Management. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8382148. [PMID: 30224904 PMCID: PMC6129362 DOI: 10.1155/2018/8382148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/25/2018] [Indexed: 01/02/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be one of the deadliest cancers for which optimal diagnostic tools are still greatly needed. Identification of PDAC-specific molecular markers would be extremely useful to improve disease diagnosis and follow-up. MT1-MMP has long been involved in pancreatic cancer, especially in tumour invasion and metastasis. In this study, we aim to ascertain the suitability of MT1-MMP as a biomarker for positron emission tomography (PET) imaging. Two probes were assessed and compared for this purpose, an MT1-MMP-specific binding peptide (MT1-AF7p) and a specific antibody (LEM2/15), labelled, respectively, with 68Ga and with 89Zr. PET imaging with both probes was conducted in patient-derived xenograft (PDX), subcutaneous and orthotopic, PDAC mouse models, and in a cancer cell line (CAPAN-2)-derived xenograft (CDX) model. Both radiolabelled tracers were successful in identifying, by means of PET imaging techniques, tumour tissues expressing MT1-MMP although they did so at different uptake levels. The 89Zr-DFO-LEM2/15 probe showed greater specific activity compared to the 68Ga-labelled peptide. The mean value of tumour uptake for the 89Zr-DFO-LEM2/15 probe (5.67 ± 1.11%ID/g, n=28) was 25-30 times higher than that of the 68Ga-DOTA-AF7p ones. Tumour/blood ratios (1.13 ± 0.51 and 1.44 ± 0.43 at 5 and 7 days of 89Zr-DFO-LEM2/15 after injection) were higher than those estimated for 68Ga-DOTA-AF7p probes (of approximately tumour/blood ratio = 0.5 at 90 min after injection). Our findings strongly point out that (i) the in vivo detection of MT1-MMP by PET imaging is a promising strategy for PDAC diagnosis and (ii) labelled LEM2/15 antibody is a better candidate than MT1-AF7p for PDAC detection.
Collapse
|
32
|
Cui G, Cai F, Ding Z, Gao L. MMP14 predicts a poor prognosis in patients with colorectal cancer. Hum Pathol 2018; 83:36-42. [PMID: 30120968 DOI: 10.1016/j.humpath.2018.03.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/07/2018] [Accepted: 03/19/2018] [Indexed: 10/28/2022]
Abstract
Matrix metalloproteinases (MMPs) are involved in most biological processes. Recently, MMP14 was reported to be up-regulated in some types of cancer and to promote cancer cell invasion and metastasis. However, there are few reports on the clinical significance of MMP14 in colorectal cancer (CRC). In this study, MMP14 expression was first investigated in The Cancer Genome Atlas (TCGA) and whole-genome expression microarray (GEO; Accession Number GSE39582) and then validated with our database. Univariate and multivariate analyses were performed to assess the association between prognostic factors and survival outcomes. MMP14 was upregulated at both the transcriptional and protein levels in cancer compared with normal tissues (P < .05), and high MMP14 expression was associated with advanced tumor stage in the 3 study cohorts. In the univariate Cox proportional hazard ratio analysis, MMP14 correlated significantly with prognosis in both the TCGA and GSE39582 databases (P < .05). In the validation cohort, patients with high MMP14 expression had lower 5-year disease-free survival (DFS; hazard ratio [HR] 6.707; 95% confidence interval [CI] 3.184, 14.128; P < .001) and overall survival (OS; HR 10.669; 95% CI 3.828, 29.737; P < .001) than those with low MMP14 expression. Multivariate survival analysis showed that MMP14 was an independent prognostic marker for both DFS (HR 5.776; 95% CI 2.719, 12.270; P < .001) and OS (HR 8.971; 95% CI 3.199, 25.156; P < .001). Clearly, MMP14 plays an important role in CRC progression and prognosis and could be a useful biomarker for prediction of survival after colectomy.
Collapse
Affiliation(s)
- Guangfei Cui
- Department of Gastrointestinal Surgery, The First People's Hospital of Shangqiu, Shangqiu 476100, Henan Province, China
| | - Feng Cai
- Department of Gastrointestinal Surgery, The First People's Hospital of Shangqiu, Shangqiu 476100, Henan Province, China
| | - Zhanwei Ding
- Department of Gastrointestinal Surgery, The First People's Hospital of Shangqiu, Shangqiu 476100, Henan Province, China
| | - Ling Gao
- Department of Gastrointestinal Surgery, The First People's Hospital of Shangqiu, Shangqiu 476100, Henan Province, China.
