1
|
Aylar D, Karatug Kacar A. Studies on Treatment Within the Scope of Medical Biotechnology for Pancreatic Diseases. Mol Biotechnol 2025; 67:1321-1335. [PMID: 38627328 DOI: 10.1007/s12033-024-01142-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/14/2024] [Indexed: 03/22/2025]
Abstract
The pancreas is made of two compartments: the exocrine pancreas, a source of digestive enzymes, and the endocrine islets which produce vital hormones. Distinct diseases could arise in the pancreas such as diabetes, neuroendocrine tumors, pancreatitis, and pancreatic cancers. Various treatment methods are being researched against these diseases. Treatment with recombinant proteins, therapeutic antibodies, vaccination, gene therapy, tissue engineering, and stem cell treatment are treatment methods. Furthermore, biomarkers are important for both treatment and diagnosis. However, some of the treatment methods mentioned above have not yet been applied to some pancreatic diseases. This review provides insights into the latest advancements in diagnosis and treatment for pancreatic diseases within the scope of medical biotechnology. In addition, some methods that are not yet used for treatment purposes for pancreatic diseases but are used in other diseases that occur in different organs due to similar reasons have been investigated. In this context, possible diagnosis and treatment methods for pancreatic diseases are interpreted. The first aim of this review is to bring together and present the current diagnosis and treatment methods for pancreatic diseases. The second aim is to highlight methods that may have treatment potential by comparing pancreatic diseases that cannot be treated with similar diseases.
Collapse
Affiliation(s)
- Dilara Aylar
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ayse Karatug Kacar
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey.
| |
Collapse
|
2
|
Zhu L, Ao L, Guo Z, Yang Y, Wang Z, Gu Z, Xin Y, Zhou L, Zhang L. Recombinant canstatin inhibits the progression of hepatocellular carcinoma by repressing the HIF-1α/VEGF signaling pathway. Biomed Pharmacother 2024; 179:117423. [PMID: 39260328 DOI: 10.1016/j.biopha.2024.117423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Hepatocellular carcinoma (HCC), a hypervascular tumor, is the most frequent primary malignant tumor of the liver. Angiogenesis inhibitors, such as endogenous angiogenesis inhibitors, are essential for HCC therapy and have generated significant interest owing to their safety, efficacy, and multitargeting attributes. Canstatin is an angiogenesis inhibitor derived from the basement membrane and exerts anti-tumor effects. However, the inhibitory effects and underlying mechanisms of action of canstatin on HCC remain unclear. Therefore, in this study, HepG2 and Huh7 cells were used to investigate the inhibitory effects of recombinant canstatin on HCC cells. Subsequently, the biosafety and inhibitory effects of recombinant canstatin on tumor growth were investigated in a xenograft animal model of liver cancer. Canstatin inhibited the growth of liver cancer cells by regulating their proliferation, apoptosis, and migration. Additionally, it suppressed the occurrence and progression of HCC by modulating the HIF-1α/VEGF signaling pathway. In mice, canstatin exerted no discernible harmful side effects and suppressed the growth of HCC subcutaneous xenograft tumors. Overall, our findings shed light on the molecular pathways underlying canstatin-induced HCC cell death that may help develop novel HCC treatments.
Collapse
Affiliation(s)
- Lingyu Zhu
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Long Ao
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Zitao Guo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yuliang Yang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Zilong Wang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Zhenghua Gu
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Yu Xin
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China
| | - Leyuan Zhou
- Department of Radiation Oncology, the Fourth Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Liang Zhang
- Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; JITRI Future Food Technol Res Inst Co Ltd, Yixing 214200, China.
