1
|
Ahmed AY, Uthirapathy S, Oghenemaro EF, M RM, Kumawat R, Mustafa YF, Kariem M, Kadhim AJ, Sharma S, Kumar MR. The SOX gene superfamily in oncogenesis: unraveling links to ncRNAs, key pathways, chemoresistance, and gene editing approaches. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04229-1. [PMID: 40392306 DOI: 10.1007/s00210-025-04229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/24/2025] [Indexed: 05/22/2025]
Abstract
While drug resistance remains the leading cause of treatment failure, chemotherapy continues to be a crucial aspect of cancer therapy. Long noncoding RNAs (lncRNAs) regulate gene expression through various methods, including transcriptional, translational, chromatin remodeling, and epigenetic mechanisms. The SRY-related high mobility group box (HMGB) family contains 20 transcription factors with a well-recognized HMG domain, and an inappropriate regulation of SOX family members is associated with many of the phenotypes of cancer, such as tumor invasion, metastasis, proliferation, apoptosis, epithelial-mesenchymal transition, stemness, and drug resistance. This association arises because SOX family members can regulate cell fate decisions. While many articles have reported on the functionalities and activities of the SOX family, it is not clear their involvement in the tumor immune microenvironment (TIME) and the seeming contrast they can have on tumors. This study elucidates the relationship between the SOX family and ncRNAs, specifically emphasizing lncRNAs. This review article highlights the potential roles of the SOX family in cancer. It presents new therapeutic options for treating cancer, outlining the physiological roles of the SOX family and the various roles they have in tumors.
Collapse
Affiliation(s)
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, PMB 1, Abraka, Delta State, Nigeria
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| |
Collapse
|
2
|
Hayasaki A, Mizuno S, Usui M, Kaluba B, Komatsubara H, Sakamoto T, Maeda K, Shinkai T, Noguchi D, Ito T, Gyoten K, Iizawa Y, Fujii T, Tanemura A, Murata Y, Kuriyama N, Watanabe M, Uchida K, Kishiwada M. Tumor Budding Is an Independent Adverse Prognostic Factor of Pancreatic Ductal Adenocarcinoma Patients Treated by Resection After Preoperative Chemoradiotherapy. Pancreas 2025; 54:e340-e348. [PMID: 39626198 DOI: 10.1097/mpa.0000000000002440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/11/2024] [Indexed: 04/24/2025]
Abstract
OBJECTIVES To examine the significance of tumor budding as a prognostic factor of resected pancreatic ductal adenocarcinoma (PDAC) specimens after preoperative chemoradiotherapy (CRT). MATERIALS AND METHODS Among 162 PDAC patients who underwent pancreatectomy after gemcitabine and S1-based CRT from 2012 to 2019, 131 were evaluated for tumor budding. Tumor buds were counted at the invasive front, where the degree of budding was the greatest (hematoxylin and eosin staining, ×20 magnification). Overall survival (OS), disease-specific survival (DSS), and recurrence-free survival (RFS) were compared between the patients without tumor budding (non-TB group) and those with tumor buddings (TB group). Multivariate Cox proportional hazards analysis was conducted to examine the significance of tumor budding as a prognostic factor. RESULTS OS, DSS, and RFS (median survival time) of the non-TB group were significantly longer than those of the TB group (OS: 50.7 vs 27.5 months, P = 0.014; DSS: 63.3 vs 33.0 months, P = 0.014; RFS: 20.3 vs 11.3 months, P = 0.028). Multivariate analysis identified adjuvant chemotherapy ( P = 0.003) as a favorable prognostic factor of OS and tumor budding ( P = 0.023) as an adverse prognostic factor of DSS. CONCLUSIONS This study revealed that the presence of tumor budding was an independent adverse prognostic factor in PDAC patients resected after gemcitabine and S1-based CRT.
Collapse
Affiliation(s)
- Aoi Hayasaki
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Shugo Mizuno
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Miki Usui
- Oncologic Pathology, Mie University, Tsu, Mie, Japan
| | - Benson Kaluba
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | | | - Tatsuya Sakamoto
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Koki Maeda
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Toru Shinkai
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Daisuke Noguchi
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Takahiro Ito
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Kazuyuki Gyoten
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Yusuke Iizawa
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Takehiro Fujii
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Akihiro Tanemura
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Yasuhiro Murata
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | - Naohisa Kuriyama
- Departments of Hepato-biliary Pancreatic and Transplant Surgery and
| | | | | | | |
Collapse
|
3
|
Lee W, Song G, Bae H. In vitro and in silico study of the synergistic anticancer effect of alpinumisoflavone with gemcitabine on pancreatic ductal adenocarcinoma through suppression of ribonucleotide reductase subunit-M1. Eur J Pharm Sci 2025; 204:106969. [PMID: 39577749 DOI: 10.1016/j.ejps.2024.106969] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
A highly aggressive neoplastic disease, pancreatic ductal adenocarcinoma (PDAC) is documented as the third chief cause of cancer-associated mortality in both sexes combined in the United States. For decades, gemcitabine-based chemotherapy has been embraced as a cornerstone drug for the treatment of PDAC. However, there have been several unsolved problems, including cytotoxicity, and chemoresistance. Gemcitabine efficacy was attributed to the attenuation of ribonucleotide reductase subunit-M1 (RRM1). Overexpression of RRM1 in PDAC is highly correlated with gemcitabine resistance and reduced gemcitabine sensitivity, resulting in a poor survival rate even after gemcitabine treatment. Moreover, the status of TP53, a tumor suppressor gene, assumes a decisive role in the response of PDAC to gemcitabine. Therefore, targeting RRM1 and P53 might be a therapeutic strategy for strengthening gemcitabine efficacy and cytotoxicity against PDAC. Alpinumisoflavone (AIF) is a prenylated isoflavone originated in Cudrania tricuspidate with versatile bioactive properties, including anticancer activity. However, there was no report whether AIF can exert anticancer effect and exhibit synergistic effect with gemcitabine against PDAC. Therefore, the anticancer properties of AIF were assessed with PANC-1 and MIA PaCa-2. In addition, synergism between AIF and gemcitabine were analyzed. Moreover, the contribution of P53 and RRM1 expression to gemcitabine resistance was assessed by comparing their protein levels in PDAC cells and normal pancreatic cells. The interactions of AIF with RRM1 protein were confirmed by molecular docking and dynamics simulation. Therefore, AIF enhances gemcitabine efficacy against PDAC through the regulation of P53 and RRM1.
Collapse
Affiliation(s)
- Woonghee Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea.
| | - Hyocheol Bae
- Department of Oriental Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, South Korea.
| |
Collapse
|
4
|
Tian L, Xiao J, Yu T. A robust statistical approach for finding informative spatially associated pathways. Brief Bioinform 2024; 25:bbae543. [PMID: 39451157 PMCID: PMC11503753 DOI: 10.1093/bib/bbae543] [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: 04/15/2024] [Revised: 08/27/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
Spatial transcriptomics offers deep insights into cellular functional localization and communication by mapping gene expression to spatial locations. Traditional approaches that focus on selecting spatially variable genes often overlook the complexity of biological pathways and the interactions among genes. Here, we introduce a novel framework that shifts the focus towards directly identifying functional pathways associated with spatial variability by adapting the Brownian distance covariance test in an innovative manner to explore the heterogeneity of biological functions over space. Unlike most other methods, this statistical testing approach is free of gene selection and parameter selection and allows nonlinear and complex dependencies. It allows for a deeper understanding of how cells coordinate their activities across different spatial domains through biological pathways. By analyzing real human and mouse datasets, the method found significant pathways that were associated with spatial variation, as well as different pathway patterns among inner- and edge-cancer regions. This innovative framework offers a new perspective on analyzing spatial transcriptomic data, contributing to our understanding of tissue architecture and disease pathology. The implementation is publicly available at https://github.com/tianlq-prog/STpathway.