| |
Collapse
|
33
|
Arkadash V, Radisky ES, Papo N. Combinatorial engineering of N-TIMP2 variants that selectively inhibit MMP9 and MMP14 function in the cell. Oncotarget 2018; 9:32036-32053. [PMID: 30174795 PMCID: PMC6112833 DOI: 10.18632/oncotarget.25885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/21/2018] [Indexed: 12/21/2022] Open
Abstract
Developing selective inhibitors for proteolytic enzymes that share high sequence homology and structural similarity is important for achieving high target affinity and functional specificity. Here, we used a combination of yeast surface display and dual-color selective library screening to obtain selective inhibitors for each of the matrix metalloproteinases (MMPs) MMP14 and MMP9 by modifying the non-specific N-terminal domain of the tissue inhibitor of metalloproteinase-2 (N-TIMP2). We generated inhibitor variants with 30- to 1175-fold improved specificity to each of the proteases, respectively, relative to wild type N-TIMP2. These biochemical results accurately predicted the selectivity and specificity obtained in cell-based assays. In U87MG cells, the activation of MMP2 by MMP14 was inhibited by MMP14-selective blockers but not MMP9-specific inhibitors. Target specificity was also demonstrated in MCF-7 cells stably expressing either MMP14 or MMP9, with only the MMP14-specific inhibitors preventing the mobility of MMP14-expressing cells. Similarly, the mobility of MMP9-expressing cells was inhibited by the MMP9-specific inhibitors, yet was not altered by the MMP14-specific inhibitors. The strategy developed in this study for improving the specificity of an otherwise broad-spectrum inhibitor will likely enhance our understanding of the basis for target specificity of inhibitors to proteolytic enzymes, in general, and to MMPs, in particular. We, moreover, envision that this study could serve as a platform for the development of next-generation, target-specific therapeutic agents. Finally, our methodology can be extended to other classes of proteolytic enzymes and other important target proteins.
Collapse
Affiliation(s)
- Valeria Arkadash
- Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA
| | - Niv Papo
- Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
34
|
Zhao P, Lan F, Zhang H, Zeng G, Liu D. Down-regulation of KIF2A inhibits gastric cancer cell invasion via suppressing MT1-MMP. Clin Exp Pharmacol Physiol 2018; 45:1010-1018. [PMID: 29781531 DOI: 10.1111/1440-1681.12974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/15/2022]
Abstract
Gastric cancer accounts for a sizeable proportion of global cancer mortality with high morbidity and poor prognosis. Kinesin superfamily proteins (KIFs) are microtubule-dependent motor proteins that function as oncogenes in cancer cells, it has been discovered in recent years. Kinesin family member 2a (KIF2A), a member of the KIFs, has received attention for its role in carcinogenesis and its prognostic value in several human cancers such as breast cancer, colorectal cancer, and squamous cell carcinoma. However, the role of KIF2A in human gastric cancer remains unknown. In this study we aimed to explore the expression and biological functions of KIF2A in human gastric cancer cells, as well as to reveal its potential action mechanism. First, we found that KIF2A was markedly increased in gastric cancer cells (MKN-28, MKN-45, NCI-N87 and SGC-7901) compared to normal gastric mucosa epithelial cells (GES-1). Then KIF2A was successfully silenced in MKN-45 and SGC-7901 cells to facilitate further research into its function. We discovered that KIF2A silencing can significantly inhibit the growth and invasion of MKN-45 and SGC-7901 cells in a time-independent manner, accompanying a decreased expression of Membrane type 1-matrix metalloproteinase (MT1-MMP). When MT1-MMP was reintroduced into MKN-45 and SGC-7901 cells in the KIF2A-siRNA group, only invasion inhibition effects on MKN-45 and SGC-7901 cells induced by KIF2A silencing can be reversed. In conclusion, our study reveals that down-regulation of KIF2A can inhibit gastric cancer cell invasion by suppressing MT1-MMP.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Fei Lan
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Hui Zhang
- Department of Internal Medicine, Jingyang County Hospital, Shaanxi, Xianyang, China
| | - Guangwei Zeng
- Department of Cardiovascular Medicine, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| | - Dong Liu
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, China
| |
Collapse
|
35
|
Decaneto E, Vasilevskaya T, Kutin Y, Ogata H, Grossman M, Sagi I, Havenith M, Lubitz W, Thiel W, Cox N. Solvent water interactions within the active site of the membrane type I matrix metalloproteinase. Phys Chem Chem Phys 2018; 19:30316-30331. [PMID: 28951896 DOI: 10.1039/c7cp05572b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Matrix metalloproteinases (MMP) are an important family of proteases which catalyze the degradation of extracellular matrix components. While the mechanism of peptide cleavage is well established, the process of enzyme regeneration, which represents the rate limiting step of the catalytic cycle, remains unresolved. This step involves the loss of the newly formed N-terminus (amine) and C-terminus (carboxylate) protein fragments from the site of catalysis coupled with the inclusion of one or more solvent waters. Here we report a novel crystal structure of membrane type I MMP (MT1-MMP or MMP-14), which includes a small peptide bound at the catalytic Zn site via its C-terminus. This structure models the initial product state formed immediately after peptide cleavage but before the final proton transfer to the bound amine; the amine is not present in our system and as such proton transfer cannot occur. Modeling of the protein, including earlier structural data of Bertini and coworkers [I. Bertini, et al., Angew. Chem., Int. Ed., 2006, 45, 7952-7955], suggests that the C-terminus of the peptide is positioned to form an H-bond network to the amine site, which is mediated by a single oxygen of the functionally important Glu240 residue, facilitating efficient proton transfer. Additional quantum chemical calculations complemented with magneto-optical and magnetic resonance spectroscopies clarify the role of two additional, non-catalytic first coordination sphere waters identified in the crystal structure. One of these auxiliary waters acts to stabilize key intermediates of the reaction, while the second is proposed to facilitate C-fragment release, triggered by protonation of the amine. Together these results complete the enzymatic cycle of MMPs and provide new design criteria for inhibitors with improved efficacy.
