1
|
Roos-Mattila M, Kaprio T, Mustonen H, Hagström J, Saharinen P, Haglund C, Seppänen H. The possible dual role of Ang-2 in the prognosis of pancreatic cancer. Sci Rep 2023; 13:18725. [PMID: 37907568 PMCID: PMC10618172 DOI: 10.1038/s41598-023-45194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features a dense desmoplastic stroma, which raises the intratumoral interstitial pressure leading to vascular collapse and hypoxia, inducing angiogenesis. Vascular growth factors, such as vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang-2), increase in PDAC. A high VEGF and a high circulating Ang-2 associate with shorter survival in PDAC. In addition to the circulatory Ang-2, PDAC endothelial and epithelial cells express Ang-2. No correlation between tumor epithelial nor endothelial cell Ang-2 expression and survival has been published. We aimed to examine Ang-2 expression and survival. This study comprised PDAC surgical patients at Helsinki University Hospital in 2000-2013. Ang-2 immunohistochemistry staining was completed on 168 PDAC patient samples. Circulating Ang-2 levels were measured using ELISA in the sera of 196 patients. Ang-2 levels were assessed against clinical data and patient outcomes. A low tumor epithelial Ang-2 expression predicted shorter disease-specific survival (DSS) compared with a high expression (p = 0.003). A high serum Ang-2 associated with shorter DSS compared with a low circulating Ang-2 (p = 0.016). Ang-2 seemingly plays a dual role in PDAC survival. Further studies are needed to determine the mechanisms causing tumor cell Ang-2 expression and its positive association with survival.
Collapse
Affiliation(s)
- Matilda Roos-Mattila
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Kaprio
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland.
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Department of Pathology, Haartmaninkatu 3 (PB 21), University of Helsinki, 00014, Helsinki, Finland.
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jaana Hagström
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Oral Pathology and Radiology, University of Turku, Turku, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
2
|
Kennedy DC, Coen B, Wheatley AM, McCullagh KJA. Microvascular Experimentation in the Chick Chorioallantoic Membrane as a Model for Screening Angiogenic Agents including from Gene-Modified Cells. Int J Mol Sci 2021; 23:452. [PMID: 35008876 PMCID: PMC8745510 DOI: 10.3390/ijms23010452] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
The chick chorioallantoic membrane (CAM) assay model of angiogenesis has been highlighted as a relatively quick, low cost and effective model for the study of pro-angiogenic and anti-angiogenic factors. The chick CAM is a highly vascularised extraembryonic membrane which functions for gas exchange, nutrient exchange and waste removal for the growing chick embryo. It is beneficial as it can function as a treatment screening tool, which bridges the gap between cell based in vitro studies and in vivo animal experimentation. In this review, we explore the benefits and drawbacks of the CAM assay to study microcirculation, by the investigation of each distinct stage of the CAM assay procedure, including cultivation techniques, treatment applications and methods of determining an angiogenic response using this assay. We detail the angiogenic effect of treatments, including drugs, metabolites, genes and cells used in conjunction with the CAM assay, while also highlighting the testing of genetically modified cells. We also present a detailed exploration of the advantages and limitations of different CAM analysis techniques, including visual assessment, histological and molecular analysis along with vascular casting methods and live blood flow observations.
Collapse
Affiliation(s)
| | | | - Antony M. Wheatley
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland, H91 W5P7 Galway, Ireland; (D.C.K.); (B.C.)
| | - Karl J. A. McCullagh
- Department of Physiology, School of Medicine, Human Biology Building, National University of Ireland, H91 W5P7 Galway, Ireland; (D.C.K.); (B.C.)
