51
|
Venkatachalam P, Nadumane VK. Purification and Characterization of a Protease Inhibitor with Anticancer Potential from Bacillus endophyticus JUPR15. CURRENT CANCER THERAPY REVIEWS 2019. [DOI: 10.2174/1573394714666180321150605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction:Introduction: Protease Inhibitors (PIs) constitute a group of proteins widely distributed among all organisms and their main function includes their ability to inhibit the proteolytic activity. PIs represent an important role in the regulation of various cellular physiological and biological processes, including cell cycle, cell death, differentiation and immune response.Material and Methods:Hence, in our search for novel anticancer compounds, we isolated microorganisms from various environmental sources and screened them for the production of protease inhibitors. Promising isolates were further checked for their protease inhibitory activity by their ability to inhibit the activity of trypsin and chymotrypsin, which were measured spectrophotometrically.Results:The isolate identified as Bacillus endophyticus JUPR15 was found to be promising with higher inhibitory activity than the other isolates. The inhibitor was purified by cold acetone precipitation and column chromatography and further subjected to characterization studies by performing 12 % SDS-PAGE to determine the molecular weight and gelatin-PAGE assay to confirm its inhibitory activity.Conclusion:The isolate exhibited promising anticancer activity on in-vitro Hela and HepG2 cancer cell lines, showing its application potentials.
Collapse
Affiliation(s)
- Prerana Venkatachalam
- Department of Biotechnology, School of Sciences, Jain University, Jayanagar, Bengaluru-560 011, India
| | | |
Collapse
|
52
|
Lin W, Ma G, Yuan Z, Qian H, Xu L, Sidransky E, Chen S. Development of Zwitterionic Polypeptide Nanoformulation with High Doxorubicin Loading Content for Targeted Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:1273-1283. [PMID: 29933695 DOI: 10.1021/acs.langmuir.8b00851] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Much attention has been drawn to targeted nanodrug delivery systems due to their high therapeutic efficacy in cancer treatment. In this work, doxorubicin (DOX) was incorporated into a zwitterionic arginyl-glycyl-aspartic acid (RGD)-conjugated polypeptide by an emulsion solvent evaporation technique with high drug loading content (45%) and high drug loading efficiency (95%). This zwitterionic nanoformulation showed excellent colloidal stability at high dilution and in serum. The pH-induced disintegration and enzyme-induced degradation of the nanoformulation were confirmed by dynamic light scattering and gel permeation chromatography. Efficient internalization of DOX in the cells and high antitumor activity in vitro was observed. Compared with the free drug, this nanoformulation showed higher accumulation in tumor and lower systemic toxicity in vivo. The DOX-loaded zwitterionic RGD-conjugated polypeptide vesicles show potential application for targeted drug delivery in the clinic.
Collapse
Affiliation(s)
- Weifeng Lin
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
| | - Guanglong Ma
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
| | - Zhefan Yuan
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
| | - Haofeng Qian
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
| | - Liangbo Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
| | - Elie Sidransky
- Department of Materials Science and Engineering, A. James Clark School of Engineering , University of Maryland , College Park , Maryland 20740 , United States
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Department of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , China
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Biomedical Materials, College of Chemistry and Materials Science , Nanjing Normal University , Nanjing 210046 , China
| |
Collapse
|
53
|
Mehner C, Radisky ES. Bad Tumors Made Worse: SPINK1. Front Cell Dev Biol 2019; 7:10. [PMID: 30778387 PMCID: PMC6369215 DOI: 10.3389/fcell.2019.00010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/16/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Christine Mehner
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States.,Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| |
Collapse
|
54
|
Yildiz T, Gu R, Zauscher S, Betancourt T. Doxorubicin-loaded protease-activated near-infrared fluorescent polymeric nanoparticles for imaging and therapy of cancer. Int J Nanomedicine 2018; 13:6961-6986. [PMID: 30464453 PMCID: PMC6217908 DOI: 10.2147/ijn.s174068] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Despite significant progress in the field of oncology, cancer remains one of the leading causes of death. Chemotherapy is one of the most common treatment options for cancer patients but is well known to result in off-target toxicity. Theranostic nanomedicines that integrate diagnostic and therapeutic functions within an all-in-one platform can increase tumor selectivity for more effective chemotherapy and aid in diagnosis and monitoring of therapeutic responses. MATERIAL AND METHODS In this work, theranostic nanoparticles were synthesized with commonly used biocompatible and biodegradable polymers and used as cancer contrast and therapeutic agents for optical imaging and treatment of breast cancer. These core-shell nanoparticles were prepared by nanoprecipitation of blends of the biodegradable and biocompatible amphiphilic copolymers poly(lactic-co-glycolic acid)-b-poly-l-lysine and poly(lactic acid)-b-poly(ethylene glycol). Poly-l-lysine in the first copolymer was covalently decorated with near-infrared fluorescent Alexa Fluor 750 molecules. RESULTS The spherical nanoparticles had an average size of 60-80 nm. The chemotherapeutic drug doxorubicin was encapsulated in the core of nanoparticles at a loading of 3% (w:w) and controllably released over a period of 30 days. A 33-fold increase in near-infrared fluorescence, mediated by protease-mediated cleavage of the Alexa Fluor 750-labeled poly-l-lysine on the surface of the nanoparticles, was observed upon interaction with the model protease trypsin. The cytocompatibility of drug-free nanoparticles and growth inhibition of drug-loaded nanoparticles on MDA-MB-231 breast cancer cells were investigated with a luminescence cell-viability assay. Drug-free nanoparticles were found to cause minimal toxicity, even at high concentrations (0.2-2,000 µg/mL), while doxorubicin-loaded nanoparticles significantly reduced cell viability at drug concentrations >10 µM. Finally, the interaction of the nanoparticles with breast cancer cells was studied utilizing fluorescence microscopy, demonstrating the potential of the nanoparticles to act as near-infrared fluorescence optical imaging agents and drug-delivery carriers. CONCLUSION Doxorubicin-loaded, enzymatically activatable nanoparticles of less than 100 nm were prepared successfully by nanoprecipitation of copolymer blends. These nanoparticles were found to be suitable as controlled drug delivery systems and contrast agents for imaging of cancer cells.
Collapse
Affiliation(s)
- Tugba Yildiz
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, TX,
| | - Renpeng Gu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC
| | - Tania Betancourt
- Materials Science, Engineering, and Commercialization Program, Texas State University, San Marcos, TX,
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA,
| |
Collapse
|
55
|
Matrix metalloproteinase-2: A key regulator in coagulation proteases mediated human breast cancer progression through autocrine signaling. Biomed Pharmacother 2018; 105:395-406. [PMID: 29870887 DOI: 10.1016/j.biopha.2018.05.155] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 05/30/2018] [Accepted: 05/30/2018] [Indexed: 01/15/2023] Open
Abstract
AIMS Cell invasion is attributed to the synthesis and secretion of proteolytically active matrix-metalloproteinases (MMPs) by tumor cells to degrade extracellular matrix (ECM) and promote metastasis. The role of protease-activated receptor 2 (PAR2) in human breast cancer migration/invasion via MMP-2 up-regulation remains ill-defined; hence we investigated whether TF-FVIIa/trypsin-mediated PAR2 activation induces MMP-2 expression in human breast cancer. MAIN METHODS MMP-2 expression and the signaling mechanisms were analyzed by western blotting and RT-PCR. MMP-2 activity was measured by gelatin zymography. Cell invasion was analyzed by transwell invasion assay whereas; wound healing assay was performed to understand the cell migratory potential. KEY FINDINGS Here, we highlight that TF-FVIIa/trypsin-mediated PAR2 activation leads to enhanced MMP-2 expression in human breast cancer cells contributing to tumor progression. Knock-down of PAR2 abrogated TF-FVIIa/trypsin-induced up-regulation of MMP-2. Again, genetic manipulation of AKT or inhibition of NF-ĸB suggested that PAR2-mediated enhanced MMP-2 expression is dependent on the PI3K-AKT-NF-ĸB pathway. We also reveal that TF, PAR2, and MMP-2 are over-expressed in invasive breast carcinoma tissues as compared to normal. Knock-down of MMP-2 significantly impeded TF-FVIIa/trypsin-induced cell invasion. Further, we report that MMP-2 activates p38 MAPK-MK2-HSP27 signaling axis that leads to actin polymerization and induces cell migration. Pharmacological inhibition of p38 MAPK or MK2 attenuates MMP-2-induced cell migration. SIGNIFICANCE The study delineates a novel signaling pathway by which PAR2-induced MMP-2 expression regulates human breast cancer cell migration/invasion. Understanding these mechanistic details will certainly help to identify crucial targets for therapeutic interventions in breast cancer metastasis.
Collapse
|
56
|
Sammarco G, Gadaleta CD, Zuccalà V, Albayrak E, Patruno R, Milella P, Sacco R, Ammendola M, Ranieri G. Tumor-Associated Macrophages and Mast Cells Positive to Tryptase Are Correlated with Angiogenesis in Surgically-Treated Gastric Cancer Patients. Int J Mol Sci 2018; 19:1176. [PMID: 29649166 PMCID: PMC5979483 DOI: 10.3390/ijms19041176] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
Mast cells and macrophages can play a role in tumor angiogenesis by stimulating microvascular density (MVD). The density of mast cells positive to tryptase (MCDPT), tumor-associated macrophages (TAMs), and MVD were evaluated in a series of 86 gastric cancer (GC) tissue samples from patients who had undergone potential curative surgery. MCDPT, TAMs, and MVD were assessed in tumor tissue (TT) and in adjacent normal tissue (ANT) by immunohistochemistry and image analysis. Each of the above parameters was correlated with the others and, in particular for TT, with important clinico-pathological features. In TT, a significant correlation between MCDPT, TAMs, and MVD was found by Pearson t-test analysis (p ranged from 0.01 to 0.02). No correlation to the clinico-pathological features was found. A significant difference in terms of mean MCDPT, TAMs, and MVD between TT and ANT was found (p ranged from 0.001 to 0.002). Obtained data suggest MCDPT, TAMs, and MVD increased from ANT to TT. Interestingly, MCDPT and TAMs are linked in the tumor microenvironment and they play a role in GC angiogenesis in a synergistic manner. The assessment of the combination of MCDPT and TAMs could represent a surrogate marker of angiogenesis and could be evaluated as a target of novel anti-angiogenic therapies in GC patients.
Collapse
Affiliation(s)
- Giuseppe Sammarco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
| | - Cosmo Damiano Gadaleta
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| | - Valeria Zuccalà
- Pathology Unit, "Pugliese-Ciaccio" Hospital, Viale Pio X, 88100 Catanzaro, Italy.
| | - Emre Albayrak
- Department of Medical Biochemistry, Gulhane Medical Faculty, Health Science University, Ankara 06010, Turkey.