Collapse
Affiliation(s)
- Elena Decaneto
- Max Planck Institute for Chemical Energy Conversion, Stiftstraße. 34-36, D-45470, Mülheim an der Ruhr, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lu Z, Chen Z, Li Y, Wang J, Zhang Z, Che Y, Huang J, Sun S, Mao S, Lei Y, Gao Y, He J. TGF-β-induced NKILA inhibits ESCC cell migration and invasion through NF-κB/MMP14 signaling. J Mol Med (Berl) 2018; 96:301-313. [PMID: 29379981 PMCID: PMC5859688 DOI: 10.1007/s00109-018-1621-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/27/2017] [Accepted: 01/15/2018] [Indexed: 12/18/2022]
Abstract
The transforming growth factor β (TGF-β) signaling pathway plays anti- and pro-tumoral roles in the vast majority of cancers, and long noncoding RNAs have been reported to play key roles in the highly contextual response process. However, the roles of long noncoding RNAs (lncRNAs) in TGF-β signaling in esophageal squamous cell carcinoma (ESCC) remain unknown. In this study, we performed RNA-seq to compare lncRNAs expression levels between TGF-β1-treated and untreated ESCC cells and observed that NF-kappaB-interacting lncRNA (NKILA) was remarkably upregulated by the classical TGF-β signaling pathway. RNA profiling of 39 pairs ESCC tumor and adjacent nontumor samples using RT-qPCR demonstrated that NKILA is significantly downregulated in ESCC tumor tissues, and NKILA expression levels were significantly decreased in advanced tumor tissues (III and IV) compared to early stages (I and II) (p < 0.01). Gain- and loss-of-function assays showed that NKILA inhibited ESCC cell metastasis in vitro and in vivo, and mechanism studies showed that NKILA repressed MMP14 expression by inhibiting IκBα phosphorylation and NF-κB activation. Collectively, these findings suggest that the TGF-β-induced lncRNA NKILA has potential as an antimetastasis therapy. KEY MESSAGES Long noncoding RNA NKILA could be remarkably upregulated by classical TGF-β signal pathway in ESCC. NKILA was significantly downregulated in esophageal squamous cell carcinoma and negatively correlated with TNM stage. NKILA inhibits ESCC cell metastasis via repressing MMP14 expression by suppressing the phosphorylation of IκBα and NF-κB activation.
Collapse
Affiliation(s)
- Zhiliang Lu
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Zhaoli Chen
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Jingnan Wang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Zhirong Zhang
- Thoracic Surgery Department, Beijing Chao-Yang Hospital, Capital University of Medical Science, Beijing, 100020, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Jianbing Huang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Shouguo Sun
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Shuangshuang Mao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, 100021, People's Republic of China.
| |
Collapse
|
37
|
Amar S, Smith L, Fields GB. Matrix metalloproteinase collagenolysis in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:1940-1951. [PMID: 28456643 PMCID: PMC5605394 DOI: 10.1016/j.bbamcr.2017.04.015] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/08/2023]
Abstract
The proteolytic processing of collagen (collagenolysis) is critical in development and homeostasis, but also contributes to numerous pathologies. Mammalian interstitial collagenolytic enzymes include members of the matrix metalloproteinase (MMP) family and cathepsin K. While MMPs have long been recognized for their ability to catalyze the hydrolysis of collagen, the roles of individual MMPs in physiological and pathological collagenolysis are less defined. The use of knockout and mutant animal models, which reflect human diseases, has revealed distinct collagenolytic roles for MT1-MMP and MMP-13. A better understanding of temporal and spatial collagen processing, along with the knowledge of the specific MMP involved, will ultimately lead to more effective treatments for cancer, arthritis, cardiovascular conditions, and infectious diseases. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- Sabrina Amar
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA.
| | - Lyndsay Smith
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA.
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA; Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
38
|
Willson JA, Muir CA, Evered CL, Cepeda MA, Damjanovski S. Stable expression of α1-antitrypsin Portland in MDA-MB-231 cells increased MT1-MMP and MMP-9 levels, but reduced tumour progression. J Cell Commun Signal 2017; 12:479-488. [PMID: 28849349 DOI: 10.1007/s12079-017-0407-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/16/2017] [Indexed: 11/29/2022] Open
Abstract
The membrane bound matrix metalloproteinase MT1-MMP plays roles in modulating cell movement, independent of its abilities to remodel the extracellular matrix. Unlike many MMPs, MT1-MMP is activated in the Golgi prior to secretion by a pro-protein convertase, primarily furin. Regulation of the activation of pro-MT1-MMP has been methodically investigated, as altering the level of the active protein has broad implications in both activating other pro-MMPs, including pro-MMP-2, and many subsequent remodelling events. Our previous work in MCF-7 cells has demonstrated that modest, and not extremely high, levels of active MT1-MMP manifests into altered cell morphology and movement. At this low but optimal amount of MT1-MMP protein, changes to MT1-MMP levels are always mirrored by MMP-9 and pERK levels, and always opposite to MMP-2 levels. In this study, stable expression of the furin inhibitor α1-antitrypsin Portland (α1-PDX) in MDA-MB-231 cells increased overall MT1-MMP levels, but cells maintained a 21% proportion of pro-MT1-MMP. The increase in MT1-MMP was mirrored by increases in MMP-9 and pERK, but a decrease in MMP-2. These changes were associated with increased NF-κB transcription. In vitro analysis showed that α1-PDX decreased cell protrusions and migration, and this manifested as decreased tumourigenesis when examined in vivo using a chick CAM assay.
