1
|
Ragab EM, Gamal DME, El-Najjar FF, Elkomy HA, Ragab MA, Elantary MA, Basyouni OM, Moustafa SM, El-Naggar SA, Elsherbiny AS. New insights into Notch signaling as a crucial pathway of pancreatic cancer stem cell behavior by chrysin-polylactic acid-based nanocomposite. Discov Oncol 2025; 16:107. [PMID: 39891818 PMCID: PMC11787125 DOI: 10.1007/s12672-025-01846-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Pancreatic cancer is an extremely deadly illness for which there are few reliable treatments. Recent research indicates that malignant tumors are highly variable and consist of a tiny subset of unique cancer cells, known as cancer stem cells (CSCs), which are responsible for the beginning and spread of tumors. These cells are typically identified by the expression of specific cell surface markers. A population of pancreatic cancer stem cells with aberrantly active developmental signaling pathways has been identified in recent studies of human pancreatic tumors. Among these Notch signaling pathway has been identified as a key regulator of CSCs self-renewal, making it an attractive target for therapeutic intervention. Chrysin-loaded polylactic acid (PLA) as polymeric nanoparticles systems have been growing interest in using as platforms for improved drug delivery. This review aims to explore innovative strategies for targeted therapy and optimized drug delivery in pancreatic CSCs by manipulating the Notch pathway and leveraging PLA-based drug delivery systems. Furthermore, we will assess the capability of PLA nanoparticles to enhance the bioavailability and effectiveness of gemcitabine in pancreatic cancer cells. The insights gained from this review have the potential to contribute to the development of novel treatment approaches that combine targeted therapy with advanced drug delivery utilizing biodegradable polymeric nanoparticles.
Collapse
Affiliation(s)
- Eman M Ragab
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Fares F El-Najjar
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hager A Elkomy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A Ragab
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mariam A Elantary
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Omar M Basyouni
- Chemistry/Zoology Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sherif M Moustafa
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa A El-Naggar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Abeer S Elsherbiny
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
Geng G, Zhang L, Yu Y, Guo X, Li Q, Ming M. ADGRL4 Promotes Cell Growth, Aggressiveness, EMT, and Angiogenesis in Neuroblastoma via Activation of ERK/STAT3 Pathway. Curr Mol Med 2025; 25:45-55. [PMID: 38178660 DOI: 10.2174/0115665240254765231117122210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Neuroblastoma (NB) is one of the most common pediatric solid tumors. Emerging evidence has indicated that ADGRL4 can act as a master regulator of tumor progression. In addition, it is well documented that the ERK/STAT3 signaling pathway can promote the proliferation, EMT, angiogenesis, and metastasis in tumors. The current study was formulated to elucidate the exact role of ADGRL4 in the malignant behaviors of NB cells and to investigate the intrinsic mechanism. METHODS In this work, expression differences of ADGRL4 in human NB cell lines and HUVECs were assessed via RT-qPCR and western blot analysis. For functional experiments, sh-ADGRL4 was transfected into SK-N-SH cells to generate ADGRL4 knockdown stable cell line. Moreover, ADGRL4 knockdown stable SK-N-SH cells were treated with LM22B-10 (an ERK activator) for rescue experiments. CCK-8, colony formation, would healing, and transwell assays determined NB cell growth, migration, and invasion. Meanwhile, proliferation-, metastasis- and EMT- associated proteins were also detected. Additionally, a tube formation assay was employed to evaluate in vitro angiogenesis. VM-cadherin, the marker of angiogenesis, was assessed using immunofluorescence staining. RESULTS Data showed notably upregulated ADGRL4 in NB cells, especially in SK-NSH cells. ADGRL4 knockdown inhibited NB cell growth, migration, invasion, EMT, and in vitro angiogenesis. ADGRL4 knockdown inactivated ERK/STAT3 signaling pathway. Activation of the ERK/STAT3 signaling pathway partially rescued the tumor suppression effects of ADGRL4 knockdown on NB cells. CONCLUSION To conclude, the downregulation of ADGRL4 may inhibit cell growth, aggressiveness, EMT, and angiogenesis in NB by inactivating the ERK/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Geng Geng
- Department of Pediatric Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong Province, P.R. China
| | - Lei Zhang
- Department of Pediatric Orthopedics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong Province, P.R. China
| | - Ying Yu
- Department of Pharmacy, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong Province, P.R. China
| | - Xingqing Guo
- Department of Pediatric Respirology and Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong Province, P.R. China
| | - Qinghao Li
- Department of Pediatric Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong Province, P.R. China
| | - Ming Ming
- Department of Pediatric Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong Province, P.R. China
| |
Collapse
|
3
|
Tolue Ghasaban F, Ghanei M, Mahmoudian RA, Taghehchian N, Abbaszadegan MR, Moghbeli M. MicroRNAs as the critical regulators of epithelial mesenchymal transition in pancreatic tumor cells. Heliyon 2024; 10:e30599. [PMID: 38726188 PMCID: PMC11079401 DOI: 10.1016/j.heliyon.2024.e30599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Pancreatic cancer (PC), as one of the main endocrine and digestive systems malignancies has the highest cancer related mortality in the world. Lack of the evident clinical symptoms and appropriate diagnostic markers in the early stages of tumor progression are the main reasons of the high mortality rate among PC patients. Therefore, it is necessary to investigate the molecular pathways involved in the PC progression, in order to introduce novel early diagnostic methods. Epithelial mesenchymal transition (EMT) is a critical cellular process associated with pancreatic tumor cells invasion and distant metastasis. MicroRNAs (miRNAs) are also important regulators of EMT process. In the present review, we discussed the role of miRNAs in regulation of EMT process during PC progression. It has been reported that the miRNAs mainly regulate the EMT process in pancreatic tumor cells through the regulation of EMT-specific transcription factors and several signaling pathways such as WNT, NOTCH, TGF-β, JAK/STAT, and PI3K/AKT. Considering the high stability of miRNAs in body fluids and their role in regulation of EMT process, they can be introduced as the non-invasive diagnostic markers in the early stages of malignant pancreatic tumors. This review paves the way to introduce a non-invasive EMT based panel marker for the early tumor detection among PC patients.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Ghanei
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reihaneh Alsadat Mahmoudian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Bodea IC, Ciocan A, Zaharie FV, Bodea R, Graur F, Ursu Ș, Ciocan RA, Al Hajjar N. HER2 Overexpression in Periampullary Tumors According to Anatomical and Histological Classification-A Systematic Review. J Pers Med 2024; 14:463. [PMID: 38793045 PMCID: PMC11122564 DOI: 10.3390/jpm14050463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive, heterogeneous, and fatal types of human cancer; therefore, more effective therapeutic drugs are urgently needed. Human epidermal growth factor receptor 2 (HER2) overexpression and amplification have been identified as a cornerstone in this pathology. The aim of this review is to identify HER2 membrane overexpression in relation to pancreatic cancer pathways that can be used in order to develop a targeted therapy. After searching the keywords, 174 articles were found during a time span of 10 years, between 2013 and 2023, but only twelve scientific papers were qualified for this investigation. The new era of biomolecular research found a significant relationship between HER2 overexpression and pancreatic cancer cells in 25-30% of cases. The variables are dependent on tumor-derived cells, with differences in receptor overexpression between PDAC (pancreatic ductal adenocarcinoma), BTC (biliary tract cancer), ampullary carcinoma, and PNETs (pancreatic neuroendocrine tumors). HER2 overexpression is frequently encountered in human pancreatic carcinoma cell lines, and the ERBB family is one of the targets in the near future of therapy, with good results in phase I, II, and III studies evaluating downregulation and tumor downstaging, respectively.
Collapse
Affiliation(s)
- Ioan Cătălin Bodea
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Andra Ciocan
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Florin Vasile Zaharie
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Raluca Bodea
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Florin Graur
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Ștefan Ursu
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Răzvan Alexandru Ciocan
- Department of Surgery-Practical Abilities, “Iuliu Hațieganu” University of Medicine and Pharmacy, Marinescu Street, No. 23, 400337 Cluj-Napoca, Romania;
| | - Nadim Al Hajjar
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| |
Collapse
|
5
|
Sun Y, Qin H, Zhang C, Xu J, Zhang T. Tetrastigma hemsleyanum (Sanyeqing) root extracts evoke S phase arrest while inhibiting the migration and invasion of human pancreatic cancer PANC-1 cells. BMC Complement Med Ther 2024; 24:133. [PMID: 38539165 PMCID: PMC10967071 DOI: 10.1186/s12906-024-04425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/01/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Ethyl acetate extracts from Tetrastigma hemsleyanum (Sanyeqing) (EFT), a member of the Vitaceae plant family, have been shown to exhibit efficacy against a variety of cancers. In this light, our current study seeks to examine the mechanism of efficacy between EFT extracts and human pancreatic cancer PANC-1 cells. METHODS The chemical components of EFT were analyzed by gas chromatography-mass spectrometry. The cytotoxicity of EFT on PANC-1 cells was measured using an MTT assay. In order to investigate EFT induction of cell cycle arrest, changes in cell-cycle distribution were monitored by flow cytometry. Wound healing and transwell assays were employed to investigate whether migration and invasion of PANC-1 cells were inhibited by EFT. Relative protein expression was detected using Western blot. RESULTS GC-MS analysis of the chemical composition of EFT revealed that the majority of constituents were organic acids and their corresponding esters. EFT exhibits measurable cytotoxicity and inhibition of PANC-1 invasion. Growth inhibition was primarily attributed to downregulation of CDK2 which induces cell cycle arrest in the S-phase. Inhibition of metastasis is achieved through downregulation of mesenchymal-associated genes/activators, including ZEB1, N-cadherin, Vimentin, and Fibronectin. Meanwhile, the expression of E-cadherin was significantly increased by EFT treatment. Furthermore, downregulation of MMP-2 and MMP-9 were observed. CONCLUSION Treatment of PANC-1 with EFT demonstrated measurable cytotoxic effects. Furthermore, EFT evoked S phase arrest while inhibiting the migration and invasion of PANC-1 cells. Additionally, EFT inhibited the epithelial to mesenchymal transition and MMPs expression in PANC-1 cells. This study serves to confirm the strong therapeutic potential of EFT while identifying the mechanisms of action.
Collapse
Affiliation(s)
- Yifan Sun
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Haiyan Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
- Nanjing Healthnice Pharmaceutical Technology Co., Ltd CN, Nanjing, 210031, People's Republic of China
| | - Chunchun Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Jian Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Ting Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, People's Republic of China.
| |
Collapse
|
6
|
Díaz-López YE, Cázares-Domínguez V, Arenas-Huertero F, Gutierrez-Aguilar R. ETV5 Silencing Produces Mesenchymal to Epithelial Transition in INS-1 (832/13) Cell Line. Horm Metab Res 2024; 56:235-243. [PMID: 38335994 DOI: 10.1055/a-2246-4778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
ETV5 has been described to be involved in the epithelial to mesenchymal transition (EMT) mainly in cancer. It is known that EMT provokes cytoskeleton remodeling, improving cellular migratory, and invasive capabilities. Moreover, overexpression of ETV5 has been correlated to cancer development and this gene has been implicated in cell proliferation. However, little is known about the downregulation of ETV5 expression in a pancreatic cell line and the inverse mesenchymal to epithelial transition (MET). Therefore, we studied the implications of ETV5 silencing over the phenotype of the insulinoma INS-1 (832/13) cell line and described the MET by partial ETV5 silencing in the INS-1 (832/13) cell line. The downregulation of ETV5 expression was obtained by using ETV5 siRNA in the insulinoma rat cell line, INS-1 (832/13). Then, ETV5 knockdown provoked a MET phenotype observed by crystal violet staining and verified by immunohistochemistry against E-cadherin. Wound healing assay showed no migration, and F-actin stain revealed rearrangement of actin microfilaments. In addition, TGFβ1 and TGFβ3 were downregulated in the absence of ETV5. ETV5 silencing induces epithelial phenotype by downregulating TGFβ1 and TGFβ3 in INS-1 (832/13) cell line.