| |
Collapse
|
3
|
Ji C, Yue S, Gu J, Kong Y, Chen H, Yu C, Sun Z, Zhao M. 2,7-Dibromocarbazole interferes with tube formation in HUVECs by altering Ang2 promoter DNA methylation status. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 697:134156. [PMID: 32380619 DOI: 10.1016/j.scitotenv.2019.134156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 06/11/2023]
Abstract
2,7-Dibromocarbazole (2,7-DBCZ) is one of the most frequently detected polyhalogenated carbazoles (PHCZs) in the environmental media. 2,7-DBCZ has attracted public attention for its potential for dioxin-like toxicity and cardiovascular toxicity. However, researches on the potential mechanism of angiogenesis inhibition by 2,7-DBCZ is still insufficient. Herein, human umbilical vein endothelial cells (HUVECs) were applied to explore the angiogenic effect of 2,7-DBCZ and the potential underlying mechanisms. 2,7-DBCZ significantly inhibited tube formation in HUVECs in the non-toxic concentration range. PCR array showed that 2,7-DBCZ reduced the expression proportion between VEGFs and Ang2, thereby inhibiting tube formation in HUVECs. Then, small RNA interference and DNA methylation assays were adopted to explore the potential mechanisms. It has been found that angiopoietin2 (Ang2)-silencing recovered the tube formation inhibited by 2,7-DBCZ. The DNA methylation status of Ang2 promoter also showed a demethylation tendency after exposure. In conclusion, 2,7-DBCZ could demethylate the Ang2 promoter to potentiate Ang2 expression, thus altering angiogenic phenotype of HUVECs by reducing the proportion between Ang2 and VEGFs. The data presented here can help to guide safety measures on the use of dioxin-like PHCZs for their potential adverse effects and provide a method for identifying the relevant biomarkers to assess their cardiovascular toxicity.
Collapse
Affiliation(s)
- Chenyang Ji
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Siqing Yue
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jinping Gu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuan Kong
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Haofeng Chen
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Chang Yu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhe Sun
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Meirong Zhao
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
4
|
Wei H, Wang F, Wang Y, Li T, Xiu P, Zhong J, Sun X, Li J. Verteporfin suppresses cell survival, angiogenesis and vasculogenic mimicry of pancreatic ductal adenocarcinoma via disrupting the YAP-TEAD complex. Cancer Sci 2017; 108:478-487. [PMID: 28002618 PMCID: PMC5378285 DOI: 10.1111/cas.13138] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/07/2016] [Accepted: 12/17/2016] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies. The Yes-associated protein-1 (YAP) plays a critical role in cell proliferation, apoptosis and angiogenesis. Verteporfin is a photosensitizer used in photodynamic therapy and also a small molecular inhibitor of the Hippo-YAP pathway. However, little is known about whether verteporfin could inhibit YAP activity in PDAC cells. Our present results showed that verteporfin suppressed the proliferation of human PDAC PANC-1 and SW1990 cells by arresting cells at the G1 phase, and inducing apoptosis in dose- and time-dependent manners. Verteporfin also inhibited the tumor growth on the PDAC xenograft model. Treatment with verteporfin led to downregulation of cyclinD1 and cyclinE1, modulation of Bcl-2 family proteins and activation of PARP. In addition, verteporfin exhibited an inhibitory effect on angiogenesis and vasculogenic mimicry via suppressing Ang2, MMP2, VE-cadherin, and α-SMA expression in vitro and in vivo. Mechanism studies demonstrated that verteporfin impaired YAP and TEAD interaction to suppress the expression of targeted genes. Our results provide a foundation for repurposing verteporfin as a promising anti-tumor drug in the treatment of pancreatic cancer by targeting the Hippo pathway.