Collapse
Affiliation(s)
- J A Willson
- Department of Biology, University of Western Ontario, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - C A Muir
- Department of Biology, University of Western Ontario, 1151 Richmond St. N, London, ON, N6A 5B7, Canada
| | - C L Evered
- Ontario Veterinary College, University of Guelph, 50 Stone Rd. E, Guelph, ON, N1G 2W1, Canada
| | - M A Cepeda
- Department of Urology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55902, USA
| | - S Damjanovski
- Department of Biology, University of Western Ontario, 1151 Richmond St. N, London, ON, N6A 5B7, Canada.
| |
Collapse
|
39
|
Radisky ES, Raeeszadeh-Sarmazdeh M, Radisky DC. Therapeutic Potential of Matrix Metalloproteinase Inhibition in Breast Cancer. J Cell Biochem 2017; 118:3531-3548. [PMID: 28585723 PMCID: PMC5621753 DOI: 10.1002/jcb.26185] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc endopeptidases that cleave nearly all components of the extracellular matrix as well as many other soluble and cell-associated proteins. MMPs have been implicated in normal physiological processes, including development, and in the acquisition and progression of the malignant phenotype. Disappointing results from a series of clinical trials testing small molecule, broad spectrum MMP inhibitors as cancer therapeutics led to a re-evaluation of how MMPs function in the tumor microenvironment, and ongoing research continues to reveal that these proteins play complex roles in cancer development and progression. It is now clear that effective targeting of MMPs for therapeutic benefit will require selective inhibition of specific MMPs. Here, we provide an overview of the MMP family and its biological regulators, the tissue inhibitors of metalloproteinases (TIMPs). We then summarize recent research from model systems that elucidate how specific MMPs drive the malignant phenotype of breast cancer cells, including acquisition of cancer stem cell features and induction of the epithelial-mesenchymal transition, and we also outline clinical studies that implicate specific MMPs in breast cancer outcomes. We conclude by discussing ongoing strategies for development of inhibitors with therapeutic potential that are capable of selectively targeting the MMPs most responsible for tumor promotion, with special consideration of the potential of biologics including antibodies and engineered proteins based on the TIMP scaffold. J. Cell. Biochem. 118: 3531-3548, 2017. © 2017 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville 32224, Florida
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville 32224, Florida
| |
Collapse
|
40
|
Bhowmick M, Tokmina-Roszyk D, Onwuha-Ekpete L, Harmon K, Robichaud T, Fuerst R, Stawikowska R, Steffensen B, Roush W, Wong HR, Fields GB. Second Generation Triple-Helical Peptide Inhibitors of Matrix Metalloproteinases. J Med Chem 2017; 60:3814-3827. [PMID: 28394608 PMCID: PMC6413923 DOI: 10.1021/acs.jmedchem.7b00018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The design of selective matrix metalloproteinase (MMP) inhibitors that also possess favorable solubility properties has proved to be especially challenging. A prior approach using collagen-model templates combined with transition state analogs produced a first generation of triple-helical peptide inhibitors (THPIs) that were effective in vitro against discrete members of the MMP family. These THPI constructs were also highly water-soluble. The present study sought improvements in the first generation THPIs by enhancing thermal stability and selectivity. A THPI selective for MMP-2 and MMP-9 was redesigned to incorporate non-native amino acids (Flp and mep), resulting in an increase of 18 °C in thermal stability. This THPI was effective in vivo in a mouse model of multiple sclerosis, reducing clinical severity and weight loss. Two other THPIs were developed to be more selective within the collagenolytic members of the MMP family. One of these THPIs was serendipitously more effective against MMP-8 than MT1-MMP and was utilized successfully in a mouse model of sepsis. The THPI targeting MMP-8 minimized lung damage, increased production of the anti-inflammatory cytokine IL-10, and vastly improved mouse survival.