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro" of Bari, Via Casamassima, 70010 Bari, Italy.
| | - Pietro Milella
- Statistic and Epidemiology Unit, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
| | - Michele Ammendola
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
- Surgery Unit, National Cancer Research Centre Istituto Tumori ''Giovanni Paolo II'', 70124 Bari, Italy.
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| |
Collapse
|
57
|
Xie Y, Chen L, Lv X, Hou G, Wang Y, Jiang C, Zhu H, Xu N, Wu L, Lou X, Liu S. The levels of serine proteases in colon tissue interstitial fluid and serum serve as an indicator of colorectal cancer progression. Oncotarget 2018; 7:32592-606. [PMID: 27081040 PMCID: PMC5078036 DOI: 10.18632/oncotarget.8693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/28/2016] [Indexed: 02/06/2023] Open
Abstract
The proteins in tissue interstitial fluids (TIFs) can spread into the blood and have been proposed as an ideal material to find blood biomarkers. The colon TIFs were collected from 8-, 13-, 18-, and 22-week ApcMin/+, a typical mouse model of colorectal cancer (CRC), and wild-type mice. iTRAQ-based quantification proteomics was conducted to survey the TIF proteins whose abundance appeared to depend on tumor progression. A total of 46 proteins that exhibited consecutive changes in abundance were identified, including six serine proteases, chymotrypsin-like elastase 1 (CELA1), chymotrypsin-like elastase 2A (CEL2A), chymopasin, chymotrypsinogen B (CTRB1), trypsin 2 (TRY2), and trypsin 4 (TRY4). The observed increases in the abundance of serine proteases were supported in another quantitative evaluation of the individual colon TIFs using a multiple reaction monitor (MRM) assay. Importantly, the increases in the abundance of serine proteases were also verified in the corresponding sera. The quantitative verification of the serine proteases was further extended to the clinical sera, revealing significantly higher levels of CELA1, CEL2A, CTRL/chymopasin, and TRY2 in CRC patients. The receiver operating characteristic analysis illustrated that the combination of CELA1 and CTRL reached the best diagnostic performance, with 90.0% sensitivity and 80.0% specificity. Thus, the quantitative target analysis demonstrated that some serine proteases are indicative of CRC progression.
Collapse
Affiliation(s)
- Yingying Xie
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lechuang Chen
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaolei Lv
- Beijing Protein Innovation, Beijing, 101318, China
| | - Guixue Hou
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Wang
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cuicui Jiang
- Beijing Protein Innovation, Beijing, 101318, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Wu
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaomin Lou
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Siqi Liu
- CAS Key Laboratory of Genome Sciences and Information, China Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Protein Innovation, Beijing, 101318, China.,Proteomics Division, BGI-Shenzhen, Shenzhen, Guangdong, 518083, China
| |
Collapse
|
58
|
Lin B, Zhou X, Lin S, Wang X, Zhang M, Cao B, Dong Y, Yang S, Wang JM, Guo M, Huang J. Epigenetic silencing of PRSS3 provides growth and metastasis advantage for human hepatocellular carcinoma. J Mol Med (Berl) 2017; 95:1237-1249. [PMID: 28844099 PMCID: PMC8171496 DOI: 10.1007/s00109-017-1578-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/14/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023]
Abstract
Protease, serine, 3 (PRSS3), a member of the trypsin family of serine proteases, has been shown to be aberrantly expressed in several cancer types and to play important roles in tumor progression and metastasis. However, the expression and function of PRSS3 gene in hepatocellular carcinoma (HCC) remain unclear. Here we found that PRSS3 expression was decreased in human HCC cell lines and HCC surgical specimens. This was associated with intragenic methylation of PRSS3 gene. Treatment with DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine and/or histone deacetylase inhibitor trichostatin A restored PRSS3 expression in HCC cell lines. Ectopic overexpression of PRSS3 gene in HCC cell lines significantly suppressed cell proliferation and colony formation and arrested cell cycle at G1/S phase, accompanied with downregulation of cyclin D1 (CCND1)/CDK4 and cyclin E1 (CCNE1)/CDK2 complexes. Moreover, PRSS3 overexpression in HCC cells inhibited HCC cell migration and invasion with downregulation of matrix metallopeptidase 2 (MMP2). Further study showed that PRSS3 overexpression diminished the phosphorylation of mitogen-activated protein kinase/extracellular-signal-regulated kinase signaling protein, mitogen-activated protein kinase kinase 1 (MEK1)/mitogen-activated protein kinase kinase 2 (MEK2) and extracellular-signal related kinase 1 (ERK1)/extracellular-signal related kinase 2 (ERK2), in HCC cells. In contrast, knockdown of PRSS3 by small interfering RNA resulted in opposite effects on an HCC cell line SNU-387 which constitutively expresses PRSS3. These results demonstrate that downregulation of PRSS3 by intragenic hypermethylation provides growth and metastasis advantage to HCC cells. The clinical relevance of PRSS3 to human HCC was shown by the intragenic methylation of PRSS3 in HCC specimens and its association with poor tumor differentiation in patients with HCC. Thus, PRSS3 is a potential prognostic biomarker and an epigenetic target for intervention of human HCC. KEY MESSAGES • PRSS3 is downregulated by intragenic hypermethylation in HCC. • Epigenetic silencing of PRSS3 facilitates growth, migration, and invasion of HCC. • PRSS3 intragenic methylation has implication in diagnosis of HCC.
Collapse
Affiliation(s)
- Bonan Lin
- College of Life Sciences & Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Xiaomeng Zhou
- College of Life Sciences & Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Shuye Lin
- College of Life Sciences & Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Xiaoyue Wang
- College of Life Sciences & Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Meiying Zhang
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Baoping Cao
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Dong
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shuai Yang
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jiaqiang Huang
- College of Life Sciences & Bioengineering, Beijing Jiaotong University, Beijing, 100044, China.
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| |
Collapse
|
59
|
Ammendola M, Gadaleta CD, Frampton AE, Piardi T, Memeo R, Zuccalà V, Luposella M, Patruno R, Zizzo N, Gadaleta P, Pessaux P, Sacco R, Sammarco G, Ranieri G. The density of mast cells c-Kit + and tryptase + correlates with each other and with angiogenesis in pancreatic cancer patients. Oncotarget 2017; 8:70463-70471. [PMID: 29050294 PMCID: PMC5642569 DOI: 10.18632/oncotarget.19716] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/24/2017] [Indexed: 12/16/2022] Open
Abstract
Literature data suggest that inflammatory cells such as mast cells (MCs) are involved in angiogenesis. MCs can stimulate angiogenesis by releasing of well identified pro-angiogenic cytokines stored in their cytoplasm. In particular, MCs can release tryptase, a potent in vivo and in vitro pro-angiogenic factor. Nevertheless, few data are available concerning the role of MCs positive to tryptase in primary pancreatic cancer angiogenesis. This study analyzed the correlation between mast cells positive to c-Kit receptor (c-Kit+ MCs), the density of MCs expressing tryptase (MCD-T) and microvascular density (MVD) in primary tumor tissue from patients affected by pancreatic ductal adenocarcinoma (PDAC). A series of 35 PDAC patients with stage T2-3N0-1M0 (by AJCC for Pancreas Cancer Staging 7th Edition) were selected and then undergone to surgery. Tumor tissue samples were evaluated by mean of immunohistochemistry and image analysis methods in terms of number of c-Kit+ MCs, MCD-T and MVD. The above parameters were related each other and with the most important main clinico-pathological features. A significant correlation between c-Kit+ MCs, MCD-T and MVD groups each other was found by Pearson t-test analysis (r ranged from 0.75 to 0.87; p-value ranged from 0.01 to 0.04). No other significant correlation was found. Our in vivo preliminary data, suggest that tumor microenvironmental MCs evaluated in terms of c-Kit+ MCs and MCD-T may play a role in PDAC angiogenesis and they could be further evaluated as a novel tumor biomarker and as a target of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Bari, Italy
| | - Adam Enver Frampton
- HPB Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Tullio Piardi
- Department of General, Digestive and Endocrine Surgery, Hopital Robert Debre, Centre Hospitalier Universitaire de Reims, Universite de Reims Champagne-Ardenne, Reims, France
| | - Riccardo Memeo
- Hepato-Biliary and Pancreatic Surgical Unit, General, Digestive and Endocrine Surgery, IRCAD, IHU Mix-Surg, Institute for Minimally Invasive Image-Guided Surgery, University of Strasbourg, 1 place de l'Hôpital, Strasbourg, France
| | - Valeria Zuccalà
- Pathology Unit, “Pugliese-Ciaccio” Hospital, Catanzaro, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, “San Giovanni di Dio” Hospital, Crotone, Italy
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University “Aldo Moro”, Bari, Italy
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, University “Aldo Moro”, Bari, Italy
| | - Pietro Gadaleta
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Bari, Italy
| | - Patrick Pessaux
- Hepato-Biliary and Pancreatic Surgical Unit, General, Digestive and Endocrine Surgery, IRCAD, IHU Mix-Surg, Institute for Minimally Invasive Image-Guided Surgery, University of Strasbourg, 1 place de l'Hôpital, Strasbourg, France
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Bari, Italy
| |
Collapse
|
60
|
Ammendola M, Sacco R, Vescio G, Zuccalà V, Luposella M, Patruno R, Zizzo N, Gadaleta C, Marech I, Ruggieri R, Kocak IF, Ozgurtas T, Gadaleta CD, Sammarco G, Ranieri G. Tryptase mast cell density, protease-activated receptor-2 microvascular density, and classical microvascular density evaluation in gastric cancer patients undergoing surgery: possible translational relevance. Therap Adv Gastroenterol 2017; 10:353-360. [PMID: 28491140 PMCID: PMC5405880 DOI: 10.1177/1756283x16673981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mast cells (MCs) can stimulate angiogenesis, releasing several proangiogenic cytokines stored in their cytoplasm. In particular, MCs can release tryptase, a potent in vivo and in vitro proangiogenic factor via protease-activated receptor-2 (PAR-2) activation and mitogen-activated protein kinase (MAPK) phosphorylation. Nevertheless, no data are available concerning the relationship among tryptase MC density (TMCD), endothelial cells (ECs) positive to PAR-2 microvascular density (PAR-2-MVD) and classical MVD (C-MVD) in gastric cancer (GC) angiogenesis. METHODS In this study, we analyzed the correlation of TMCD, PAR-2-MVD, C-MVD with each other and with the main clinicopathological features in GC patients who underwent surgery. A series of 77 GC patients with stage T2-3N2-3M0 (classified by the American Joint Committee on Cancer for Gastric Cancer, 7th edition) were selected and then underwent surgery. RESULTS Tumour tissue samples were evaluated by mean of immunohistochemistry and image analysis methods in terms of numbers of TMCD, PAR-2-MVD and C-MVD. A significant correlation between the TMCD, PAR-2-MVD and C-MVD groups with each other was found by Pearson t-test analysis (r ranged from 0.64 to 0.76; p value ranged from 0.02 to 0.03). There was no other significant correlation between the above parameters and clinicopathological features. CONCLUSIONS Our in vivo preliminary data suggest that TMCD and PAR-2-MVD may play a role in GC angiogenesis and they could be further evaluated as a target of antiangiogenic therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Viale Europa – Germaneto, 88100, Catanzaro, Italy
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Giuseppina Vescio
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Valeria Zuccalà
- Health Science Department, Pathology Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, ‘San Giovanni di Dio’ Hospital, Crotone, Italy
| | - Rosa Patruno
- Chair of Pathology, University ‘Aldo Moro’ Veterinary Medical School, Bari, Italy
| | - Nicola Zizzo
- Chair of Pathology, University ‘Aldo Moro’ Veterinary Medical School, Bari, Italy
| | - Claudia Gadaleta
- Chair of Pathology, University ‘Aldo Moro’ Veterinary Medical School, Bari, Italy
| | - Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| | - Roberta Ruggieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| | - Ibrahim Furkan Kocak
- Department of Biochemistry, Gulhane Military Medical Academy Etlik, Ankara, Turkey
| | - Taner Ozgurtas
- Department of Biochemistry, Gulhane Military Medical Academy Etlik, Ankara, Turkey
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| |
Collapse
|
61
|
Ma G, Lin W, Yuan Z, Wu J, Qian H, Xu L, Chen S. Development of ionic strength/pH/enzyme triple-responsive zwitterionic hydrogel of the mixed l-glutamic acid and l-lysine polypeptide for site-specific drug delivery. J Mater Chem B 2017; 5:935-943. [DOI: 10.1039/c6tb02407f] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Environmentally responsive hydrogels for drug delivery.