Collapse
Affiliation(s)
- Honglong Wei
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Fuhai Wang
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Yong Wang
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Tao Li
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Peng Xiu
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Jingtao Zhong
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Xueying Sun
- Department of Molecular Medicine and PathologyFaculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Jie Li
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| |
Collapse
|
5
|
Wang H, Hang C, Ou XL, Nie JS, Ding YT, Xue SG, Gao H, Zhu JX. MiR-145 functions as a tumor suppressor via regulating angiopoietin-2 in pancreatic cancer cells. Cancer Cell Int 2016; 16:65. [PMID: 27570490 PMCID: PMC5000507 DOI: 10.1186/s12935-016-0331-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/23/2016] [Indexed: 12/21/2022] Open
Abstract
Background Pancreatic cancer is currently one of the leading causes of cancer deaths without any effective therapies. Mir-145 has been found to be tumor-suppressive in various types of cancers. The aim of this study is to investigate the role of miR-145 in pancreatic cancer cells and explore its underlying mechanism. Methods Quantitative real time PCR was used to determine the expression level of miR-145 and angiopoietin-2 (Ang-2) mNRA, and the expression level of Ang-2 protein was measured by western blotting. The anti-cancer activities of miR-145 were tested both in in vitro by using cell invasion and colony formation assay and in vivo by using xenograft assay. The direct action of miR-145 on Ang-2 was predicted by TargetScan and confirmed by luciferase report assay. The vascularization of xenografts were performed by immunohistochemical analysis. Results The expression level of miR-145 was significantly lower and the expression levels of Ang-2 mRNA and protein was significantly higher in the more aggressive pancreatic cancer cells (MiaPaCa-2 and Panc-1) when compared to that in BxPC3 cells. Overexpression of miR-145 in the BxPC3, MiaPaCa-2 and Panc-1 cells suppressed the cell invasion and colony formation ability, and the expression level of Ang-2 protein in MiaPaCa-2 and Panc-1 cells was also suppressed after pre-miR-145 transfection. Intratumoral delivery of miR-145 inhibited the growth of pancreatic cancer xenografts and angiogenesis in vivo, and also suppressed the expression level of angiopoietin-2 protein. Luciferase report assay showed that Ang-2 is a direct target of miR-145, and down-regulation of angiopoietin-2 by treatment with Ang-2 siRNA in the BxPC3, MiaPaCa-2 and Panc-1 cells suppressed cell invasion and colony formation ability. The reverse transcription PCR results also showed that Tie1 and Tie2 were expressed in BxPC3, MiaPaCa-2 and Panc-1 cells. Conclusion MiR-145 functions as a tumor suppressor in pancreatic cancer cells by targeting Ang-2 for translation repression and thus suppresses pancreatic cancer cell invasion and growth, which suggests that restoring of miR-145 may be a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery, DrumTower Clinical Medical College of Nanjing Medical University, Nanjing, 210008 Jiangsu Province China
| | - Cheng Hang
- Department of Gastroenterology, The First People's Hospital of Taicang, Suzhou, 215400 Jiangsu Province China
| | - Xi-Long Ou
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009 Jiangsu Province China
| | - Jin-Shan Nie
- Department of Gastroenterology, The First People's Hospital of Taicang, Suzhou, 215400 Jiangsu Province China
| | - Yi-Tao Ding
- Department of General Surgery, DrumTower Clinical Medical College of Nanjing Medical University, Nanjing, 210008 Jiangsu Province China
| | - Shi-Gui Xue
- Department of Gastroenterology, The First People's Hospital of Taicang, Suzhou, 215400 Jiangsu Province China
| | - Hua Gao
- Department of Gastroenterology, The First People's Hospital of Taicang, Suzhou, 215400 Jiangsu Province China
| | - Jian-Xin Zhu
- Department of Gastroenterology, The First People's Hospital of Taicang, Suzhou, 215400 Jiangsu Province China
| |
Collapse
|
6
|
Büning H, Hacker UT. Inhibitors of Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:261-85. [DOI: 10.1007/978-3-319-32805-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
7
|
