1
|
Alshahrani MY, Oghenemaro EF, Rizaev J, Kyada A, Roopashree R, Kumar S, Taha ZA, Yadav G, Mustafa YF, Abosaoda MK. Exploring the modulation of TLR4 and its associated ncRNAs in cancer immunopathogenesis, with an emphasis on the therapeutic implications and mechanisms underlying drug resistance. Hum Immunol 2025; 86:111188. [PMID: 39631102 DOI: 10.1016/j.humimm.2024.111188] [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: 08/28/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
This study provides an in-depth analysis of the pathogenic relevance, therapeutic implications, and mechanisms of treatment resistance associated with TLR4 and its ncRNAs in cancer immunopathogenesis. TLR4, a pivotal component of the innate immune response, has been implicated in promoting inflammation, tumorigenesis, and immune evasion across various malignancies, including gastric, ovarian, and hepatocellular carcinoma. The interactions between TLR4 and specific ncRNAs, such as lncRNAs and miRNAs, play a crucial role in modulating TLR4 signaling pathways, influencing immune cell dynamics, and contributing to chemoresistance. These ncRNAs facilitate tumor-promoting processes, including macrophage polarization, dendritic cell suppression, and T-cell regulation, effectively establishing an immunosuppressive tumor microenvironment that further enhances therapeutic resistance. A comprehensive understanding of the complex interplay between TLR4 and ncRNAs unveils potential avenues for identifying predictive biomarkers and discovering novel therapeutic targets in cancer. Future research initiatives should prioritize the development of personalized therapeutic strategies that specifically target TLR4 signaling and its ncRNA regulators to counteract drug resistance and improve clinical outcomes. This review extensively evaluates the role of TLR4 in cancer biology, emphasizing its critical importance in developing innovative cancer management strategies.
Collapse
Affiliation(s)
- Mohammad Y Alshahrani
- Central Labs, King Khalid University, AlQura 'a, Abha, P.O. Box 960, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Abraka, Delta State, Nigeria.
| | - Jasur Rizaev
- Professor, Doctor of Medical Sciences, Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan.
| | - Ashishkumar Kyada
- Marwadi University, Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Zahraa Ahmed Taha
- Medical Laboratory Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001 Babylon, Iraq.
| | - Geeta Yadav
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, -41001, Iraq.
| | - Munthar Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq; College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq.
| |
Collapse
|
2
|
Zhang Y, Zhang C, Peng C, Jia J. Unraveling the crosstalk: circRNAs and the wnt signaling pathway in cancers of the digestive system. Noncoding RNA Res 2024; 9:853-864. [PMID: 38586314 PMCID: PMC10995981 DOI: 10.1016/j.ncrna.2024.03.004] [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: 01/04/2024] [Revised: 03/02/2024] [Accepted: 03/03/2024] [Indexed: 04/09/2024] Open
Abstract
Circular RNA (circRNA) is a unique type of noncoding RNA molecule characterized by its closed-loop structure. Functionally versatile, circRNAs play pivotal roles in gene expression regulation, protein activity modulation, and participation in cell signaling processes. In the context of cancers of the digestive system, the Wnt signaling pathway holds particular significance. Anomalous activation of the Wnt pathway serves as a primary catalyst for the development of colorectal cancer. Extensive research underscores the notable participation of circRNAs associated with the Wnt pathway in the progression of digestive system tumors. These circRNAs exhibit pronounced dysregulation across esophageal cancer, gastric cancer, liver cancer, colorectal cancer, pancreatic cancer, and cholangiocarcinoma. Furthermore, the altered expression of circRNAs linked to the Wnt pathway correlates with prognostic factors in digestive system tumors. Additionally, circRNAs related to the Wnt pathway showcase potential as diagnostic, therapeutic, and prognostic markers within the realm of digestive system tumors. This comprehensive review outlines the interplay between circRNAs and the Wnt signaling pathway in cancers of the digestive system. It seeks to provide a comprehensive perspective on their association while delving into ongoing research that explores the clinical applications of circRNAs associated with the Wnt pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chuanhui Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Junjun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Dai J, Hao Y, Chen X, Yu Q, Wang B. miR‑122/SENP1 axis confers stemness and chemoresistance to liver cancer through Wnt/β‑catenin signaling. Oncol Lett 2023; 26:390. [PMID: 37559577 PMCID: PMC10407855 DOI: 10.3892/ol.2023.13976] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/22/2023] [Indexed: 08/11/2023] Open
Abstract