| |
Collapse
|
3
|
Fuller AM, Eisinger-Mathason TSK. Context Matters: Response Heterogeneity to Collagen-Targeting Approaches in Desmoplastic Cancers. Cancers (Basel) 2022; 14:cancers14133132. [PMID: 35804902 PMCID: PMC9264969 DOI: 10.3390/cancers14133132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary A common feature of tumor types such as breast cancer, prostate cancer, pancreatic cancer, and soft-tissue sarcoma is the deposition of collagen-rich tissue called desmoplasia. However, efforts to control tumor growth by disrupting desmoplasia, collectively known as “collagen-targeting approaches”, have had mixed and contradictory results, sometimes even within the same cancer type. We believe that this phenomenon may be due—at least partially—to the fact that “collagen” is not a single molecule, but rather a diverse molecular family composed of 28 unique collagen types. Therefore, in this review, we discuss the diversity of collagen molecules in normal and cancer tissue, and explore how collagen heterogeneity relates to the mixed efficacy of collagen-targeting approaches for cancer therapy. Abstract The deposition of collagen-rich desmoplastic tissue is a well-documented feature of the solid tumor microenvironment (TME). However, efforts to target the desmoplastic extracellular matrix (ECM) en masse, or collagen molecules more specifically, have been met with mixed and sometimes paradoxical results. In this review, we posit that these discrepancies are due—at least in part—to the incredible diversity of the collagen superfamily. Specifically, whereas studies of “collagen-targeting” approaches frequently refer to “collagen” as a single molecule or relatively homogeneous molecular family, 28 individual collagens have been identified in mammalian tissues, each with a unique structure, supramolecular assembly pattern, tissue distribution, and/or function. Moreover, some collagen species have been shown to exert both pro- and anti-neoplastic effects in the desmoplastic TME, even within the same cancer type. Therefore, herein, we describe the diversity of the collagen family in normal tissues and highlight the context-specific roles of individual collagen molecules in desmoplastic tumors. We further discuss how this heterogeneity relates to the variable efficacy of “collagen-targeting” strategies in this setting and provide guidance for future directions in the field.
Collapse
|
4
|
Chamani R, Taleqani MH, Imanpour A, Khatami M. New insights into short peptides derived from the collagen NC1 α1, α2, and α3 (IV) domains: An experimental and MD simulations study. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140769. [PMID: 35151893 DOI: 10.1016/j.bbapap.2022.140769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
Abstract
Proteolytically cleavage of the collagen NC1 α1 to α3 (IV) domains leads to antiangiogenic proteins called Arresten, Canstatin, and Tumstatin, respectively. The research identified that the two overlapping peptides derived from Tumstatin are more effective than other fragments and amino acids L78, V82, and D84 are essential for their activity. In the present study, the efficacy of a nine amino acid peptide derived from Tumstatin (Tum), containing amino acids L78, V82, and D84 was compared to the corresponding sequence in Arresten (Ars) and Canstatin (Can) in vitro and in vivo. Moreover, CD spectroscopy, MD, and docking simulations were performed to evaluate the structure and the interaction of peptides to integrin αvβ3, respectively. Results demonstrated that peptides inhibit viability, migration, and tube formation in vitro, as well as the growth of tumor in vivo and Canstatin-derived peptide was more potent than others. CD measurement and DSSP calculation revealed that Can had more coil conformation. According to MD simulations, Can had more fluctuation, less intramolecular interactions, and less structural compactness compared to Tum and Ars. It can be assumed that amino acid variations lead to a more flexible and loose structure compared to the other peptides. The Canstatin-derived peptide interacts with the integrin αvβ3 extremely close to RGD binding site by the most negative binding energy and more interactions. In conclusion, we for the first time identified an active peptide derived from Canstatin and showed that the sequence affected structure and thereby interaction of peptide to its receptor.
Collapse
Affiliation(s)
- Reyhane Chamani
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran.
| | | | - Aylar Imanpour
- Department of Fisheries, Faculty of Natural Resources, Urmia University, Urmia, Iran
| | - Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| |
Collapse
|
5
|
Sugiyama A, Okada M, Otani K, Yamawaki H. [Development of basic research toward clinical application of cleaved fragment of type IV collagen]. Nihon Yakurigaku Zasshi 2021; 156:282-287. [PMID: 34470932 DOI: 10.1254/fpj.21016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Basement membrane is a dense sheet-like extracellular matrix (ECM), which separates cells from surrounding interstitium. Type IV collagen is a major component of basement membrane and three of six α chains (namely α1-α6 chains) form a triple-helix structure. Recently, endogenous bioactive factors called "matricryptins" or "matrikines", which are produced by degrading and cleaving C-terminal domain of type IV collagen, attract attentions as a novel therapeutic target or a candidate for biomarkers. In all type IV collagens, matricryptins called arresten (α1 chain), canstatin (α2), tumstatin (α3), tetrastatin (α4), pentastatin (α5), and hexastatin (α6), have been identified. The type IV collagen-derived matricryptins have been previously studied as new therapeutic targets for neoplastic diseases since they exert anti-angiogenic and/or anti-tumor effects. On the other hand, we have recently demonstrated the cardioprotective effects of matricryptins in addition to the altered expression levels in cardiac diseases. In this review, we introduce the results of fundamental studies for the type IV collagen-derived matricryptins in various diseases, such as neoplastic diseases and cardiac diseases, and discuss the potential clinical application as novel therapeutic agents and biomarkers.