Fan G, Bo J, Wan R, Peng M, Luan Y, Deng M, Xu L. The effect of lentiviral vector-mediated RNA interference targeting hypoxia-inducible factor 1α on the uptake of fluorodeoxyglucose ((18)f) in the human pancreatic cancer cell line, patu8988. Cancer Biother Radiopharm 2015; 30:160-8. [PMID: 25853522 DOI: 10.1089/cbr.2014.1700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hypoxia can stimulate (18)F-fluorodeoxyglucose ((18)F-FDG) uptake in cultured tumor cells. This study has investigated the effect of lentiviral vector-mediated RNA interference (RNAi) targeting hypoxia-inducible factor 1α (HIF-1α) on the changes in HIF-1 and glucose transporter 1 (Glut-1) expression, the cell growth, and the uptake of (18)F-FDG in the human pancreatic cancer cell line, Patu8988. Lentiviral RNAi vector targeting the HIF-1α gene (LV-HIF-1αRNAi) was constructed and used to treat cells at various concentrations (25-200 nM). The expression changes of HIF-1α and Glut-1 in hypoxic Patu8988 cells after RNAi treatment were determined using real time reverse transcription-polymerase chain reaction (real-time PCR). The inhibition rate of cell proliferation 48 hours after the addition of 10 μL of different concentrations of LV-HIF-1αRNAi (25-200 nM) was assayed using the MTT method. Meanwhile, the cell uptake of (18)F-FDG was also assessed. After RNAi transfection, the relative expression levels of HIF-1α mRNA and Glut-1 under hypoxia were reduced and the relative expression levels of HIF-1α protein also decreased. Compared with the control group, the inhibition rates of cell proliferation under different viral dosages were 5.98%, 15.65%, 26.42%, and 40.81%, respectively, positively correlated with the viral doses (r=0.558, p<0.05). Under hypoxia, Glut-1 mRNA expression in Patu8988 cells treated with 200 nM of LV-HIF-1αRNAi for 24, 48, and 72 hours, respectively, was positively correlated with the inhibition rate of cell proliferation (r=0.618, p<0.05) as well as the inhibition rate of (18)F-FDG uptake (r=0.664, p<0.05), while the latter two displayed a positive correlation with each other too (r=0.582, p<0.05). Under hypoxia, RNAi targeting HIF-1α significantly inhibited the expression of Glut-1 mRNA in Patu8988 pancreatic cancer cells and their uptake of (18)F-FDG. These results suggest that LV-HIF-1αRNAi may form a new treatment for pancreatic cancer, and the effectiveness of the treatment can be readily assessed with (18)F-FDG imaging.
Collapse
Affiliation(s)
- Guanglei Fan
- Department of Nuclear Medicine, Changzhou Second People's Hospital, Nanjing Medical University , Changzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
8
|
Yuan Z, Pastoriza J, Quinn T, Libutti SK. Targeting Tumor Vasculature Using Adeno-Associated Virus Phage Vectors Coding Tumor Necrosis Factor-α. GENE THERAPY OF CANCER 2014:19-33. [DOI: 10.1016/b978-0-12-394295-1.00002-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Knockdown of angiopoietin-2 suppresses metastasis in human pancreatic carcinoma by reduced matrix metalloproteinase-2. Mol Biotechnol 2013; 53:336-44. [PMID: 22457202 DOI: 10.1007/s12033-012-9532-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Angiopoietin-2 (Ang2) has been shown highly expressed in resected human pancreatic carcinoma samples, and has tightly combination with tumor angiogenesis, but the role in metastasis of it is less clear. We were, therefore, interested in exploring the effects of Ang2 silencing on the invasion and metastasis of pancreatic carcinoma. Lentivirus (LV)-mediated Ang2 small hairpin RNA (LV-RNAi) and mock lentivirus (LV-NC) were transfected into pancreatic carcinoma cell line MIA PaCa-2. Groups were designed in this study: the control group (MIA PaCa-2 cells), the LV-NC group (cells transfected with the LV-NC), the LV-RNAi-KD1 group (cells transfected with LV-RNAi of knock down sequence (1) and the LV-RNAi-KD2 group (cells transfected with LV-RNAi of knock down sequence (2). Boyden chamber transwell assay was used to detect the cell invasion change. The protein levels of Ang2, MMP-2, and MMP-9 gene and mRNA level of MMP-2, MMP-9 were detected by Western blot and real-time polymerase chain reaction, respectively. Orthotopic pancreatic carcinoma xenotransplantation model were successfully built with MIA PaCa-2 cells injection. After treatment with intraperitoneal injection of LV-RNAi-KD2 (LV-RNAi), mice growth, liver function test, tumor volume and peritoneal metastatic numbers were observed and counted. Moreover, expression of Ang2, MMP-2, MMP-9 were measured by immunohistochemistry. Ang2 expression were successfully knocked down in two LV-RNAi groups, especially in the LV-RNAi-KD2group. Compared with the control group and the LV-NC group, the mRNA and protein level of MMP-2 gene were downregulated significantly in LV-RNAi groups, also the invasion cell number decreased in boyden chamber transwell assay after LV-RNAi transfection. Meanwhile, no obvious MMP-9 gene expression changes were found among all the groups. LV-RNAi injection inhibited pancreatic carcinoma metastasis and growth in vivo by downregulating the expression of MMP-2 not MMP-9. Most importantly, LV-mediated gene therapy with Ang2 knockdown exhibited almost no toxicity in vivo. These findings demonstrate that Ang2 gene silencing exert an anti-metastasis effect in vitro and in vivo, and Ang2 targeted gene therapy has the potential to serve as a novel way for pancreatic carcinoma treatment.