The property of inherent stemness of tumor cells coupled with the development of chemoresistance results in a poor prognosis for patients with liver cancer. Therefore, the present study focused on microRNA (miR)-122, a potential tumor suppressor, the expression of which has been previously shown to be significantly decreased and negatively associated with cancer cell stemness in liver cancer. The present study aimed to identify the molecular targets of miR-122 whilst uncovering the mechanism underlying chemoresistance and stemness of HepG2 cells in liver cancer. Bioinformatics online tools, such as ENCORI, coupled with dual-luciferase reporter assays in HepG2 cells, were used to identify and validate small ubiquitin-like modifier (SUMO) specific peptidase 1 (SENP1) as a potential target of miR-122 in liver cancer. The liver cancer stem cell population was determined using sphere formation assays and flow cytometry, whilst stem cell markers (Oct3/4, Nanog, B lymphoma Mo-MLV insertion region 1 homolog and Notch1) were detected by reverse transcription-quantitative PCR. Chemoresistance, cell proliferation and migratory ability of HepG2 cells were monitored using Cell Counting Kit-8, colony formation and Transwell assays, respectively. The overexpression of miR-122 by mimic transfection led to a significant decrease in the number spheres, downregulation of stem cell marker expression, the number of CD24+ cells, drug-resistance protein levels (P-glycoprotein and multidrug resistance protein), impaired chemoresistance, proliferation and migration of HepG2 cells. The transfection of SENP1 overexpression vector resulted in contrasting functions to miR-122 mimics, by partially reversing the effects induced by miR-122 mimic transfection in HepG2 cells. Wnt/β-catenin signaling has been proven to be involved in cancer stemness and malignant behavior. Western blotting analysis in HepG2 cells showed that the expression levels of both Wnt1 and β-catenin were significantly reduced after overexpressing miR-122, but increased after overexpressing SENP1. Co-transfection with the SENP1 overexpression vector reversed the suppression induced by the miR-122 mimics on Wnt1 and β-catenin expression. Co-immunoprecipitation, SUMOylation and half-life assays showed SENP1 interacted with β-catenin and decreased the SUMOylation of β-catenin, thereby enhancing its stability. Finally, tumor xenograft analyses revealed that HepG2 cells transfected with Agomir-122 exerted significantly lower tumor initiation frequency and growth rate, and a superior response to DOX in vivo, compared with those transfected with Agomir NC. Taken together, data from the present study miR-122/SENP1 axis can regulate β-catenin stability through de-SUMOylation, thereby promoting stemness and chemoresistance in liver cancer.
Collapse
Affiliation(s)
- Jianbo Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400060, P.R. China
- Department of General Surgery, Nan'an District People's Hospital of Chongqing, Chongqing 400060, P.R. China
| | - Yaqin Hao
- Department of Gastroenterology, The Fifth People's Hospital of Chongqing, Chongqing 400060, P.R. China
| | - Xun Chen
- Department of Anesthesiology, Nan'an District People's Hospital of Chongqing, Chongqing 400060, P.R. China
| | - Qingsan Yu
- Department of General Surgery, Nan'an District People's Hospital of Chongqing, Chongqing 400060, P.R. China
| | - Bin Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400060, P.R. China
- Department of General Surgery, Chongqing Hospital of Integrated Traditional Chinese and Western Medicine, Chongqing 400060, P.R. China
| |
Collapse
|
4
|
Tang Y, Guo C, Chen C, Zhang Y. Characterization of cellular senescence patterns predicts the prognosis and therapeutic response of hepatocellular carcinoma. Front Mol Biosci 2022; 9:1100285. [PMID: 36589233 PMCID: PMC9800843 DOI: 10.3389/fmolb.2022.1100285] [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: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a prevalent malignancy with a high mortality rate. Cellular senescence, an irreversible state of cell cycle arrest, plays a paradoxical role in cancer progression. Here, we aimed to identify Hepatocellular carcinoma subtypes by cellular senescence-related genes (CSGs) and to construct a cellular senescence-related gene subtype predictor as well as a novel prognostic scoring system, which was expected to predict clinical outcomes and therapeutic response of Hepatocellular carcinoma. Methods: RNA-seq data and clinical information of Hepatocellular carcinoma patients were derived from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC). The "multi-split" selection was used to screen the robust prognostic cellular senescence-related genes. Unsupervised clustering was performed to identify CSGs-related subtypes and a discriminant model was obtained through multiple statistical approaches. A CSGs-based prognostic model-CSGscore, was constructed by LASSO-Cox regression and stepwise regression. Immunophenoscore (IPS) and Tumor Immune Dysfunction and Exclusion (TIDE) were utilized to evaluate the immunotherapy response. Tumor stemness indices mRNAsi and mDNAsi were used to analyze the relationship between CSGscore and stemness. Results: 238 robust prognostic differentially expressed cellular senescence-related genes (DECSGs) were used to categorize all 336 hepatocellular carcinoma patients of the TCGA-LIHC cohort into two groups with different survival. Two hub genes, TOP2A and KIF11 were confirmed as key indicators and were used to form a precise and concise cellular senescence-related gene subtype predictor. Five genes (PSRC1, SOCS2, TMEM45A, CCT5, and STC2) were selected from the TCGA training dataset to construct the prognostic CSGscore signature, which could precisely predict the prognosis of hepatocellular carcinoma patients both in the training and validation datasets. Multivariate analysis verified it as an independent prognostic factor. Besides, CSGscore was also a valuable predictor of therapeutic responses in hepatocellular carcinoma. More downstream analysis revealed the signature genes were significantly associated with stemness and tumor progression. Conclusion: Two subtypes with divergent outcomes were identified by prognostic cellular senescence-related genes and based on that, a subtype indicator was established. Moreover, a prognostic CSGscore system was constructed to predict the survival outcomes and sensitivity of therapeutic responses in hepatocellular carcinoma, providing novel insight into hepatocellular carcinoma biomarkers investigation and design of tailored treatments depending on the molecular characteristics of individual patients.