Collapse
Affiliation(s)
- Akira Sugiyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Kosuke Otani
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University
| |
Collapse
|
6
|
Zhang F, Duan J, Song H, Yang L, Zhou M, Wang X. Combination of canstatin and arsenic trioxide suppresses the development of hepatocellular carcinoma. Drug Dev Res 2020; 82:430-439. [PMID: 33244794 DOI: 10.1002/ddr.21766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/06/2020] [Accepted: 11/18/2020] [Indexed: 11/05/2022]
Abstract
Complication of arsenic trioxide (ATO) and other drugs in cancer treatment has attracted much focus, but is limitedly investigated in hepatocellular carcinoma (HCC). This study aimed to explore the role of ATO combined with canstatin in HCC. HepG2 cells were treated with different concentrations of ATO with or without canstatin, CCK-8, flow cytometry, Transwell assays were conducted to determine cell proliferation, apoptosis, adhesion, migration, and invasion abilities. Besides, the protein expression or mRNA level of caspase-3, PCNA, and MMP-2 was measured using western blotting or qRT-PCR. BALB/c-nu/nu mice were used to establish nude mouse transplantation tumor model, and received ATO or canstatin treatment for 3 weeks. The results showed that ATO inhibited cell proliferation, adhesion, migration and invasion, and promoted cell apoptosis with a concentration-dependent way. Canstatin had a significantly inhibitory effect on cell proliferation, but had limited effects on the other cellular behaviors. Besides, combination with ATO and canstatin strengthened the effects of ATO alone on cell proliferation inhibition and cell apoptosis promotion. Moreover, both of ATO and canstatin increased the protein expression of caspase-3, while decreased PCNA and MMP-2, which was further strengthened upon their combination. Furthermore, both of ATO and canstatin inhibited tumor growth in vivo, which was also strengthened upon their combination. Collectively, we found that combined canstatin and ATO significantly inhibited cell proliferation, migration and adhesion abilities, and promoted cell apoptosis, and inhibited tumor growth, thus suppressed the progression of HCC.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| | - Jingjing Duan
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| | - Hailin Song
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| | - Li Yang
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| | - Ming Zhou
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| | - Xuewen Wang
- Department of Pathophysiology, Shihezi University, the Xinjiang Uygur Autonomous Region, Shihezi, China
| |
Collapse
|
7
|
Decreased Expression of Canstatin in Rat Model of Monocrotaline-Induced Pulmonary Arterial Hypertension: Protective Effect of Canstatin on Right Ventricular Remodeling. Int J Mol Sci 2020; 21:ijms21186797. [PMID: 32947968 PMCID: PMC7554857 DOI: 10.3390/ijms21186797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease which causes right ventricular (RV) failure. Canstatin, a C-terminal fragment of type IV collagen α2 chain, is expressed in various rat organs. However, the expression level of canstatin in plasma and organs during PAH is still unclear. We aimed to clarify it and further investigated the protective effects of canstatin in a rat model of monocrotaline-induced PAH. Cardiac functions were assessed by echocardiography. Expression levels of canstatin in plasma and organs were evaluated by enzyme-linked immunosorbent assay and Western blotting, respectively. PAH was evaluated by catheterization. RV remodeling was evaluated by histological analyses. Real-time polymerase chain reaction was performed to evaluate RV remodeling-related genes. The plasma concentration of canstatin in PAH rats was decreased, which was correlated with a reduction in acceleration time/ejection time ratio and an increase in RV weight/body weight ratio. The protein expression of canstatin in RV, lung and kidney was decreased in PAH rats. While recombinant canstatin had no effect on PAH, it significantly improved RV remodeling, including hypertrophy and fibrosis, and prevented the increase in RV remodeling-related genes. We demonstrated that plasma canstatin is decreased in PAH rats and that administration of canstatin exerts cardioprotective effects.