Collapse
Affiliation(s)
- Guanglong Ma
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| | - Weifeng Lin
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| | - Zhefan Yuan
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| | - Jiang Wu
- School of Pharmaceutical Sciences Key Laboratory of Biotechnology and Pharmaceutical Engineering
- Wenzhou Medical University
- Wenzhou
- China
| | - Haofeng Qian
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| | - Liangbo Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- Department of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- China
| |
Collapse
|
62
|
Ammendola M, Sacco R, Zuccalà V, Luposella M, Patruno R, Gadaleta P, Zizzo N, Gadaleta CD, De Sarro G, Sammarco G, Oltean M, Ranieri G. Mast Cells Density Positive to Tryptase Correlate with Microvascular Density in both Primary Gastric Cancer Tissue and Loco-Regional Lymph Node Metastases from Patients That Have Undergone Radical Surgery. Int J Mol Sci 2016; 17:1905. [PMID: 27854307 PMCID: PMC5133903 DOI: 10.3390/ijms17111905] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/05/2016] [Accepted: 11/10/2016] [Indexed: 12/27/2022] Open
Abstract
Mast Cells (MCs) play a role in immune responses and more recently MCs have been involved in tumoral angiogenesis. In particular MCs can release tryptase, a potent in vivo and in vitro pro-angiogenic factor via proteinase-activated receptor-2 (PAR-2) activation and mitogen-activated protein kinase (MAPK) phosphorylation. MCs can release tryptase following c-Kit receptor activation. Nevertheless, no data are available concerning the relationship among MCs Density Positive to Tryptase (MCDPT) and Microvascular Density (MVD) in both primary gastric cancer tissue and loco-regional lymph node metastases. A series of 75 GC patients with stage T2-3N2-3M₀ (by AJCC for Gastric Cancer Seventh Edition) undergone to radical surgery were selected for the study. MCDPT and MVD were evaluated by immunohistochemistry and by image analysis system and results were correlated each to other in primary tumor tissue and in metastatic lymph nodes harvested. Furthermore, tissue parameters were correlated with important clinico-pathological features. A significant correlation between MCDPT and MVD was found in primary gastric cancer tissue and lymph node metastases. Pearson t-test analysis (r ranged from 0.74 to 0.79; p-value ranged from 0.001 to 0.003). These preliminary data suggest that MCDPT play a role in angiogenesis in both primary tumor and in lymph node metastases from GC. We suggest that MCs and tryptase could be further evaluated as novel targets for anti-angiogenic therapies.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
- Surgery Unit, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy.
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
| | - Valeria Zuccalà
- Pathology Unit, "Pugliese-Ciaccio" Hospital, Viale Pio X, 88100 Catanzaro, Italy.
| | - Maria Luposella
- Cardiovascular Disease Unit, "San Giovanni di Dio" Hospital, 88900 Crotone, Italy.
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro" of Bari, Via Casamassima, 70010 Bari, Italy.
| | - Pietro Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro" of Bari, Via Casamassima, 70010 Bari, Italy.
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University "Magna Graecia" Medical School, Viale Europa, Germaneto, 88100 Catanzaro, Italy.
| | - Mihai Oltean
- The Institute for Clinical Sciences, Department of Transplantation, University Hospital, Sahlgrenska Academy at the University of Gothenburg, 41345 Gothenburg, Sweden.
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori "Giovanni Paolo II", viale Orazio Flacco 65, 70124 Bari, Italy.
| |
Collapse
|
63
|
Forrest CM, McNair K, Vincenten MCJ, Darlington LG, Stone TW. Selective depletion of tumour suppressors Deleted in Colorectal Cancer (DCC) and neogenin by environmental and endogenous serine proteases: linking diet and cancer. BMC Cancer 2016; 16:772. [PMID: 27716118 PMCID: PMC5054602 DOI: 10.1186/s12885-016-2795-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/21/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The related tumour suppressor proteins Deleted in Colorectal Cancer (DCC) and neogenin are absent or weakly expressed in many cancers, whereas their insertion into cells suppresses oncogenic behaviour. Serine proteases influence the initiation and progression of cancers although the mechanisms are unknown. METHODS The effects of environmental (bacterial subtilisin) and endogenous mammalian (chymotrypsin) serine proteases were examined on protein expression in fresh, normal tissue and human neuroblastoma and mammary adenocarcinoma lines. Cell proliferation and migration assays (chemoattraction and wound closure) were used to examine cell function. Cells lacking DCC were transfected with an ectopic dcc plasmid. RESULTS Subtilisin and chymotrypsin selectively depleted DCC and neogenin from cells at nanomolar concentrations without affecting related proteins. Cells showed reduced adherence and increased migration, but after washing they re-attached within 24 h, with recovery of protein expression. These effects are induced by chymotryptic activity as they are prevented by chymostatin and the soybean Bowman-Birk inhibitor typical of many plant protease inhibitors. CONCLUSIONS Bacillus subtilis, which secretes subtilisin is widely present in soil, the environment and the intestinal contents, while subtilisin itself is used in meat processing, animal feed probiotics and many household cleaning agents. With chymotrypsin present in chyme, blood and tissues, these proteases may contribute to cancer development by depleting DCC and neogenin. Blocking their activity by Bowman-Birk inhibitors may explain the protective effects of a plant diet. Our findings identify a potential non-genetic contribution to cancer cell behaviour which may explain both the association of processed meats and other factors with cancer incidence and the protection afforded by plant-rich diets, with significant implications for cancer prevention.
Collapse
Affiliation(s)
- Caroline M Forrest
- College of Medical, Veterinary and Life Sciences, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Kara McNair
- College of Medical, Veterinary and Life Sciences, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Maria C J Vincenten
- College of Medical, Veterinary and Life Sciences, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | - Trevor W Stone
- College of Medical, Veterinary and Life Sciences, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
64
|
Abstract
Most cancer patients experience severe pain during their disease course, and the management of cancer pain is a major challenge for patients and the healthcare team. Many diverse translational models of cancer pain in recent years have improved our understanding of cancer-related pain. Cancer and associated cells in the cancer microenvironment may release various peripheral mediators, including ATP, formaldehyde, protons, proteases, endothelin, bradykinin, TNF and NGF, that result in the activation and/or sensitization of peripheral and central neurons, that contribute to the clinical manifestations of cancer-related pain. Identification of these mediators and the peripheral and central mechanisms by which they contribute to cancer-related pain may provide novel therapeutic targets to alleviate cancer patient suffering.
Collapse
Affiliation(s)
- David K Lam
- Oral & Maxillofacial Surgery, University of Toronto, Toronto, ON, Canada
- University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, ON, Canada
- Dental Oncology, Maxillofacial & Ocular Prosthetics, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Wasser Pain Management Centre, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
65
|
Abstract
Although many studies have demonstrated that components of the hemostatic system may be involved in signaling leading to cancer progression, the potential mechanisms by which they contribute to cancer dissemination are not yet precisely understood. Among known coagulant factors, tissue factor (TF) and thrombin play a pivotal role in cancer invasion. They may be generated in the tumor microenvironment independently of blood coagulation and can induce cell signaling through activation of protease-activated receptors (PARs). PARs are transmembrane G-protein-coupled receptors (GPCRs) that are activated by a unique proteolytic mechanism. They play important roles in vascular physiology, neural tube closure, hemostasis, and inflammation. All of these agents (TF, thrombin, PARs—mainly PAR-1 and PAR-2) are thought to promote cancer invasion and metastasis at least in part by facilitating tumor cell migration, angiogenesis, and interactions with host vascular cells, including platelets, fibroblasts, and endothelial cells lining blood vessels. Here, we discuss the role of PARs and their activators in cancer progression, focusing on TF- and thrombin-mediated actions. Therapeutic options tailored specifically to inhibit PAR-induced signaling in cancer patients are presented as well.