Collapse
Affiliation(s)
- Yael Efrén Díaz-López
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Investigación en Enfermedades Metabólicas, Obesidad y Diabetes, Hospital Infantil de México Federico Gomez, Mexico City, Mexico
| | - Vicenta Cázares-Domínguez
- Laboratorio de Investigación en Enfermedades Metabólicas, Obesidad y Diabetes, Hospital Infantil de México Federico Gomez, Mexico City, Mexico
| | - Francisco Arenas-Huertero
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Ruth Gutierrez-Aguilar
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Investigación en Enfermedades Metabólicas, Obesidad y Diabetes, Hospital Infantil de México Federico Gomez, Mexico City, Mexico
| |
Collapse
|
7
|
Wang SSY. Advancing biomarker development for diagnostics and therapeutics using solid tumour cancer stem cell models. TUMORI JOURNAL 2024; 110:10-24. [PMID: 36964664 DOI: 10.1177/03008916231158411] [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] [Indexed: 03/26/2023]
Abstract
The cancer stem cell model hopes to explain solid tumour carcinogenesis, tumour progression and treatment failure in cancers. However, the cancer stem cell model has led to minimal clinical translation to cancer stem cell biomarkers and targeted therapies in solid tumours. Many reasons underlie the challenges, one being the imperfect understanding of the cancer stem cell model. This review hopes to spur further research into clinically translatable cancer stem cell biomarkers through first defining cancer stem cells and their associated models. With a better understanding of these models there would be a development of more accurate biomarkers. Making the clinical translation of biomarkers into diagnostic tools and therapeutic agents more feasible.
Collapse
|
8
|
Uehara M, Domoto T, Takenaka S, Takeuchi O, Shimasaki T, Miyashita T, Minamoto T. Glycogen synthase kinase 3β: the nexus of chemoresistance, invasive capacity, and cancer stemness in pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:4. [PMID: 38318525 PMCID: PMC10838383 DOI: 10.20517/cdr.2023.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
The treatment of pancreatic cancer remains a significant clinical challenge due to the limited number of patients eligible for curative (R0) surgery, failures in the clinical development of targeted and immune therapies, and the pervasive acquisition of chemotherapeutic resistance. Refractory pancreatic cancer is typified by high invasiveness and resistance to therapy, with both attributes related to tumor cell stemness. These malignant characteristics mutually enhance each other, leading to rapid cancer progression. Over the past two decades, numerous studies have produced evidence of the pivotal role of glycogen synthase kinase (GSK)3β in the progression of over 25 different cancer types, including pancreatic cancer. In this review, we synthesize the current knowledge on the pathological roles of aberrant GSK3β in supporting tumor cell proliferation and invasion, as well as its contribution to gemcitabine resistance in pancreatic cancer. Importantly, we discuss the central role of GSK3β as a molecular hub that mechanistically connects chemoresistance, tumor cell invasion, and stemness in pancreatic cancer. We also discuss the involvement of GSK3β in the formation of desmoplastic tumor stroma and in promoting anti-cancer immune evasion, both of which constitute major obstacles to successful cancer treatment. Overall, GSK3β has characteristics of a promising therapeutic target to overcome chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Masahiro Uehara
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Takahiro Domoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Satoshi Takenaka
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Osamu Takeuchi
- Biomedical Laboratory, Department of Research, Kitasato University Kitasato Institute Hospital, Tokyo 108-8642, Japan
| | - Takeo Shimasaki
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Tomoharu Miyashita
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Toshinari Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
| |
Collapse
|
9
|
Ma Y, Yu Y, Yin Y, Wang L, Yang H, Luo S, Zheng Q, Pan Y, Zhang D. Potential role of epithelial-mesenchymal transition induced by periodontal pathogens in oral cancer. J Cell Mol Med 2024; 28:e18064. [PMID: 38031653 PMCID: PMC10805513 DOI: 10.1111/jcmm.18064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
With the increasing incidence of oral cancer in the world, it has become a hotspot to explore the pathogenesis and prevention of oral cancer. It has been proved there is a strong link between periodontal pathogens and oral cancer. However, the specific molecular and cellular pathogenic mechanisms remain to be further elucidated. Emerging evidence suggests that periodontal pathogens-induced epithelial-mesenchymal transition (EMT) is closely related to the progression of oral cancer. Cells undergoing EMT showed increased motility, aggressiveness and stemness, which provide a pro-tumour environment and promote malignant metastasis of oral cancer. Plenty of studies proposed periodontal pathogens promote carcinogenesis via EMT. In the current review, we discussed the association between the development of oral cancer and periodontal pathogens, and summarized various mechanisms of EMT caused by periodontal pathogens, which are supposed to play an important role in oral cancer, to provide targets for future research in the fight against oral cancer.
Collapse
Affiliation(s)
- Yiwei Ma
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yingyi Yu
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yuqing Yin
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Liu Wang
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Huishun Yang
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Shiyin Luo
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Qifan Zheng
- Department of Periodontics, School of StomatologyChina Medical UniversityShenyangChina
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of StomatologyChina Medical UniversityShenyangChina
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of StomatologyChina Medical UniversityShenyangChina
| |
Collapse
|
10
|
Huang W, Lin T, Huang L, Wu J, Hong J, Qiu F, Tian Y, Wang Y. miR-24-3p Regulates Epithelial-Mesenchymal Transition and the Malignant Phenotype of Pancreatic Adenocarcinoma by Regulating ASF1B Expression. Biochem Genet 2023; 61:742-761. [PMID: 36114946 PMCID: PMC10060286 DOI: 10.1007/s10528-022-10278-5] [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: 04/13/2022] [Accepted: 08/20/2022] [Indexed: 12/24/2022]
Abstract
Anti-silencing function protein 1 homolog B (ASF1B) has been implicated in the occurrence and development of cancers. The present work explored the functional role and the expression regulation of ASF1B in pancreatic ductal adenocarcinoma (PDAC). Based on the real-time quantitative PCR (qRT-PCR) and immunohistochemistry (IHC), ASF1B was significantly upregulated in PDAC tissues. High expression of ASF1B was associated with a poor overall survival (OS) and recurrence-free survival (DFS) in the PDAC patients. ASF1B also showed a relatively higher expression in PDAC cells (AsPC-1, PANC-1) when compared with human pancreatic ductal epithelial cells (HPDFe-6). CCK8 and clone formation assay demonstrated that silencing ASF1B impaired the proliferation in PANC-1 and AsPC-1 cells, and Annexin V-PI staining showed an increased level of apoptosis upon ASF1B silencing. ASF1B silencing also suppressed the migration and invasion in PDAC cells, as revealed by Transwell assays. We further showed that miR-24-3p was downregulated in PDAC tissues and cells, which functionally interacted with ASF1B by dual-luciferase reporter assay. miR-24-3p negatively regulated ASF1B expression to modulate the malignant phenotype of PDAC cells. ASF1B shows high expression in PDAC, which promotes the malignancy and EMT process of PDAC cells. miR-24-3p is a negative regulator of ASF1B and is downregulated in PDAC cells. Our data suggest that targeting ASF1B/miR-24-3p axis may serve as an intervention strategy for the management of PDAC.
Collapse
Affiliation(s)
- Wentao Huang
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China.
| | - Tiansheng Lin
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Long Huang
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Junyi Wu
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Jiaming Hong
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Funan Qiu
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Yifeng Tian
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| | - Yaodong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, NO.134, Dongjie Street, Gulou District, Fuzhou, 350001, Fujian, China
| |
Collapse
|
11
|
Sun L, Liu J, Guo M, Xu J, Wang D. First diagnosed invasive lobular carcinoma of the breast combined with gastric metastasis and bone metastasis: a case report and review of the literature. BMC Womens Health 2023; 23:133. [PMID: 36966290 PMCID: PMC10040113 DOI: 10.1186/s12905-023-02267-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
RATIONALE Chinese women topped the list of new breast cancers, the first diagnosed gastric metastasis and bone metastasis is extremely infrequent. The clinical and pathological diagnosis of metastatic breast cancer is difficult. To our knowledge, this is the first reported case of the first diagnosis of breast cancer with both gastric metastasis and bone metastasis. CASE REPORT The female patient was found to have abdominal distension for 15 days with nausea and vomiting. The patient underwent a gastroscopy at an outside hospital 4 days ago, showing: duodenal bulb changes, gastric retention and chronic non-atrophic gastritis. Gastroscopic biopsy showed chronic inflammation and edema of the duodenal mucosa with glandular hyperplasia. Conservative treatment was given with no relief of symptoms. She was seen in our hepatobiliary and pancreatic surgery department. After admission, palliative surgery was performed, and the swelling and surrounding involved tissues were taken for examination during surgery. The rapid pathological return could not exclude tumor lesions, and the postoperative pathology confirmed the diagnosis of invasive lobular carcinoma of the breast with gastric metastases, and the systemic examination revealed combined bone metastases. DIAGNOSIS Pathology and immunohistochemistry(IHC), a whole-body bone scan confirmed the first diagnosis of breast cancer with both gastric and bone metastases. INTERVENTIONS Palliative treatment with bisphosphonates and CDK4/6i (Palbociclib) in combination with AI (Exemestane) was administered. OUTCOMES The patient is currently under regular evaluation and is being followed up.
Collapse
Affiliation(s)
- Lin Sun
- Department of Breast Surgery, Second Affiliated Hospital of Jilin University, Changchun, 130041, China
| | - Jiajia Liu
- Department of Breast Surgery, Second Affiliated Hospital of Jilin University, Changchun, 130041, China
| | - Meng Guo
- Department of Breast Surgery, Second Affiliated Hospital of Jilin University, Changchun, 130041, China
| | - Jiaqi Xu
- Department of Breast Surgery, Second Affiliated Hospital of Jilin University, Changchun, 130041, China
| | - Dan Wang
- Department of Breast Surgery, Second Affiliated Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
12
|
Lian Y, Zeng S, Wen S, Zhao X, Fang C, Zeng N. Review and Application of Integrin Alpha v Beta 6 in the Diagnosis and Treatment of Cholangiocarcinoma and Pancreatic Ductal Adenocarcinoma. Technol Cancer Res Treat 2023; 22:15330338231189399. [PMID: 37525872 PMCID: PMC10395192 DOI: 10.1177/15330338231189399] [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: 02/14/2023] [Revised: 06/13/2023] [Accepted: 06/28/2023] [Indexed: 08/02/2023] Open
Abstract
Integrin Alpha v Beta 6 is expressed primarily in solid epithelial tumors, such as cholangiocarcinoma, pancreatic cancer, and colorectal cancer. It has been considered a potential and promising molecular marker for the early diagnosis and treatment of cancer. Cholangiocarcinoma and pancreatic ductal adenocarcinoma share genetic, histological, and pathophysiological similarities due to the shared embryonic origin of the bile duct and pancreas. These cancers share numerous clinicopathological characteristics, including growth pattern, poor response to conventional radiotherapy and chemotherapy, and poor prognosis. This review focuses on the role of integrin Alpha v Beta 6 in cancer progression. It addition, it reviews how the marker can be used in molecular imaging and therapeutic targets. We propose further research explorations and questions that need to be addressed. We conclude that integrin Alpha v Beta 6 may serve as a potential biomarker for cancer disease progression and prognosis.