Collapse
|
8
|
Long-term administration of recombinant canstatin prevents adverse cardiac remodeling after myocardial infarction. Sci Rep 2020; 10:12881. [PMID: 32732948 PMCID: PMC7393096 DOI: 10.1038/s41598-020-69736-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/15/2020] [Indexed: 11/08/2022] Open
Abstract
Myocardial infarction (MI) still remains a leading cause of mortality throughout the world. An adverse cardiac remodeling, such as hypertrophy and fibrosis, in non-infarcted area leads to uncompensated heart failure with cardiac dysfunction. We previously demonstrated that canstatin, a C-terminus fragment of type IV collagen α2 chain, exerted anti-remodeling effect against isoproterenol-induced cardiac hypertrophy model rats. In the present study, we examined whether a long-term administration of recombinant canstatin exhibits a cardioprotective effect against the adverse cardiac remodeling in MI model rats. Left anterior descending artery of male Wistar rats was ligated and recombinant mouse canstatin (20 μg/kg/day) was intraperitoneally injected for 28 days. Long-term administration of canstatin improved survival rate and significantly inhibited left ventricular dilatation and dysfunction after MI. Canstatin significantly inhibited scar thinning in the infarcted area and significantly suppressed cardiac hypertrophy, nuclear translocation of nuclear factor of activated T-cells, interstitial fibrosis and increase of myofibroblasts in the non-infarcted area. Canstatin significantly inhibited transforming growth factor-β1-induced differentiation of rat cardiac fibroblasts into myofibroblasts. The present study for the first time demonstrated that long-term administration of recombinant canstatin exerts cardioprotective effects against adverse cardiac remodeling in MI model rats.
Collapse
|
9
|
A current perspective of canstatin, a fragment of type IV collagen alpha 2 chain. J Pharmacol Sci 2018; 139:59-64. [PMID: 30580971 DOI: 10.1016/j.jphs.2018.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 01/09/2023] Open
Abstract
Type IV collagen is a main component of basement membrane extracellular matrix. Canstatin, a non-collagenous C-terminal fragment of type IV collagen α2 chain, was firstly identified as an endogenous anti-angiogenic and anti-tumor factor, which also has an anti-lymphangiogenic effect. Then, canstatin has been widely investigated as a novel target molecule for cancer therapy. The anti-angiogenic effect of canstatin may be also useful for the treatment of ocular neovascularization. Recently, we have demonstrated that canstatin, which is abundantly expressed in the heart tissue, exerts various biological activities in cardiac cells. In rat H9c2 cardiomyoblasts, canstatin inhibits isoproterenol- or hypoxia-induced apoptosis. Canstatin plays an important role in modulating voltage-dependent calcium channel activity in rat cardiomyocytes. Canstatin also regulates various biological functions in rat cardiac fibroblasts and myofibroblasts. The expression of canstatin decreases in the infarcted area after myocardial infarction. This review focuses on a current perspective for the roles of canstatin in tumorigenesis, ocular neovascularization and cardiac pathology.