Collapse
Affiliation(s)
- Yuqin Tang
- Clinical Bioinformatics Experimental Center, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Chengbin Guo
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Chuanliang Chen
- Clinical Bioinformatics Experimental Center, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Yongqiang Zhang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Demir AB, Benvenuto D, Karacicek B, Erac Y, Spoto S, Angeletti S, Ciccozzi M, Tosun M. Implications of Possible HBV-Driven Regulation of Gene Expression in Stem Cell-like Subpopulation of Huh-7 Hepatocellular Carcinoma Cell Line. J Pers Med 2022; 12:jpm12122065. [PMID: 36556285 PMCID: PMC9786676 DOI: 10.3390/jpm12122065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Elevated levels of STIM1, an endoplasmic reticulum Ca2+ sensor/buffering protein, appear to be correlated with poor cancer prognosis in which microRNAs are also known to play critical roles. The purpose of this study is to investigate possible HBV origins of specific microRNAs we identified in a stem cell-like subpopulation of Huh-7 hepatocellular carcinoma (HCC) cell lines with enhanced STIM1 and/or Orai1 expression that mimicked poor cancer prognosis. Computational strategies including phylogenetic analyses were performed on miRNome data we obtained from an EpCAM- and CD133-expressing Huh-7 HCC stem cell-like subpopulation with enhanced STIM1 and/or Orai1 expression originally cultured in the present work. Results revealed two putative regions in the HBV genome based on the apparent clustering pattern of stem loop sequences of microRNAs, including miR3653. Reciprocal analysis of these regions identified critical human genes, of which their transcripts are among the predicted targets of miR3653, which was increased significantly by STIM1 or Orai1 enhancement. Briefly, this study provides phylogenetic evidence for a possible HBV-driven epigenetic remodeling that alters the expression pattern of Ca2+ homeostasis-associated genes in STIM1- or Orai1 overexpressing liver cancer stem-like cells for a possible mutual survival outcome. A novel region on HBV-X protein may affect liver carcinogenesis in a genotype-dependent manner. Therefore, detection of the viral genotype would have a clinical impact on prognosis of HBV-induced liver cancers.
Collapse
Affiliation(s)
- Ayse Banu Demir
- Department of Medical Biology, Faculty of Medicine, Izmir University of Economics, 35330 Izmir, Turkey
| | - Domenico Benvenuto
- Faculty of Medicine, University Campus Bio-Medico of Rome (UCBM), 200 Rome, Italy
| | - Bilge Karacicek
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, 35340 Izmir, Turkey
| | - Yasemin Erac
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35100 Izmir, Turkey
| | - Silvia Spoto
- Diagnostic and Therapeutic Medicine Division, Fondazione Policlinico Universitario Campus Bio-Medico, 200 Rome, Italy
| | - Silvia Angeletti
- Clinical Laboratory Science Unit, Faculty of Medicine, University Campus Bio-Medico of Rome (UCBM), 200 Rome, Italy
- Clinical Laboratory Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico Via Alvaro del Portillo, 200 Rome, Italy
- Correspondence: (S.A.); (M.T.); Tel.: +39-06225411461 (S.A.); +90-2324889843 (M.T.)
| | - Massimo Ciccozzi
- Medical Statistics and Molecular Epidemiology Unit, Faculty of Medicine, University Campus Bio-Medico of Rome (UCBM), 200 Rome, Italy
| | - Metiner Tosun
- Department of Medical Pharmacology, Faculty of Medicine, Izmir University of Economics, 35330 Izmir, Turkey
- Correspondence: (S.A.); (M.T.); Tel.: +39-06225411461 (S.A.); +90-2324889843 (M.T.)
| |
Collapse
|
6
|
Su D, Lin Z. Dichloroacetate attenuates the stemness of hepatocellular carcinoma cells via promoting nucleus-cytoplasm translocation of YAP. ENVIRONMENTAL TOXICOLOGY 2021; 36:975-983. [PMID: 33405312 DOI: 10.1002/tox.23098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 06/12/2023]
Abstract
The antitumor effects of dichloroacetate (DCA) have been widely explored, however, its roles in hepatocellular carcinoma (HCC) progression are still unclear. In the current work, we found that DCA had little effects on HCC cell viability, but could attenuate the stemness of HCC cells, which is evident by decreasing the tumor sphere-formation ability, ALDH activity and the expression of stemness critical regulators. Mechanistic studies based on RNA-sequencing data showed that DCA activated the Hippo pathway. Furthermore, we indicated that DCA promoted the nucleus-cytoplasm translocation of YAP, but not TAZ, another critical executor of Hippo pathway. Moreover, suppressing of Hippo pathway using XMU-MP-1, an inhibitor of Hippo pathway, partially abrogated DCA-induced inhibitory effects on HCC cell stemness. This work suggests that DCA might be a potential inhibitor for HCC progression.