Collapse
Affiliation(s)
- Yunyu Lian
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Sai Wen
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Chihua Fang
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Ning Zeng
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| |
Collapse
|
13
|
Husanie H, Abu-Remaileh M, Maroun K, Abu-Tair L, Safadi H, Atlan K, Golan T, Aqeilan RI. Loss of tumor suppressor WWOX accelerates pancreatic cancer development through promotion of TGFβ/BMP2 signaling. Cell Death Dis 2022; 13:1074. [PMID: 36572673 PMCID: PMC9792466 DOI: 10.1038/s41419-022-05519-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer is one of the most lethal cancers, owing to its late diagnosis and resistance to chemotherapy. The tumor suppressor WW domain-containing oxidoreductase (WWOX), one of the most active fragile sites in the human genome (FRA16D), is commonly altered in pancreatic cancer. However, the direct contribution of WWOX loss to pancreatic cancer development and progression remains largely unknown. Here, we report that combined conditional deletion of Wwox and activation of KRasG12D in Ptf1a-CreER-expressing mice results in accelerated formation of precursor lesions and pancreatic carcinoma. At the molecular level, we found that WWOX physically interacts with SMAD3 and BMP2, which are known activators of the TGF-β signaling pathway. In the absence of WWOX, TGFβ/BMPs signaling was enhanced, leading to increased macrophage infiltration and enhanced cancer stemness. Finally, overexpression of WWOX in patient-derived xenografts led to diminished aggressiveness both in vitro and in vivo. Overall, our findings reveal an essential role of WWOX in pancreatic cancer development and progression and underscore its role as a bona fide tumor suppressor.
Collapse
Affiliation(s)
- Hussam Husanie
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Muhannad Abu-Remaileh
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kian Maroun
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Abu-Tair
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hazem Safadi
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karine Atlan
- grid.17788.310000 0001 2221 2926Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Talia Golan
- grid.12136.370000 0004 1937 0546Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Rami I. Aqeilan
- grid.9619.70000 0004 1937 0538The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
Chen S, Zhou S, Huang YE, Yuan M, Lei W, Chen J, Lin K, Jiang W. Estimating Metastatic Risk of Pancreatic Ductal Adenocarcinoma at Single-Cell Resolution. Int J Mol Sci 2022; 23:ijms232315020. [PMID: 36499343 PMCID: PMC9736800 DOI: 10.3390/ijms232315020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by intra-tumoral heterogeneity, and patients are always diagnosed after metastasis. Thus, finding out how to effectively estimate metastatic risk underlying PDAC is necessary. In this study, we proposed scMetR to evaluate the metastatic risk of tumor cells based on single-cell RNA sequencing (scRNA-seq) data. First, we identified diverse cell types, including tumor cells and other cell types. Next, we grouped tumor cells into three sub-populations according to scMetR score, including metastasis-featuring tumor cells (MFTC), transitional metastatic tumor cells (TransMTC), and conventional tumor cells (ConvTC). We identified metastatic signature genes (MSGs) through comparing MFTC and ConvTC. Functional enrichment analysis showed that up-regulated MSGs were enriched in multiple metastasis-associated pathways. We also found that patients with high expression of up-regulated MSGs had worse prognosis. Spatial mapping of MFTC showed that they are preferentially located in the cancer and duct epithelium region, which was enriched with the ductal cells' associated inflammation. Further, we inferred cell-cell interactions, and observed that interactions of the ADGRE5 signaling pathway, which is associated with metastasis, were increased in MFTC compared to other tumor sub-populations. Finally, we predicted 12 candidate drugs that had the potential to reverse expression of MSGs. Taken together, we have proposed scMetR to estimate metastatic risk in PDAC patients at single-cell resolution which might facilitate the dissection of tumor heterogeneity.
Collapse
|
15
|
Nakayama F, Miyoshi M, Kimoto A, Kawano A, Miyashita K, Kamoshida S, Shimizu K, Hori Y. Pancreatic cancer cell-derived exosomes induce epithelial-mesenchymal transition in human pancreatic cancer cells themselves partially via transforming growth factor β1. Med Mol Morphol 2022; 55:227-235. [PMID: 35475918 PMCID: PMC9043512 DOI: 10.1007/s00795-022-00321-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/10/2022] [Indexed: 12/03/2022]
Abstract
Distant metastasis is a dismal prognostic factor of pancreatic cancer. Metastasis is established in several steps, but the mechanism underlying the very early stages remains unclear. Epithelial-mesenchymal transition (EMT) is involved in these stages. Although signaling molecules have been reported to induce EMT, the mechanism underlying their origin is unclear. In this study, we hypothesized that pancreatic cancer cell-derived exosomes induce EMT in cancer cells themselves, a notion we entertained because we found EMT in in vitro three-dimensional colonies of cancer cells, with vimentin-positive cells observed in some of the budding pancreatic cancer cells and in single cells outside the colony as well. First, we clarified that pancreatic cancer cell-derived exosomes induce EMT in cancer cells themselves. Next, we examined the involvement of transforming growth factor-β1 (TGF-β1), and TGF-β1 knock-down in pancreatic cancer cells with TGF-β1 siRNA significantly suppressed TGF-β1 gene expression in cancer cells, and exosomal TGF-β1 was significantly reduced in the secretory exosomes. Exosomes from TGF-β1 knock-down cells suppressed EMT induction in cancer cells themselves and TGF-β1 protein expression in target cells. Taken together, these findings suggest that TGF-β1 is involved in EMT induction via exosomes, results that may support the production of effective metastasis inhibitors.
Collapse
Affiliation(s)
- Fumiya Nakayama
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Makoto Miyoshi
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Ai Kimoto
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Akari Kawano
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Kumiko Miyashita
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Shingo Kamoshida
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Kazuya Shimizu
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
- Department of Internal Medicine, National Hospital Organization Kobe Medical Center, 3-1-1 Nishiochiai, Suma-ku, Kobe, 654-0155, Japan
| | - Yuichi Hori
- Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan.
| |
Collapse
|
16
|
Wang J, Yu C, Jiang X, Wu X, Jia Y, Zhang H, Li Z. [Vasohibin-2 promotes proliferation and metastasis of cervical cancer cells by regulating epithelial-mesenchymal transition]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:966-975. [PMID: 35869758 DOI: 10.12122/j.issn.1673-4254.2022.07.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the role of vasohibin-2 (VASH2) in regulation of proliferation and metastasis of cervical cancer cells. METHODS We analyzed the differentially expressed genes between cervical cancer cells with flotillin-1 overexpression and knockdown by RNA-seq combined with analysis of public databases. The expression levels of VASH2 were examined in normal cervical epithelial cells (HcerEpic), cervical cancer cell lines (HeLa, C-33A, Ca ski, SiHa and MS751) and fresh cervical cancer tissues with different lymph node metastasis status. We further tested the effects of lentivirus-mediated overexpression and interference of VASH2 on proliferation, migration, invasion and lymphatic vessel formation of the cervical cancer cells and detected the expression levels of key epithelial-mesenchymal transition (EMT) markers and TGF-β mRNA. RESULTS RNA-seq and analysis of public databases showed that VASH2 expression was significantly upregulated in cervical cancer cells exogenously overexpressing flotillin-1 (P < 0.05) and downregulated in cells with flotillin-1 knockdown (P < 0.05), and was significantly higher in cervical cancer tissues with lymph node metastasis than in those without lymph node metastasis (P < 0.01). In cervical cancer cell lines Ca Ski, SiHa, and MS751 and cervical cancer tissue specimens with lymph node metastasis, VASH2 expression was also significantly upregulated as compared with HcerEpic cells and cervical cancer tissues without lymph node metastasis (P < 0.05). Exogenous overexpression of VASH2 significantly promoted proliferation, migration, invasion and lymphatic vessel formation of cervical cancer cells, whereas these abilities were significantly inhibited in cells with VASH2 knockdown (P < 0.05). The cervical cancer cells overexpressing VASH2 showed significant down- regulation of e-cadherin and up- regulation of N-cadherin, Vimentin and VEGF-C, while the reverse changes were detected in cells with VASH2 knockdown (P < 0.05). TGF-β mRNA expression was significantly up-regulated in cervical cancer cells overexpressing VASH2 and down-regulated in cells with VASH2 knockdown (P < 0.001). CONCLUSION Flotillin-1 may participate in TGF-β signaling pathway-mediated EMT through its down-stream target gene VASH2 to promote the proliferation, migration, invasion and lymphatic vessel formation of cervical cancer cells in vitro.
Collapse
Affiliation(s)
- J Wang
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - C Yu
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - X Jiang
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - X Wu
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - Y Jia
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - H Zhang
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| | - Z Li
- Department of Gynecology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, China
| |
Collapse
|
17
|
Yang X, Qin C, Zhao B, Li T, Wang Y, Li Z, Li T, Wang W. Long Noncoding RNA and Circular RNA: Two Rising Stars in Regulating Epithelial-Mesenchymal Transition of Pancreatic Cancer. Front Oncol 2022; 12:910678. [PMID: 35719940 PMCID: PMC9204003 DOI: 10.3389/fonc.2022.910678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with especially poor prognosis. However, the molecular mechanisms of pancreatic oncogenesis and malignant progression are not fully elucidated. Epithelial-mesenchymal transition (EMT) process is important to drive pancreatic carcinogenesis. Recently, long noncoding RNAs (lncRNAs) and circular RNAs(circRNAs) have been characterized to participate in EMT in PDAC, which can affect the migration and invasion of tumor cells by playing important roles in epigenetic processes, transcription, and post-transcriptional regulation. LncRNAs can act as competing endogenous RNAs (ceRNA) to sequester target microRNAs(miRNAs), bind to the genes which localize physically nearby, and directly interact with EMT-related proteins. Currently known circRNAs mostly regulate the EMT process in PDAC also by acting as a miRNA sponge, directly affecting the protein degradation process. Therefore, exploring the functions of lncRNAs and circRNAs in EMT during pancreatic cancer might help pancreatic cancer treatments.
Collapse
Affiliation(s)
- Xiaoying Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Qin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bangbo Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianhao Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanyang Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeru Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weibin Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Begum H, Chirra N, Kumar D, Murugesan P, Kantevari S, Tangutur AD. Autophagic and apoptotic cell death induced by the quinoline derivative 2-(6-methoxynaphthalen-2-yl)quinolin-4-amine in pancreatic cancer cells is via ER stress and inhibition of Akt/mTOR signaling pathway. Drug Dev Res 2022; 83:910-926. [PMID: 35092073 DOI: 10.1002/ddr.21916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2023]
Abstract
Pancreatic cancer (PC) is among the most lethal cancers and is resistant to existing therapies, which highlights the need for new and alternative therapeutic treatments. Autophagy is emerging as one of the alternative cell death mechanisms and is well known to cross-talk with apoptosis. Autophagy can act as a viable option to treat highly resistant PC. The current study investigates and provides insight into the autophagic and apoptotic cell death induced by quinoline derivative 2-(6-methoxynaphthalen-2-yl)quinolin-4-amine (6MN-4-AQ) in PC cell lines PANC-1 and MIA PaCa-2. Treatment with 6MN-4-AQ reduced cell viability in concentration dependent manner (2-16 μM) and inhibited the clonogenic potential of PC cells at a concentration of 4 μM for 24 h. Further, we found that 6MN-4-AQ induced both apoptosis and autophagic cell death simultaneously. We identified that 6MN-4-AQ induced autophagic cell death by forming cytoplasmic vacuoles, the elevation of autophagy flux, increase in LC3-II, Beclin-1 protein expression, and degradation of p62. Moreover, 6MN-4-AQ induced apoptosis via Caspase-3 activation and cleavage of PARP in PC cells. Upon investigating the underlying mechanism associated with 6MN-4-AQ induced cell death, it was observed that 6MN-4-AQ treatment is able to suppress the Akt/mTOR pathway and induced ER stress leading to the induction of autophagy. Also, 6MN-4-AQ treatment suppressed epithelial to mesenchymal transition by reducing the protein expression of SLUG, snail, and vimentin. Subsequently, 6MN-4-AQ inhibited cell migration and invasion by down regulating MMP-7 and MMP-9 protein expression, suggesting that 6MN-4-AQ may serve as a plausible therapeutic agent for PC.