Collapse
|
66
|
Ammendola M, Sacco R, Sammarco G, Piardi T, Zuccalà V, Patruno R, Zullo A, Zizzo N, Nardo B, Marech I, Crovace A, Gadaleta CD, Pessaux P, Ranieri G. Mast cells positive to tryptase, endothelial cells positive to protease-activated receptor-2, and microvascular density correlate among themselves in hepatocellular carcinoma patients who have undergone surgery. Onco Targets Ther 2016; 9:4465-4471. [PMID: 27499640 PMCID: PMC4959580 DOI: 10.2147/ott.s105368] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mast cells (MCs) can stimulate angiogenesis, releasing several proangiogenic cytokines stored in their cytoplasm. In particular MCs can release tryptase, a potent in vivo and in vitro proangiogenic factor via proteinase-activated receptor-2 (PAR-2) activation and mitogen-activated protein kinase phosphorylation. Nevertheless, no data are available concerning the relationship between MC density positive to tryptase (MCDPT), endothelial cells positive to PAR-2 forming microvascular density (PAR-2-MVD), and classical MVD (C-MVD) in hepatocellular carcinoma (HCC) angiogenesis. This study analyzed the correlation between MCDPT, PAR-2-MVD, and C-MVD, each correlated to the others and to the main clinicopathological features, in early HCC patients who underwent surgery. METHODS A series of 53 HCC patients with early stage (stage 0 according to the Barcelona Clinic Liver Cancer Staging Classification) were selected and then underwent surgery. Tumor tissue samples were evaluated by means of immunohistochemistry and image analysis methods in terms of number of MCDPT, PAR-2-MVD, and C-MVD. RESULTS A significant correlation between MCDPT, PAR-2-MVD, and C-MVD groups, each correlated to the others, was found by Pearson t-test analysis (r ranged from 0.67 to 0.81; P-value ranged from 0.01 to 0.03). No other significant correlation was found. CONCLUSION Our in vivo pilot data suggest that MCDPT and PAR-2-MVD may play a role in HCC angiogenesis and could be further evaluated as a target of antiangiogenic therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Catanzaro, Italy
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Catanzaro, Italy
| | - Tullio Piardi
- Department of General, Digestive and Endocrine Surgery, Hopital Robert Debre, Centre Hospitalier Universitaire de Reims, Universite de Reims Champagne-Ardenne, Reims, France
| | - Valeria Zuccalà
- Department of Health Science, Pathology Unit, University of Catanzaro "Magna Graecia" Medical School, Catanzaro
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro", Bari
| | - Alessandra Zullo
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Catanzaro, Italy
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro", Bari
| | - Bruno Nardo
- Department of Medical and Surgery Sciences, S Orsola Hospital, University of Bologna, Bologna
| | - Ilaria Marech
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II"
| | - Alberto Crovace
- Department of Emergency and Organ Transplantation (DETO), Veterinary Medical School, University "Aldo Moro", Bari, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II"
| | - Patrick Pessaux
- Hepato-Biliary and Pancreatic Surgical Unit, General, Digestive and Endocrine Surgery, IRCAD, IHU Mix-Surg, Institute for Minimally Invasive Image-Guided Surgery, University of Strasbourg, Strasbourg, France
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II"
| |
Collapse
|
67
|
Ammendola M, Sacco R, Sammarco G, Luposella M, Patruno R, Gadaleta CD, Sarro GD, Ranieri G. Mast Cell-Targeted Strategies in Cancer Therapy. Transfus Med Hemother 2016; 43:109-113. [PMID: 27330532 PMCID: PMC4909357 DOI: 10.1159/000444942] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/18/2016] [Indexed: 02/06/2023] Open
Abstract
Mast cells (MCs) are cells that originate in the bone marrow from pluripotent CD34+ hematopoietic stem cells. Precursors of MCs migrate through the circulation to their target tissues, completing their maturation process into granulated cells under the influence of several microenvironment growth factors. The most important of these factors is the ligand for the c-Kit receptor (c-Kit-R) namely stem cell factor (SCF), secreted mainly by fibroblasts and endothelial cells (ECs). SCF also regulates development, survival and de novo proliferation of MCs. It has already been demonstrated that gain-of-function mutations of gene c-Kit encoding c-Kit-R result in the development of some tumors. Furthermore, MCs are able also to modulate both innate and adaptive immune response and to express the high-affinity IgE receptor following IgE activation. Among the other IgE-independent MC activation mechanisms, a wide variety of other surface receptors for cytokines, chemokines, immunoglobulins, and complement are also described. Interestingly, MCs can stimulate angiogenesis by releasing of several pro-angiogenic cytokines stored in their cytoplasm. Studies published in the last year suggest that angiogenesis stimulated by MCs may play an important role in tumor growth and progression. Here, we aim to focus several biological features of MCs and to summarize new anti-cancer MC-targeted strategies with potential translation in human clinical trials.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, ‘San Giovanni di Dio’ Hospital, Crotone, Italy
| | - Rosa Patruno
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro ‘Magna Graecia’ Medical School, Catanzaro, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, ‘Giovanni Paolo II’, Bari, Italy
| |
Collapse
|
68
|
Zhu Q, Yang KL. Polyethylene glycol (PEG) gel arrays for differentiating oligopeptide fragments and on-chip protease assays. Biosens Bioelectron 2016; 77:1126-33. [DOI: 10.1016/j.bios.2015.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/21/2015] [Accepted: 11/02/2015] [Indexed: 11/16/2022]
|
69
|
Yang L, Ma Y, Han W, Li W, Cui L, Zhao X, Tian Y, Zhou Z, Wang W, Wang H. Proteinase-activated receptor 2 promotes cancer cell migration through RNA methylation-mediated repression of miR-125b. J Biol Chem 2015; 290:26627-26637. [PMID: 26354435 PMCID: PMC4646319 DOI: 10.1074/jbc.m115.667717] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/05/2015] [Indexed: 01/08/2023] Open
Abstract
Proteinase activated-receptor 2 (PAR2) participates in cancer metastasis promoted by serine proteinases. The current study aimed to test the molecular mechanism by which PAR2 promotes cancer cell migration. In different cancer cells, activation of PAR2 by activating peptide (PAR2-AP) dramatically increased cell migration, whereas knock down of PAR2 inhibited cellular motility. The PAR2 activation also repressed miR-125b expression while miR-125b mimic successfully blocked PAR2-induced cell migration. Moreover, Grb associated-binding protein 2 (Gab2) was identified as a novel target gene of miR-125b and it mediated PAR2-induced cell migration. The correlation of PAR2 with miR-125b and Gab2 was further supported by the findings obtained from human colorectal carcinoma specimens. Remarkably, knock down of NOP2/Sun domain family, member 2 (NSun2), a RNA methyltransferase, blocked the reduction in miR-125b induced by PAR2. Furthermore, PAR2 activation increased the level of N(6)-methyladenosine (m(6)A)-containing pre-miR-125b in NSun2-dependent manner. Taken together, our results demonstrated that miR-125b mediates PAR2-induced cancer cell migration by targeting Gab2 and that NSun2-dependent RNA methylation contributes to the down-regulation of miR-125b by PAR2 signaling. These findings suggest a novel epigenetic mechanism by which microenvironment regulates cancer cell migration by altering miRNA expression.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Binding Sites
- Cell Line, Tumor
- Cell Movement/drug effects
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- HCT116 Cells
- HT29 Cells
- Humans
- Methylation/drug effects
- Methyltransferases/antagonists & inhibitors
- Methyltransferases/genetics
- Methyltransferases/metabolism
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Oligopeptides/pharmacology
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Protein Binding
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
- Signal Transduction
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Lan Yang
- From the State Key Laboratory of Molecular Oncology
| | - Yiming Ma
- From the State Key Laboratory of Molecular Oncology
| | - Wenxiao Han
- From the State Key Laboratory of Molecular Oncology
| | - Weiwei Li
- From the State Key Laboratory of Molecular Oncology
| | - Liang Cui
- Department of Abdominal Surgical Oncology, and
| | - Xinhua Zhao
- From the State Key Laboratory of Molecular Oncology
| | - Yantao Tian
- Department of Abdominal Surgical Oncology, and
| | - Zhixiang Zhou
- Department of Gastrointestinal Surgical Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 100021 Beijing, PR China and
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191 Beijing, PR China
| | - Hongying Wang
- From the State Key Laboratory of Molecular Oncology,
| |
Collapse
|
70
|
Thompson ML, Jimenez-Andrade JM, Chartier S, Tsai J, Burton EA, Habets G, Lin PS, West BL, Mantyh PW. Targeting cells of the myeloid lineage attenuates pain and disease progression in a prostate model of bone cancer. Pain 2015; 156:1692-1702. [PMID: 25993548 PMCID: PMC4545688 DOI: 10.1097/j.pain.0000000000000228] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tumor cells frequently metastasize to bone where they can generate cancer-induced bone pain (CIBP) that can be difficult to fully control using available therapies. Here, we explored whether PLX3397, a high-affinity small molecular antagonist that binds to and inhibits phosphorylation of colony-stimulating factor-1 receptor, the tyrosine-protein kinase c-Kit, and the FMS-like tyrosine kinase 3, can reduce CIBP. These 3 targets all regulate the proliferation and function of a subset of the myeloid cells including macrophages, osteoclasts, and mast cells. Preliminary experiments show that PLX3397 attenuated inflammatory pain after formalin injection into the hind paw of the rat. As there is an inflammatory component in CIBP, involving macrophages and osteoclasts, the effect of PLX3397 was explored in a prostate model of CIBP where skeletal pain, cancer cell proliferation, tumor metastasis, and bone remodeling could be monitored in the same animal. Administration of PLX3397 was initiated on day 14 after prostate cancer cell injection when the tumor was well established, and tumor-induced bone remodeling was first evident. Over the next 6 weeks, sustained administration of PLX3397 attenuated CIBP behaviors by approximately 50% and was equally efficacious in reducing tumor cell growth, formation of new tumor colonies in bone, and pathological tumor-induced bone remodeling. Developing a better understanding of potential effects that analgesic therapies have on the tumor itself may allow the development of therapies that not only better control the pain but also positively impact disease progression and overall survival in patients with bone cancer.
Collapse
Affiliation(s)
- Michelle L. Thompson
- Department of Pharmacology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| | - Juan Miguel Jimenez-Andrade
- Department of Pharmacology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| | - Stephane Chartier
- Department of Pharmacology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| | - James Tsai
- Plexxikon, Inc., 91 Bolivar Drive, Berkeley, CA 94710, USA
| | | | - Gaston Habets
- Plexxikon, Inc., 91 Bolivar Drive, Berkeley, CA 94710, USA
| | - Paul S. Lin
- Plexxikon, Inc., 91 Bolivar Drive, Berkeley, CA 94710, USA
| | - Brian L. West
- Plexxikon, Inc., 91 Bolivar Drive, Berkeley, CA 94710, USA
| | - Patrick W. Mantyh
- Department of Pharmacology, Arizona Cancer Center, University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA
| |
Collapse
|
71
|
Nemati M, Santos A, Kumeria T, Losic D. Label-Free real-time quantification of enzyme levels by interferometric spectroscopy combined with gelatin-modified nanoporous anodic alumina photonic films. Anal Chem 2015; 87:9016-24. [PMID: 26259031 DOI: 10.1021/acs.analchem.5b02225] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Herein, we present an interferometric sensor based on the combination of chemically functionalized nanoporous anodic alumina photonic films (NAA-PFs) and reflectometric interference spectroscopy (RIfS) aimed to detect trace levels of enzymes by selective digestion of gelatin. The fabrication and sensing performance of the proposed sensor were characterized in real-time by estimating the changes in effective optical thickness (i.e., sensing principle) of gelatin-modified NAA-PFs (i.e., sensing element) during enzymatic digestion. The working range (WR), sensitivity (S), low limit of detection (LLoD), and linearity (R(2)) of this enzymatic sensor were established by a series of experiments with different concentrations of gelatin (i.e., specific chemical sensing element) and trypsin (i.e., analyte), a model protease enzyme with relevant implications as a biomarker in the diagnosis of several diseases. The chemical selectivity of the sensor was demonstrated by comparison of gelatin digestion by other nonspecific enzyme models such as chymotrypsin and horseradish peroxidase. Furthermore, the role of the chemical sensing element (i.e., gelatin) was assessed by using hemoglobin instead of gelatin. Finally, we demonstrated that this sensor can be readily used to establish the kinetic parameters of enzymatic reactions. The obtained results revealed that the presented sensor has a promising potential to be used as a point-of-care system for fast detection of gastrointestinal diseases at early stages.