Collapse
Affiliation(s)
- Duanyu Su
- Department of Cancer Radiotherapy, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Zhian Lin
- Department of Cancer Radiotherapy, Zhongshan Hospital Xiamen University, Xiamen, China
| |
Collapse
|
7
|
Popescu GDA, Scheau C, Badarau IA, Dumitrache MD, Caruntu A, Scheau AE, Costache DO, Costache RS, Constantin C, Neagu M, Caruntu C. The Effects of Capsaicin on Gastrointestinal Cancers. Molecules 2020; 26:94. [PMID: 33379302 PMCID: PMC7794743 DOI: 10.3390/molecules26010094] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal (GI) cancers are a group of diseases with very high positions in the ranking of cancer incidence and mortality. While they show common features regarding the molecular mechanisms involved in cancer development, organ-specific pathophysiological processes may trigger distinct signaling pathways and intricate interactions with inflammatory cells from the tumoral milieu and mediators involved in tumorigenesis. The treatment of GI cancers is a topic of increasing interest due to the severity of these diseases, their impact on the patients' survivability and quality of life, and the burden they set on the healthcare system. As the efficiency of existing drugs is hindered by chemoresistance and adverse reactions when administered in high doses, new therapies are sought, and emerging drugs, formulations, and substance synergies are the focus of a growing number of studies. A class of chemicals with great potential through anti-inflammatory, anti-oxidant, and anti-tumoral effects is phytochemicals, and capsaicin in particular is the subject of intensive research looking to validate its position in complementing cancer treatment. Our paper thoroughly reviews the available scientific evidence concerning the effects of capsaicin on major GI cancers and its interactions with the molecular pathways involved in the course of these diseases.
Collapse
Affiliation(s)
| | - Cristian Scheau
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.A.B.); (C.C.)
| | - Ioana Anca Badarau
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.A.B.); (C.C.)
| | - Mihai-Daniel Dumitrache
- Departament of Pneumology IV, “Marius Nasta” Institute of Pneumophtysiology, 050159 Bucharest, Romania;
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania;
- Department of Preclinical Sciences, Faculty of Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Daniel Octavian Costache
- Department of Dermatology, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania;
| | - Raluca Simona Costache
- Gastroenterology and Internal Medicine Clinic, “Carol Davila” Central Military Emergency Hospital, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Carolina Constantin
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.N.)
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.N.)
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
- Faculty of Biology, University of Bucharest, 76201 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.A.B.); (C.C.)
- Department of Dermatology, Prof. N.C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| |
Collapse
|
8
|
Zhong Y, Qi H, Li X, An M, Shi Q, Qi J. Tumor supernatant derived from hepatocellular carcinoma cells treated with vincristine sulfate have therapeutic activity. Eur J Pharm Sci 2020; 155:105557. [PMID: 32946955 DOI: 10.1016/j.ejps.2020.105557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/03/2020] [Accepted: 09/13/2020] [Indexed: 12/11/2022]
Abstract
Vincristine sulfate (VCR), a commonly used chemotherapeutic agent, kills cancer cells as well as the normal cells for its cytotoxicity. But it is still unclear whether it can exert therapeutic effect on untreated cancer cells by changing the supernatant of cancer cells. Here, we explored the subsequent cascade effects of the supernatant of cancer cells that were transiently treated with VCR on untreated tumor cells and its responsible mechanisms. VCR and three different hepatocellular carcinoma (HCC) cell lines were used for an experiment. The experiment was conducted in vitro to eliminate the body's internal factors and the effects of the immune system. The results suggested that drug-free tumor supernatant (TSN) could promote the differentiation, repress the transcription of liver cancer stem cell's markers and the proliferation in SMMC-7721, Bel-7402 and Huh7 cells. Furthermore, we found that the TSN could abolish YAP1 transcriptional activity to inhibit the proliferation and increase the transcriptional activity of HNF4α to promote the differentiation in SMMC-7721 and Bel-7402 cells. In conclusion, the TSN could inhibit the proliferation and induce differentiation in different HCC cells.