Collapse
Affiliation(s)
- Habeebunnisa Begum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nagaraju Chirra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Fluoro & Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Dinesh Kumar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Periyasamy Murugesan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Srinivas Kantevari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Fluoro & Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
19
|
Modeling Pancreatic Cancer with Patient-Derived Organoids Integrating Cancer-Associated Fibroblasts. Cancers (Basel) 2022; 14:cancers14092077. [PMID: 35565206 PMCID: PMC9103557 DOI: 10.3390/cancers14092077] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/10/2021] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Pancreatic cancer tissue is resistant to anticancer drugs because of its complex microenvironment. Cancer-associated fibroblasts (CAFs) are an important source of extracellular matrix components, which alter the physical and chemical properties of pancreatic tissue, thus impairing effective intratumoral drug delivery and resulting in resistance to conventional chemotherapy. In this study, we developed a novel CAF-integrated pancreatic cancer organoid (CIPCO) model that can mimic the tumor microenvironment and confirmed that the gene expression and pathological characteristics of CIPCO are similar to those of human cancer tissue. The organoid model could serve as a preclinical model for developing individualized therapies. Abstract Pancreatic cancer is a devastating disease and is highly resistant to anticancer drugs because of its complex microenvironment. Cancer-associated fibroblasts (CAFs) are an important source of extracellular matrix (ECM) components, which alter the physical and chemical properties of pancreatic tissue, thus impairing effective intratumoral drug delivery and resulting in resistance to conventional chemotherapy. The objective of this study was to develop a new cancer organoid model, including a fibrous tumor microenvironment (TME) using CAFs. The CAF-integrated pancreatic cancer organoid (CIPCO) model developed in this study histologically mimicked human pancreatic cancer and included ECM production by CAFs. The cancer cell–CAF interaction in the CIPCO promoted epithelial–mesenchymal transition of cancer cells, which was reversed by CAF inhibition using all-trans retinoic acid. Deposition of newly synthesized collagen I in the CIPCO disturbed the delivery of gemcitabine to cancer cells, and treatment with collagenase increased the cytotoxic effect of gemcitabine. This model may lead to the development of next-generation cancer organoid models recapitulating the fibrous TME.
Collapse
|
20
|
Gulla A, Andriusaityte U, Zdanys GT, Babonaite E, Strupas K, Kelly H. The Impact of Epithelial-Mesenchymal Transition and Metformin on Pancreatic Cancer Chemoresistance: A Pathway towards Individualized Therapy. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:467. [PMID: 35454306 PMCID: PMC9032206 DOI: 10.3390/medicina58040467] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/26/2022]
Abstract
Globally, pancreatic ductal adenocarcinoma remains among the most aggressive forms of neoplastic diseases, having a dismal prognostic outcome. Recent findings elucidated that epithelial-mesenchymal transition (EMT) can play an important role in pancreatic tumorigenic processes, as it contributes to the manifestation of malignant proliferative masses, which impede adequate drug delivery. An organized literature search with PubMed, Scopus, Microsoft Academic and the Cochrane library was performed for articles published in English from 2011 to 2021 to review and summarize the latest updates and knowledge on the current understanding of EMT and its implications for tumorigenesis and chemoresistance. Furthermore, in the present paper, we investigate the recent findings on metformin as a possible neoadjuvant chemotherapy agent, which affects EMT progression and potentially provides superior oncological outcomes for PDAC patients. Our main conclusions indicate that selectively suppressing EMT in pancreatic cancer cells has a promising therapeutic utility by selectively targeting the chemotherapy-resistant sub-population of cancer stem cells, inhibiting tumor growth via EMT pathways and thereby improving remission in PDAC patients. Moreover, given that TGF-β1-driven EMT generates the migration of tumor-initiating cells by directly linking the acquisition of abnormal cellular motility with the maintenance of tumor initiating potency, the chemoprevention of TGF-β1-induced EMT may have promising clinical applications in the therapeutic management of PDAC outcomes.
Collapse
Affiliation(s)
- Aiste Gulla
- Institute of Clinical Medicine, Clinic of Gastroenterology, Surgery, Nephrology, Faculty of Medicine, Vilnius University, Santariskiu Str. 2, 08661 Vilnius, Lithuania;
- Center of Visceral Medicine and Translational Research, Department of Surgery, Georgetown University Hospital, 3800 Reservoir Road Northwest BLES Building 1st. Floor, Washington, DC 20007, USA
| | - Urte Andriusaityte
- Faculty of Medicine, Vilnius University, M. K. Čiurlionio Str. 21, 03101 Vilnius, Lithuania; (U.A.); (G.T.Z.); (E.B.)
| | - Gabrielius Tomas Zdanys
- Faculty of Medicine, Vilnius University, M. K. Čiurlionio Str. 21, 03101 Vilnius, Lithuania; (U.A.); (G.T.Z.); (E.B.)
| | - Elena Babonaite
- Faculty of Medicine, Vilnius University, M. K. Čiurlionio Str. 21, 03101 Vilnius, Lithuania; (U.A.); (G.T.Z.); (E.B.)
| | - Kestutis Strupas
- Institute of Clinical Medicine, Clinic of Gastroenterology, Surgery, Nephrology, Faculty of Medicine, Vilnius University, Santariskiu Str. 2, 08661 Vilnius, Lithuania;
| | - Helena Kelly
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephen’s Green, D02 YN77 Dublin, Ireland;
| |
Collapse
|
21
|
Chen S, Guo W, Meng M, Wu D, Zhou T, Wang L, Xu J. LncRNA SNHG1 Promotes the Progression of Pancreatic Cancer by Regulating FGFR1 Expression via Competitively Binding to miR-497. Front Oncol 2022; 12:813850. [PMID: 35141164 PMCID: PMC8818711 DOI: 10.3389/fonc.2022.813850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundLong noncoding RNA small nucleolar RNA host gene 1 (SNHG1) is dysregulated in a variety of tumors. However, little is known of its role in pancreatic cancer (PC).MethodsThe role of SNHG1 on PC cell proliferation, migration, invasion, apoptosis, and the epithelial-mesenchymal transition (EMT) were assessed in vitro using MTT, EDU, wound healing, and Transwell assays, as well as flow cytometry and western blotting. Luciferase reporter assay, western blotting, and qRT-PCR were used to examine SNHG1 regulation. Tumor growth in mice was also investigated.ResultsDownregulation of SNHG1 blocked cell proliferation, migration and invasion, and induced apoptosis in vitro, while also inhibiting the EMT, shown by changes in the biomarkers E-cadherin, N-cadherin, and Vimentin. The opposite results were observed on upregulation of SNHG1. In vivo experiments showed that downregulation of SNHG1 inhibited tumor development in nude mice. Furthermore, experiments investigating the regulatory mechanism of SNHG1 indicated that SNHG1 acted as a competitive endogenous RNA, positively regulating the expression of fibroblast growth factor receptor 1 (FGFR1) through sponging miR-497. Rescue experiments demonstrated that the effects of SNHG1 downregulation on PC cells were attenuated when simultaneously inhibiting the levels of miR-497.ConclusionsSNHG1 upregulates FGFR1 expression by sponging miR-497, which promotes the progression of PC. SNHG1 may thus be a novel target for treating PC.
Collapse
Affiliation(s)
- Shihong Chen
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Wenyi Guo
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Mingyang Meng
- Department of General Medicine, Xiangyang NO.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Dong Wu
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Tao Zhou
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Lei Wang
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
- *Correspondence: Lei Wang, ; Jianwei Xu,
| | - Jianwei Xu
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
- *Correspondence: Lei Wang, ; Jianwei Xu,
| |
Collapse
|
22
|
Wang T, Chen P, Dong R, Weir S, Baltezor M, Schoenen FJ, Chen Q. Novel Compound C150 Inhibits Pancreatic Cancer Cell Epithelial-to-Mesenchymal Transition and Tumor Growth in Mice. Front Oncol 2022; 11:773350. [PMID: 34976816 PMCID: PMC8714879 DOI: 10.3389/fonc.2021.773350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer cell epithelial-to-mesenchymal transition (EMT) is an important contributor to cell invasion and tumor progression. Therefore, targeting EMT may be beneficial for pancreatic cancer treatment. The aim of the present study was to report on the inhibitory effect of the novel compound C150 on the EMT of pancreatic cancer cells. C150 inhibited cell proliferation in multiple pancreatic cancer cells with IC50 values of 1-2.5 μM, while in an non-cancerous pancreatic epithelial cell line hTERT-HPNE the IC50 value was >12.5 μM. C150 significantly inhibited pancreatic cancer cell migration and invasion, as demonstrated by 3-dimensional cell invasion, wound healing and Boyden chamber Transwell migration-invasion assays. Moreover, C150 treatment decreased MMP-2 gene expression in PANC-1 cells and reduced MMP-2 activity in gelatin zymography assay. In an orthotopic mouse model of pancreatic cancer, C150 significantly reduced tumor growth at the dose of 15 mg/kg by intraperitoneal injection three times per week. Furthermore, C150 enhanced protein degradation of Snail, an important EMT-promoting transcription factor, and decreased the expression of the mesenchymal marker N-cadherin, while it increased the expression of the epithelial markers zonula occludens-1 and claudin-1. The findings of the present study suggested that C150 is a novel EMT inhibitor that may be promising for inhibiting pancreatic cancer growth and metastasis.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ping Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ruochen Dong
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Scott Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Michael Baltezor
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, KS, United States
| | - Frank J Schoenen
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, United States
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
23
|
Gupta S, Kumar A, Tejavath KK. A pharmacognostic approach for mitigating pancreatic cancer: emphasis on herbal extracts and phytoconstituents. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00246-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Pancreatic cancer is studied as one of the most lethal cancers with currently no control of its lethality, mainly due to its late diagnosis and lack of foolproof treatment processes. Despite continuous efforts being made in looking for therapies to deal with cancer, it keeps on being a labyrinth for the researchers. Efforts like discovering new treatment options, repurposing existing drugs, are continuously made to deal with this cancer.
Main body
With the urge to get answers and the fact that nature has all roots of therapeutics, efforts are made in the direction of finding those answers for providing ministrations for pancreatic cancer from plant products. Plant products are used as treatment options either directly in the form of extracts or an alternative to them is individual phytochemicals that are either isolated from the plants or are commercially synthesized for various purposes. In this review, we put forward such pharmacognostic initiatives made in combating pancreatic cancer, focusing mainly on plant extracts and various phytochemicals; along with the mechanisms which they triggered to fulfill the need for cytotoxicity to pancreatic cancer cells (in vitro and in vivo).
Conclusion
This study will thus provide insights into new combination therapy that can be used and also give a clue on which plant product and phytoconstituent can be used in dealing with pancreatic cancer.
Graphical abstract
Collapse
|
24
|
Huang YK, Busuttil RA, Boussioutas A. The Role of Innate Immune Cells in Tumor Invasion and Metastasis. Cancers (Basel) 2021; 13:5885. [PMID: 34884995 PMCID: PMC8656477 DOI: 10.3390/cancers13235885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis is considered one of the hallmarks of cancer and enhanced tumor invasion and metastasis is significantly associated with cancer mortality. Metastasis occurs via a series of integrated processes involving tumor cells and the tumor microenvironment. The innate immune components of the microenvironment have been shown to engage with tumor cells and not only regulate their proliferation and survival, but also modulate the surrounding environment to enable cancer progression. In the era of immune therapies, it is critical to understand how different innate immune cell populations are involved in this process. This review summarizes recent literature describing the roles of innate immune cells during the tumor metastatic cascade.
Collapse
Affiliation(s)
- Yu-Kuan Huang
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; (Y.-K.H.); (R.A.B.)
| | - Rita A. Busuttil
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; (Y.-K.H.); (R.A.B.)
| | - Alex Boussioutas
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; (Y.-K.H.); (R.A.B.)