Collapse
Affiliation(s)
- Leqi Tian
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, Guangdong 518172, P.R. China
- Shenzhen Research Institute of Big Data, Shenzhen, Guangdong 518172, P.R. China
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen, Guangdong 518172, P.R. China
| | - Tianwei Yu
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen, Guangdong 518172, P.R. China
- Shenzhen Research Institute of Big Data, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
5
|
Benke M, Zeöld A, Kittel Á, Khamari D, Hritz I, Horváth M, Keczer B, Borka K, Szücs Á, Wiener Z. MiR-200b categorizes patients into pancreas cystic lesion subgroups with different malignant potential. Sci Rep 2023; 13:19820. [PMID: 37963969 PMCID: PMC10646105 DOI: 10.1038/s41598-023-47129-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
Extracellular vesicles (EV) carry their cargo in a membrane protected form, however, their value in early diagnostics is not well known. Although pancreatic cysts are heterogeneous, they can be clustered into the larger groups of pseudocysts (PC), and serous and mucinous pancreatic cystic neoplasms (S-PCN and M-PCN, respectively). In contrast to PCs and S-PCNs, M-PCNs may progress to malignant pancreatic cancers. Since current diagnostic tools do not meet the criteria of high sensitivity and specificity, novel methods are urgently needed to differentiate M-PCNs from other cysts. We show that cyst fluid is a rich source of EVs that are positive and negative for the EV markers CD63 and CD81, respectively. Whereas we found no difference in the EV number when comparing M-PCN with other pancreatic cysts, our EV-based biomarker identification showed that EVs from M-PCNs had a higher level of miR-200b. We also prove that not only EV-derived, but also total cyst fluid miR-200b discriminates patients with M-PCN from other pancreatic cysts with a higher sensitivity and specificity compared to other diagnostic methods, providing the possibility for clinical applications. Our results show that measuring miR-200b in cyst fluid-derived EVs or from cyst fluid may be clinically important in categorizing patients.
Collapse
Affiliation(s)
- Márton Benke
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Anikó Zeöld
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Ágnes Kittel
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
- HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell and Immunobiology, and HUN-REN-SU Translational Extracellular Vesicle Research Group, Semmelweis University, Budapest, Hungary
| | - István Hritz
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Miklós Horváth
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Bánk Keczer
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Katalin Borka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Ákos Szücs
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, Budapest, Hungary.
| | - Zoltán Wiener
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
6
|
Zhou X, Wang W, Li Z, Chen L, Wen C, Ruan Q, Xu Z, Liu R, Xu J, Bai Y, Deng J. Rosmarinic Acid Decreases the Malignancy of Pancreatic Cancer Through Inhibiting Gli1 Signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153861. [PMID: 34864627 DOI: 10.1016/j.phymed.2021.153861] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Rosmarinic acid (RA) has been shown to exert anti-tumor effects on various types of cancer. However, its roles in the treatment of pancreatic ductal adenocarcinoma (PDAC) and the underlying mechanisms remain elusive. PURPOSE The present study aimed to investigate the therapeutic effects of RA on PDAC as well as the underlying mechanisms. STUDY DESIGN Evaluation of the effects of RA on PDAC malignancy both in vitro and in vivo. METHODS Cell counting kit 8 (CCK8) assay, colony formation assay, 5-Ethynyl-2'-deoxyuridine (EDU) incorporation assay, cell cycle analysis, and apoptosis assay were conducted to assess the inhibitory effect of RA on PDAC cell proliferation. Meanwhile, western blotting and RT-qPCR assay were performed to detect the target gene expression at protein and mRNA levels, respectively. Moreover, the in vivo anti-tumor activities of RA were assayed in an xenograft mouse model of PDAC. RESULTS RA dramatically down-regulated Gli1 and its downstream targets. Further studies showed that RA prevents the nuclear translocation of Gli1, while promoting the degradation of cytosolic Gli1 via the proteasome pathway. Moreover, we observed that RA induced G1/S cell cycle arrest and apoptosis in the PDAC cells through regulating the expression of P21, P27, CDK2, Cyclin E, Bax, and Bcl-2, it inhibited the PDAC cell migration and invasion via E-cadherin and MMP-9. Notably, Gli1 overexpression markedly reversed the above RA-induced effects on PDAC cells, whereas Gli1 knockdown enhanced the effects. Additionally, the in vivo assays demonstrated that RA suppresses the tumor growth of PDAC presumably by inhibiting Gli1. CONCLUSION We provided evidence that RA restrained the nuclear translocation of Gli1 and facilitates Gli1 degradation via proteasome pathway, reducing the malignancy of PDAC cells. These findings implicated RA as a therapeutic agent for PDAC.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weiming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhaofeng Li
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Lin Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325035, China
| | - Chunmei Wen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qingqing Ruan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zheng Xu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Rongdiao Liu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jinzhong Xu
- Department of Clinical Pharmacy, The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling 317500, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Center for Health Assessment, Wenzhou Medical University, Wenzhou 325000, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Center for Health Assessment, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
7
|
van Roey R, Brabletz T, Stemmler MP, Armstark I. Deregulation of Transcription Factor Networks Driving Cell Plasticity and Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:753456. [PMID: 34888306 PMCID: PMC8650502 DOI: 10.3389/fcell.2021.753456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a very aggressive disease with 5-year survival rates of less than 10%. The constantly increasing incidence and stagnant patient outcomes despite changes in treatment regimens emphasize the requirement of a better understanding of the disease mechanisms. Challenges in treating pancreatic cancer include diagnosis at already progressed disease states due to the lack of early detection methods, rapid acquisition of therapy resistance, and high metastatic competence. Pancreatic ductal adenocarcinoma, the most prevalent type of pancreatic cancer, frequently shows dominant-active mutations in KRAS and TP53 as well as inactivation of genes involved in differentiation and cell-cycle regulation (e.g. SMAD4 and CDKN2A). Besides somatic mutations, deregulated transcription factor activities strongly contribute to disease progression. Specifically, transcriptional regulatory networks essential for proper lineage specification and differentiation during pancreas development are reactivated or become deregulated in the context of cancer and exacerbate progression towards an aggressive phenotype. This review summarizes the recent literature on transcription factor networks and epigenetic gene regulation that play a crucial role during tumorigenesis.
Collapse
Affiliation(s)
- Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
CDKN2A-Mutated Pancreatic Ductal Organoids from Induced Pluripotent Stem Cells to Model a Cancer Predisposition Syndrome. Cancers (Basel) 2021; 13:cancers13205139. [PMID: 34680288 PMCID: PMC8533699 DOI: 10.3390/cancers13205139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide a unique platform to study hereditary disorders and predisposition syndromes by resembling germline mutations of affected individuals and by their potential to differentiate into nearly every cell type of the human body. We employed plucked human hair from two siblings with a family history of cancer carrying a pathogenic CDKN2A variant, P16-p.G101W/P14-p.R115L, to generate patient-specific iPSCs in a cancer-prone ancestry for downstream analytics. The differentiation capacity to pancreatic progenitors and to pancreatic duct-like organoids (PDLOs) according to a recently developed protocol remained unaffected. Upon inducible expression of KRASG12Dusing a piggyBac transposon system in CDKN2A-mutated PDLOs, we revealed structural and molecular changes in vitro, including disturbed polarity and epithelial-to-mesenchymal (EMT) transition. CDKN2A-mutated KRASG12DPDLO xenotransplants formed either a high-grade precancer lesion or a partially dedifferentiated PDAC-like tumor. Intriguingly, P14/P53/P21 and P16/RB cell-cycle checkpoint controls have been only partly overcome in these grafts, thereby still restricting the tumorous growth. Hereby, we provide a model for hereditary human pancreatic cancer that enables dissection of tumor initiation and early development starting from patient-specific CDKN2A-mutated pluripotent stem cells.