Collapse
Affiliation(s)
- Mahdieh Nemati
- School of Chemical Engineering, The University of Adelaide , Engineering North Building, 5005 Adelaide, South Australia, Australia
| | - Abel Santos
- School of Chemical Engineering, The University of Adelaide , Engineering North Building, 5005 Adelaide, South Australia, Australia
| | - Tushar Kumeria
- School of Chemical Engineering, The University of Adelaide , Engineering North Building, 5005 Adelaide, South Australia, Australia
| | - Dusan Losic
- School of Chemical Engineering, The University of Adelaide , Engineering North Building, 5005 Adelaide, South Australia, Australia
| |
Collapse
|
72
|
Radhakrishnan K, Thomas MB, Pulakkat S, Gnanadhas DP, Chakravortty D, Raichur AM. Stimuli-responsive protamine-based biodegradable nanocapsules for enhanced bioavailability and intracellular delivery of anticancer agents. JOURNAL OF NANOPARTICLE RESEARCH 2015; 17:341. [DOI: 10.1007/s11051-015-3145-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
|
73
|
Chi L, Liu Q, Xu S, Xiao Z, Ma D, Li J. Maternally derived trypsin may have multiple functions in the early development of turbot (Scopthalmus maximus). Comp Biochem Physiol A Mol Integr Physiol 2015; 188:148-55. [PMID: 26159647 DOI: 10.1016/j.cbpa.2015.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/30/2015] [Accepted: 06/30/2015] [Indexed: 11/18/2022]
Abstract
Trypsin is an important serine protease that is considered to be involved in digestion of protein in teleost fish. Nevertheless, studies on trypsin/trypsinogen in fish embryos are very limited. In this study, the trypsinogen of turbot (Scophthalmus maximus) (tTG) was identified and the expression patterns and activity of trypsinogen/trypsin were investigated. The results showed that the tTG mRNA was evenly distributed in the oocytes and was also expressed along the yolk periphery in early embryos. At later embryo stages and 1 days after hatching (dph), the tTG mRNA concentrated at the alimentary tract and head. Quantitative expression analysis showed that the tTG transcripts decreased after fertilization until the gastrula stage, then increased with the embryo and larvae development. This result was also confirmed by the specific activity analysis of trypsin and in-situ-hybridization (ISH). All of the results indicated that tTG in early embryo stages was maternally derived and expressed by itself after gastrula stages. Additionally, location of tTG mRNA in embryos and larvae was investigated; we considered that trypsin may have multiple functions during the embryo development process. Based on our results regarding trypsinogen in embryos and early development, we concluded that the trypsin/trypsinogen in turbot embryos was inherited from a maternal source and we suggested that trypsin in early development has multiple functions in the process of development.
Collapse
Affiliation(s)
- Liang Chi
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Qinghua Liu
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China.
| | - Shihong Xu
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Zhizhong Xiao
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Daoyuan Ma
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Jun Li
- Center of Biotechnology R&D, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China.
| |
Collapse
|
74
|
Zeng T, Zhang T, Wei W, Li Z, Wu D, Wang L, Guo J, He X, Ma N. Compact, Programmable, and Stable Biofunctionalized Upconversion Nanoparticles Prepared through Peptide-Mediated Phase Transfer for High-Sensitive Protease Sensing and in Vivo Apoptosis Imaging. ACS APPLIED MATERIALS & INTERFACES 2015; 7:11849-56. [PMID: 25970768 DOI: 10.1021/acsami.5b01446] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Protease represents an important class of biomarkers for disease diagnostics and drug screening. Conventional fluorescence-based probes for in vivo protease imaging suffer from short excitation wavelengths and poor photostability. Upconversion nanoparticles (UCNPs) hold great promise for biosensing and bioimaging because of their deep-tissue excitability, robust photostability, and minimal imaging background. However, producing highly stable and compact biofunctionalized UCNP probes with optimal bioresponsivity for in vivo imaging of protease activities still remains challenging and has not been previously demonstrated. Herein, we report facile preparation of highly compact and stable biofunctionalized UCNPs through peptide-mediated phase transfer for high-sensitive detection of protease in vitro and in vivo. We demonstrate that the polyhistidine-containing chimeric peptides could displace oleic acid molecules capped on UCNPs synthesized in organic solvents and, thereby, directly transfer UCNPs from the chloroform phase to the water phase. The resulting UCNPs possess high stability, programmable surface properties, and a compact coating layer with minimized thickness for efficient luminescence resonance energy transfer (LRET). On the basis of this strategy, we prepared LRET-based UCNP probes with optimal bioresponsivity for in vitro high-sensitive detection of trypsin and in vivo imaging of apoptosis for chemotherapy efficacy evaluation. The reported strategy could be extended to construct a variety of peptide-functionalized UCNPs for various biomedical applications.
Collapse
Affiliation(s)
- Tao Zeng
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Tao Zhang
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Wei Wei
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Zhi Li
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Dan Wu
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Li Wang
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Jun Guo
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Xuewen He
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Nan Ma
- †Key Laboratory of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, and ‡Testing and Analysis Center, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| |
Collapse
|
75
|
Cui F, Yang K, Li Y. Investigate the binding of catechins to trypsin using docking and molecular dynamics simulation. PLoS One 2015; 10:e0125848. [PMID: 25938485 PMCID: PMC4418572 DOI: 10.1371/journal.pone.0125848] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/25/2015] [Indexed: 01/31/2023] Open
Abstract
To explore the inhibitory mechanism of catechins for digestive enzymes, we investigated the binding mode of catechins to a typical digestive enzyme-trypsin and analyzed the structure-activity relationship of catechins, using an integration of molecular docking, molecular dynamics simulation and binding free energy calculation. We found that catechins with different structures bound to a conservative pocket S1 of trypsin, which is comprised of residues 189–195, 214–220 and 225–228. In the trypsin-catechin complexes, Asp189 by forming strong hydrogen bonding, and Gln192, Trp215 and Gly216 through hydrophobic interactions, all significantly contribute to the binding of catechins. The number and the position of hydroxyl and aromatic groups, the structure of stereoisomers, and the orientation of catechins in the binding pocket S1 of trypsin all affect the binding affinity. The binding affinity is in the order of Epigallocatechin gallate (EGCG) > Epicatechin gallate (ECG) > Epicatechin (EC) > Epigallocatechin (EGC), and 2R-3R EGCG shows the strongest binding affinity out of other stereoisomers. Meanwhile, the synergic conformational changes of residues and catechins were also analyzed. These findings will be helpful in understanding the knowledge of interactions between catechins and trypsin and referable for the design of novel polyphenol based functional food and nutriceutical formulas.
Collapse
Affiliation(s)
- Fengchao Cui
- Key Laboratory of Synthetic Rubber & Laboratory of Advanced Power Sources, Changchun Institute of Applied Chemistry (CIAC), Chinese Academy of Sciences, Changchun, P. R. China
| | - Kecheng Yang
- Key Laboratory of Synthetic Rubber & Laboratory of Advanced Power Sources, Changchun Institute of Applied Chemistry (CIAC), Chinese Academy of Sciences, Changchun, P. R. China
| | - Yunqi Li
- Key Laboratory of Synthetic Rubber & Laboratory of Advanced Power Sources, Changchun Institute of Applied Chemistry (CIAC), Chinese Academy of Sciences, Changchun, P. R. China
- * E-mail:
| |
Collapse
|
76
|
Ida S, Ozaki N, Araki K, Hirashima K, Zaitsu Y, Taki K, Sakamoto Y, Miyamoto Y, Oki E, Morita M, Watanabe M, Maehara Y, Yamamura KI, Baba H, Ohmuraya M. SPINK1 Status in Colorectal Cancer, Impact on Proliferation, and Role in Colitis-Associated Cancer. Mol Cancer Res 2015; 13:1130-8. [PMID: 25804623 DOI: 10.1158/1541-7786.mcr-14-0581] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/05/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Colorectal cancer is a major cause of deaths due to cancer; therefore, research into its etiology is urgently needed. Although it is clear that chronic inflammation is a risk factor for colorectal cancer, the details remain uncertain. Serine protease inhibitor, Kazal type 1 (SPINK1) is mainly produced in pancreatic acinar cells. However, SPINK1 is expressed in various cancers and in inflammatory states, such as colon cancer and inflammatory bowel disease. There are structural similarities between SPINK1 and epidermal growth factor (EGF). Hence, it was hypothesized that SPINK1 functions as a growth factor for tissue repair in inflammatory states, and if prolonged, acts as a promoter for cell proliferation in cancerous tissues. Here, immunohistochemical staining for SPINK1 was observed in a high percentage of colorectal cancer patient specimens and SPINK1 induced proliferation of human colon cancer cell lines. To clarify its role in colon cancer in vivo, a mouse model exposed to the colon carcinogen azoxymethane and nongenotoxic carcinogen dextran sodium sulfate revealed that Spink3 (mouse homolog of SPINK1) is overexpressed in cancerous tissues. In Spink3 heterozygous mice, tumor multiplicity and tumor volume were significantly decreased compared with wild-type mice. These results suggest that SPINK1/Spink3 stimulates the proliferation of colon cancer cells and is involved in colorectal cancer progression. IMPLICATIONS Evidence suggests that SPINK1 is an important growth factor that connects chronic inflammation and cancer.
Collapse
Affiliation(s)
- Satoshi Ida
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan. Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobuyuki Ozaki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimi Araki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Kotaro Hirashima
- Department of Digestive and General Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoko Zaitsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsunobu Taki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan. Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Morita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-Ichi Yamamura
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Ohmuraya
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
77
|
Xie L, Duan Z, Liu C, Zheng Y, Zhou J. Protease-activated receptor 2 agonist increases cell proliferation and invasion of human pancreatic cancer cells. Exp Ther Med 2014; 9:239-244. [PMID: 25452809 PMCID: PMC4247309 DOI: 10.3892/etm.2014.2052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 08/20/2014] [Indexed: 01/18/2023] Open
Abstract
The aim of this study was to determine the expression of protease-activated receptor 2 (PAR-2) in the human pancreatic cancer cell line SW1990, and to evaluate its effect on cell proliferation and invasion. The expression of PAR-2 protein and mRNA in SW1990 cells was determined by immunocytochemistry and reverse transcription polymerase chain reaction (PCR), respectively. MTT and cell invasion and migration assays, as well as semi-quantitative PCR and zymography analysis, were additionally performed. PAR-2 mRNA was significantly upregulated in the cells treated with trypsin or the PAR-2 activating peptide Ser-Leu-Ile-Gly-Lys-Val (SLIGKV) (P<0.01), but not in the Val-Lys-Gly-Ile-Leu-Ser group (P>0.05). Trypsin and SLIGKV significantly promoted SW1990 cell proliferation in a dose- and time-dependent manner (P<0.05). Compared with the control group, trypsin and SLIGKV significantly increased the mRNA expression (P<0.01) and gelatinolytic activity (P<0.01) of matrix metalloproteinase (MMP)-2. In conclusion, PAR-2 is expressed in SW1990 cells. PAR-2 activation may promote the invasion and migration of human pancreatic cancer cells by increasing MMP-2 expression.