- Department of Gastroenterology, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
25
|
Miquel M, Zhang S, Pilarsky C. Pre-clinical Models of Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:748631. [PMID: 34778259 PMCID: PMC8578999 DOI: 10.3389/fcell.2021.748631] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a hostile solid malignancy coupled with an extremely high mortality rate. Metastatic disease is already found in most patients at the time of diagnosis, resulting in a 5-year survival rate below 5%. Improved comprehension of the mechanisms leading to metastasis is pivotal for the development of new targeted therapies. A key field to be improved are modeling strategies applied in assessing cancer progression, since traditional platforms fail in recapitulating the complexity of PDAC. Consequently, there is a compelling demand for new preclinical models that mirror tumor progression incorporating the pressure of the immune system, tumor microenvironment, as well as molecular aspects of PDAC. We suggest the incorporation of 3D organoids derived from genetically engineered mouse models or patients as promising new tools capable to transform PDAC pre-clinical modeling and access new frontiers in personalized medicine.
Collapse
Affiliation(s)
- Maria Miquel
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Shuman Zhang
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
26
|
Xiong Y, Huang J. Anti-malarial drug: the emerging role of artemisinin and its derivatives in liver disease treatment. Chin Med 2021; 16:80. [PMID: 34407830 PMCID: PMC8371597 DOI: 10.1186/s13020-021-00489-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Artemisinin and its derivatives belong to a family of drugs approved for the treatment of malaria with known clinical safety and efficacy. In addition to its anti-malarial effect, artemisinin displays anti-viral, anti-inflammatory, and anti-cancer effects in vivo and in vitro. Recently, much attention has been paid to the therapeutic role of artemisinin in liver diseases. Several studies suggest that artemisinin and its derivatives can protect the liver through different mechanisms, such as those pertaining to inflammation, proliferation, invasion, metastasis, and induction of apoptosis and autophagy. In this review, we provide a comprehensive discussion of the underlying molecular mechanisms and signaling pathways of artemisinin and its derivatives in treating liver diseases. Further pharmacological research will aid in determining whether artemisinin and its derivatives may serve as promising medicines for the treatment of liver diseases in the future. ![]()
Collapse
Affiliation(s)
- Ye Xiong
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Jianrong Huang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
27
|
Li H, Pan W, Xu L, Yin D, Cheng S, Zhao F. Prognostic Significance of Microvascular Invasion in Pancreatic Ductal Adenocarcinoma: A Systematic Review and Meta-Analysis. Med Sci Monit 2021; 27:e930545. [PMID: 34393219 PMCID: PMC8378224 DOI: 10.12659/msm.930545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/19/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The incidence, pathogenesis, and prognostic effect of microvascular invasion on pancreatic ductal adenocarcinoma (PDAC) remain controversial. This study aimed to summarize the incidence, pathogenesis, role in clinical management, recurrence, and prognostic significance of microvascular invasion in PDAC. MATERIAL AND METHODS A literature review and meta-analysis were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement. Systematic literature searches were conducted using PubMed and Google Scholar up to February 2021. RESULTS Seventeen studies were included in the meta-analysis. The incidence of microvascular invasion was 49.0% (95% confidence interval [CI], 43.8-54.5%) among PDAC patients who underwent surgery. The weighted multivariate Cox proportional hazards model hazard ratio for disease-free survival of 8 studies was 1.78 (95% CI 1.53-2.08, P<0.001), and there was no statistically significant difference between the subgroups (P=0.477). The hazard ratio for overall survival of 14 studies was 1.49 (95% CI 1.27-1.74, P<0.001), and there was no statistically significant difference between the subgroups (P=0.676). CONCLUSIONS Microvascular invasion occurred in nearly half of PDAC patients after surgery and was closely related to disease-free and overall survival. Understanding the role of microvascular invasion in PDAC will help provide more personalized and effective preoperative or postoperative strategies to achieve better survival outcomes.
Collapse
Affiliation(s)
- Huangbao Li
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, PR China
| | - Weiwei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, PR China
| | - Liu Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, PR China
| | - Dong Yin
- Department of Oncology, First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, PR China
| | - Shuqun Cheng
- Department of Oncology, First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, PR China
- Department of Hepatic Surgery IV, The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, PR China
| | - Fengqing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, PR China
| |
Collapse
|
28
|
Wang P, Zhu Z. Prognostic and Clinicopathological Significance of E-Cadherin in Pancreatic Cancer Patients: A Meta-Analysis. Front Oncol 2021; 11:627116. [PMID: 33912451 PMCID: PMC8074677 DOI: 10.3389/fonc.2021.627116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background Several recent studies have investigated the prognostic and clinicopathological significance of epithelial cadherin (E-cadherin) in pancreatic cancer; however, conclusions from these studies remain inconsistent. Therefore, we performed a meta-analysis to evaluate the effects of E-cadherin expression on the prognosis and clinicopathological characteristics of pancreatic cancer. Methods Embase, PubMed, and Web of Science were searched to identify articles associated with E-cadherin and pancreatic cancer. Hazard ratios (HRs) and odds ratios (ORs) with corresponding confidence intervals (CIs) were calculated and summarized. All eligible studies were searched until May 20, 2020. Heterogeneity among studies was assessed using the Chi-square test and I2 statistic. Results Overall, 25 studies were identified, of which 12 reports with 1,032 cases concerned the prognosis of pancreatic cancer, and 22 involved the risk and clinical characteristics of pancreatic cancer. The overall results revealed that E-cadherin expression was significantly related to overall survival, gender, tumor grade, lymph node metastasis, tumor differentiation, and risk of pancreatic cancer. In the subgroup analysis, no significant heterogeneity or publication bias was observed. Conclusions E-cadherin expression is strongly associated with the risk, clinical features, and prognosis of pancreatic cancer, suggesting that E-cadherin may be an effective biomarker for the clinical assessments and predicting prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Pengbo Wang
- Radiotherapy Department I, Yantaishan Hospital, Yantai, China
| | - Zengkuan Zhu
- Department of Oncology, Shengli Oilfield Central Hospital, Dongying, China
| |
Collapse
|
29
|
van de Merbel AF, van Hooij O, van der Horst G, van Rijt-van de Westerlo CCM, van der Mark MH, Cheung H, Kroon J, Verhaegh GW, Tijhuis J, Wellink A, Maas P, Viëtor H, Schalken JA, van der Pluijm G. The Identification of Small Molecule Inhibitors That Reduce Invasion and Metastasis of Aggressive Cancers. Int J Mol Sci 2021; 22:ijms22041688. [PMID: 33567533 PMCID: PMC7915539 DOI: 10.3390/ijms22041688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Transformed epithelial cells can activate programs of epithelial plasticity and switch from a sessile, epithelial phenotype to a motile, mesenchymal phenotype. This process is linked to the acquisition of an invasive phenotype and the formation of distant metastases. The development of compounds that block the acquisition of an invasive phenotype or revert the invasive mesenchymal phenotype into a more differentiated epithelial phenotype represent a promising anticancer strategy. In a high-throughput assay based on E-cadherin (re)induction and the inhibition of tumor cell invasion, 44,475 low molecular weight (LMW) compounds were screened. The screening resulted in the identification of candidate compounds from the PROAM02 class. Selected LMW compounds activated E-cadherin promoter activity and inhibited cancer cell invasion in multiple metastatic human cancer cell lines. The intraperitoneal administration of selected LMW compounds reduced the tumor burden in human prostate and breast cancer in vivo mouse models. Moreover, selected LMW compounds decreased the intra-bone growth of xenografted human prostate cancer cells. This study describes the identification of the PROAM02 class of small molecules that can be exploited to reduce cancer cell invasion and metastases. Further clinical evaluation of selected candidate inhibitors is warranted to address their safety, bioavailability and antitumor efficacy in the management of patients with aggressive cancers.
Collapse
Affiliation(s)
- Arjanneke F. van de Merbel
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
| | - Onno van Hooij
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (O.v.H.); (C.C.M.v.R.-v.d.W.); (G.W.V.); (J.A.S.)
| | - Geertje van der Horst
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
| | - Cindy C. M. van Rijt-van de Westerlo
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (O.v.H.); (C.C.M.v.R.-v.d.W.); (G.W.V.); (J.A.S.)
- Oncodrone BV, 6525 GA Nijmegen, The Netherlands; (A.W.); (H.V.)
| | - Maaike H. van der Mark
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
| | - Henry Cheung
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
| | - Jan Kroon
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
- Department of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gerald W. Verhaegh
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (O.v.H.); (C.C.M.v.R.-v.d.W.); (G.W.V.); (J.A.S.)
| | - Johan Tijhuis
- Specs, 2712 PB Zoetermeer, The Netherlands; (J.T.); (P.M.)
| | - Antoine Wellink
- Oncodrone BV, 6525 GA Nijmegen, The Netherlands; (A.W.); (H.V.)
| | - Peter Maas
- Specs, 2712 PB Zoetermeer, The Netherlands; (J.T.); (P.M.)
| | - Henk Viëtor
- Oncodrone BV, 6525 GA Nijmegen, The Netherlands; (A.W.); (H.V.)
| | - Jack A. Schalken
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (O.v.H.); (C.C.M.v.R.-v.d.W.); (G.W.V.); (J.A.S.)
- Oncodrone BV, 6525 GA Nijmegen, The Netherlands; (A.W.); (H.V.)
| | - Gabri van der Pluijm
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.F.v.d.M.); (G.v.d.H.); (M.H.v.d.M.); (H.C.); (J.K.)
- Correspondence: ; Tel.: +31-715265255
| |
Collapse
|
30
|
Yan Q, Chen BJ, Hu S, Qi SL, Li LY, Yang JF, Zhou H, Yang CC, Chen LJ, Du J. Emerging role of RNF2 in cancer: From bench to bedside. J Cell Physiol 2021; 236:5453-5465. [PMID: 33400276 DOI: 10.1002/jcp.30260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/03/2020] [Accepted: 12/22/2020] [Indexed: 01/16/2023]
Abstract
RNF2 (also known as ding, Ring1B or Ring2) is a member of the Ring finger protein family, which functions as E3 ubiquitin ligase for monoubiquitination of histone H2A at lysine 119 (H2AK119ub). RNF2 gene is located at the 1q25.3 site of human chromosome and the coding region is composed of 9 exons, encoding 336 amino acids in total. Many studies have demonstrated that overexpressed RNF2 was involved in the pathological progression of multiple cancers and has an impact on their clinical features. For instance, the upregulated expression level of RNF2 is positively correlated with the occurrence and progression of hepatocellular carcinoma, melanoma, prostate cancer, breast cancer, pancreatic cancer, gastric cancer, and bladder urothelial carcinoma, as well as with the radioresistance of lung cancer and chemoresistance of ovarian cancer. This review provides an up-to-date perspective on the relationship between RNF2 and several cancers and highlights recent studies on RNF2 regulation. In particular, the relevant cellular signaling pathways and potential clinical value of RNF2 in cancers are also discussed, suggesting its potential as an epigenetic biomarker and therapeutic target for these cancers.
Collapse
Affiliation(s)
- Qi Yan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Bang-Jie Chen
- First Clinical Medical College of Anhui Medical university, Hefei, China
| | - Shuang Hu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Shun-Li Qi
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liang-Yun Li
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun-Fa Yang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hong Zhou
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Chen-Chen Yang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Li-Jian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jian Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Li T, Ren T, Huang C, Li Y, Yang P, Che G, Luo L, Chen Y, Peng S, Lin Y, Zeng L. S100A16 induces epithelial-mesenchymal transition in human PDAC cells and is a new therapeutic target for pancreatic cancer treatment that synergizes with gemcitabine. Biochem Pharmacol 2020; 189:114396. [PMID: 33359364 DOI: 10.1016/j.bcp.2020.114396] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers, with a poor 5-year survival rate of approximately 6%, mostly due to poor treatment response and early progression. The S100 gene family participates in various pathophysiological processes in various malignancies. S100A16 is a member of the S100 family, which is abnormally expressed in PDAC; however, its biological functions and mechanisms of action remain unclear. We analysed the Gene Expression Omnibus (GEO) public database and the gene ChIP data collected in our previous study of human PDAC cell line PANC-1 cocultured with M2 macrophages to identify differentially expressed genes (DEGs). Twenty-three overexpressed genes were identified by screening. Then, the selected genes were analysed using The Cancer Genome Atlas (TCGA) database to assess whether they have significant impact on the overall survival (OS) of PDAC patients. Of the 14 DEGs identified, S100A16 was associated with poor prognosis and was selected for further investigation; the results indicate that S100A16 is positively correlated with epithelial-mesenchymal transition (EMT)-related genes in the TCGA dataset. Subsequent in vitro and in vivo experiments demonstrated that S100A16 induces the EMT to promote the metastasis of human PDAC cells and that the effect is mediated by the enhanced expression of TWIST1 and activation of the STAT3 signalling pathway. The antitumour effect of gemcitabine (GEM) was enhanced in combination with S100A16 downregulation. In conclusion, our findings suggest that S100A16 is a novel potential therapeutic target for human PDAC treatment.