Collapse
|
9
|
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, Kong B, Fu DL, Bruns C, Zhao Y, Qin LX, Dong QZ. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther 2021; 6:249. [PMID: 34219130 PMCID: PMC8255319 DOI: 10.1038/s41392-021-00659-4] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an increasingly common cause of cancer mortality with a tight correspondence between disease mortality and incidence. Furthermore, it is usually diagnosed at an advanced stage with a very dismal prognosis. Due to the high heterogeneity, metabolic reprogramming, and dense stromal environment associated with pancreatic cancer, patients benefit little from current conventional therapy. Recent insight into the biology and genetics of pancreatic cancer has supported its molecular classification, thus expanding clinical therapeutic options. In this review, we summarize how the biological features of pancreatic cancer and its metabolic reprogramming as well as the tumor microenvironment regulate its development and progression. We further discuss potential biomarkers for pancreatic cancer diagnosis, prediction, and surveillance based on novel liquid biopsies. We also outline recent advances in defining pancreatic cancer subtypes and subtype-specific therapeutic responses and current preclinical therapeutic models. Finally, we discuss prospects and challenges in the clinical development of pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Qiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhe-Fang Wang
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Xiao-Lin Wu
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Cheng-Hui Zhou
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Jia-Yan Yan
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
- Key laboratory of whole-period monitoring and precise intervention of digestive cancer, Shanghai Municipal Health Commission (SMHC), Shanghai, China.
| |
Collapse
|
10
|
Wang H, Shi X, Wu S. miR-550a-3/NFIC plays a driving role in esophageal squamous cell cancer cells proliferation and metastasis partly through EMT process. Mol Cell Biochem 2020; 472:115-123. [PMID: 32567032 DOI: 10.1007/s11010-020-03790-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
In this study, the functional role of miR-550a-3 and its direct target nuclear factor IC (NFIC) in esophageal squamous cell cancer (ESCC) cells were explored. Differential expression of miR-550a-3 in ESCC tissues was acquired from TCGA database, and Kaplan-Meier method was used to determine the relationship between miR-550a-3 expression and survival time of ESCC patients. Expression level of miR-550a-3 in several ESCC cell lines was measured by qRT-PCR. Two cell lines including Eca109 and JAR were used to perform proliferation, cloning, invasion and migration experiments. Targeted relationship between miR-550a-3 and NFIC was speculated by predication software and confirmed by dual luciferase assay. Additionally, potential relationship between miR-550a-3 and NFIC was analyzed by Spearman rank correlation analysis and western blot. Rescue assays were performed to explore the function of miR-550a-3/NFIC in ESCC cells biological behaviors. Expression levels of key proteins involved in epithelial-to-mesenchymal transition (EMT) process were determined by western blot. By consulting TCGA database, we found that high expression of miR-550a-3 was positively connected with the poor prognosis of patients with ESCC. In addition, overexpression of miR-550a-3 promoted the proliferation, colony formation and metastasis of ESCC cells. Moreover, rescue assays revealed that overexpression of NFIC attenuated the promoting effects of miR-550a-3 on ESCC cells malignant behaviors. While the promoting effects of miR-550a-3 on EMT process were inhibited by NFIC. Our results illustrate the importance of miR-550a-3/NFIC in regulation of ESCC cells growth and metastasis, which could contribute to developing novel target for early diagnosis or neoteric therapeutic target for ESCC.
Collapse
Affiliation(s)
- Huiqing Wang
- Gastroenterology, The Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Dalian, Liaoning, China
| | - Xiaoyu Shi
- Thoracic Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Shanbin Wu
- Gastroenterology, The Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Dalian, Liaoning, China.
| |
Collapse
|
11
|
Peperstraete E, Lecerf C, Collette J, Vennin C, Raby L, Völkel P, Angrand PO, Winter M, Bertucci F, Finetti P, Lagadec C, Meignan S, Bourette RP, Bourhis XL, Adriaenssens E. Enhancement of Breast Cancer Cell Aggressiveness by lncRNA H19 and its Mir-675 Derivative: Insight into Shared and Different Actions. Cancers (Basel) 2020; 12:cancers12071730. [PMID: 32610610 PMCID: PMC7407157 DOI: 10.3390/cancers12071730] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is a major public health problem and the leading world cause of women death by cancer. Both the recurrence and mortality of breast cancer are mainly caused by the formation of metastasis. The long non-coding RNA H19, the precursor of miR-675, is involved in breast cancer development. The aim of this work was to determine the implication but, also, the relative contribution of H19 and miR-675 to the enhancement of breast cancer metastatic potential. We showed that both H19 and miR-675 increase the invasive capacities of breast cancer cells in xenografted transgenic zebrafish models. In vitro, H19 and miR-675 enhance the cell migration and invasion, as well as colony formation. H19 seems to induce the epithelial-to-mesenchymal transition (EMT), with a decreased expression of epithelial markers and an increased expression of mesenchymal markers. Interestingly, miR-675 simultaneously increases the expression of both epithelial and mesenchymal markers, suggesting the induction of a hybrid phenotype or mesenchymal-to-epithelial transition (MET). Finally, we demonstrated for the first time that miR-675, like its precursor H19, increases the stemness properties of breast cancer cells. Altogether, our data suggest that H19 and miR-675 could enhance the aggressiveness of breast cancer cells through both common and different mechanisms.
Collapse
Affiliation(s)
- Evodie Peperstraete
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Clément Lecerf
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Jordan Collette
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Constance Vennin
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Ludivine Raby
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Pamela Völkel
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Pierre-Olivier Angrand
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Marie Winter
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - François Bertucci
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Département d’Oncologie Médicale, Institut Paoli-Calmettes, 13009 Marseille, France; (F.B.); (P.F.)
| | - Pascal Finetti
- Laboratoire d’Oncologie Prédictive, CRCM, Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR7258, Aix-Marseille Université, Département d’Oncologie Médicale, Institut Paoli-Calmettes, 13009 Marseille, France; (F.B.); (P.F.)
| | - Chann Lagadec
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Samuel Meignan
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Roland P. Bourette
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Xuefen Le Bourhis
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
| | - Eric Adriaenssens
- University Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020–UMR 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.P.); (C.L.); (J.C.); (C.V.); (L.R.); (P.V.); (P.-O.A.); (M.W.); (C.L.); (S.M.); (R.P.B.); (X.L.B.)
- Correspondence: ; Tel.: +33-(0)3-20-33-64-06
| |
Collapse
|
12
|
Jin Y, Weng Y, Wang Y, Lin J, Deng X, Shen B, Zhan Q, Lu X. miR-934 as a Prognostic Marker Facilitates Cell Proliferation and Migration of Pancreatic Tumor by Targeting PROX1. Onco Targets Ther 2020; 13:3389-3399. [PMID: 32368095 PMCID: PMC7183785 DOI: 10.2147/ott.s249662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/06/2020] [Indexed: 01/09/2023] Open
Abstract
Background Pancreatic cancer is an extremely lethal digestive cancer with late diagnosis and poor prognosis. miR-934 has been reported to serve as an oncogene in multiple cancers, such as ovarian cancer and bladder cancer. However, its role in pancreatic cancer remains undiscovered. Materials and Methods The expression data of miR-934 were obtained from the Gene Expression Omnibus database and from our own patient samples. The clinicopathological data and corresponding follow-up data were retrieved from The Cancer Genome Atlas database. CCK8 and colony formation assays were conducted to measure cell proliferation capacity in vitro. Wound healing and transwell assays were performed to detect the migration ability of pancreatic cancer cell. Results We found that miR-934 was significantly upregulated in pancreatic tumor samples and cell lines. The expression of miR-934 was related to pathological stages. Upregulated miR-934 was associated with poor prognosis in patients with pancreatic cancer. Mir-934 inhibition reduced, while overexpression promoted, cell proliferation and migration. Mechanically, we found miR-934 could directly bind to 3'-UTR of PROX1 leading to mRNA derogation. Furthermore, increased cell proliferation and migration caused by miR-934 overexpression could be reversed by forced PROX1 expression. Conclusion miR-934 is an oncogene in pancreatic cancer and could serve as a prognosis indicator for patients with pancreatic cancer, suggesting that miR-934 is a promising therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Yangbing Jin
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Yuanchi Weng
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Yue Wang
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Jiewei Lin
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Xiaxing Deng
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Baiyong Shen
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Qian Zhan
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Xiongxiong Lu
- Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.,Research Institute of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| |
Collapse
|
13
|
Thomas D, Radhakrishnan P. Pancreatic Stellate Cells: The Key Orchestrator of The Pancreatic Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:57-70. [PMID: 32040855 DOI: 10.1007/978-3-030-37184-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer is one of the most challenging adenocarcinomas due to its hostile molecular behavior and complex tumor microenvironment. It has been recently postulated that pancreatic stellate cells (PSCs), the resident lipid-storing cells of the pancreas, are important components of the tumor microenvironment as they can transdifferentiate into highly proliferative myofibroblasts in the context of tissue injury. Targeting tumor-stromal crosstalk in the tumor microenvironment has emerged as a promising therapeutic strategy against pancreatic cancer progression and metastasis. This chapter brings a broad view on the biological and pathological role of PSCs in the pancreas, activated stellate cells in the onset of tissue fibrosis, and tumor progression with particular emphasis on the bidirectional interactions between tumor cells and PSCs. Further, potential therapeutic regimens targeting activated PSCs in the pre-clinical and clinical trials are discussed.