Collapse
Affiliation(s)
- Yan Zhong
- School of Pharmaceutical Sciences, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China
| | - Huanli Qi
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xuejiao Li
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengyang An
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Qingwen Shi
- School of Pharmaceutical Sciences, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jinsheng Qi
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
9
|
Hemati H, Kaur J, Sobti RC, Trehanpati N. Inhibition of NOTCH signaling pathway chemosensitizes HCC CD133 + cells to vincristine and 5-fluorouracil through upregulation of BBC3. Biochem Biophys Res Commun 2020; 525:941-947. [PMID: 32173531 DOI: 10.1016/j.bbrc.2020.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/01/2020] [Indexed: 02/08/2023]
Abstract
In hepatocellular carcinoma (HCC), the poor response to the chemotherapeutic agents is partially attributed to the chemoresistance property of cancer stem cells (CSCs). NOTCH signaling pathway plays a crucial role in the chemoresistance through the maintenance of the CSCs. We observed that the NOTCH pathway was activated in HCC CD133+ cells treated with vincristine (VIN)1 and 5-fluorouracil (5-FU)2. Therefore, we examined whether inhibition of the NOTCH can improve sensitization of HCC CD133+ cells to VIN and 5-FU. The Huh7 cell line was pre-incubated γ-secretase DAPT, as a NOTCH inhibitor, and then treated with IC50 dose of VIN or 5-FU. The CD133+ cells were then isolated and analyzed for the cell viability, apoptosis, migration and spheroid formation capacities, and gene and protein expression. It was observed that pre-incubation with DAPT significantly downregulated the expression of NOTCH-related genes and led to a significant reduction in VIN- and 5-FU-CD133+ population. In addition, DAPT pre-incubated VIN- and 5-FU-treated-CD133+ cells formed fewer spheroids in 3D culture and had a lesser migration capacity in 2D culture. Importantly, DAPT enhanced the apoptosis rate of VIN- and 5-FU-treated CD133+ cells for 3- and 2-fold, which was correlated with the enhanced expression of pro-apoptotic BBC3 (BCL-2-binding component 3) and decreased expression of HES1 that was reported to regulate BBC3 negatively. Collectively, it was observed that NOTCH inhibition sensitized the HCC CD133+ cells to VIN and 5-FU through enhancing BBC3-mediated apoptosis. The results highlighted the role of NOTCH/HES1/BBC3 axis in resistance of CD133+ cells to VIN and 5-FU. Understanding the molecular mechanisms underlying chemoresistance in HCC CD133+ cells may help in designing the novel targeted therapies to chemosensitize them.
Collapse
Affiliation(s)
- Hamed Hemati
- Department of Biotechnology, Panjab University, Chandigarh, India; Department of Cellular & Molecular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jagdeep Kaur
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - R C Sobti
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Nirupma Trehanpati
- Department of Cellular & Molecular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
10
|
Steichen C, Hannoun Z, Luce E, Hauet T, Dubart-Kupperschmitt A. Genomic integrity of human induced pluripotent stem cells: Reprogramming, differentiation and applications. World J Stem Cells 2019; 11:729-747. [PMID: 31692979 PMCID: PMC6828592 DOI: 10.4252/wjsc.v11.i10.729] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/13/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Ten years after the initial generation of induced pluripotent stem cells (hiPSCs) from human tissues, their potential is no longer questioned, with over 15000 publications listed on PubMed, covering various fields of research; including disease modeling, cell therapy strategies, pharmacology/toxicology screening and 3D organoid systems. However, despite evidences that the presence of mutations in hiPSCs should be a concern, publications addressing genomic integrity of these cells represent less than 1% of the literature. After a first overview of the mutation types currently reported in hiPSCs, including karyotype abnormalities, copy number variations, single point mutation as well as uniparental disomy, this review will discuss the impact of reprogramming parameters such as starting cell type and reprogramming method on the maintenance of the cellular genomic integrity. Then, a specific focus will be placed on culture conditions and subsequent differentiation protocols and how their may also trigger genomic aberrations within the cell population of interest. Finally, in a last section, the impact of genomic alterations on the possible usages of hiPSCs and their derivatives will also be exemplified and discussed. We will also discuss which techniques or combination of techniques should be used to screen for genomic abnormalities with a particular focus on the necessary quality controls and the potential alternatives.