Collapse
Affiliation(s)
- Manishabrata Bhowmick
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
- Sigma-Aldrich Corporation, 3 Strathmore Road, Natick, Massachusetts 01760, United States
| | - Dorota Tokmina-Roszyk
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Lillian Onwuha-Ekpete
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Kelli Harmon
- Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, United States
| | - Trista Robichaud
- University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio Texas 78229, United States
| | - Rita Fuerst
- The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Roma Stawikowska
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
| | - Bjorn Steffensen
- University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio Texas 78229, United States
- School of Dental Medicine, Tufts University, 1 Kneeland Street, Boston, Massachusetts 02111, United States
| | - William Roush
- The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Hector R. Wong
- Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, United States
| | - Gregg B. Fields
- Florida Atlantic University, 5353 Parkside Drive, Jupiter, Florida 33458, United States
- The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
41
|
Knapinska AM, Estrada CA, Fields GB. The Roles of Matrix Metalloproteinases in Pancreatic Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:339-354. [PMID: 28662827 DOI: 10.1016/bs.pmbts.2017.03.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Matrix metalloproteinases (MMPs) have long been implicated for roles in cancer initiation, tumor growth, and metastasis. However, pancreatic cancer clinical trials using broad-based MMP inhibitors were discouraging. To better evaluate the use of MMP inhibitors in pancreatic cancer, (a) more precise roles of individual MMPs in pancreatic cancer needed to be determined and (b) animal models that more accurately represented human pancreatic cancer needed to be developed. The last decade has seen substantial progress in both areas. MT1-MMP has been recognized as a critical mediator of several steps in pancreatic cancer progression, while MMP-9 appears to be an antitarget when considering pancreatic cancer therapies.
Collapse
Affiliation(s)
| | | | - Gregg B Fields
- Florida Atlantic University, Jupiter, FL, United States; The Scripps Research Institute/Scripps Florida, Jupiter, FL, United States.
| |
Collapse
|
42
|
Inhibition of MT1-MMP proteolytic function and ERK1/2 signalling influences cell migration and invasion through changes in MMP-2 and MMP-9 levels. J Cell Commun Signal 2017; 11:167-179. [PMID: 28070797 DOI: 10.1007/s12079-016-0373-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/19/2016] [Indexed: 10/25/2022] Open
Abstract
Membrane type-1 matrix metalloproteinase (MT1-MMP, MMP-14) is a unique protease that cleaves extracellular proteins, activates proMMPs, and initiates intracellular signalling. MCF-7 cells are non-invasive and deficient in MT1-MMP, MMP-2, and MMP-9 expression. We created an MCF-7 cell line (C2) that stably produces active MT1-MMP and demonstrated increased ERK1/2 phosphorylation. MAPK inhibition in this cell line showed an inverse relationship in MMP-2 and MMP-9 transcripts where levels of these genes increased and decreased, respectively. Using invasive MDA-MB 231 cells that endogenously produce MT1-MMP and have naturally high pERK levels, we demonstrated the identical inverse relationship between MMP-2 and -9 transcript and protein levels, suggesting that this novel relationship is conserved amongst MT1-MMP positive breast cancer cells. To further analyze the relationship between MMP-2 and -9 levels, we chemically inhibited activation and catalytic activity of MT1-MMP using a furin and MMP inhibitor, respectively, to show that interference with the functions of MT1-MMP induced changes in MMP-2 and 9 transcript levels that were always inverse of each other, and likely mediated by differential transcriptional activity of the NF-κB transcription factor. Furthermore, we analyzed the functional consequences of these expression changes to show MMP, and in particular ERK, inhibition decreased migration and invasion using 2D culture, and inhibits the formation of an invasive phenotype in Matrigel 3D culture. This study demonstrated a novel inverse transcriptional relationship between MMP-2 and -9 levels and MT1-MMP activity that have functional consequences, and also showed that increases in the levels of MMPs does not necessarily correlate with an invasive phenotype.
Collapse
|
43
|
Decaneto E, Suladze S, Rosin C, Havenith M, Lubitz W, Winter R. Pressure and Temperature Effects on the Activity and Structure of the Catalytic Domain of Human MT1-MMP. Biophys J 2016; 109:2371-81. [PMID: 26636948 DOI: 10.1016/j.bpj.2015.10.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/12/2015] [Accepted: 10/19/2015] [Indexed: 11/19/2022] Open
Abstract
Membrane type 1-matrix metalloproteinase (MT1-MMP or MMP-14) is a zinc-transmembrane metalloprotease involved in the degradation of extracellular matrix and tumor invasion. While changes in solvation of MT1-MMP have been recently studied, little is known about the structural and energetic changes associated with MT1-MMP while interacting with substrates. Steady-state kinetic and thermodynamic data (including activation energies and activation volumes) were measured over a wide range of temperatures and pressures by means of a stopped-flow fluorescence technique. Complementary temperature- and pressure-dependent Fourier-transform infrared measurements provided corresponding structural information of the protein. MT1-MMP is stable and active over a wide range of temperatures (10-55 °C). A small conformational change was detected at 37 °C, which is responsible for the change in activity observed at the same temperature. Pressure decreases the enzymatic activity until complete inactivation occurs at 2 kbar. The inactivation is associated with changes in the rate-limiting step of the reaction caused by additional hydration of the active site upon compression and/or minor conformational changes in the active site region. Based on these data, an energy and volume diagram could be established for the various steps of the enzymatic reaction.