Collapse
Affiliation(s)
- Ting Li
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Tianyi Ren
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Chumei Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province 518107, China
| | - Yufang Li
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Pengfei Yang
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Gang Che
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Lisi Luo
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yutong Chen
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Siqi Peng
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yujing Lin
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Linjuan Zeng
- Department of Abdominal Oncology, The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| |
Collapse
|
32
|
Novizio N, Belvedere R, Pessolano E, Tosco A, Porta A, Perretti M, Campiglia P, Filippelli A, Petrella A. Annexin A1 Released in Extracellular Vesicles by Pancreatic Cancer Cells Activates Components of the Tumor Microenvironment, through Interaction with the Formyl-Peptide Receptors. Cells 2020; 9:cells9122719. [PMID: 33353163 PMCID: PMC7767312 DOI: 10.3390/cells9122719] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive cancers in the world. Several extracellular factors are involved in its development and metastasis to distant organs. In PC, the protein Annexin A1 (ANXA1) appears to be overexpressed and may be identified as an oncogenic factor, also because it is a component in tumor-deriving extracellular vesicles (EVs). Indeed, these microvesicles are known to nourish the tumor microenvironment. Once we evaluated the autocrine role of ANXA1-containing EVs on PC MIA PaCa-2 cells and their pro-angiogenic action, we investigated the ANXA1 paracrine effect on stromal cells like fibroblasts and endothelial ones. Concerning the analysis of fibroblasts, cell migration/invasion, cytoskeleton remodeling, and the different expression of specific protein markers, all features of the cell switching into myofibroblasts, were assessed after administration of wild type more than ANXA1 Knock-Out EVs. Interestingly, we demonstrated a mechanism by which the ANXA1-EVs complex can stimulate the activation of formyl peptide receptors (FPRs), triggering mesenchymal switches and cell motility on both fibroblasts and endothelial cells. Therefore, we highlighted the importance of ANXA1/EVs-FPR axes in PC progression as a vehicle of intercommunication tumor cells-stroma, suggesting a specific potential prognostic/diagnostic role of ANXA1, whether in soluble form or even if EVs are captured in PC.
Collapse
Affiliation(s)
- Nunzia Novizio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
| | - Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
| | - Emanuela Pessolano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende 43, 84081 Baronissi, Italy;
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (N.N.); (R.B.); (E.P.); (A.T.); (A.P.); (P.C.)
- Correspondence: ; Tel.: +39-089-969-762; Fax: +39-089-969-602
| |
Collapse
|
33
|
Expression of POU2F3 Transcription Factor Control Inflammation, Immunological Recruitment and Metastasis of Pancreatic Cancer in Mice. BIOLOGY 2020; 9:biology9100341. [PMID: 33086657 PMCID: PMC7603360 DOI: 10.3390/biology9100341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022]
Abstract
Simple Summary The presence and the role of TUFT cells in pancreatic ductal adenocarcinoma (PDAC) is discussed. Therefore, we decided to inactivate the POU2F3 gene, which is essential for TUFT cells development, in an aggressive PDAC mice model known as PDX1-Cre;LSL-KrasG12D;Ink4afl/fl. Morphological and molecular analysis of POU2F3-deleted PDAC show not significant changes in tumors growth and survival of animals although it promotes EMT. Remarkably, we observed that in POU2F3-deleted animals the lack of TUFT cells prevents metastasis formation and strongly modifies the immunological and inflammatory landscape. Abstract TUFT cells have been described as strong modulators of inflammatory cells in several tissues including pancreas. TUFT cells, also known as DCLK1+ cells, are dependent of the transcriptional factor POU2F3. Several works report DCLK1+ cells in early stages of PDAC development suggesting an important role of TUFT cells in PDAC development. Therefore, we developed a mice model (PDX1-Cre;KrasG12D;Ink4afl/fl), known as PKI model, deficient or not of POU2F3. In this animal model, deficiency of POU2F3 results in the absence of TUFT cells in PDAC as expected. Although, tumor development and growth are not significantly influenced, the development of liver metastasis was almost completely inhibited in POU2F3-deficient mice. Surprisingly, the absence of metastasis was associated with a higher expression of epithelial-to-mesenchymal transition markers, but to a lower inflammatory microenvironment suggesting that inflammation influences metastasis production more than epithelial-to-mesenchymal transition in this animal model. We can conclude that POU2F3 could be a new therapeutic target for control PDAC progression.
Collapse
|
34
|
Xu J, Liu S, Yang X, Cao S, Zhou Y. Paracrine HGF promotes EMT and mediates the effects of PSC on chemoresistance by activating c-Met/PI3K/Akt signaling in pancreatic cancer in vitro. Life Sci 2020; 263:118523. [PMID: 33039386 DOI: 10.1016/j.lfs.2020.118523] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/19/2020] [Accepted: 09/27/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Pancreatic stellate cells (PSCs) play key roles in the pancreatic tumor microenvironment and are considered to contribute to chemoresistance. PSCs can participate in malignant behaviors of pancreatic carcinoma (PC) by secreting hepatocyte growth factor (HGF). The objective of this research was to explore the potential molecular mechanism of HGF on gemcitabine (GEM) chemoresistance of PC. MATERIALS AND METHODS HGF, c-Met, E-Cadherin and Vimentin levels were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The changes of HGF level were detected by ELISA. The half maximal inhibitory concentration, the growth inhibitions and apoptosis of pancreatic cancer cells (PCCs) were respectively assayed using CCK-8 and flow cytometry. Associated proteins were measured using western blot and cell immunofluorescence assay. KEY FINDINGS PSCs strongly expressed HGF, and its receptor c-Met was expressed in PCCs. PCCs exerted a positive regulative effect on HGF production. HGF neutralizing antibody AMG102 could effectively reduce the HGF level in PSC-conditioned medium (PSC-CM). PSC-CM promoted chemoresistance in PCCs. When exposed to PSC-CM, PCCs underwent epithelial-to-mesenchymal transition (EMT), and c-Met was also activated. Recombinant human HGF had the same protective effect. Blocking the HGF/c-Met axis with a c-Met inhibitor PHA665752 and AMG102 reduced the phosphorylation level of c-Met (p-c-Met) and attenuated EMT and chemoresistance. P-c-Met overexpression resulted in activation of the PI3K/Akt pathway, and inhibition of PI3K/Akt signaling with LY294002 reversed chemoresistance and EMT. SIGNIFICANCE PSCs can activate the c-Met/PI3K/Akt pathway in PCCs via paracrine HGF, induce EMT of PCCs and inhibit cancer cell apoptosis, thus enhance chemoresistance to Gem in PCCs.
Collapse
Affiliation(s)
- Jianfei Xu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaopeng Yang
- Department of Gastrointestinal Surgery, Yidu Central Hospital, Weifang, China
| | - Shougen Cao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
35
|
Shah VM, Sheppard BC, Sears RC, Alani AW. Hypoxia: Friend or Foe for drug delivery in Pancreatic Cancer. Cancer Lett 2020; 492:63-70. [PMID: 32822815 DOI: 10.1016/j.canlet.2020.07.041] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal solid tumors with an overall five-year survival rate of that has only just reached 10%. The tumor microenvironment of PDAC is characterized by desmoplasia, which consist of dense stroma of fibroblasts and inflammatory cells, resulting in a hypoxic environment due to limited oxygen diffusion through the tumor. Hypoxia contributes to the aggressive tumor biology by promoting tumor progression, malignancy, and promoting resistance to conventional and targeted therapeutic agents. In depth research in the area has identified that hypoxia modulates the tumor biology through hypoxia inducible factors (HIFs), which not only are the key determinant of pancreatic malignancy but also an important target for therapy. In this review, we summarize the recent advances in understanding hypoxia driven phenotypes, which are responsible for the highly aggressive and metastatic characteristics of pancreatic cancer, and how hypoxia can be exploited as a target for drug delivery.
Collapse
Affiliation(s)
- Vidhi M Shah
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, 2730 SW Moody Ave., Portland, OR, 97201, USA; Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Brett C Sheppard
- Department of Surgery, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA; OHSU Knight Cancer Institute at Oregon Health & Science University, Portland, OR, 97239, USA
| | - Rosalie C Sears
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 S.W Sam Jackson Park Road, Portland, OR, 97239, USA; Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Rd., Portland, OR, 97239, USA; OHSU Knight Cancer Institute at Oregon Health & Science University, Portland, OR, 97239, USA
| | - Adam Wg Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University/OHSU, 2730 SW Moody Ave., Portland, OR, 97201, USA; OHSU Knight Cancer Institute at Oregon Health & Science University, Portland, OR, 97239, USA; Department of Biomedical Engineering, School of Medicine at Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
36
|
Takahashi H, Katsuta E, Yan L, Tokumaru Y, Katz MH, Takabe K. Transcriptomic Profile of Lymphovascular Invasion, a Known Risk Factor of Pancreatic Ductal Adenocarcinoma Metastasis. Cancers (Basel) 2020; 12:E2033. [PMID: 32722116 PMCID: PMC7465682 DOI: 10.3390/cancers12082033] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022] Open
Abstract
Lymphovascular invasion (LVI) is an aggressive pathologic feature and considered a risk factor for distant metastasis. We hypothesized that pancreatic ductal adenocarcinomas (PDACs) with LVI are associated with shorter survival, as well as aggressive cancer biology and lymphangiogenesis in transcriptomic analysis. Utilizing the cancer genome atlas (TCGA)-PDAC cohort, we found that positive LVI was significantly associated with positive perineural invasion (PNI) (p = 0.023), and higher American Joint Committee on Cancer (AJCC) T (p = 0.017) and N (p < 0.001) categories. Furthermore, positive LVI was associated with shorter overall survival (OS) (p = 0.014) and was an independent risk factor of poor OS. Although there was no association between LVI status and lymphangiogenesis, epithelial-mesenchymal transition (EMT), or metastasis-related genes, Gene Set Enrichment Analysis revealed a strong association with cell-proliferation-related gene sets such as mitotic spindles (Normalized enrichment score (NES) = 1.76, p = 0.016) and G2/M checkpoints (NES = 1.75, p = 0.036), as well as with transforming growth factor beta (TGF-beta) signaling (NES = 1.61, p = 0.043), which is a known mechanism of metastasis in PDACs. In conclusion, positive LVI was an independent risk factor of poor OS in PDACs. We found that PDACs with LVI were possibly associated with accelerated cell proliferation and enhanced TGF-beta signaling independent of lymphangiogenesis. Transcriptomic profiling elucidates more precise tumor biology of LVI-positive PDACs.
Collapse
Affiliation(s)
- Hideo Takahashi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (H.T.); (E.K.); (Y.T.)
| | - Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (H.T.); (E.K.); (Y.T.)
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (H.T.); (E.K.); (Y.T.)
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Matthew H.G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (H.T.); (E.K.); (Y.T.)