Collapse
Affiliation(s)
- Clara Steichen
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers F-86021, France
- Université de Poitiers, Faculté de Médecine et Pharmacie, Bâtiment D1, 6 rue de la milétrie, TSA 51115, 86073 Poitiers Cedex 9, France
| | - Zara Hannoun
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94800, France
- UMR_S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif F-94800, France
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Eléanor Luce
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94800, France
- UMR_S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif F-94800, France
- Département Hospitalo-Universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| | - Thierry Hauet
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers F-86021, France
- Université de Poitiers, Faculté de Médecine et Pharmacie, Bâtiment D1, 6 rue de la milétrie, TSA 51115, 86073 Poitiers Cedex 9, France
- Service de Biochimie, Pôle Biospharm, CHU de Poitiers, Poitiers F-86021, France
- Fédération Hospitalo-Universitaire SUPORT, CHU de Poitiers, Poitiers F-86021, France
| | - Anne Dubart-Kupperschmitt
- INSERM U1193, Hôpital Paul Brousse, Villejuif F-94800, France
- UMR_S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif F-94800, France
- Département Hospitalo-Universitaire Hepatinov, Hôpital Paul Brousse, Villejuif F-94807, France
| |
Collapse
|
11
|
Karacicek B, Erac Y, Tosun M. Functional consequences of enhanced expression of STIM1 and Orai1 in Huh-7 hepatocellular carcinoma tumor-initiating cells. BMC Cancer 2019; 19:751. [PMID: 31366337 PMCID: PMC6668110 DOI: 10.1186/s12885-019-5947-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022] Open
Abstract
Background The endoplasmic reticulum (ER) Ca2+ sensor, stromal interaction molecule1 (STIM1) activates the plasma membrane (PM) channel Orai1 in order to mediate store-operated Ca2+ entry (SOCE) in response to ER store depletion. Enhanced expression of STIM1 in cancer tissue has been associated with poor patient prognosis. Therefore, this study investigated the functional consequences of enhanced expression of STIM1 and Orai1 in a tumor-initiating subpopulation of Huh-7 hepatocellular carcinoma (HCC) cells that express epithelial cell adhesion molecule (EpCAM) and Prominin 1 (CD133). Methods We performed qRT-PCR, intracellular Ca2+ monitoring, protein analyses, and real-time cell proliferation assays on EpCAM(+)CD133(+) subpopulation of tumor-initiating Huh-7 HCC cells expressing high levels of STIM1 and/or Orai1. Statistical significance between the means of two groups was evaluated using unpaired Student’s t-test. Results Enhanced STIM1 expression significantly increased ER Ca2+ release and proliferation rate of EpCAM(+)CD133(+) cells. Conclusion STIM1 overexpression may facilitate cancer cell survival by increasing ER Ca2+-buffering capacity, which makes more Ca2+ available for the cytosolic events, on the other hand, possibly preventing Ca2+-dependent enzymatic activity in mitochondria whose Ca2+ uniporter requires much higher cytosolic Ca2+ levels.
Collapse
Affiliation(s)
- B Karacicek
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University, 35340, Izmir, Turkey
| | - Y Erac
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35100, Izmir, Turkey
| | - M Tosun
- Department of Pharmacology, School of Medicine, Izmir University of Economics, 35330, Izmir, Turkey.
| |
Collapse
|
12
|
Scheau C, Badarau IA, Caruntu C, Mihai GL, Didilescu AC, Constantin C, Neagu M. Capsaicin: Effects on the Pathogenesis of Hepatocellular Carcinoma. Molecules 2019; 24:E2350. [PMID: 31247901 PMCID: PMC6651067 DOI: 10.3390/molecules24132350] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent cancers, and to date, there have been very few drugs available that can improve survival, the most well-known being sorafenib. The pathogenesis of HCC is complex, involving multiple processes including abnormal cell and tissue regeneration, angiogenesis, genomic instability, cellular proliferation, and signaling pathway alterations. Capsaicin is a substance that holds increasingly high interest and is studied as a therapeutic option in a wide array of diseases. Several studies have investigated capsaicin roles in various stages of HCC oncogenesis. This paper aims to thoroughly detail the available information on the individual effects of capsaicin on the cellular mechanisms and pathways involved in HCC development, as well as investigate their possible cooperation and interferences. The synergistic antitumor effects of capsaicin and sorafenib are also addressed.
Collapse
Affiliation(s)
- Cristian Scheau
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Constantin Caruntu
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
- Department of Dermatology, Prof. N.C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania.
| | - Gratiela Livia Mihai
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Andreea Cristiana Didilescu
- Department of Embryology, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Carolina Constantin
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
- Faculty of Biology, University of Bucharest, 76201 Bucharest, Romania
| |
Collapse
|
13
|
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, Lehen'kyi V. Monoclonal Antibodies Targeting Ion Channels and Their Therapeutic Potential. Front Pharmacol 2019; 10:606. [PMID: 31231216 PMCID: PMC6561378 DOI: 10.3389/fphar.2019.00606] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) represent a rapidly growing pharmaceutical class of protein drugs that becomes an important part of the precision therapy. mAbs are characterized by their high specificity and affinity for the target antigen, which is mostly present on the cell surface. Ion channels are a large family of transmembrane proteins that control ion transport across the cell membrane. They are involved in almost all biological processes in both health and disease and are widely considered as prospective targets. However, no antibody-based drug targeting ion channel has been developed so far that has progressed to clinical use. Thus, we provide a comprehensive review of the elaborated mAbs against ion channels, describe their mechanisms of action, and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Aline Hantute-Ghesquier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - V'yacheslav Lehen'kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France.,FONDATION ARC, Villejuif, France
| |
Collapse
|
14
|
MiR-122 Targets SerpinB3 and Is Involved in Sorafenib Resistance in Hepatocellular Carcinoma. J Clin Med 2019; 8:jcm8020171. [PMID: 30717317 PMCID: PMC6406326 DOI: 10.3390/jcm8020171] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
The only first-line treatment approved for advanced hepatocellular carcinoma (HCC) is sorafenib. Since many patients experience drug resistance, the discovery of more effective therapeutic strategies represents an unmet clinical need. MicroRNA (MiR)-122 is downregulated in most HCCs, while oncogenic SerpinB3 is upregulated. Here, we assessed the relationship between miR-122 and SerpinB3 and their influence on cell phenotype and sorafenib resistance in HCC. A bioinformatics analysis identified SerpinB3 among hypothetical miR-122 targets. In SerpinB3-overexpressing HepG2 cells, miR-122 transfection decreased SerpinB3 mRNA and protein levels, whereas miR-122 inhibition increased SerpinB3 expression. Luciferase assay demonstrated the interaction between miR-122 and SerpinB3 mRNA. In an HCC rat model, high miR-122 levels were associated with negative SerpinB3 expression, while low miR-122 levels correlated with SerpinB3 positivity. A negative correlation between miR-122 and SerpinB3 or stem cell markers was found in HCC patients. Anti-miR-122 transfection increased cell viability in sorafenib-treated Huh-7 cells, while miR-122 overexpression increased sorafenib sensitivity in treated cells, but not in those overexpressing SerpinB3. In conclusion, we demonstrated that miR-122 targets SerpinB3, and its low levels are associated with SerpinB3 positivity and a stem-like phenotype in HCC. MiR-122 replacement therapy in combination with sorafenib deserves attention as a possible therapeutic strategy in SerpinB3-negative HCCs.