Collapse
|
10
|
Wang SC, Lee TH, Hsu CH, Chang YJ, Chang MS, Wang YC, Ho YS, Wen WC, Lin RK. Antroquinonol D, isolated from Antrodia camphorata, with DNA demethylation and anticancer potential. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:5625-35. [PMID: 24784321 DOI: 10.1021/jf4056924] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
DNA methyltransferase 1 (DNMT1) catalyzes DNA methylation and is overexpressed in various human diseases, including cancer. A rational approach to preventing tumorigenesis involves the use of pharmacologic inhibitors of DNA methylation; these inhibitors should reactivate tumor suppressor genes (TSGs) in tumor cells and restore tumor suppressor pathways. Antroquinonol D (3-demethoxyl antroquinonol), a new DNMT1 inhibitor, was isolated from Antrodia camphorata and identified using nuclear magnetic resonance. Antroquinonol D inhibited the growth of MCF7, T47D, and MDA-MB-231 breast cancer cells without harming normal MCF10A and IMR-90 cells. The SRB assay showed that the 50% growth inhibition (GI50) in MCF7, T47D, and MDA-MB-231 breast cancer cells following treatment with antroquinonol D was 8.01, 3.57, and 25.08 μM, respectively. d-Antroquinonol also inhibited the migratory ability of MDA-MB-231 breast cancer cells in wound healing and Transwell assays. In addition, antroquinonol D inhibited DNMT1 activity, as assessed by the DNMT1 methyltransferase activity assay. As the cofactor SAM level increased, the inhibitory effects of d-antroquinonol on DNMT1 gradually decreased. An enzyme activity assay and molecular modeling revealed that antroquinonol D is bound to the catalytic domain of DNMT1 and competes for the same binding pocket in the DNMT1 enzyme as the cofactor SAM, but does not compete for the binding pocket in the DNMT3B enzyme. An Illumina Methylation 450 K array-based assay and real-time PCR assay revealed that antroquinonol D decreased the methylation status and reactivated the expression of multiple TSGs in MDA-MB-231 breast cancer cells. In conclusion, we showed that antroquinonol D induces DNA demethylation and the recovery of multiple tumor suppressor genes, while inhibiting breast cancer growth and migration potential.
Collapse
Affiliation(s)
- Sheng-Chao Wang
- Graduate Institute of Pharmacognosy, Taipei Medical University , 250 Wu-Hsing Street Taipei, TW 110, Taiwan, R. O. C
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Feng S, Li S, Li Q, Shi K, Xue L. Preparation of recombinant human canstatin using transgenic Dunaliella salina. Acta Biochim Biophys Sin (Shanghai) 2014; 46:428-30. [PMID: 24777497 DOI: 10.1093/abbs/gmu009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Shuying Feng
- Department of Immunology, Medical College, Henan University of Science and Technology, Luoyang 471003, China
| | | | | | | | | |
Collapse
|
12
|
Monboisse JC, Sénéchal K, Thevenard J, Ramont L, Brassart-Pasco S, Maquart FX. [Matrikines: a new anticancer therapeutic strategy]. Biol Aujourdhui 2012; 206:111-123. [PMID: 22748049 DOI: 10.1051/jbio/2012017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Indexed: 06/01/2023]
Abstract
Tumor microenvironment is a complex system composed of a largely altered extracellular matrix (ECM) with different cell types that determine the angiogenic response. Upon the influence of hypoxia, tumor cells secrete cytokines that activate stromal cells to produce proteases and angiogenic factors. The proteases degrade the stromal ECM and participate in the release of various ECM fragments, named matrikines or matricryptins, capable to control tumor invasion and metastasis dissemination. We will focus on the matrikines derived from the NC1 domains of the different constitutive chains of basement membrane-associated collagens and mainly collagen IV. The putative targets of the matrikine action are the proliferation and invasive properties of tumor or inflammatory cells, and the angiogenic and lymphangiogenic responses. For example, canstatin, tumstatin and tetrastatin, respectively derived from the NC1 domains of α2, α3 and α4 chains of collagen IV, inhibit in vivo tumor growth in various experimental cancer models. Their anti-cancer activity comprises an anti-proliferative effect on tumor cells and on endothelial cells by induction of cell apoptosis or cell cycle blockade and the induction of a loss of their migratory phenotype. Matrikines constitute a new family of potent anticancer agents that could be used under various therapeutic strategies: i) induction of their overexpression by cancer cells or by the host cells, ii) use of recombinant proteins or synthetic peptides or structural analogues designed from the structure of the active sequences. These matrikines could be used in combination with conventional chemotherapy or radiotherapy to limit tumor progression.
Collapse
Affiliation(s)
- Jean Claude Monboisse
- FRE CNRS/URCA 3481, Université Reims Champagne Ardenne, UFR Médecine, 51 Rue Cognacq-Jay, 51095 Reims Cedex, France.