Collapse
Affiliation(s)
- Divya Thomas
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
14
|
Monkman JH, Thompson EW, Nagaraj SH. Targeting Epithelial Mesenchymal Plasticity in Pancreatic Cancer: A Compendium of Preclinical Discovery in a Heterogeneous Disease. Cancers (Basel) 2019; 11:E1745. [PMID: 31703358 PMCID: PMC6896204 DOI: 10.3390/cancers11111745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a particularly insidious and aggressive disease that causes significant mortality worldwide. The direct correlation between PDAC incidence, disease progression, and mortality highlights the critical need to understand the mechanisms by which PDAC cells rapidly progress to drive metastatic disease in order to identify actionable vulnerabilities. One such proposed vulnerability is epithelial mesenchymal plasticity (EMP), a process whereby neoplastic epithelial cells delaminate from their neighbours, either collectively or individually, allowing for their subsequent invasion into host tissue. This disruption of tissue homeostasis, particularly in PDAC, further promotes cellular transformation by inducing inflammatory interactions with the stromal compartment, which in turn contributes to intratumoural heterogeneity. This review describes the role of EMP in PDAC, and the preclinical target discovery that has been conducted to identify the molecular regulators and effectors of this EMP program. While inhibition of individual targets may provide therapeutic insights, a single 'master-key' remains elusive, making their collective interactions of greater importance in controlling the behaviours' of heterogeneous tumour cell populations. Much work has been undertaken to understand key transcriptional programs that drive EMP in certain contexts, however, a collaborative appreciation for the subtle, context-dependent programs governing EMP regulation is needed in order to design therapeutic strategies to curb PDAC mortality.
Collapse
Affiliation(s)
- James H. Monkman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Erik W. Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Shivashankar H. Nagaraj
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| |
Collapse
|
15
|
Lee J, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol 2019; 13:1623-1650. [PMID: 31243883 PMCID: PMC6670020 DOI: 10.1002/1878-0261.12537] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/07/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Reliable biomarkers are required to evaluate and manage pancreatic ductal adenocarcinoma. Circulating tumor cells and circulating tumor DNA are shed into blood and can be relatively easily obtained from minimally invasive liquid biopsies for serial assays and characterization, thereby providing a unique potential for early diagnosis, forecasting disease prognosis, and monitoring of therapeutic response. In this review, we provide an overview of current technologies used to detect circulating tumor cells and circulating tumor DNA and describe recent advances regarding the multiple clinical applications of liquid biopsy in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jee‐Soo Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Sung Sup Park
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Young Kyung Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineHallym University College of MedicineAnyangKorea
| | - Jeffrey A. Norton
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | | |
Collapse
|
16
|
Chen X, Cheng F, Liu Y, Zhang L, Song L, Cai X, You T, Fan X, Wang D, Gong A, Zhu H. Toll-like receptor 2 and Toll-like receptor 4 exhibit distinct regulation of cancer cell stemness mediated by cell death-induced high-mobility group box 1. EBioMedicine 2019; 40:135-150. [PMID: 30679086 PMCID: PMC6413584 DOI: 10.1016/j.ebiom.2018.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND High-mobility group box 1 (HMGB1), a common extracellular damage associated molecular pattern molecule, is overexpressed in several solid tumors including pancreatic carcinoma. We previously observed that radiotherapy induced dying cells secrete HMGB1 and accelerate pancreatic carcinoma progression through an unclear mechanism. METHODS Using the Millicell system as an in vitro co-culture model, we performed quantitative reverse transcriptase-polymerase chain reaction, western blot and sphere forming ability analyses to access the effect of dying-cell-derived HMGB1 on CD133+ cancer cell stemness in vitro and in vivo. Interactions between HMGB1 and Toll-like receptor 2(TLR2)/TLR4 were studied by co- immunoprecipitation. Western blot and short-hairpin RNA-based knockdown assays were conducted to detect HMGB1 and TLR2/TLR4 signaling activity. FINDINGS Radiation-associated, dying-cell-derived HMGB1 maintained stemness and contributed to CD133+ cancer stem cell self-renewal in vitro and in vivo. In overexpressing and silencing experiments, we demonstrated that the process was activated by TLR2 receptor, whereas TLR4 antagonized HMGB1-TLR2 signaling. Wnt/β-catenin signaling supported the HMGB1-TLR2 mediated stemness of CD133+ cancer cells. INTERPRETATION Our results show how irradiation-induced cell death might enhance the stemness of resident cancer cells, and indicate HMGB1-TLR2 signaling as a potential therapeutic target for preventing pancreatic cancer recurrence.
Collapse
Affiliation(s)
- Xuelian Chen
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Fang Cheng
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- School of Pharmaceutical Sciences (Shenzhen), SYSU, 510006, China
- Faculty of Science and Engineering, Åbo Akademi University and Turku Centre for Biotechnology, Turku FI-20520, Finland
| | - Yanfang Liu
- Department of Central Laboratory, The First People's Hospital of Zhenjiang, Zhenjiang 212001, China
| | - Lirong Zhang
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Lian Song
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xiaojie Cai
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Tao You
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xin Fan
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Haitao Zhu
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
17
|
Samandari M, Julia MG, Rice A, Chronopoulos A, Del Rio Hernandez AE. Liquid biopsies for management of pancreatic cancer. Transl Res 2018; 201:98-127. [PMID: 30118658 DOI: 10.1016/j.trsl.2018.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/17/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the main causes of cancer-related deaths worldwide. It is asymptomatic at an early stage, and most diagnosis occurs when the disease is already at a late stage, by which time the tumor is nonresectable. In order to increase the overall survival of patients with pancreatic cancer, as well as to decrease the cancer burden, it is necessary to perform early diagnosis, prognosis stratifications and cancer monitoring using accurate, minimally invasive, and cost-effective methods. Liquid biopsies seek to detect tumor-associated biomarkers in a variety of extractable body fluids and can help to monitor treatment response and disease progression, and even predict patient outcome. In patients with pancreatic cancer, tumor-derived materials, primarily circulating tumor DNA, circulating tumor cells and exosomes, are being studied for inclusion in the management of the disease. This review focuses on describing the biology of these biomarkers, methods for their enrichment and detection, as well as their potential for clinical application. Moreover, we discuss the future direction of liquid biopsies and introduce how they can be exploited toward point of care personalized medicine for the management of pancreatic cancer.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - María Gil Julia
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
18
|
Li ZY, Sun XY. Molecular targets regulating invasion and metastasis of pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2018; 26:1651-1659. [DOI: 10.11569/wcjd.v26.i28.1651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zi-Yi Li
- The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xue-Ying Sun
- The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
19
|
Huang Y, Li G, Wang K, Mu Z, Xie Q, Qu H, Lv H, Hu B. Collagen Type VI Alpha 3 Chain Promotes Epithelial-Mesenchymal Transition in Bladder Cancer Cells via Transforming Growth Factor β (TGF-β)/Smad Pathway. Med Sci Monit 2018; 24:5346-5354. [PMID: 30066698 PMCID: PMC6085978 DOI: 10.12659/msm.909811] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Collagen type VI alpha 3 chain (COL6A3) has been proven to be a biomarker in the occurrence and development of bladder cancer, which is the most common malignant tumor in the urinary system. This study aimed to explore the effect and molecular mechanism of COL6A3 on EMT in vitro induced by TGF-β/Smad in bladder carcinoma. Material/Methods There were 42 patients included in the Kaplan-Meier survival analysis. A cell counting kit-8 (CCK-8) assay and an angiogenesis assay were used to measure cell proliferation and tube formation, respectively. Western blot analysis and quantitative reverse transcription-polymerase chain reaction (qPCR) were conducted for the proteins and mRNAs expression. Results COL6A3 was highly expressed in tissues and cells of bladder cancer. COL6A3 silencing could inhibit the cell proliferation and angiopoiesis. In addition, COL6A3 silencing obviously suppressed the levels of matrix metalloproteinase-2 (MMP2), Matrix metalloproteinase-9 (MMP9), and vimentin. On the contrary, the levels of epithelium-specific cell-cell adhesion molecule (E-cadherin) and tumor inhibitor of metalloproteinase-1 (TIMP-1) were significantly increased. Furthermore, we found that COL6A3 silencing reduced the activity of p-Smad2, p-Smad3, and transforming growth factor β (TGF-β). Conclusions COL6A3 could influence the viability and angiogenesis of bladder cancer cells. COL6A3 may have a certain relationship with the TGF-β/Smad-induced EMT process.