Collapse
|
15
|
Li N, Zhu Y. Targeting liver cancer stem cells for the treatment of hepatocellular carcinoma. Therap Adv Gastroenterol 2019; 12:1756284818821560. [PMID: 30719075 PMCID: PMC6348509 DOI: 10.1177/1756284818821560] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 11/15/2018] [Indexed: 02/04/2023] Open
Abstract
Liver cancer is one of the most common malignant tumors and prognosis remains poor. It has been increasingly recognized that liver cancer stem cells (LCSCs) are responsible for the carcinogenesis, recurrence, metastasis and chemoresistance of hepatocellular carcinoma (HCC). Targeting LCSCs is promising to be a new direction for the treatment of HCC. Herein, we summarize the potentially therapeutic targets in LCSCs at the level of genes, molecules and cells, such as knockout of oncogenes or oncoproteins, restoring the silent tumor suppressor genes, inhibition of the transcription factors and regulation of noncoding RNAs (including microRNAs and long noncoding RNAs) in LCSCs at the genetic level; inhibition of markers and blockade of the key signaling pathways of LCSCs at the molecular level; and inhibiting autophagy and application of oncolytic adenoviruses in LCSCs at the cellular level. Moreover, we analyze the potential targets in LCSCs to eliminate chemoresistance of HCC. Thereinto, the suppression of autophagy and Nanog by chloroquine and shRNA respectively may be the most promising targeting approaches. These targets may provide novel therapeutic strategies for the treatment of HCC by targeting LCSCs.
Collapse
Affiliation(s)
- Na Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | | |
Collapse
|
16
|
Huang WC, Tung SL, Chen YL, Chen PM, Chu PY. IFI44L is a novel tumor suppressor in human hepatocellular carcinoma affecting cancer stemness, metastasis, and drug resistance via regulating met/Src signaling pathway. BMC Cancer 2018; 18:609. [PMID: 29848298 PMCID: PMC5977745 DOI: 10.1186/s12885-018-4529-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/18/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. The disease recurrent rate is relatively high resulted in poor 5-year survival in advanced HCC. Cancer stem cells (CSCs) have been considered to be one of the main mechanisms for chemoresistance, metastasis, and recurrent disease. Interferon-induced protein 44-like (IFI44L) gene is a type I interferon-stimulated gene (ISG) and belongs to the IFI44 family. Previous reports indicated antiviral activity against HCV in IFI44L, however, its precise role and function in HCC has not been unveiled. METHODS To explore the characteristics of hepatic CSCs, we successfully enriched hepatic cancer stem-like cells from three established liver cancer cell lines (Hep3B, HepG2, and PLC lines). Parental Hep3B and HepG2 cells and their sphere cells were treated with doxorubicin for 48 h and cell viability was measured by MTT assay. HCC tissue blocks from 217 patients were sampled for tissue microarray (TMA). Follow-up information and histopathological and clinical data including age, gender, tumor grade, advanced stages, HBV, HCV, tumor number, tumor size, relapse-free survival, and overall survival were obtained from the cancer registry and medical charts. The liver TMA was evaluated for IFI44L expression using immunohistochemical staining and scores. RESULTS These hepatic cancer stem-like cells possess important cancer stemness characteristics including sphere-forming abilities, expressing important HCC cancer stem cell markers, and more chemoresistant. Interestingly, we found that overexpression of IFI44L decreased chemoresistance towards doxorubicin and knockdown of IFI44L restored chemoresistance as well as promoted sphere formation. Furthermore, we found that depletion of IFI44L enhanced migration, invasion, and pulmonary metastasis through activating Met/Src signaling pathway. Clinically, the expression level of IFI44L significantly reduced in HCC tumor tissues. Low expression of IFI44L levels also correlated with larger tumor size, disease relapse, advanced stages, and poor clinical survival in HCC patients. CONCLUSION Taken together, we first demonstrated that IFI44L is a novel tumor suppressor to affect cancer stemness, metastasis, and drug resistance via regulating Met/Src signaling pathway in HCC and can be serve as an important prognostic marker.