Collapse
Affiliation(s)
- Elena Decaneto
- Max Planck Institute for Chemical Energy Conversion, Mülheim a. d. Ruhr, Germany; Department of Physical Chemistry II, Ruhr-University Bochum, Bochum, Germany
| | - Saba Suladze
- Department of Chemistry and Chemical Biology, Physical Chemistry, Technische Universität Dortmund, Dortmund, Germany
| | - Christopher Rosin
- Department of Chemistry and Chemical Biology, Physical Chemistry, Technische Universität Dortmund, Dortmund, Germany
| | - Martina Havenith
- Department of Physical Chemistry II, Ruhr-University Bochum, Bochum, Germany
| | - Wolfgang Lubitz
- Max Planck Institute for Chemical Energy Conversion, Mülheim a. d. Ruhr, Germany
| | - Roland Winter
- Department of Chemistry and Chemical Biology, Physical Chemistry, Technische Universität Dortmund, Dortmund, Germany.
| |
Collapse
|
44
|
Naseh G, Mohammadifard M, Mohammadifard M. Upregulation of cyclin-dependent kinase 7 and matrix metalloproteinase-14 expression contribute to metastatic properties of gastric cancer. IUBMB Life 2016; 68:799-805. [PMID: 27562173 DOI: 10.1002/iub.1543] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/10/2016] [Indexed: 01/01/2023]
Abstract
The aim of this study was to evaluate the protein and mRNA expressions of matrix metalloproteinase-14 (MMP14) and CDK7 in gastric cancer (GC) tissues. Upregulation of MMP14 mRNA level was observed in GC tissues when compared with the matched normal tissues (mean ± SD: 3.92 ± 1.15 vs. 1.35 ± 0.81, P < 0.001). This study indicated that mRNA levels of CDK7 were statistically overexpressed in GC when compared with matched normal tissues (4.12 ± 0.84 vs. 1.43 ± 0.71, P < 0.001). The protein levels of MMP14 were found to be increased in GC (60.41%; P < 0.001). The expression of CDK7 was higher in GC tissues than matched normal tissues (70.83; P < 0.001). We found that high MMP14 expression was related to advanced TNM stage (P = 0.004), tumor grade (P = 0.002), and lymph node metastasis (P = 0.015), but no association with other clinical variables (P > 0.05). In addition, high expression of CDK7 was significantly linked to advanced TNM stage (P = 0.001), pathological grade (P = 0.012), and presence of lymph node metastasis (P = 0.009), while no correlation between CDK7 expression and other clinical variables, such as age and gender, distance metastasis. The patients with high expression of MMP14 and CDK7 exhibited worse survival time than those with higher levels. Cox multivariate regression analysis clearly showed that high expression of MMP14 and CDK7 was independent prognostic factors for overall survival in patients with GC. Taken together, these results indicated the overexpression of above markers in the progression and the tumorigenesis of GC and overall patient survival. © 2016 IUBMB Life, 68(10):799-805, 2016.
Collapse
Affiliation(s)
- Ghodratolah Naseh
- Department of Surgery, Imam Reza Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahyar Mohammadifard
- Department of Radiology, Imam Reza Hospital, Birjand University of Medical Sciences, Birjand, Iran.
| | - Mahtab Mohammadifard
- Department of Pathology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Kim TW, Ryu HH, Li SY, Li CH, Lim SH, Jang WY, Jung S. PDIA6 regulation of ADAM17 shedding activity and EGFR-mediated migration and invasion of glioblastoma cells. J Neurosurg 2016; 126:1829-1838. [DOI: 10.3171/2016.5.jns152831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVEIn patients with glioblastoma, local invasion of tumor cells causes recurrence and shortens survival. The goal of this study was to determine whether protein disulfide isomerase (PDI) A6 regulates migration and invasion of glioblastoma cells and the associated factors.METHODSU87MG cells were treated with either PDIA6 or ADAM17 small interfering RNA (siRNA) fragments or with both types of siRNA fragments, and expression was confirmed by reverse transcription–polymerase chain reaction and Western blot. Migration and invasion were assessed using a wound-healing assay, a Matrigel assay, and an organotypic culture system. After the U87MG cells were treated with siRNAs and epidermal growth factor receptor (EGFR) inhibitors, the expression of matrix metalloproteinase–2 (MMP-2), membrane Type 1-matrix metalloproteinase (MT1-MMP), integrin, phosphorylated focal adhesion kinase (pFAK), and phosphorylated EGFR (pEGFR) was detected by Western blotting and zymography.RESULTSU87MG cell migration and invasion increased significantly after inhibition of PDIA6. The MMP-2 activation ratio and ADAM17 activity (as a sheddase of the proligand) increased, and expression of pEGFR, pFAK, integrin α5β3, and MT1-MMP was induced, compared with control levels. Furthermore, heparin-binding epidermal growth factor (EGFR signaling ligand) was highly expressed in PDIA6-knockdown cells. After siPDIA6-transfected U87MG cells were treated with EGFR signaling inhibitors, expression of pFAK, MMP-2, and MT1-MMP decreased and invasion decreased significantly. Simultaneous double-knockdown of PDIA6 and ADAM17 reduced pEGFR and pFAK expression, compared with control levels.CONCLUSIONSThe authors propose that inhibiting PDIA6 could transduce EGFR signaling by activating and inducing ADAM17 during migration and invasion of U87MG glioblastoma cells. The results of this study suggest that PDIA6 is an important component of EGFR-mediated migration and invasion of U87MG cells. This is the first report of the effects of PDIA6 on migration and invasion in glioblastoma.