Collapse
|
10
|
The angiopoietin:Tie 2 interaction: a potential target for future therapies in human vascular disease. Cytokine Growth Factor Rev 2013; 24:579-92. [PMID: 23838360 DOI: 10.1016/j.cytogfr.2013.05.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 01/06/2023]
Abstract
Angiopoietin-1 and -2 are endogenous ligands for the vascular endothelial receptor tyrosine kinase Tie2. Signalling by angiopoietin-1 promotes vascular endothelial cell survival and the sprouting and reorganisation of blood vessels, as well as inhibiting activation of the vascular endothelial barrier to reduce leakage and leucocyte migration into tissues. Angiopoietin-2 generally has an opposing action, and is released naturally at times of vascular growth and inflammation. There is a significant body of emerging evidence that promoting the actions of angiopoietin-1 through Tie2 is of benefit in pathologies of vascular activation, such as sepsis, stroke, diabetic retinopathy and asthma. Similarly, methods to inhibit the actions of angiopoietin-2 are emerging and have been demonstrated to be of preclinical and clinical benefit in reducing tumour angiogenesis. Here the author reviews the evidence for potential benefits of modulation of the interaction of angiopoietins with Tie2, and the potential applications. Additionally, methods for delivery of the complex protein angiopoietin-1 are discussed, as well as potentially deleterious consequences of administering angiopoietin-1.
Collapse
|
11
|
Aravindan S, Delma CR, Thirugnanasambandan SS, Herman TS, Aravindan N. Anti-pancreatic cancer deliverables from sea: first-hand evidence on the efficacy, molecular targets and mode of action for multifarious polyphenols from five different brown-algae. PLoS One 2013; 8:e61977. [PMID: 23613993 PMCID: PMC3628576 DOI: 10.1371/journal.pone.0061977] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 03/15/2013] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer (PC) remains the fourth leading cause of cancer death with an unacceptable survival that has remained relatively unchanged over the past 25 years. The presence of occult or clinical metastases at the time of diagnosis together with the lack of effective chemotherapies pose a dire need for designing new and targeted therapeutic deliverables that favors the clinical outcome. Herein, we investigated the anti-tumorigenic potential of polyphenols from five different brown-algae in human PC cells (MiaPaCa-2, Panc-1, BXPC-3 and Panc-3.27). Total anti-oxidant capacity (TAC) analysis on stepwise polyphenol separations with increasing polarity (Hexane-DCM-EA-methanol) identified high levels of TAC in DCM and EA extractions across all seaweeds assessed. All DCM and EA separated polyphenols induced a dose-dependent and sustained (time-independent) inhibition of cell proliferation and viability. Further, these polyphenols profoundly enhanced DNA damage (acridine orange/Ethidium bromide staining and DNA fragmentation) in all the cell lines investigated. More importantly, luciferase reporter assay revealed a significant inhibition of NFκB transcription in cells treated with polyphenols. Interestingly, QPCR analysis identified a differential yet definite regulation of pro-tumorigenic EGFR, VEGFA, AKT, hTERT, kRas, Bcl2, FGFα and PDGFα transcription in cells treated with DCM and EA polyphenols. Immunoblotting validates the inhibitory potential of seaweed polyphenols in EGFR phosphorylation, kRas, AurKβ and Stat3. Together, these data suggest that intermediate polarity based fractions of seaweed polyphenols may significantly potentiate tumor cell killing and may serve as potential drug deliverable for PC cure. More Studies dissecting out the active constituents in potent fractions, mechanisms of action and synergism, if any, are warranted and are currently in process.