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY 14260, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Surgery, Yokohama City University, Yokohama 236-0004, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| |
Collapse
|
37
|
Ji X, Guo X, Wang Y, Li X, Li H. Rab18 Regulates Proliferation, Invasion and Cisplatin Sensitivity Through STAT3 Signaling in Head and Neck Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:4123-4134. [PMID: 32494165 PMCID: PMC7231766 DOI: 10.2147/ott.s238503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/14/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction The clinical significance, biological roles and potential mechanism of Rab18 remain unknown in most human cancers, including head and neck squamous cell carcinoma (HNSCC). Methods We used immunohistochemistry to examine Rab18 protein expression in 112 cases of HNSCC specimens. We overexpressed and knockdown Rab18 in FaDu and Detroit562 cancer cell lines. Biological roles and mechanisms of Rab18 were examined using MTT, colony formation, Matrigel invasion assay, Western blotting, Annexin V and JC1 staining. Results Rab18 was upregulated in 45/112 (40.2%) cases of HNSCC tissues, which correlated with advanced T classification, positive nodal metastasis and tumor node metastasis (TNM) stage. The Oncomine and The Cancer Genome Atlas (TCGA) analyses indicated that Rab18 was elevated in human HNSCC tissues and correlated with poor patient survival. Functionally, Rab18 overexpression increased growth rate, colony numbers, cell cycle progression and invading ability in FaDu cells. Rab18 downregulated cisplatin-induced apoptosis and upregulated the mitochondrial membrane potential (Δψm). Western blot revealed that Rab18 overexpression induced epithelial-to-mesenchymal transition, with downregulation of E-cadherin and upregulation of N-cadherin, Vimentin and Twist. Rab18 overexpression also upregulated Survivin protein and Rab18 knockdown showed the opposite effects on these proteins. Treatment of STAT3 inhibitor, SH-4-54, inhibited cell invasion, increased E-cadherin and downregulated N-cadherin, Twist and Survivin. SH-4-54 also abolished the effects of BCAT1 on these proteins, as well as cell invasion. Conclusion In summary, our data showed that Rab18 was overexpressed in human HNSCC and functioned as an oncoprotein. Rab18 regulated HNSCC cell proliferation, invasion and cisplatin sensitivity through STAT3 signaling in HNSCC.
Collapse
Affiliation(s)
- Xu Ji
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xing Guo
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yan Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiaotian Li
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hong Li
- Department of Otolaryngology, The Fourth Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
38
|
Sommariva M, Gagliano N. E-Cadherin in Pancreatic Ductal Adenocarcinoma: A Multifaceted Actor during EMT. Cells 2020; 9:E1040. [PMID: 32331358 PMCID: PMC7226001 DOI: 10.3390/cells9041040] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a step-wise process observed in normal and tumor cells leading to a switch from epithelial to mesenchymal phenotype. In tumors, EMT provides cancer cells with a metastatic phenotype characterized by E-cadherin down-regulation, cytoskeleton reorganization, motile and invasive potential. E-cadherin down-regulation is known as a key event during EMT. However, E-cadherin expression can be influenced by the different experimental settings and environmental stimuli so that the paradigm of EMT based on the loss of E-cadherin determining tumor cell behavior and fate often becomes an open question. In this review, we aimed at focusing on some critical points in order to improve the knowledge of the dynamic role of epithelial cells plasticity in EMT and, specifically, address the role of E-cadherin as a marker for the EMT axis.
Collapse
Affiliation(s)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| |
Collapse
|
39
|
Wang B, Yang X, Zhao T, Du H, Wang T, Zhong S, Yang B, Li H. Upregulation of contactin-1 expression promotes prostate cancer progression. Oncol Lett 2019; 19:1611-1618. [PMID: 32002038 PMCID: PMC6960391 DOI: 10.3892/ol.2019.11244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 11/14/2019] [Indexed: 12/31/2022] Open
Abstract
Contactin-1 (CNTN-1) has been reported to serve an oncogenic role in several cancer types. However, detailed mechanisms describing the influence of CNTN-1 in prostate cancer progression have not yet been elucidated. The present study aimed to determine the clinical significance of CNTN-1 expression in prostate cancer progression, and also to investigate the regulatory role of CNTN-1 in the proliferation, migration and invasive ability of prostate cancer cells. The results of the present study indicated that expression levels of CNTN-1 were significantly higher in prostate cancer tissues compared with adjacent normal tissues. Moreover, a high expression level of CNTN-1 was positively correlated with tumor size, stage and metastasis, as well as a poorer prognosis in patients with prostate cancer. Furthermore, CNTN-1-knockdown in prostate cancer cells (using short hairpin RNA) resulted in the significant inhibition of cancer cell proliferation, colony formation, migration and invasiveness. Silencing of CNTN-1 expression also suppressed epithelial-mesenchymal transition in prostate cancer cells via the upregulation of E-cadherin, and the downregulation of N-cadherin and vimentin expression. Inhibition of CNTN-1 expression also reduced the activity of the PI3K/AKT signaling pathway in prostate cancer cells. Thus, it was demonstrated that CNTN-1 expression is upregulated, and plays an oncogenic role, in prostate cancer cells. The results of the current study suggest that CNTN-1 may represent a promising therapeutic target, potentially improving the treatment of patients with prostate cancer.
Collapse
Affiliation(s)
- Boren Wang
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Xi Yang
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Ting Zhao
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Hanghang Du
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Tong Wang
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Suping Zhong
- Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Bo Yang
- Department of Pathology, First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Hui Li
- Department of Microbiology and Immunology, Medical School of Jishou University, Jishou, Hunan 416000, P.R. China
| |
Collapse
|
40
|
Zinn R, Otterbein H, Lehnert H, Ungefroren H. RAC1B: A Guardian of the Epithelial Phenotype and Protector Against Epithelial-Mesenchymal Transition. Cells 2019; 8:cells8121569. [PMID: 31817229 PMCID: PMC6952788 DOI: 10.3390/cells8121569] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
The small GTPase Ras-related C3 botulinum toxin substrate 1B (RAC1B) has been shown to potently inhibit transforming growth factor (TGF)-β1-induced cell migration and epithelial-mesenchymal transition (EMT) in pancreatic and breast epithelial cells, but the underlying mechanism has remained obscure. Using a panel of pancreatic ductal adenocarcinoma (PDAC)-derived cell lines of different differentiation stages, we show that RAC1B is more abundantly expressed in well differentiated as opposed to poorly differentiated cells. Interestingly, RNA interference-mediated knockdown of RAC1B decreased expression of the epithelial marker protein E-cadherin, encoded by CDH1, and enhanced its TGF-β1-induced downregulation, whereas ectopic overexpression of RAC1B upregulated CDH1 expression and largely prevented its TGF-β1-induced silencing of CDH1. Conversely, knockdown of RAC1B, or deletion of the RAC1B-specific exon 3b by CRISPR/Cas-mediated genomic editing, enhanced basal and TGF-β1-induced upregulation of mesenchymal markers like Vimentin, and EMT-associated transcription factors such as SNAIL and SLUG. Moreover, we demonstrate that knockout of RAC1B enhanced the cells’ migratory activity and derepressed TGF-β1-induced activation of the mitogen-activated protein kinase ERK2. Pharmacological inhibition of ERK1/2 activation in RAC1B-depleted cells rescued cells from the RAC1B knockdown-induced enhancement of cell migration, TGF-β1-induced downregulation of CDH1, and upregulation of SNAI1. We conclude that RAC1B promotes epithelial gene expression and suppresses mesenchymal gene expression by interfering with TGF-β1-induced MEK-ERK signaling, thereby protecting cells from undergoing EMT and EMT-associated responses like acquisition of cell motility.
Collapse
Affiliation(s)
- Rabea Zinn
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hannah Otterbein
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Ungefroren
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Correspondence: ; Tel.: +49-451-3101-7866
| |
Collapse
|
41
|
Negoi I, Beuran M, Hostiuc S, Sartelli M, El-Hussuna A, de-Madaria E. Glycosylation alterations in acute pancreatitis and pancreatic cancer: CA19-9 expression is involved in pathogenesis and maybe targeted by therapy. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S306. [PMID: 32016025 DOI: 10.21037/atm.2019.10.72] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ionut Negoi
- Carol Davila University of Medicine and Pharmacy Bucharest, Bucharest, Romania.,Department of General Surgery, Emergency Hospital of Bucharest, Bucharest, Romania
| | - Mircea Beuran
- Carol Davila University of Medicine and Pharmacy Bucharest, Bucharest, Romania.,Department of General Surgery, Emergency Hospital of Bucharest, Bucharest, Romania
| | - Sorin Hostiuc
- Carol Davila University of Medicine and Pharmacy Bucharest, Bucharest, Romania.,Department of Legal Medicine and Bioethics, National Institute of Legal Medicine Mina Minovici, Bucharest, Romania
| | | | - Alaa El-Hussuna
- Department of Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Enrique de-Madaria
- Gastroenterology Department, Alicante University General Hospital, ISABIAL, Alicante, Spain
| |
Collapse
|
42
|
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
|
43
|
Hoca M, Becer E, Kabadayı H, Yücecan S, Vatansever HS. The Effect of Resveratrol and Quercetin on Epithelial-Mesenchymal Transition in Pancreatic Cancer Stem Cell. Nutr Cancer 2019; 72:1231-1242. [DOI: 10.1080/01635581.2019.1670853] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mustafa Hoca
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Near East University, Nicosia, Cyprus
| | - Eda Becer
- Faculty of Pharmacy, Department of Biochemistry, Near East University, Nicosia, Cyprus
- Research Center of Experimental Health Sciences (DESAM), Near East University, Nicosia, Cyprus
| | - Hilal Kabadayı
- Faculty of Medicine, Department of Histology and Embryology, Manisa Celal Bayar University, Manisa, Turkey
| | - Sevinç Yücecan
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Near East University, Nicosia, Cyprus
| | - Hafize Seda Vatansever
- Research Center of Experimental Health Sciences (DESAM), Near East University, Nicosia, Cyprus
- Faculty of Medicine, Department of Histology and Embryology, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
44
|
Liu X, Guo XZ, Li HY, Chen J. KAI1 reverses the epithelial-mesenchymal transition in human pancreatic cancer cells. Hepatobiliary Pancreat Dis Int 2019; 18:471-477. [PMID: 30948327 DOI: 10.1016/j.hbpd.2019.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) plays an important role in pancreatic cancer (PC). In the present study, we investigated the effects of KAI1 gene overexpression on the EMT of human PC cell lines, MIA PaCa-2 and PACN-1. METHODS Plasmids overexpressing KAI1 and pCMV were transfected into MIA PaCa-2 and PACN-1 cells, respectively. After selection of differently transfected cells by G418, KAI1 protein levels were examined by Western blotting, and transfected cells were renamed as MIA PaCa-2-K, MIA PaCa-2-p, PACN-1-K and PACN-1-p. Wound healing and Transwell migration assays were then performed comparing the two groups of cells. EMT-related markers were analyzed by Western blotting. RESULTS The percentage of wound closure significantly decreased in MIA PaCa-2-K cells compared with MIA PaCa-2-p and MIA PaCa-2 cells after 24, 48 and 72 h (P < 0.05). In PACN-1-K cells, the percentage of wound closure significantly decreased as well (P < 0.05). Numbers of invading MIA PaCa-2, MIA PaCa-2-p and MIA PaCa-2-K cells were determined as 48.0 ± 15.4, 50.0 ± 12.4, and 12.0 ± 3.8, respectively. The corresponding numbers of invading PACN-1, PACN-1-p and PACN-1-K cells were 29.0 ± 10.6, 31.0 ± 11.4, and 8.0 ± 4.2, respectively. KAI1 overexpression induced a significant upregulation of E-cadherin and also significant downregulation of Snail, vimentin, matrix metalloproteinase 2 (MMP2) and MMP9 (all P < 0.05) in PC cells. CONCLUSIONS KAI1 reversed EMT-related marker expression and inhibited migration and invasion of PC cells. Thus, KAI1 might represent a novel potential therapeutic target for PC.