| | | | | | | | | | | |
Collapse
|
13
|
Hwang-Bo J, Yoo KH, Park JH, Jeong HS, Chung IS. Recombinant canstatin inhibits angiopoietin-1-induced angiogenesis and lymphangiogenesis. Int J Cancer 2011; 131:298-309. [PMID: 21823121 DOI: 10.1002/ijc.26353] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 07/21/2011] [Indexed: 01/25/2023]
Abstract
We describe the effect of recombinant canstatin, the NC1 domain of the α2 chain of Type IV collagen, on suppression of angiogenesis and lymphangiogenesis both in vitro and in vivo. Recombinant canstatin produced from stably transformed Drosophila S2 cells reduced the expression of angiopoietin-1 in hypoxia mimetic agent, CoCl(2) -treated CT-26 cells. Recombinant canstatin inhibited proliferation, tube formation and migration of human angiopoietin-1 (rhAngpt-1)-treated human umbilical vein endothelial cells (HUVEC) and lymphatic endothelial cells (LEC). Recombinant canstatin suppressed the expression of Tie-2 and vascular endothelial growth factor-3 (VEGFR-3) transcripts in rhAngpt-1-treated HUVEC and LEC, respectively. The inhibitory effect of recombinant canstatin on tumor growth was also investigated using a heterotopic CT-26 colon carcinoma animal (BALB/c mice) model. Recombinant canstatin reduced the final volume and weight of tumors, and blood and lymphatic vessel densities of tumors, which were evaluated by CD-31 and LYVE-1 immunostaining. Immunohistochemical analysis showed that recombinant canstatin dramatically reduced the expression of angiopoietin-1 in CT-26 colon carcinoma-induced tumor, but not the expression of VEGF-C. Tie-2 and VEGFR-3 expressions were also reduced in recombinant canstatin-treated tumors. These results indicate that recombinant canstatin has anti-tumoral activities against CT-26 colon carcinoma cells. Recombinant canstatin reduces the expression of angiopoietin-1 in hypoxia-induced CT-26 cells and inhibits the angiogenic and lymphangiogenic signaling induced by angiopoietin-1. Recombinant canstatin probably inhibits angiogenesis and lymphangiogenesis via suppression of the integrin-dependent FAK signaling induced by angiopoietin-1/Tie-2 and/or VEGFR-3.
Collapse
Affiliation(s)
- Jeon Hwang-Bo
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin, Korea
| | | | | | | | | |
Collapse
|
14
|
Abstract
Endogenous inhibitors of angiogenesis are proteins or fragments of proteins that are formed in the body, which can inhibit the angiogenic process. These molecules can be found both in the circulation and sequestered in the extracellular matrix (ECM) surrounding cells. Many matrix-derived inhibitors of angiogenesis, such as endostatin, tumstatin, canstatin and arresten, are bioactive fragments of larger ECM molecules. These substances become released upon proteolysis of the ECM and the vascular basement membrane (VBM) by enzymes of the tumor microenvironment. Although the role of matrix-derived angiogenesis inhibitors is well studied in animal models of cancer, their role in human cancers is less established. In this review we discuss the current knowledge about these molecules and their potential use as cancer therapeutics and biomarkers.
Collapse
|
15
|
E1B-55kD-deleted oncolytic adenovirus armed with canstatin gene yields an enhanced anti-tumor efficacy on pancreatic cancer. Cancer Lett 2009; 285:89-98. [PMID: 19481338 DOI: 10.1016/j.canlet.2009.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 05/02/2009] [Accepted: 05/06/2009] [Indexed: 11/20/2022]
Abstract
Conditionally-replicating adenovirus (CRAd) therapy is currently being tested against pancreatic cancer and has shown some promise. To improve the efficacy, a novel virus CRAd-Cans was designed by deletion of E1B-55kDa gene for selective replication in tumor cells, as well as carrying a new angiogenesis inhibitor gene, canstatin. CRAd-Cans mediated higher expression of canstatin in BxPC-3 pancreatic cancer cell line compared to the replication-deficient adenovirus Ad5-Cans. The modified CRAd-Cans manifested the same selective replication and cytocidal effects in pancreatic cancer cells as ONYX-015 in vitro, yet showed greater reduction of tumor growth in nude mice with markedly prolonged survival rate in vivo (P<0.05), compared to that of either ONYX-015 or Ad5-Cans. Pathological examination revealed viral replication, decreased microvessel density and increased cancer cell apoptosis in CRAd-Cans-treated xenografts. The results suggest that the novel oncolytic virus CRAd-Cans, showing synergistic effects of oncolytic therapy and anti-angiogenesis therapy, is a new promising therapeutics for pancreatic cancer.
Collapse
|