Collapse
Affiliation(s)
- Tae-Wan Kim
- 1Department of Neurosurgery, Brain Tumor Clinic and Gamma Knife Center, and
| | - Hyang-Hwa Ryu
- 2Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Song-Yuan Li
- 2Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Chun-Hao Li
- 2Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Sa-Hoe Lim
- 1Department of Neurosurgery, Brain Tumor Clinic and Gamma Knife Center, and
- 2Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Woo-Youl Jang
- 1Department of Neurosurgery, Brain Tumor Clinic and Gamma Knife Center, and
| | - Shin Jung
- 1Department of Neurosurgery, Brain Tumor Clinic and Gamma Knife Center, and
- 2Brain Tumor Research Laboratory, Chonnam National University Research Institute of Medical Sciences, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| |
Collapse
|
46
|
Cerofolini L, Amar S, Lauer JL, Martelli T, Fragai M, Luchinat C, Fields GB. Bilayer Membrane Modulation of Membrane Type 1 Matrix Metalloproteinase (MT1-MMP) Structure and Proteolytic Activity. Sci Rep 2016; 6:29511. [PMID: 27405411 PMCID: PMC4942797 DOI: 10.1038/srep29511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/20/2016] [Indexed: 11/23/2022] Open
Abstract
Cell surface proteolysis is an integral yet poorly understood physiological process. The present study has examined how the pericellular collagenase membrane-type 1 matrix metalloproteinase (MT1-MMP) and membrane-mimicking environments interplay in substrate binding and processing. NMR derived structural models indicate that MT1-MMP transiently associates with bicelles and cells through distinct residues in blades III and IV of its hemopexin-like domain, while binding of collagen-like triple-helices occurs within blades I and II of this domain. Examination of simultaneous membrane interaction and triple-helix binding revealed a possible regulation of proteolysis due to steric effects of the membrane. At bicelle concentrations of 1%, enzymatic activity towards triple-helices was increased 1.5-fold. A single mutation in the putative membrane interaction region of MT1-MMP (Ser466Pro) resulted in lower enzyme activation by bicelles. An initial structural framework has thus been developed to define the role(s) of cell membranes in modulating proteolysis.
Collapse
Affiliation(s)
- Linda Cerofolini
- Giotto Biotech S.R.L., Via Madonna del Piano 6, 50019 Sesto Fiorentino (FI), Italy
| | - Sabrina Amar
- Department of Chemistry &Biochemistry, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Janelle L Lauer
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Tommaso Martelli
- CERM, University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino (FI), Italy.,Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy
| | - Marco Fragai
- CERM, University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino (FI), Italy.,Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy
| | - Claudio Luchinat
- CERM, University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino (FI), Italy.,Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy
| | - Gregg B Fields
- Department of Chemistry &Biochemistry, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA.,Department of Chemistry, The Scripps Research Institute/Scripps Florida, 130 Scripps Way, Jupiter, FL 33458, USA.,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, 33458, Port St. Lucie, FL 34987, USA
| |
Collapse
|
47
|
Xue Z, Wu X, Chen X, Luo Q. MT3-MMP down-regulation promotes tumorigenesis and correlates to poor prognosis in esophageal squamous cell carcinoma. Cancer Med 2016; 5:2459-68. [PMID: 27292876 PMCID: PMC5055189 DOI: 10.1002/cam4.790] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 04/30/2016] [Accepted: 05/09/2016] [Indexed: 01/13/2023] Open
Abstract
The membrane‐type matrix metalloproteinases (MT‐MMPs) play an important role in degrading the extracellular matrix (ECM) and facilitating protease‐dependent tumor progression and invasion. Here, we report that unlike MT1‐MMP, MT3‐MMP was down‐regulated in esophageal squamous cell carcinoma (ESCC) as detected by real‐time PCR (qPCR), Western blot analysis, and immunohistochemistry (IHC). Down‐regulation of MT3‐MMP was observed at protein level in 66.3% of ESCC specimens (by IHC, n = 86) for routine pathologic diagnosis, as well as at mRNA level in 63.3% of surgically resected ESCC tumors paired with surrounding nontumor tissues (by qPCR, n = 30). Notably, MT3‐MMP down‐regulation significantly correlated with lymph node metastasis and poor overall survival of patients with ESCC (median 5‐year survival = 50.69 vs. 30.77 months for patients with MT3‐MMP‐negative and ‐positive ESCC, respectively). Mechanistically, MT3‐MMP negatively regulated proliferation, colony formation, and migration of ESCC cells, in association with cell cycle arrest at G1, due to up‐regulation of p21Cip1 and p27Kip1. Together, as a tumor suppressor in ESCC, MT3‐MMP down‐regulation represents an unfavorable factor for prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Zengfu Xue
- Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Xiumin Wu
- Department of Pharmacy, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiong Chen
- Department of Medical Oncology, The Affiliated Dongfang Hospital of Xiamen University, Fuzhou, Fujian, China
| | - Qi Luo
- Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
48
|
Camodeca C, Nuti E, Tepshi L, Boero S, Tuccinardi T, Stura EA, Poggi A, Zocchi MR, Rossello A. Discovery of a new selective inhibitor of A Disintegrin And Metalloprotease 10 (ADAM-10) able to reduce the shedding of NKG2D ligands in Hodgkin's lymphoma cell models. Eur J Med Chem 2016; 111:193-201. [PMID: 26871660 DOI: 10.1016/j.ejmech.2016.01.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 11/29/2022]
Abstract
Hodgkin's lymphoma (HL) is the most common malignant lymphoma in young adults in the western world. This disease is characterized by an overexpression of ADAM-10 with increased release of NKG2D ligands, involved in an impaired immune response against tumor cells. We designed and synthesized two new ADAM-10 selective inhibitors, 2 and 3 based on previously published ADAM-17 selective inhibitor 1. The most promising compound was the thiazolidine derivative 3, with nanomolar activity for ADAM-10, high selectivity over ADAM-17 and MMPs and good efficacy in reducing the shedding of NKG2D ligands (MIC-B and ULBP3) in three different HL cell lines at non-toxic doses. Molecular modeling studies were used to drive the design and X-ray crystallography studies were carried out to explain the selectivity of 3 for ADAM-10 over MMPs.