Collapse
Affiliation(s)
- Xu Liu
- Department of Gastroenterology, General Hospital of Shenyang Military Area, 83 Wenhua Road, Shenyang 110840, China
| | - Xiao-Zhong Guo
- Department of Gastroenterology, General Hospital of Shenyang Military Area, 83 Wenhua Road, Shenyang 110840, China.
| | - Hong-Yu Li
- Department of Gastroenterology, General Hospital of Shenyang Military Area, 83 Wenhua Road, Shenyang 110840, China
| | - Jiang Chen
- Department of Gastroenterology, General Hospital of Shenyang Military Area, 83 Wenhua Road, Shenyang 110840, China
| |
Collapse
|
45
|
Wang XH, Wu HY, Gao J, Wang XH, Gao TH, Zhang SF. IGF1R facilitates epithelial-mesenchymal transition and cancer stem cell properties in neuroblastoma via the STAT3/AKT axis. Cancer Manag Res 2019; 11:5459-5472. [PMID: 31354352 PMCID: PMC6580139 DOI: 10.2147/cmar.s196862] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
Background Neuroblastoma (NB) displays the most heterogeneity in clinical manifestation. The insulin-like growth factor 1 receptor (IGF1R) has long been recognized for its role in tumourigenesis and growth. The IGF/IGF1R pathway is important in maintaining cell survival. It is reported that IGF1R participates in the occurrence of NB, but the mechanism is still unclear. Methods Human NB cell lines IMR-32 and SH-SY5Y were recruited in this study. IGF1R was knocked down by transfection with short hairpin RNA. Signal transducer and activator of transcription 3 (STAT3) expression was inhibited by Cryptotanshinone treatment. Cell proliferation, migration, and invasion were determined by MTT assay, wound healing assay, and cell invasion assay, respectively. The cancer stem cell properties were characterized by tumour sphere formation assay and colony formation assay. The mRNA and protein expression levels of related proteins were detected by RT-PCR and Western blot, respectively. Results The knockdown of IGF1R inhibits NB cell tumourigenesis and the epithelial-mesenchymal transition (EMT) of NB cells. Additionally, IGF1R was found to stimulate cancer stem cell-like properties in NPC cells. The knockdown of IGF1R significantly reduced the phosphorylation of AKT, and STAT3, indicating that the activation of the AKT and STAT3 pathways was inhibited by IGF1R knockdown. Furthermore, IGF1R was demonstrated to stimulate cancer stem cell-like properties in NB cells via the regulation of the STAT3/AKT axis. Conclusion IGF1R promotes cancer stem cell properties to facilitate EMT in neuroblastoma via the STAT3/AKT axis.
Collapse
Affiliation(s)
- Xiao-Hui Wang
- Department of Pediatric Surgery, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| | - Hai-Ying Wu
- Department of Obstetrics, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| | - Jian Gao
- Department of Pediatric Surgery, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| | - Xu-Hui Wang
- Department of Pediatric Surgery, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| | - Tian-Hui Gao
- Department of Medical Oncology, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| | - Shu-Feng Zhang
- Department of Pediatric Surgery, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou 450003, People's Republic of China
| |
Collapse
|
46
|
Negoi I, Beuran M, Hostiuc S, El-Hussuna A, de-Madaria E. Platelet-to-lymphocyte ratio and CA19-9 are simple and informative prognostic factors in patients with resected pancreatic cancer. Hepatobiliary Pancreat Dis Int 2019; 18:203-205. [PMID: 31000337 DOI: 10.1016/j.hbpd.2019.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/26/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Ionut Negoi
- Carol Davila University of Medicine and Pharmacy Bucharest, Romania; Department of General Surgery, Emergency Hospital of Bucharest, Romania.
| | - Mircea Beuran
- Carol Davila University of Medicine and Pharmacy Bucharest, Romania; Department of General Surgery, Emergency Hospital of Bucharest, Romania
| | - Sorin Hostiuc
- Carol Davila University of Medicine and Pharmacy Bucharest, Romania; Department of Legal Medicine and Bioethics, National Institute of Legal Medicine Mina Minovici, Romania
| | - Alaa El-Hussuna
- Department of Surgery, Aalborg University Hospital, Hobrovej 18-22, 9000 Aalborg, Denmark
| | - Enrique de-Madaria
- Servicio de Aparato Digestivo, Unidad de Patología Pancreática, Gastroenterology Department, Pancreatic Unit, Hospital General Universitario de Alicante, Alicante, Spain
| |
Collapse
|
47
|
Wang RQ, Geng J, Sheng WJ, Liu XJ, Jiang M, Zhen YS. The ionophore antibiotic gramicidin A inhibits pancreatic cancer stem cells associated with CD47 down-regulation. Cancer Cell Int 2019; 19:145. [PMID: 31139022 PMCID: PMC6532126 DOI: 10.1186/s12935-019-0862-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
Background Pancreatic cancer stem cells (CSCs), a special population of cells, renew themselves infinitely and resist to various treatment. Gramicidin A (GrA), an ionophore antibiotic derived from microorganism, can form channels across the cell membrane and disrupt cellular ionic homeostasis, leading to cell dysfunction and death. As reported, the ionophore antibiotic salinomycin (Sal) has been proved to kill CSCs effectively. Whether GrA owns the potential as a therapeutic drug for CSCs still remains unknown. This study investigated the effect of GrA on pancreatic CSCs and the mechanism. Methods Tumorsphere formation assay was performed to assess pancreatic CSCs self-renewal potential. In vitro hemolysis assay was determined to test the borderline concentration of GrA. CCK-8 assay was used to detect pancreatic cancer cell proliferation capability. Flow cytometry was performed to detect cell apoptosis and mitochondrial membrane potential. Scanning and transmission electron microscopy was used to observe ultrastructural morphological changes on cell membrane surface and mitochondria, respectively. Western blot analysis was used to determine relative protein expression levels. Immunofluorescence staining was performed to observe CD47 re-distribution. Results GrA at 0.05 μM caused tumorspheres disintegration and decrease in number of pancreatic cancer BxPC-3 and MIA PaCa-2 cells. GrA and Sal both inhibited cancer cell proliferation. The IC50 values of GrA and Sal for BxPC-3 cells were 0.025 μM and 0.363 μM; while for MIA PaCa-2 cells were 0.032 μM and 0.163 μM, respectively. Compared on equal concentrations, the efficacy of GrA was stronger than that of Sal. GrA at 0.1 μM or lower did not cause hemolysis. GrA induced ultrastructural changes, such as the decrease of microvilli-like protrusions on cell surface membrane and the swelling of mitochondria. GrA down-regulated the expression levels of CD133, CD44, and CD47; in addition, CD47 re-distribution was observed on cell surface. Moreover, GrA showed synergism with gemcitabine in suppressing cancer cell proliferation. Conclusions The study found that GrA was highly active against pancreatic CSCs. It indicates that GrA exerts inhibitory effects against pancreatic CSCs associated with CD47 down-regulation, implying that GrA might play a positive role in modulating the interaction between macrophages and tumor cells.
Collapse
Affiliation(s)
- Rui-Qi Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Jing Geng
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Wei-Jin Sheng
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Xiu-Jun Liu
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Min Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| | - Yong-Su Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Tiantan Xili, Beijing, 100050 China
| |
Collapse
|
48
|
Shan M, Yang D, Dou H, Zhang L. Fucosylation in cancer biology and its clinical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 162:93-119. [PMID: 30905466 DOI: 10.1016/bs.pmbts.2019.01.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fucosylation is the process of transferring fucose from GDP-fucose to their substrates, which includes certain proteins, N- and O-linked glycans in glycoprotein or glycolipids, by fucosyltransferases in all mammalian cells. Fucosylated glycans play vital role in selectin-mediated leukocyte extravasation, lymphocyte homing, and pathogen-host interactions, whereas fucosylated proteins are essential for signaling transduction in numerous ontogenic events. Aberrant fucosylation due to the availability of high energy donor GDP-fucose, abnormal expression of FUTs and/or α-fucosidase, and the availability of their substrates leads to different fucosylated glycan or protein structures. Accumulating evidence demonstrates that aberrant fucosylation plays important role in all aspects of cancer biology. In this review, we will summarize the current knowledge about fucosylation in different physiological and pathological processes with a focus on their roles not only in cancer cell proliferation, invasion, and metastasis but also in tumor immune surveillance. Furthermore, the clinical potential and applications of fucosylation in cancer diagnosis and treatment will also be discussed.
Collapse
Affiliation(s)
- Ming Shan
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Dandan Yang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huaiqian Dou
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lijuan Zhang
- Systems Biology and Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
49
|
Varillas JI, Zhang J, Chen K, Barnes II, Liu C, George TJ, Fan ZH. Microfluidic Isolation of Circulating Tumor Cells and Cancer Stem-Like Cells from Patients with Pancreatic Ductal Adenocarcinoma. Theranostics 2019; 9:1417-1425. [PMID: 30867841 PMCID: PMC6401494 DOI: 10.7150/thno.28745] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) requires multimodal therapeutic approaches and disease monitoring for effective treatment. Liquid biopsy biomarkers, including circulating tumor cells (CTCs) and cancer stem-like cells (CSCs), hold promise for evaluating treatment response promptly and guiding therapeutic modifications. Methods: From 24 patients with metastatic PDAC (stage IV, M1) undergoing active systemic treatment, we collected 78 blood samples at different time points for CTC and CSC isolation using a microfluidic platform functionalized with antibodies against a CTC biomarker, epithelial cell adhesion molecule (EpCAM), or a CSC biomarker, CD133. These isolated cells were further verified, via fluorescent staining and imaging, using cytokeratin (CK), CD45, and nucleic acid stain 4',6-diamidino-2-phenylindole (DAPI). Results: The majority (84.4%) of patient blood samples were positive for CTCs (EpCAM+CK+CD45-DAPI+) and 70.8% of patient blood samples were positive for CSCs (CD133+CK+CD45-DAPI+), using the highest baseline value of healthy samples as threshold. The CTC subtypes (EpCAM+CK+CD45-DAPI+CD133+ and EpCAM+CK+CD45-DAPI+CD133-) and CSC subtypes (CD133+CK+CD45-DAPI+EpCAM+ and CD133+CK+CD45-DAPI+EpCAM-) were also analyzed using immunochemical methods. In several cases, CSCs exhibited cytokeratin expression that did not express EpCAM, indicating that they will not be detected using EpCAM-based isolation. Conclusion: The microfluidic platform enabled the reliable isolation of CTCs and CSCs from PDAC patient samples, as well as their subtypes. Complementary assessment of both CTCs and CSCs appears advantageous to assess the profile of tumor progressing in some cases. This research has important implications for the application and interpretation of approved methods to detect CTCs.
Collapse
|
50
|
Hui DHF, Tam KJ, Jiao IZF, Ong CJ. Semaphorin 3C as a Therapeutic Target in Prostate and Other Cancers. Int J Mol Sci 2019; 20:E774. [PMID: 30759745 PMCID: PMC6386986 DOI: 10.3390/ijms20030774] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 12/21/2022] Open
Abstract
The semaphorins represent a large family of signaling molecules with crucial roles in neuronal and cardiac development. While normal semaphorin function pertains largely to development, their involvement in malignancy is becoming increasingly evident. One member, Semaphorin 3C (SEMA3C), has been shown to drive a number of oncogenic programs, correlate inversely with cancer prognosis, and promote the progression of multiple different cancer types. This report surveys the body of knowledge surrounding SEMA3C as a therapeutic target in cancer. In particular, we summarize SEMA3C's role as an autocrine andromedin in prostate cancer growth and survival and provide an overview of other cancer types that SEMA3C has been implicated in including pancreas, brain, breast, and stomach. We also propose molecular strategies that could potentially be deployed against SEMA3C as anticancer agents such as biologics, small molecules, monoclonal antibodies and antisense oligonucleotides. Finally, we discuss important considerations for the inhibition of SEMA3C as a cancer therapeutic agent.
Collapse
Affiliation(s)
- Daniel H F Hui
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| | - Kevin J Tam
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| | - Ivy Z F Jiao
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| | - Christopher J Ong
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|