Collapse
Affiliation(s)
- Yan Huang
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Gang Li
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Kai Wang
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Zhongyi Mu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Qingpeng Xie
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Hongchen Qu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Hang Lv
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Bin Hu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
20
|
Jiang W, Yuan Q, Jiang Y, Huang L, Chen C, Hu G, Wan R, Wang X, Yang L. Identification of Sox6 as a regulator of pancreatic cancer development. J Cell Mol Med 2018; 22:1864-1872. [PMID: 29369542 PMCID: PMC5824410 DOI: 10.1111/jcmm.13470] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/26/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer (PC) is an aggressive malignancy associated with a poor prognosis and low responsiveness to chemotherapy and radiotherapy. Most patients with PC have metastatic disease at diagnosis, which partly accounts for the high mortality from this disease. Here, we explored the role of the transcription factor sex‐determining region Y‐box (Sox) 6 in the invasiveness of PC cells. We showed that Sox6 is down‐regulated in patients with PC in association with metastatic disease. Sox6 overexpression suppressed PC cell proliferation and migration in vitro and tumour growth and liver metastasis in vivo. Sox6 inhibited epithelial‐mesenchymal transition (EMT), and Akt signalling. Sox6 was shown to interact with the promoter of Twist1, a helix–loop–helix transcription factor involved in the induction of EMT, and to modulate the expression of Twist1 by recruiting histone deacetylase 1 to the promoter of the Twist1 gene. Twist1 overexpression reversed the effect of Sox6 on inhibiting EMT, confirming that the effect of Sox6 on suppressing tumour invasiveness is mediated by the modulation of Twist1 expression. These results suggest a novel mechanism underlying the aggressive behaviour of PC cells and identify potential therapeutic targets for the treatment of PC.
Collapse
Affiliation(s)
- Weiliang Jiang
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Qiongying Yuan
- Department of Gastroenterology, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yuanye Jiang
- Department of Gastroenterology, The Central Hospital of Putuo District, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Huang
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyong Hu
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Wan
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Xingpeng Wang
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Yang
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital/First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Disease, School of Medicine, Institute of Pancreatic Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Zhou HS, Su XF, Fu XL, Wu GZ, Luo KL, Fang Z, Yu F, Liu H, Hu HJ, Chen LS, Cai B, Tian ZQ. Mesenchymal stem cells promote pancreatic adenocarcinoma cells invasion by transforming growth factor-β1 induced epithelial-mesenchymal transition. Oncotarget 2018; 7:41294-41305. [PMID: 27191496 PMCID: PMC5173060 DOI: 10.18632/oncotarget.9319] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/25/2016] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) could be ideal delivery vehicles for antitumor biological agents in pancreatic adenocarcinoma (PA). While the role of MSCs in tumor growth is elusive. Inflammation is an important feature of PA. In this study, we reported that MSCs pre-stimulated with the combination of TNF-α and IFN-γ promote PA cells invasion. The invasion of PA cell lines were evaluate by wound healing assay and transwell assay in vitro and liver metastasis in nude mice. We observed MSCs pre-stimulated with the combination of TNF-α and IFN-γ promoted PA cells invasion in vitro and in vivo. Consistent with MSCs promoting PA cells invasion, PA cells were found undergo epithelial-mesenchymal transition (EMT). We demonstrated that MSCs pre-stimulated with both of TNF-α and IFN-γ provoked expression transforming growth factor-β1 (TGF-β1). MSCs promoting EMT-mediated PA cells invasion could be reversed by short interfering RNA of TGF-β1. Our results suggest that MSCs could promote PA cells invasion in inflammation microenvironment and should be cautious as delivery vehicles in molecular target therapy.
Collapse
Affiliation(s)
- Hai-Sen Zhou
- Nanjing Lishui People's Hospital, Nanjing 211200, P.R. China
| | - Xiao-Fang Su
- Department of Rehabilitation Medicine, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Xing-Li Fu
- Health Science Center, Jiangsu University, Zhenjiang 212013, P.R. China
| | - Guo-Zhong Wu
- Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Kun-Lun Luo
- Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Zheng Fang
- Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Feng Yu
- Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Hong Liu
- Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| | - Hong-Juan Hu
- Nanjing Lishui People's Hospital, Nanjing 211200, P.R. China
| | - Liu-Sheng Chen
- Nanjing Lishui People's Hospital, Nanjing 211200, P.R. China
| | - Bing Cai
- Department of General Surgery, Wuxi People's Hospital, Nanjing Medical University, Wuxi 214023, P.R. China
| | - Zhi-Qiang Tian
- Department of General Surgery, Wuxi People's Hospital, Nanjing Medical University, Wuxi 214023, P.R. China.,Department of General Surgery, The 101st Hospital of Chinese PLA, Wuxi 214044, P.R. China
| |
Collapse
|
22
|
Chouat E, Zehani A, Chelly I, Njima M, Maghrebi H, Bani MA, Njim L, Zakhama A, Haouet S, Kchir N. Tumor budding is a prognostic factor linked to epithelial mesenchymal transition in pancreatic ductal adenocarcinoma. Study report and literature review. Pancreatology 2018; 18:79-84. [PMID: 29233500 DOI: 10.1016/j.pan.2017.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/04/2017] [Accepted: 11/19/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a devastatingly poor prognosis. Surgical resection is undertaken in only 20% of patients. Most of well-known prognostic factors reflect tumor stage more than its biology. So it is important to identify new biological indicators related to survival in order to develop new therapies. OBJECTIVE To determine the relation between tumor budding and Epithelial Mesenchymal Transition (EMT) and to evaluate their impact on survival for patients after resection of PDAC. METHODS We herein report a retrospective study of 50 patients with resected PDAC. Tumor budding, immunohistochemical expression of vimentin and other standard factors were correlated with survival using the Kaplan-Meier method and Cox multivariable survival analysis. For tumor budding assessment, an inter-observer variability study was performed using 100 images of tumor slides stained with Hematoxylin & Eosin and Pan-Cytokeratin. RESULTS Tumor budding was present in all tumors. A substantial agreement between six pathologists was established in distinguishing high-grade from low-grade budding (κ = 0.6 and 0.73 for H&E and PCK images respectively). High-grade budding was identified in 56% of tumors (28/50). It was an adverse prognostic factor independent of tumor size, resection margins status, nodal status and vascular invasion (p = 0.008). Tumor budding was significantly associated with vimentin expression (p = 0.002). CONCLUSIONS The association of tumor budding with vimentin expression supported the idea that EMT is a key process in PDAC responsible for progression and drug resistance. Consequently, the elucidation of EMT molecular biology and development of new targeted therapy may improve disease outcome.