Collapse
Affiliation(s)
- Liqun Xie
- Department of Gastroenterology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Zexing Duan
- Department of Gastroenterology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China ; Hunan Provincial Corps Hospital, Chinese People's Armed Police Forces, Changsha, Hunan 410006, P.R. China
| | - Caiju Liu
- Department of Gastroenterology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Yanmin Zheng
- Department of Gastroenterology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Jing Zhou
- Department of Gastroenterology, Affiliated Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| |
Collapse
|
78
|
Clemente A, Arques MDC. Bowman-Birk inhibitors from legumes as colorectal chemopreventive agents. World J Gastroenterol 2014; 20:10305-10315. [PMID: 25132747 PMCID: PMC4130838 DOI: 10.3748/wjg.v20.i30.10305] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/21/2014] [Accepted: 04/23/2014] [Indexed: 02/06/2023] Open
Abstract
Aberrant functioning of serine proteases in inflammatory and carcinogenic processes within the gastrointestinal tract (GIT) has prompted scientists to investigate the potential of serine protease inhibitors, both natural and synthetic, as modulators of their proteolytic activities. Protease inhibitors of the Bowman-Birk type, a major protease inhibitor family in legume seeds, which inhibit potently and specifically trypsin- and chymotrypsin-like proteases, are currently being investigated as colorectal chemopreventive agents. Physiologically relevant amounts of Bowman-Birk inhibitors (BBI) can reach the large intestine in active form due to their extraordinary resistance to extreme conditions within the GIT. Studies in animal models have proven that dietary BBI from several legume sources, including soybean, pea, lentil and chickpea, can prevent or suppress carcinogenic and inflammatory processes within the GIT. Although the therapeutic targets and the action mechanism of BBI have not yet been elucidated, the emerging evidence suggests that BBI exert their preventive properties via protease inhibition; in this sense, serine proteases should be considered as primary targets in early stages of carcinogenesis. The validation of candidate serine proteases as therapeutic targets together with the identification, within the wide array of natural BBI variants, of the most potent and specific protease inhibitors, are necessary to better understand the potential of this protein family as colorectal chemopreventive agents.
Collapse
|
79
|
Fan Y, Qu X, Ma Y, Qu J, Liu Y, Hu X. Cbl-b accelerates trypsin-induced cell detachment through ubiquitination and degradation of proline-rich tyrosine kinase 2. Tumour Biol 2014; 35:11129-35. [DOI: 10.1007/s13277-014-2296-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/30/2014] [Indexed: 11/30/2022] Open
|
80
|
Kim H, Ng CYW, Algar WR. Quantum dot-based multidonor concentric FRET system and its application to biosensing using an excitation ratio. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:5676-5685. [PMID: 24810095 DOI: 10.1021/la501102x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
A plethora of semiconductor quantum dot (QD)-based probes that rely on Förster resonance energy transfer (FRET) have been developed for the optical detection of a wide array of biological targets. To date, the vast majority of these probes have utilized one-step energy transfer between individual donor-acceptor pairs. Here, we report a new multidonor concentric FRET configuration that comprised two fluorescent dyes assembled around a central CdSeS/ZnS QD through peptide linkers. One of these dyes, either Alexa Fluor 555 (A555) or Alexa Fluor 647 (A647), served as an acceptor for both the central QD and the other coassembled dye, Alexa Fluor 488 (A488). The unresolved emission between the A488 and the QD precluded a standard analysis of FRET efficiency from quenching of donor emission intensity or decay time, instead necessitating an analysis of the two energy transfer pathways from deconvolved excitation spectra. When A647 was the terminal acceptor, both the QD-to-A647 and A488-to-A647 energy transfer pathways could be interrogated with blue light, but only the former could be interrogated with violet light. The different degrees of A647 sensitization between these two excitation wavelengths was a predictable function of the above energy transfer efficiencies and dye stoichiometry, and was exploited for quantitative bioanalysis through an excitation ratio, which is in contrast to the conventional use of an emission ratio with FRET-based probes. Detection of the activity of nanomolar concentrations of trypsin, a model protease that hydrolyzed the A488-labeled peptide linker, was demonstrated using both a fluorescence plate reader and a low-cost, compact device that used two low-power light-emitting diodes (LEDs) as excitation sources and a silicon photodiode to detect A647 emission. This multidonor concentric FRET configuration represents a new modality for ratiometric biosensing with QDs and is potentially useful for portable in vitro diagnostics.
Collapse
Affiliation(s)
- Hyungki Kim
- Department of Chemistry, University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | | | | |
Collapse
|
81
|
Malfettone A, Silvestris N, Saponaro C, Ranieri G, Russo A, Caruso S, Popescu O, Simone G, Paradiso A, Mangia A. High density of tryptase-positive mast cells in human colorectal cancer: a poor prognostic factor related to protease-activated receptor 2 expression. J Cell Mol Med 2014; 17:1025-37. [PMID: 23991686 PMCID: PMC3780541 DOI: 10.1111/jcmm.12073] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 04/01/2013] [Indexed: 12/11/2022] Open
Abstract
Tryptase(+) mast cells (MCs), abundant in the invasive front of tumours, contribute to tissue remodelling. Indeed, protease-activated receptor-2 (PAR-2) activation by MC-tryptase is considered an oncogenic event in colorectal cancer (CRC). Recently, we have suggested NHERF1 as a potential new marker in CRC. In this study, we aimed to determine the distribution of tryptase(+) MCs and PAR-2 and to examine the relationship between PAR-2 and NHERF1, investigating their reputed usefulness as tumour markers. We studied a cohort of 115 CRC specimens including primary cancer (C) and adjacent normal mucosa (NM) by immunohistochemical double staining, analyzing the protein expression of MC-tryptase, PAR-2 and cytoplasmic NHERF1. MC density was higher in NM than in C. Tumours with high TNM stage and poor grade showed the highest MC density. A higher PAR-2 immunoreactivity characterized tumours most infiltrated by MCs compared with samples with low MC density. Furthermore, PAR-2 overexpression was associated with advanced TNM stage, poor grade and lymphovascular invasion (LVI). A positive correlation existed between tryptase(+) MC density and PAR-2 expression. Cytoplasmic NHERF1 was higher in C than in NM and overexpressing tumours resulted associated with nodal and distant metastases, poor grade and LVI. PAR-2 correlated with cytoplasmic NHERF1 and the PAR-2(+)/cytoplasmic NHERF1(+) expression immunophenotype identified tumours associated with unfavourable prognosis and aggressive clinical parameters. Our data indicate that the high density of tryptase(+) MCs at invasive margins of tumours was associated with advanced stages of CRC and was strongly correlated with PAR-2 expression.
Collapse
Affiliation(s)
- Andrea Malfettone
- Functional Biomorphology Laboratory, National Cancer Research Centre, Giovanni Paolo II, Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Genovese L, Zawada L, Tosoni A, Ferri A, Zerbi P, Allevi R, Nebuloni M, Alfano M. Cellular localization, invasion, and turnover are differently influenced by healthy and tumor-derived extracellular matrix. Tissue Eng Part A 2014; 20:2005-18. [PMID: 24498848 DOI: 10.1089/ten.tea.2013.0588] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The interplay between tumor cells and the microenvironment has been recognized as one of the hallmarks of cancer biology. To assess the role of extracellular matrix (ECM) in the modulation of tissue homeostasis and tumorigenesis, we developed a protocol for the purification of tissue-derived ECM using mucosae from healthy human colon, perilesional area, and colorectal carcinoma (CRC). Matched specimens were collected from the left colon of patients undergoing CRC resection surgery. ECMs were obtained from tissues that were decellularized with hypotonic solutions containing ionic and nonionic detergents, hypertonic solution, and endonuclease in the absence of denaturing agents. Mucosae-derived ECMs maintained distribution and localization of proteins and glycoproteins typical of the original tissues, and showed different three-dimensional (3D) structures among normal versus perilesional and tumor-derived stroma. The three types of ECM differentially regulated the localization and organization of seeded monocytes and cancer cells that were located and organized as in the original tissue. Specifically, healthy, perilesional, and CRC-derived ECMs sustained differentiation and polarization of cancer epithelial cells. In addition, healthy, but not perilesional and CRC-derived ECM constrained invasion of cancer cells. All three ECMs sustained turnover between cell proliferation and death up to 40 days of culture, although each ECM showed different ability in supporting cell proliferation, with tumor>perilesional>healthy-derived ECMs. Healthy-, perilesional- and CRC-derived ECM differently modulated cell homeostasis, spreading in the stroma and turnover between proliferation and death, and equally supported differentiation and polarization of cancer epithelial cells, thus highlighting the contribution of different ECMs modulating some features of tissue homeostasis and tumorigenesis. Moreover, these ECMs provide competent scaffolds useful to assess efficacy of antitumor drugs in a 3D setting that more closely recapitulates the native microenvironment. Further, ECM-based scaffolds may also be beneficial for future studies seeking prognostic and diagnostic stromal markers and targets for antineoplastic drugs.
Collapse
Affiliation(s)
- Luca Genovese
- 1 School of Medicine, Vita-Salute San Raffaele University , Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Proteinase-activated receptors differentially modulate in vitro invasion of human pancreatic adenocarcinoma PANC-1 cells in correlation with changes in the expression of CDC42 protein. Pancreas 2014; 43:103-8. [PMID: 23921961 PMCID: PMC3843996 DOI: 10.1097/mpa.0b013e31829f0b81] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Proteinase-activated receptor-1 (PAR-1) and PAR-2 have been associated with increased invasiveness and metastasis in human malignancies. The role of PAR-3 has been less investigated. We examined the role of PARs in a human pancreatic adenocarcinoma PANC-1 cell line phenotype in vitro. METHODS We knocked down PAR-1, PAR-2, or PAR-3, whereas empty vector-infected cells served as controls. Specific peptide agonists of PARs were used to stimulate the receptors. In vitro assays of colony formation, migration, and invasion were used to characterize the phenotypes, and Western analysis was used to follow cell division control protein 42 homolog (CDC42) expression. RESULTS PAR-1 and PAR-2 knockdowns (KDs) were markedly less, whereas PAR-3 KDs were robustly more migratory and invasive than the controls. Stimulation of PAR-1 or PAR-2 by their peptide agonists increased, whereas PAR-3 agonist reduced the invasion of the control cells. Knockdowns of all three PARs exhibited changes in the expression of CDC42, which correlated with the changes in their invasion. Conversely, stimulation of vector-control cells with PAR-1 or PAR-2 agonists enhanced, whereas PAR-3 agonist reduced the expression of CDC42. In the respective KD cells, the effects of the agonists were abrogated. CONCLUSION The expression and/or activation of PARs is linked to the invasiveness of PANC-1 cells in vitro, probably via modulation of the expression of CDC42.