Collapse
Affiliation(s)
- Caterina Camodeca
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elisa Nuti
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Livia Tepshi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy; CEA, iBiTec-S, Service d'Ingenierie Moleculaire des Proteines, CE-Saclay, 91191 Gif sur Yvette Cedex, France
| | - Silvia Boero
- Unit of Molecular Oncology and Angiogenesis, IRCCS AOU San Martino-IST, 16132 Genoa, Italy
| | - Tiziano Tuccinardi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Enrico A Stura
- CEA, iBiTec-S, Service d'Ingenierie Moleculaire des Proteines, CE-Saclay, 91191 Gif sur Yvette Cedex, France
| | - Alessandro Poggi
- Unit of Molecular Oncology and Angiogenesis, IRCCS AOU San Martino-IST, 16132 Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Armando Rossello
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy.
| |
Collapse
|
49
|
Zhang YH, Wang JJ, Li M, Zheng HX, Xu L, Chen YG. Matrix Metallopeptidase 14 Plays an Important Role in Regulating Tumorigenic Gene Expression and Invasion Ability of HeLa Cells. Int J Gynecol Cancer 2016; 26:600-6. [PMID: 26825836 DOI: 10.1097/igc.0000000000000652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The objectives of this study were to investigate the functional effect of matrix metallopeptidase 14 (MMP14) on cell invasion in cervical cancer cells (HeLa line) and to study the underlying molecular mechanisms. METHODS Expression vector of short hairpin RNA targeting MMP14 was treated in HeLa cells, and then, transfection efficiency was verified by a florescence microscope. Transwell assay was used to investigate cell invasion ability in HeLa cells. Quantitative polymerase chain reaction and Western blotting analysis were used to detect the expression of MMP14 and relative factors in messenger RNA and protein levels, respectively. RESULTS Matrix metallopeptidase 14 short hairpin RNA expression vector transfection obviously decreased MMP14 expression in messenger RNA and protein levels. Down-regulation of MMP14 suppressed invasion ability of HeLa cells and reduced transforming growth factor β1 and vascular endothelial growth factor B expressions. Furthermore, MMP14 knockdown decreased bone sialoprotein and enhanced forkhead box protein L2 expression in both RNA and protein levels. CONCLUSION Matrix metallopeptidase 14 plays an important role in regulating invasion of HeLa cells. Matrix metallopeptidase 14 knockdown contributes to attenuating the malignant phenotype of cervical cancer cell.
Collapse
Affiliation(s)
- Ying-Hui Zhang
- *Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China; and †Department of Biochemical and Molecular of Medical College, Soochow University, Suzhou, China
| | | | | | | | | | | |
Collapse
|
50
|
UCA1 functions as a competing endogenous RNA to suppress epithelial ovarian cancer metastasis. Tumour Biol 2016; 37:10633-41. [PMID: 26867765 DOI: 10.1007/s13277-016-4917-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/27/2016] [Indexed: 02/05/2023] Open
Abstract
Urothelial cancer associated 1 (UCA1) is an example of functional long noncoding RNAs involved in many biologic processes. However, little is known about the association between UCA1 expression and metastasis in epithelial ovarian cancer (EOC). Findings of this study confirmed that not only UCA1 was aberrantly upregulated in EOC tissues and cells, but also correlated with status of lymph node metastasis and FIGO stage. Furthermore, univariate and multivariate analyses showed that UCA1 was a prognostic factor for overall survival in EOC patients. In vitro, knockdown of UCA1 reduced the invasion and migration ability of EOC cells. The results showed that UCA1 could function as an endogenous sponge by directly binding to miR-485-5p. Depletion of UCA1 was involved in the downregulation of matrix metallopeptidase 14 (MMP14) expression, a target gene of miR-485-5p. In conclusion, our work indicates that UCA1 is a new prognostic biomarker for EOC, establishing a novel connection among UCA1, miR-485-5p, and MMP14 in EOC metastasis.
Collapse
|