Collapse
Affiliation(s)
- Ezzeddine Chouat
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia.
| | - Alia Zehani
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| | - Ines Chelly
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| | - Manel Njima
- Department of Pathology, Fattouma Bourguiba University Hospital, 1st June 1955 Street, 5000 Monastir, Tunisia
| | - Houcine Maghrebi
- Department of Surgery A, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| | - Mohammed Amine Bani
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| | - Leila Njim
- Department of Pathology, Fattouma Bourguiba University Hospital, 1st June 1955 Street, 5000 Monastir, Tunisia
| | - Abdelfatteh Zakhama
- Department of Pathology, Fattouma Bourguiba University Hospital, 1st June 1955 Street, 5000 Monastir, Tunisia
| | - Slim Haouet
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| | - Nidhameddine Kchir
- Department of Pathology, Rabta University Hospital, Rabta Jebbari, 1007 Tunis, Tunisia
| |
Collapse
|
23
|
Wu Q, Tian Y, Zhang J, Zhang H, Gu F, Lu Y, Zou S, Chen Y, Sun P, Xu M, Sun X, Xia C, Chi H, Ying Zhu A, Tang D, Wang D. Functions of pancreatic stellate cell-derived soluble factors in the microenvironment of pancreatic ductal carcinoma. Oncotarget 2017; 8:102721-102738. [PMID: 29254283 PMCID: PMC5731993 DOI: 10.18632/oncotarget.21970] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer with poor prognosis because it is highly resistant to traditional chemotherapy and radiotherapy and it has a low rate of surgical resection eligibility. Pancreatic stellate cells (PSC) have become a research hotspot in recent years, and play a vital role in PDAC microenvironment by secreting soluble factors such as transforming growth factor β, interleukin-6, stromal cell-derived factor-1, hepatocyte growth factor and galectin-1. These PSC-derived cytokines and proteins contribute to PSC activation, participating in PDAC cell proliferation, migration, fibrosis, angiogenesis, immunosuppression, epithelial-mesenchymal transition, and chemoradiation resistance, leading to malignant outcome. Consequently, targeting these cytokines and proteins or their downstream signaling pathways is promising for treating PDAC.
Collapse
Affiliation(s)
- Qi Wu
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Ying Tian
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Jingqiu Zhang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Hongpeng Zhang
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Fengming Gu
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Yongdie Lu
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Shengnan Zou
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Yuji Chen
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Pengxiang Sun
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Mengyue Xu
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Xiaoming Sun
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Chao Xia
- Nanjing Medical University, Nanjing, P.R. China
| | - Hao Chi
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - A Ying Zhu
- Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| |
Collapse
|
24
|
Cai J, Du S, Wang H, Xin B, Wang J, Shen W, Wei W, Guo Z, Shen X. Tenascin-C induces migration and invasion through JNK/c-Jun signalling in pancreatic cancer. Oncotarget 2017; 8:74406-74422. [PMID: 29088796 PMCID: PMC5650351 DOI: 10.18632/oncotarget.20160] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/20/2017] [Indexed: 12/21/2022] Open
Abstract
Tenascin-C (TNC), a large extracellular matrix glycoprotein, has been reported to be associated with metastasis and poor prognosis in pancreatic cancer. However, the effects and mechanisms of TNC in pancreatic cancer metastasis largely remain unclear. We performed Transwell assays to investigate the effects of TNC on Capan-2, AsPC-1 and PANC-1 cells. In addition, western blot and RT-qPCR assays were used to examine potential TNC metastasis-associated targets, such as JNK/c-Jun, Paxillin/FAK, E-cadherin, N-cadherin, Vimentin, and MMP9/2. Lastly, we utilized a variety of methods, such as immunofluorescence, gelatin zymography and immunoprecipitation, to determine the molecular mechanisms of TNC in pancreatic cancer cell motility. The present study showed that TNC induced migration and invasion in pancreatic cancer cells and regulated a number of metastasis-associated proteins, including the EMT markers, MMP9 and Paxillin. Moreover, our data showed that TNC induced pancreatic cancer cells to generate an EMT phenotype and acquire motility potential through the activation of JNK/c-Jun signalling. In addition, TNC increased the DNA binding activity of c-Jun to the MMP9 promoter, an action likely resulting in increased MMP9 expression and activity. TNC/JNK also markedly induced the phosphorylation of Paxillin on serine 178, which is critical for the association between FAK and Paxillin and promoted the formation of focal adhesions. TNC/JNK initiates cell migration and invasion of pancreatic cancer cells through the promotion of EMT, the transactivation of MMP9 and the phosphorylation of Paxillin on serine 178. TNC may be a potential therapeutic target for treating pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Jun Cai
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shaoxia Du
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Hui Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Beibei Xin
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Wenyuan Shen
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Wei Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhongkui Guo
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaohong Shen
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
25
|
Khalafalla FG, Khan MW. Inflammation and Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma: Fighting Against Multiple Opponents. CANCER GROWTH AND METASTASIS 2017; 10:1179064417709287. [PMID: 28579826 PMCID: PMC5436837 DOI: 10.1177/1179064417709287] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and one of the most lethal human cancers. Inflammation is a critical component in PDAC initiation and progression. Inflammation also contributes to the aggressiveness of PDAC indirectly via induction of epithelial-mesenchymal transition (EMT), altogether leading to enhanced resistance to chemotherapy and poor survival rates. This review gives an overview of the key pro-inflammatory signaling pathways involved in PDAC pathogenesis and discusses the role of inflammation in induction of EMT and development of chemoresistance in patients with PDAC.
Collapse
|
26
|
Qiu Z, Chu Y, Xu B, Wang Q, Jiang M, Li X, Wang G, Yu P, Liu G, Wang H, Kang H, Liu J, Zhang Y, Jin JP, Wu K, Liang J. Increased expression of calponin 2 is a positive prognostic factor in pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:56428-56442. [PMID: 28915602 PMCID: PMC5593573 DOI: 10.18632/oncotarget.17701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/10/2017] [Indexed: 02/06/2023] Open
Abstract
Calponin 2 plays an important role in regulating actin cytoskeleton, which is critical for cell division and migration. Previous studies have demonstrated that calponin 2 inhibits prostate cancer cell proliferation and metastasis. However, the role of calponin 2 in pancreatic tumor growth, metastasis and patient survival remains unclear. Here, we demonstrate that the level of calponin 2 is a positive prognostic factor for patients with pancreatic ductal adenocarcinoma (PDAC). Patients with high calponin 2 expression in the tumor presented less lymph node metastasis and longer survival. Knockdown of calponin 2 facilitated pancreatic cancer cell proliferation and metastasis. Further experiments suggested that PI3K/AKT, NF-κB, Vimentin, Fibronectin, Snail and Slug were upregulated and E-cadherin was downregulated after calponin 2 was knocked down, implicating altered functions in PDAC proliferation and metastasis. In addition, we verified that calponin 2 functioned through inhibiting PI3K/AKT and NF-κB pathways. Our study suggests that the upregulation of calponin 2 in PDAC correlates to lower malignancy and presents a novel target for the development of new treatment.
Collapse
Affiliation(s)
- Zhaoyan Qiu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bing Xu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qian Wang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaowei Li
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Pengfei Yu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guoxiao Liu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hua Wang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Huijie Kang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiayu Liu
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Yu Zhang
- Department of Cardiovascular Surgery, General Hospital of Lanzhou Military Area Command, Lanzhou, China
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Liang
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Mody HR, Hung SW, Pathak RK, Griffin J, Cruz-Monserrate Z, Govindarajan R. miR-202 Diminishes TGFβ Receptors and Attenuates TGFβ1-Induced EMT in Pancreatic Cancer. Mol Cancer Res 2017; 15:1029-1039. [PMID: 28373289 DOI: 10.1158/1541-7786.mcr-16-0327] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 10/24/2016] [Accepted: 03/29/2017] [Indexed: 12/15/2022]
Abstract
Previous studies in our laboratory identified that 3-deazaneplanocin A (DZNep), a carbocyclic adenosine analog and histone methyl transferase inhibitor, suppresses TGFβ-induced epithelial-to-mesenchymal (EMT) characteristics. In addition, DZNep epigenetically reprograms miRNAs to regulate endogenous TGFβ1 levels via miR-663/4787-mediated RNA interference (Mol Cancer Res. 2016 Sep 13. pii: molcanres.0083.2016) (1). Although DZNep also attenuates exogenous TGFβ-induced EMT response, the mechanism of this inhibition was unclear. Here, DZNep induced miR-202-5p to target both TGFβ receptors, TGFBR1 and TGFBR2, for RNA interference and thereby contributes to the suppression of exogenous TGFβ-induced EMT in pancreatic cancer cells. Lentiviral overexpression of miR-202 significantly reduced the protein levels of both TGFβ receptors and suppressed TGFβ signaling and EMT phenotypic characteristics of cultured parenchymal pancreatic cancer cells. Consistently, transfection of anti-miRNAs against miR-202-5p resulted in increased TGFBR1 and TGFBR2 protein expressions and induced EMT characteristics in these cells. In stellate pancreatic cells, miR-202 overexpression slowed growth as well as reduced stromal extracellular membrane matrix protein expression. In orthotopic pancreatic cancer mouse models, both immunodeficient and immunocompetent, miR-202 reduced tumor burden and metastasis. Together, these findings demonstrate an alternative mechanism of DZNep in suppressing TGFβ signaling at the receptor level and uncover the EMT-suppressing role of miR-202 in pancreatic cancer.Implications: These findings support the possibility of combining small molecule-based (e.g., DZNep analogs) or large molecule-based (e.g., miRNAs) epigenetic modifiers with conventional nucleoside analogs (e.g., gemcitabine, capecitabine) to improve the antimetastatic potential of current pancreatic cancer therapy. Mol Cancer Res; 15(8); 1029-39. ©2017 AACR.