Collapse
Affiliation(s)
- Sheeja Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Marine Sciences, Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, India
| | - Caroline R. Delma
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Marine Sciences, Center of Advanced Study in Marine Biology, Annamalai University, Parangipettai, India
| | | | - Terence S. Herman
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
12
|
Tang D, Wang D, Yuan Z, Xue X, Zhang Y, An Y, Chen J, Tu M, Lu Z, Wei J, Jiang K, Miao Y. Persistent activation of pancreatic stellate cells creates a microenvironment favorable for the malignant behavior of pancreatic ductal adenocarcinoma. Int J Cancer 2013; 132:993-1003. [PMID: 22777597 DOI: 10.1002/ijc.27715] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/20/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common malignant tumors with poor prognosis due to extremely high malignancy, low rate of eligibility for surgical resection and chemoradiation resistance. Increasing evidence indicate that the interaction between activated pancreatic stellate cells (PSCs) and PDAC cells plays an important role in the development of PDAC. By producing high levels of cytokines, chemotactic factors, growth factors and excessive extracellular matrix (ECM), PSCs create desmoplasia and a hypoxic microenvironment that promote the initiation, development, evasion of immune surveillance, invasion, metastasis and resistance to chemoradiation of PDAC. Therefore, targeting the interaction between PSCs and PDAC cells may represent a novel therapeutic approach to advanced PDAC, especially therapies that target PSCs of the pancreatic tumor microenvironment.
Collapse
Affiliation(s)
- Dong Tang
- Department of Gastrointestinal Surgery, Subei People's Hospital of Jiangsu Province (Clinical Medical College of Yangzhou University), Yangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhao X, Li DC, Zhu XG, Gan WJ, Li Z, Xiong F, Zhang ZX, Zhang GB, Zhang XG, Zhao H. B7-H3 overexpression in pancreatic cancer promotes tumor progression. Int J Mol Med 2012; 31:283-91. [PMID: 23242015 PMCID: PMC4042878 DOI: 10.3892/ijmm.2012.1212] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/26/2012] [Indexed: 12/25/2022] Open
Abstract
B7-H3, a member of the B7-family molecules, plays an important role in adaptive immune
responses. In addition, B7-H3 is also expressed in several types of human cancers and is
correlated with the poor outcome of cancer patients. However, its exact role in cancer is
not known. In the present study, we compared B7-H3 expression in normal pancreas and
pancreatic cancer tissue specimens, and determined the effects of low B7-H3 expression on
the human pancreatic cancer cell line Patu8988 using lentivirus-mediated RNA interference.
B7-H3 expression in pancreatic specimens was determined by enzyme-linked immunosorbent
assay (ELISA). A Patu8988 cell line with low B7-H3 expression was established by
lentivirus-mediated RNA interference to investigate the effect of B7-H3 on cell
proliferation, migration and invasion in vitro. By establishing
subcutaneous transplantation tumor and orthotopic transplantation pancreatic cancer mouse
models, the effect of B7-H3 on cell proliferation, migration and invasion was studied
in vivo. B7-H3 in tissue samples was significantly higher in the
pancreatic cancer group than in the normal pancreas group (mean ± SD,
193.6±9.352 vs. 87.74±7.433 ng/g; P<0.0001). B7-H3 knockdown by
RNA interference decreased cell migration and Transwell invasion up to 50%
in vitro. No apparent impact was observed on cell proliferation
in vitro. In the subcutaneous transplantation tumor mouse model, the
tumor growth rate was reduced by the knockdown of B7-H3. In the orthotopic transplantation
pancreatic cancer mouse model, the effect of inhibiting metastasis by knocking down B7-H3
was assessed in terms of the average postmortem abdominal visceral metastatic tumor
weight. This demonstrated that inhibition of B7-H3 expression reduced pancreatic cancer
metastasis in vivo. In conclusion, B7-H3 is aberrantly expressed in
pancreatic cancer. In addition to modulating tumor immunity, B7-H3 may have a novel role
in regulating pancreatic tumor progression.
Collapse
Affiliation(s)
- Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|