Collapse
|
84
|
He X, Ma N. Biomimetic synthesis of fluorogenic quantum dots for ultrasensitive label-free detection of protease activities. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:2527-2531. [PMID: 23420767 DOI: 10.1002/smll.201202570] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/06/2013] [Indexed: 06/01/2023]
Abstract
A biomimetic quantum dot synthesis-based strategy for ultrasensitive label-free detection of protease activities is reported. A dithiol peptide substrate can be activated by the protease through cleavage to form monothiol peptides, which then triggers QD growth and generates a photoluminescence signal readout. As low as 0.8 nM trypsin can be detected directly in buffer and serum and 4 pM trypsin can be detected via trypsinogen amplification with high signal to background ratios.
Collapse
Affiliation(s)
- Xuewen He
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | | |
Collapse
|
85
|
White MJV, Glenn M, Gomer RH. Trypsin potentiates human fibrocyte differentiation. PLoS One 2013; 8:e70795. [PMID: 23951012 PMCID: PMC3737277 DOI: 10.1371/journal.pone.0070795] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 06/22/2013] [Indexed: 02/07/2023] Open
Abstract
Trypsin-containing topical treatments can be used to speed wound healing, although the mechanism of action is unknown. To help form granulation tissue and heal wounds, monocytes leave the circulation, enter the wound tissue, and differentiate into fibroblast-like cells called fibrocytes. We find that 20 to 200 ng/ml trypsin (concentrations similar to those used in wound dressings) potentiates the differentiation of human monocytes to fibrocytes in cell culture. Adding trypsin inhibitors increases the amount of trypsin needed to potentiate fibrocyte differentiation, suggesting that the potentiating effect is dependent on trypsin proteolytic activity. Proteases with other site specificities such as pepsin, endoprotease GluC, and chymotrypsin do not potentiate fibrocyte differentiation. This potentiation requires the presence of albumin in the culture medium, and tryptic fragments of human or bovine albumin also potentiate fibrocyte differentiation. These results suggest that topical trypsin speeds wound healing by generating tryptic fragments of albumin, which in turn potentiate fibrocyte differentiation.
Collapse
Affiliation(s)
- Michael J. V. White
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Melissa Glenn
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
86
|
Vihinen P, Ala-Aho R, Kähäri VM. Diagnostic and prognostic role of matrix metalloproteases in cancer. ACTA ACUST UNITED AC 2013; 2:1025-39. [PMID: 23495924 DOI: 10.1517/17530059.2.9.1025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Matrix metalloproteases (MMPs) are key players in the progression and metastasis of cancer. MMPs cleave extracellular matrix components and in this way promote tumor growth, invasion and vascularization. MMPs also affect tumor progression by regulating availability and activity of growth factors, inflammatory cytokines and chemokines. Accordingly, several MMPs have been found to serve as prognostic indicators in solid tumors. Usually the increased levels of MMPs in patients' tumor tissue or serum/plasma are associated with poor outcome. Interestingly, recent results show that certain MMPs also serve as tumor suppressors. OBJECTIVE This review discusses the latest view on MMPs as diagnostic and prognostic indicators in cancer patients. METHODS Studies with clinical samples of 70 or more patients are included in particular. In addition, the possible roles of MMPs in future molecular diagnostics and in the evaluation of therapeutic responses are discussed. CONCLUSION MMP-9 in particular has shown prognostic value in various types of tumor, and its measurement in circulation, urine or tumor tissue might help in clinical surveillance of otherwise problematic patient cases. There is upcoming new knowledge on MMPs in therapy response evaluation, in which MMPs might be useful together with CT scans and other clinically more established prognostic factors. Certain MMPs have a dual role in terms of cancer-modulating properties and thus it is essential to evaluate their expression and function in tumor cells and host environment to select validated therapy targets but spare MMP antitargets.
Collapse
Affiliation(s)
- Pia Vihinen
- Turku University Hospital, Department of Oncology and Radiotherapy, POB 52, FIN-20521 Turku, Finland +358 2 313 0804 ; +358 2 313 2809 ;
| | | | | |
Collapse
|
87
|
Gam LH. Breast cancer and protein biomarkers. World J Exp Med 2012; 2:86-91. [PMID: 24520539 PMCID: PMC3905586 DOI: 10.5493/wjem.v2.i5.86] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 06/28/2012] [Accepted: 10/07/2012] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is a healthcare concern of women worldwide. Despite procedures being available for diagnosis, prognosis and treatment of breast cancer, researchers are working intensively on the disease in order to improve the life quality of breast cancer patients. At present, there is no single treatment known to bring a definite cure for breast cancer. One of the possible solutions for combating breast cancer is through identification of reliable protein biomarkers that can be effectively used for early detection, prognosis and treatments of the cancer. Therefore, the task of identification of biomarkers for breast cancer has become the focus of many researchers worldwide.
Collapse
Affiliation(s)
- Lay-Harn Gam
- Lay-Harn Gam, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| |
Collapse
|
88
|
Hirota CL, Moreau F, Iablokov V, Dicay M, Renaux B, Hollenberg MD, MacNaughton WK. Epidermal growth factor receptor transactivation is required for proteinase-activated receptor-2-induced COX-2 expression in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2012; 303:G111-9. [PMID: 22517768 DOI: 10.1152/ajpgi.00358.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Proteinase-activated receptor (PAR)(2), a G protein-coupled receptor activated by serine proteinases, has been implicated in both intestinal inflammation and epithelial proliferation. Cyclooxygenase (COX)-2 is overexpressed in the gut during inflammation as well as in colon cancer. We hypothesized that PAR(2) drives COX-2 expression in intestinal epithelial cells. Treatment of Caco-2 colon cancer cells with the PAR(2)-activating peptide 2-furoyl-LIGRLO-NH(2) (2fLI), but not by its reverse-sequence PAR(2)-inactive peptide, for 3 h led to an increase in intracellular COX-2 protein expression accompanied by a COX-2-dependent increase in prostaglandin E(2) production. 2fLI treatment for 30 min significantly increased metalloproteinase activity in the culture supernatant. Increased epidermal growth factor receptor (EGFR) phosphorylation was observed in cell lysates following 40 min of treatment with 2fLI. The broad-spectrum metalloproteinase inhibitor marimastat inhibited both COX-2 expression and EGFR phosphorylation. The EGFR tyrosine kinase inhibitor PD153035 also abolished 2fLI-induced COX-2 expression. Although PAR(2) activation increased ERK MAPK phosphorylation, neither ERK pathway inhibitors nor a p38 MAPK inhibitor affected 2fLI-induced COX-2 expression. However, inhibition of either Src tyrosine kinase signaling by PP2, Rho kinase signaling by Y27632, or phosphatidylinositol 3 (PI3) kinase signaling by LY294002 prevented 2fLI-induced COX-2 expression. Trypsin increased COX-2 expression through PAR(2) in Caco-2 cells and in an EGFR-dependent manner in the noncancerous intestinal epithelial cell-6 cell line. In conclusion, PAR(2) activation drives COX-2 expression in Caco-2 cells via metalloproteinase-dependent EGFR transactivation and activation of Src, Rho, and PI3 kinase signaling. Our findings provide a mechanism whereby PAR(2) can participate in the progression from chronic inflammation to cancer in the intestine.
Collapse
Affiliation(s)
- Christina L Hirota
- Dept. of Physiology and Pharmacology, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada
| | | | | | | | | | | | | |
Collapse
|
89
|
Molecular alterations associated with osteosarcoma development. Sarcoma 2012; 2012:523432. [PMID: 22448123 PMCID: PMC3289857 DOI: 10.1155/2012/523432] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/02/2011] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma is the most frequent malignant primary bone tumor characterized by a high potency to form lung metastases which is the main cause of death. Unfortunately, the conventional chemotherapy is not fully effective on osteosarcoma metastases. The progression of a primary tumor to metastasis requires multiple processes, which are neovascularization, proliferation, invasion, survival in the bloodstream, apoptosis resistance, arrest at a distant organ, and outgrowth in secondary sites. Consequently, recent studies have revealed new insights into the molecular mechanisms of metastasis development. The understanding of the mechanism of molecular alterations can provide the identification of novel therapeutic targets and/or prognostic markers for osteosarcoma treatment to improve the clinical outcome.
Collapse
|
90
|
Targeting proteinase-activated receptors: therapeutic potential and challenges. Nat Rev Drug Discov 2012; 11:69-86. [PMID: 22212680 DOI: 10.1038/nrd3615] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.
Collapse
|
91
|
Viljoen A, Patrick JT. In search for a better marker of acute pancreatitis: third time lucky? Clin Chem 2011; 57:1471-3. [PMID: 21920916 DOI: 10.1373/clinchem.2011.173385] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
92
|
Gratio V, Loriot C, Virca GD, Oikonomopoulou K, Walker F, Diamandis EP, Hollenberg MD, Darmoul D. Kallikrein-related peptidase 14 acts on proteinase-activated receptor 2 to induce signaling pathway in colon cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2625-36. [PMID: 21907696 DOI: 10.1016/j.ajpath.2011.07.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 07/07/2011] [Accepted: 07/26/2011] [Indexed: 12/16/2022]
Abstract
Serine proteinases participate in tumor growth and invasion by cleaving and activating proteinase-activated receptors (PARs). Recent studies have implicated PAR-1 and PAR-4 (activated by thrombin) and PAR-2 (activated by trypsin but not by thrombin) in human colon cancer growth. The endogenous activators of PARs in colon tumors, however, are still unknown. We hypothesize that the kallikrein-related peptidase (KLK) family member KLK14, a known tumor biomarker, is produced by colonic tumors and signals to human colon cancer cells by activating PARs. We found that i) KLK14 mRNA was present in 16 human colon cancer cell lines, ii) KLK14 protein was expressed and secreted in colon cancer cell lines, and iii) KLK14 (0.1 μmol/L) induced increases in intracellular calcium in HT29, a human colon cancer-derived cell line. KLK14-induced calcium flux was associated with internalization of KLK14-mediated activation of PAR-2. Furthermore, KLK14 induced significant extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation and HT29 cell proliferation, presumably by activating PAR-2. A PAR-2 cleavage and activation-blocking antibody dramatically reduced KLK14-induced ERK1/2 signaling. Finally, ectopic expression of KLK14 in human colon adenocarcinomas and its absence in normal epithelia was demonstrated by IHC analysis. These results demonstrate, for the first time, the aberrant expression of KLK14 in colon cancer and its involvement in PAR-2 receptor signaling. Thus, KLK14 and its receptor, PAR-2, may represent therapeutic targets for colon tumorigenesis.