Collapse
Affiliation(s)
- Hardik R Mody
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, Ohio.,Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Sau Wai Hung
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Rakesh K Pathak
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, Ohio
| | - Jazmine Griffin
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Zobeida Cruz-Monserrate
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, Ohio. .,Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia.,The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
28
|
Chirumbolo S, Bjørklund G. The sesquiterpene α-bisabolol in the adipocyte-cancer desmoplastic crosstalk: does it have an action on epithelial-mesenchymal transition mechanisms? Int J Clin Oncol 2017; 22:222-228. [PMID: 27942879 DOI: 10.1007/s10147-016-1072-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
Alpha-bisabolol is a plant-derived sesquiterpene alcohol recently associated with a supposed anti-cancer action due to its ability to induce BID-related apoptosis. The molecule, which enters the cell through lipid rafts, may also interact with kisspeptin receptor 1, which has recently been associated with tumor mobility and invasiveness. This evidence suggests the possibility that α-bisabolol might act on the epithelial-mesenchymal transition mechanism, closely associated with the desmoplastic reaction of adipose tissue surrounding a pancreatic ductal adenocarcinoma. This review addresses the issue on the basis of the most recent reported literature in the field.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Strada Le Grazie 9, Verona, Italy.
- CONEM Scientific Secretary, Mo i Rana, Norway.
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| |
Collapse
|
29
|
Miura K, Kimura K, Amano R, Yamazoe S, Ohira G, Nishio K, Kametani N, Hirakawa K, Ohira M. Analysis of the origin of anaplastic pancreatic cancer and the mechanism of its dedifferentiation. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2017; 24:176-184. [PMID: 28064441 DOI: 10.1002/jhbp.429] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND We researched the origin and progression of anaplastic pancreatic cancer (APC) from the viewpoints of cell lineage, epithelial-mesenchymal transition (EMT) and cancer stem-like cells (CSC). METHODS Using specimens from patients with APC and differentiated pancreatic ductal adenocarcinoma (PDAC), expression of sex-determining region Y-box 9 (SOX9), E-cadherin, vimentin, ZEB1, Snail, N-cadherin, CD24 and CD44 was estimated using immunohistochemistry. RESULTS Almost all cases were positive for SOX9 expression. APC cases were negative, but many PDAC cases were positive for the expression of E-cadherin. A much higher number of APC cases than PDAC cases were positive for the expression of other EMT related proteins and for the expression of CSC related proteins. The ductal cancerous component of APC accounted for an average of 12% of the cancerous lesion and the expression of each marker in this component was similar to that of PDAC cases. CONCLUSIONS Anaplastic pancreatic cancer had pancreatic duct cell like features and might gain dedifferentiate components through EMT and the acquisition of CSC properties.
Collapse
Affiliation(s)
- Kotaro Miura
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kenjiro Kimura
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ryosuke Amano
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Sadaaki Yamazoe
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Go Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kohei Nishio
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Naoki Kametani
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
30
|
Zinovyeva MV, Kuzmich AI, Monastyrskaya GS, Sverdlov ED. The role of FOXA subfamily factors in embryonic development and carcinogenesis of the pancreas. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2017. [DOI: 10.3103/s0891416816030113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Mody HR, Hung SW, AlSaggar M, Griffin J, Govindarajan R. Inhibition of S-Adenosylmethionine-Dependent Methyltransferase Attenuates TGFβ1-Induced EMT and Metastasis in Pancreatic Cancer: Putative Roles of miR-663a and miR-4787-5p. Mol Cancer Res 2016; 14:1124-1135. [PMID: 27624777 PMCID: PMC5107158 DOI: 10.1158/1541-7786.mcr-16-0083] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/29/2016] [Accepted: 08/27/2016] [Indexed: 01/25/2023]
Abstract
The identification of epigenetic reversal agents for use in combination chemotherapies to treat human pancreatic ductal adenocarcinomas (PDAC) remains an unmet clinical need. Pharmacologic inhibitors of Enhancer of Zeste Homolog 2 (EZH2) are emerging as potential histone methylation reversal agents for the treatment of various solid tumors and leukemia; however, the surprisingly small set of mRNA targets identified with EZH2 knockdown suggests novel mechanisms contribute to their antitumorigenic effects. Here, 3-deazaneplanocin-A (DZNep), an inhibitor of S-adenosyl-L-homocysteine hydrolase and EZH2 histone lysine-N-methyltransferase, significantly reprograms noncoding microRNA (miRNA) expression and dampens TGFβ1-induced epithelial-to-mesenchymal (EMT) signals in pancreatic cancer. In particular, miR-663a and miR-4787-5p were identified as PDAC-downregulated miRNAs that were reactivated by DZNep to directly target TGFβ1 for RNA interference. Lentiviral overexpression of miR-663a and miR-4787-5p reduced TGFβ1 synthesis and secretion in PDAC cells and partially phenocopied DZNep's EMT-resisting effects, whereas locked nucleic acid (LNA) antagomiRNAs counteracted them. DZNep, miR-663a, and miR-4787-5p reduced tumor burden in vivo and metastases in an orthotopic mouse pancreatic tumor model. Taken together, these findings suggest the epigenetic reprogramming of miRNAs by synthetic histone methylation reversal agents as a viable approach to attenuate TGFβ1-induced EMT features in human PDAC and uncover putative miRNA targets involved in the process. IMPLICATIONS The findings support the potential for synthetic histone methylation reversal agents to be included in future epigenetic-chemotherapeutic combination therapies for pancreatic cancer. Mol Cancer Res; 14(11); 1124-35. ©2016 AACR.
Collapse
Affiliation(s)
- Hardik R Mody
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, Ohio
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Sau Wai Hung
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Mohammad AlSaggar
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Jazmine Griffin
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| | - Rajgopal Govindarajan
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, Ohio
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia, Athens, Georgia
| |
Collapse
|
32
|
Gutiérrez ML, Corchete L, Teodosio C, Sarasquete ME, del Mar Abad M, Iglesias M, Esteban C, Sayagues JM, Orfao A, Muñoz-Bellvis L. Identification and characterization of the gene expression profiles for protein coding and non-coding RNAs of pancreatic ductal adenocarcinomas. Oncotarget 2016; 6:19070-86. [PMID: 26053098 PMCID: PMC4662476 DOI: 10.18632/oncotarget.4233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022] Open
Abstract
Significant advances have been achieved in recent years in the identification of the genetic and the molecular alterations of pancreatic ductal adenocarcinoma (PDAC). Despite this, at present the understanding of the precise mechanisms involved in the development and malignant transformation of PDAC remain relatively limited. Here, we evaluated for the first time, the molecular heterogeneity of PDAC tumors, through simultaneous assessment of the gene expression profile (GEP) for both coding and non-coding genes of tumor samples from 27 consecutive PDAC patients. Overall, we identified a common GEP for all PDAC tumors, characterized by an increased expression of genes involved in PDAC cell proliferation, local invasion and metastatic capacity, together with a significant alteration of the early steps of the cellular immune response. At the same time, we confirm and extend on previous observations about the genetic complexity of PDAC tumors as revealed by the demonstration of two clearly distinct and unique GEPs (e.g. epithelial-like vs. mesenchymal-like) reflecting the alteration of different signaling pathways involved in the oncogenesis and progression of these tumors. Our results also highlight the potential role of the immune system microenvironment in these tumors, with potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- María Laura Gutiérrez
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL (University of Salamanca), Salamanca, Spain
| | - Luis Corchete
- Cancer Research Center and Service of Hematology (University Hospital of Salamanca), Salamanca, Spain
| | - Cristina Teodosio
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL (University of Salamanca), Salamanca, Spain
| | - María Eugenia Sarasquete
- Cancer Research Center and Service of Hematology (University Hospital of Salamanca), Salamanca, Spain
| | - María del Mar Abad
- Department of Pathology (University Hospital of Salamanca), Salamanca, Spain
| | - Manuel Iglesias
- Service of General and Gastrointestinal Surgery and IBSAL (University Hospital of Salamanca), Salamanca, Spain
| | - Carmen Esteban
- Service of General and Gastrointestinal Surgery and IBSAL (University Hospital of Salamanca), Salamanca, Spain
| | - José María Sayagues
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL (University of Salamanca), Salamanca, Spain
| | - Alberto Orfao
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL (University of Salamanca), Salamanca, Spain
| | - Luis Muñoz-Bellvis
- Service of General and Gastrointestinal Surgery and IBSAL (University Hospital of Salamanca), Salamanca, Spain
| |
Collapse
|
33
|
Gao Y, Zhu Y, Zhang Z, Zhang C, Huang X, Yuan Z. Clinical significance of pancreatic circulating tumor cells using combined negative enrichment and immunostaining-fluorescence in situ hybridization. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:66. [PMID: 27066900 PMCID: PMC4828870 DOI: 10.1186/s13046-016-0340-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/04/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circulating tumor cells (CTCs) hold great potential in both clinical application and basic research for the managements of cancer. However, it remains to be an enormous challenge to obtain efficient detection of pancreatic CTCs. New detection platforms for the detection of pancreatic CTCs are urgently required. METHODS In the present study, we applied a newly-developed platform integrated subtraction enrichment and immunostaining-fluorescence in situ hybridization (SE-iFISH) to analyze clinical significance of pancreatic CTCs. Immunostaining of CK, CD45, DAPI and FISH with the centromere of chromosome 8 (CEP8) were utilized to identify CTCs. Cells with features of CK+/CD45-/DAPI+/CEP8 = 2, CK+/CD45-/DAPI+/CEP8 > 2, CK-/CD45-/DAPI+/CEP8 > 2 were defined as pancreatic CTCs. The Kaplan-Meier method and Cox proportional hazards model were used to analyze the relationship of CTC level and other clinicopathological factors with pancreatic cancer clinical outcomes. RESULTS CTC count in pancreatic cancer was higher than healthy individuals (median, 3 vs. 0 per 7.5 ml; P < 0.001). SE-iFISH platform yielded a sensitivity of 88% and specificity of 90% in pancreatic cancer at the cutoff value of 2 cells/7.5 ml. Pancreatic cancer patients with lower CTC count (<3/7.5 ml) had substantially better overall survival (OS) compared with these with higher CTC count (≥3/7.5 ml) (15.2 vs. 10.2 months, P = 0.023). Multivariate analysis indicated that higher CTC count was a strong indicator for worse OS (HR = 4.547, P = 0.016). CONCLUSION Our current data showed that CTCs could be detected in pancreatic cancer patients in various stages, whether localized, locally advanced and metastatic. Besides, CTCs have shown the potential implication in predicting prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Yang Gao
- Department of General Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233, China
| | - Yayun Zhu
- Department of General Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233, China
| | - Zhenzhen Zhang
- Biotecan Medical Diagnostics Co., Ltd, Zhangjiang Center for Translational Medicine, Shanghai, China
| | - Cheng Zhang
- Biotecan Medical Diagnostics Co., Ltd, Zhangjiang Center for Translational Medicine, Shanghai, China
| | - Xinyu Huang
- Department of General Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233, China
| | - Zhou Yuan
- Department of General Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, No.600, Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
34
|
Wu X, Wu G, Wu Z, Yao X, Li G. MiR-200a Suppresses the Proliferation and Metastasis in Pancreatic Ductal Adenocarcinoma through Downregulation of DEK Gene. Transl Oncol 2016; 9:25-31. [PMID: 26947878 PMCID: PMC4800058 DOI: 10.1016/j.tranon.2015.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/10/2015] [Accepted: 11/10/2015] [Indexed: 12/24/2022] Open
Abstract
MiR-200a has been reported to be able to suppress the epithelial-mesenchymal transition process in pancreatic cancer stem cells, suggesting that miR-200a could suppress the metastasis of pancreatic ductal adenocarcinoma (PDAC). However, its role in proliferation and metastasis of PDAC and the underlying mechanism by which miR-200a works in PDAC have not been elucidated. In our study, we for the first time identified that DEK gene is a direct downstream target of miR-200a. It was found that overexpression of miR-200a decreased DEK expression, suppressing the proliferation, migration, and invasion of PDAC cells. Meanwhile, knockdown of miR-200a can increase DEK level, promoting the proliferation, migration, and invasion of PDAC cells. Our study demonstrated that miR-200a suppresses the metastasis in pancreatic PDAC through downregulation of DEK, suggesting that miR-200a may be used as a novel potential marker in prediction of metastasis of PDAC.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, PR China
| | - Guannan Wu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, PR China
| | - Zhenfeng Wu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, PR China
| | - Xuequan Yao
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, No. 155 Hanzhong Road, Nanjing 210029, PR China
| | - Gang Li
- Department of General Surgery, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting Road, Nanjing 210009, PR China
| |
Collapse
|
35
|
Hasegawa S, Nagano H, Konno M, Eguchi H, Tomokuni A, Tomimaru Y, Asaoka T, Wada H, Hama N, Kawamoto K, Marubashi S, Nishida N, Koseki J, Mori M, Doki Y, Ishii H. A crucial epithelial to mesenchymal transition regulator, Sox4/Ezh2 axis is closely related to the clinical outcome in pancreatic cancer patients. Int J Oncol 2015; 48:145-52. [PMID: 26648239 DOI: 10.3892/ijo.2015.3258] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/07/2015] [Indexed: 11/05/2022] Open
Abstract
Pancreatic cancer has a poor prognosis because of its high invasiveness and recurrence, and these properties closely link to the phenomenon of epithelial-mesenchymal transition (EMT). Recently, it has been reported that Sox4 is indispensable for EMT in vitro and in vivo and regulates various master regulators of EMT including Zeb, Twist and Snail. Moreover, Sox4 induces the transcription of Ezh2 which is the histone methyltransferase, and reprograms the cancer epigenome to promote EMT and metastasis. Therefore, the present study evaluated the importance of Sox4, Ezh2 and miR-335, which regulate Sox4 expression epigenetically, in clinical samples with pancreatic cancer. This retrospective analysis included data from 36 consecutive patients who underwent complete surgical resection for pancreatic cancer and did not undergo any preoperative therapies. We assessed the clinical significance of Sox4/Ezh2 axis and miR-335 expression, using immunohistochemistry and qRT-PCR with laser captured microdissection (LCM). The Sox4 positive patients had significantly worse prognosis as for disease-free survival (DFS) (P=0.0154) and the Ezh2-positive patients had significantly worse prognosis as for overall survival (OS) (P=0.0347). The miR-335 expression was inversely correlated with Sox4 expression in the identical clinical specimens, but it was not related to the prognosis. Sox4/Ezh2 axis was closely associated with the prognosis in pancreatic cancer patients.
Collapse
Affiliation(s)
- Shinichiro Hasegawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Akira Tomokuni
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiroshi Wada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naoki Hama
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Koichi Kawamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shigeru Marubashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naohiro Nishida
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jun Koseki
- Department of Cancer Profiling Discovery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|