Collapse
Affiliation(s)
- Valérie Gratio
- Institut National de la Santé et de la Recherche Médicale (INSERM) U773, Centre de Recherche Biomédicale Bichat-Beaujon, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Kuang L, Wang L, Wang Q, Zhao Q, Du B, Li D, Luo J, Liu M, Hou A, Qian M. Cudratricusxanthone G inhibits human colorectal carcinoma cell invasion by MMP-2 down-regulation through suppressing activator protein-1 activity. Biochem Pharmacol 2011; 81:1192-200. [DOI: 10.1016/j.bcp.2011.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 02/23/2011] [Accepted: 02/23/2011] [Indexed: 12/16/2022]
|
94
|
Global expression study in colorectal cancer on proteins with alkaline isoelectric point by two-dimensional difference gel electrophoresis. J Proteomics 2011; 74:858-73. [PMID: 21385629 DOI: 10.1016/j.jprot.2011.02.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 02/24/2011] [Accepted: 02/26/2011] [Indexed: 01/01/2023]
Abstract
Colorectal cancer is one of the leading causes of cancer death worldwide. To identify candidates for biomarkers and therapeutic targets, we investigated the proteome of colorectal cancer tissues. Using 2D-DIGE in combination with our original large format electrophoresis apparatus, we compared surgically resected normal and tumor tissues from 53 patients with colorectal cancer. We focused on proteins with an alkaline pI using IPG gels for the alkaline range. We observed 1687 protein spots, and found 100 spots with statistical (p<0.01) and significant (>2-fold) differences between the normal and the tumor tissue groups. Among these 100 protein spots, five showed a different intensity between tumor tissues from the stage-II and the stage-III patients. MS experiments revealed that these 100 protein spots corresponded to 58 unique proteins. These included six proteins which had not been previously reported to be associated with colorectal cancer. Among these proteins, five were not reported in any type of malignancy. IEF/western blotting confirmed the differences in protein expression between the normal and the tumor tissues. These results may provide an insight for biomarker development and drug target discovery in colorectal cancer.
Collapse
|
95
|
Abstract
Ongoing and breakthrough pain is a primary concern for the cancer patient. Although the etiology of cancer pain remains unclear, animal models of cancer pain have allowed investigators to unravel some of the cancer-induced neuropathologic processes that occur in the region of tumor growth and in the dorsal horn of the spinal cord. Within the cancer microenvironment, cancer and immune cells produce and secrete mediators that activate and sensitize primary afferent nociceptors. Pursuant to these peripheral changes, nociceptive secondary neurons in spinal cord exhibit increased spontaneous activity and enhanced responsiveness to three modes of noxious stimulation: heat, cold, and mechanical stimuli. As our understanding of the peripheral and central mechanisms that underlie cancer pain improves, targeted analgesics for the cancer patient will likely follow.
Collapse
Affiliation(s)
- Brian L Schmidt
- Department of Oral and Maxillofacial Surgery, School of Dentistry, University of California San Francisco, USA.
| | | | | | | |
Collapse
|
96
|
Olszewski U, Zeillinger R, Geissler K, Hamilton G. Genome-wide gene expression analysis of chemoresistant pulmonary carcinoid cells. LUNG CANCER-TARGETS AND THERAPY 2010; 1:107-117. [PMID: 28210111 DOI: 10.2147/lctt.s12874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
PURPOSE Carcinoids are highly chemoresistant tumors associated with a dismal prognosis. This study involved a comparison of the genome-wide gene expression pattern of a chemoresistant and a chemosensitive pulmonary carcinoid cell line to reveal factors that contribute to the resistant phenotype. MATERIALS AND METHODS Gene expression of UMC-11 chemoresistant carcinoid cells as assessed by 32 K microarray was compared with H835 chemosensitive carcinoid cells, and the genes that were differentially expressed and expected to be related to chemoresistance were selected. RESULTS Drug-resistant UMC-11 cells exhibited increased expression of transcripts known to confer resistance to different cytostatics such as P-glycoprotein, multidrug resistance-associated proteins 2 and 3, effectors of the glutathione detoxification and xenobiotics degradation pathways, and ion transporters including Na+/K+-adenosine triphosphatase. In addition, enhanced transcription of several S100 proteins, capable of suppressing apoptosis, regulation of topoisomerase I (topo I) expression by antisense transcripts from TOPO1 pseudogenes, and alterations of the cytoskeleton seem to contribute to the multidrug-resistant phenotype. A multitude of epidermal growth factor (EGF)-related and neuropeptide growth factors, overexpression of proteases, and appearance of aerobic glycolytic metabolism complement the malignant phenotype of the UMC-11 cells. CONCLUSION The multidrug-resistant phenotype of the UMC-11 pulmonary carcinoid cell line seems to be mediated by classical efflux pumps, drug metabolization or conjugation systems, and, possibly, modulation of apoptotic cell death by S100 proteins and topo I expression by pseudogene transcripts. Autocrine or paracrine stimulation by a host of EGF-related and neuropeptide growth factors, as well as high metastatic potency indicated by increased expression of components of aerobic glycolysis and proteolytic enzymes, may furthermore account for the failure of therapeutic interventions.
Collapse
Affiliation(s)
- Ulrike Olszewski
- Ludwig Boltzmann Cluster of Translational Oncology, Ludwig Boltzmann Society, Vienna, Austria
| | - Robert Zeillinger
- Ludwig Boltzmann Cluster of Translational Oncology, Ludwig Boltzmann Society, Vienna, Austria
| | - Klaus Geissler
- Ludwig Boltzmann Cluster of Translational Oncology, Ludwig Boltzmann Society, Vienna, Austria
| | - Gerhard Hamilton
- Ludwig Boltzmann Cluster of Translational Oncology, Ludwig Boltzmann Society, Vienna, Austria
| |
Collapse
|
97
|
Expression of protease activated receptor-2 in human colorectal cancer and its association with tumor progression. Dis Colon Rectum 2010; 53:1202-8. [PMID: 20628286 DOI: 10.1007/dcr.0b013e3181d536f6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE Protease activated receptor-2 has been reported to promote the proliferation of colorectal cancer cells. The aim of this study was to investigate the relationship of protease activated receptor-2 expression to clinicopathologic factors to clarify its role in tumor progression and patient survival in human colorectal cancer. METHODS Immunohistochemical staining with anti-protease activated receptor-2 antibody was carried out and evaluated on samples from 295 patients with colorectal cancer obtained during surgical treatment at Kangnam St. Mary's Hospital, Seoul, Republic of Korea. Medical records were reviewed and relationships between immunostaining and clinicopathologic findings were evaluated. RESULTS Positive staining for protease activated receptor-2 was observed in samples from 100 patients (33.9%). Expression of protease activated receptor-2 was significantly associated with depth of tumor invasion (P = .013), liver metastasis (P = .008), TNM stage (P = .009), and tumor location (P = .011). However, protease activated receptor-2 expression did not significantly correlate with overall survival of patients and was not a significant negative prognostic factor for survival in univariate or multivariate analyses. CONCLUSIONS The expression of protease activated receptor-2 is related to tumor invasion and metastasis in human colorectal cancer. However, further studies are needed to clarify the role of protease activated receptor-2 in the prognosis of patients with colorectal cancer.
Collapse
|
98
|
Tahara K, Mimori K, Iinuma H, Iwatsuki M, Yokobori T, Ishii H, Anai H, Kitano S, Mori M. Serum matrix-metalloproteinase-1 is a bona fide prognostic marker for colorectal cancer. Ann Surg Oncol 2010; 17:3362-9. [PMID: 20617463 DOI: 10.1245/s10434-010-1149-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) are involved in the degradation of extracellular matrix components and are associated with invasion and metastasis. MMP proteins could be serum tumor markers or molecular targets in the treatment of malignancy. The purpose of the current study was to identify a prognostic serum marker in cases of colorectal cancer (CRC) prior to surgical intervention. MATERIALS AND METHODS Laser microdissection and microarray analysis were used to characterize gene expression in 73 cases of CRC. We then focused on expression of MMP-1. We examined serum MMP-1 activity before resection in another subset of 75 cases of CRC to validate the clinical significance of MMP-1 as a prognostic marker in CRC after surgically curative operation. RESULTS Disease-free survival was 51% in the MMP-1 high expression group and 81% in the low-expression group (P < .05). Survival was 52% in the MMP-1 high expression group and 90% in the low group (P < .05). In multivariate analysis for disease-free survival, MMP-1 and lymph node metastasis were significant independent prognostic indicators. In multivariate analysis of overall survival, serum MMP-1 level was the only significant independent indicator among factors. CONCLUSIONS Within the MMP family of proteins, MMP-1 is not a cancer-specific protease. However, MMP-1 activity does predict the future course of progression of malignant cells. Thus, MMP-1, which is activated at the primary lesion and is found in serum, assists in the clinical diagnosis of CRC. It is also an important molecule for understanding the underlying mechanism of invasion and metastasis of CRC.
Collapse
Affiliation(s)
- Kouichirou Tahara
- Department of Surgical Oncology, Medical Institute of Bioregulation, Kyushu University, Beppu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Array comparative genomic hybridization-based characterization of genetic alterations in pulmonary neuroendocrine tumors. Proc Natl Acad Sci U S A 2010; 107:13040-5. [PMID: 20615970 DOI: 10.1073/pnas.1008132107] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The goal of this study was to characterize and classify pulmonary neuroendocrine tumors based on array comparative genomic hybridization (aCGH). Using aCGH, we performed karyotype analysis of 33 small cell lung cancer (SCLC) tumors, 13 SCLC cell lines, 19 bronchial carcinoids, and 9 gastrointestinal carcinoids. In contrast to the relatively conserved karyotypes of carcinoid tumors, the karyotypes of SCLC tumors and cell lines were highly aberrant. High copy number (CN) gains were detected in SCLC tumors and cell lines in cytogenetic bands encoding JAK2, FGFR1, and MYC family members. In some of those samples, the CN of these genes exceeded 100, suggesting that they could represent driver alterations and potential drug targets in subgroups of SCLC patients. In SCLC tumors, as well as bronchial carcinoids and carcinoids of gastrointestinal origin, recurrent CN alterations were observed in 203 genes, including the RB1 gene and 59 microRNAs of which 51 locate in the DLK1-DIO3 domain. These findings suggest the existence of partially shared CN alterations in these tumor types. In contrast, CN alterations of the TP53 gene and the MYC family members were predominantly observed in SCLC. Furthermore, we demonstrated that the aCGH profile of SCLC cell lines highly resembles that of clinical SCLC specimens. Finally, by analyzing potential drug targets, we provide a genomics-based rationale for targeting the AKT-mTOR and apoptosis pathways in SCLC.
Collapse
|
100
|
Lefkowitz RB, Schmid-Schönbein GW, Heller MJ. Whole blood assay for trypsin activity using polyanionic focusing gel electrophoresis. Electrophoresis 2010; 31:2442-51. [DOI: 10.1002/elps.201000011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|