Collapse
Affiliation(s)
- Wei-Chieh Huang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Shiao-Lin Tung
- Department of Hematology and Oncology, Ton-Yen General Hospital, Hsinchu, Taiwan
| | - Yao-Li Chen
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of General Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Po-Ming Chen
- Taiwan Agricultural Chemicals and Toxic Substances Research Institute, Council of Agriculture, Taichung, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan. .,Department of Pathology, Show Chwan Memorial Hospital, No.542, Sec.1, Chung-Shang Road, Changhua City, Changhua County, 50008, Taiwan, Republic of China. .,National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
| |
Collapse
|
17
|
Chen Y, Meng L, Shang H, Dou Q, Lu Z, Liu L, Wang Z, He X, Song Y. β2 spectrin-mediated differentiation repressed the properties of liver cancer stem cells through β-catenin. Cell Death Dis 2018; 9:424. [PMID: 29555987 PMCID: PMC5859291 DOI: 10.1038/s41419-018-0456-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 01/01/2018] [Accepted: 02/27/2018] [Indexed: 12/23/2022]
Abstract
βII-Spectrin (β2SP), a Smad3/4 adaptor protein during transforming growth factor (TGF) β/Smad signal pathway, plays a critical role in suppressing hepatocarcinogenesis. Dedifferentiation is a distinctive feature of cancer progression. Therefore, we investigated whether the disruption of β2SP contributed to tumorigenesis of hepatocellular carcinoma (HCC) through the dedifferentiation. Down-regulation of β2SP in hepatocytes was observed in cirrhotic liver and HCC. The level of β2SP expression was closely associated with the differentiation status of hepatocytes in rat model of hepatocarcinogenesis and clinical specimens. Transgenic expression of β2SP in HCC cells promoted the differentiation of HCC cells and suppressed the growth of HCC cells in vitro. Efficient transduction of β2SP into liver CSCs resulted in a reduction in colony formation ability, spheroid formation capacity, invasive activity, chemo-resistance properties, tumorigenicity in vivo. In addition, β2 spectrin exerted its effect through β catenin in liver CSCs. In conclusion, β2 spectrin repressed the properties of liver CSCs through inducing differentiation; thus, strategies to restore its levels and activities would be a novel strategy for HCC prevention and differentiation therapy.
Collapse
Affiliation(s)
- Yuhua Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lingling Meng
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haitao Shang
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Dou
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Lu
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liping Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingxing He
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuhu Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
18
|
Nrf2-p62 autophagy pathway and its response to oxidative stress in hepatocellular carcinoma. Transl Res 2018; 193:54-71. [PMID: 29274776 DOI: 10.1016/j.trsl.2017.11.007] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Deregulation of autophagy is proposed to play a key pathogenic role in hepatocellular carcinoma (HCC), the most common primary malignancy of the liver and the third leading cause of cancer death. Autophagy is an evolutionarily conserved catabolic process activated to degrade and recycle cell's components. Under stress conditions, such as oxidative stress and nutrient deprivation, autophagy is an essential survival pathway that operates in harmony with other stress response pathways. These include the redox-sensitive transcription complex Nrf2-Keap1 that controls groups of genes with roles in detoxification and antioxidant processes, intermediary metabolism, and cell cycle regulation. Recently, a functional association between a dysfunctional autophagy and Nrf2 pathway activation has been identified in HCC. This appears to occur through the physical interaction of the autophagy adaptor p62 with the Nrf2 inhibitor Keap1, thus leading to increased stabilization and transcriptional activity of Nrf2, a key event in reprogramming metabolic and stress response pathways of proliferating hepatocarcinoma cells. These emerging molecular mechanisms and the therapeutic perspective of targeting Nrf2-p62 interaction in HCC are discussed in this paper along with the prognostic value of autophagy in this type of cancer.
Collapse
|
19
|
Cripto-1 contributes to stemness in hepatocellular carcinoma by stabilizing Dishevelled-3 and activating Wnt/β-catenin pathway. Cell Death Differ 2018; 25:1426-1441. [PMID: 29445127 PMCID: PMC6113239 DOI: 10.1038/s41418-018-0059-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 01/10/2023] Open
Abstract
Identification and characterization of functional molecular targets conferring stemness properties in hepatocellular carcinoma (HCC) offers crucial insights to overcome the major hurdles of tumor recurrence, metastasis and chemoresistance in clinical management. In the current study, we investigated the significance of Cripto-1 in contributing to HCC stemness. Cripto-1 was upregulated in the sorafenib-resistant clones derived from HCC cell lines and patient-derived xenograft that we previously developed, suggesting an association between Cripto-1 and stemness. By in vitro experiments, Cripto-1 fostered cell proliferation, migration, and invasion. It also enhanced self-renewal ability and conferred chemoresistance of HCC cells. Consistently, silencing of Cripto-1 suppressed in vivo tumorigenicity on serial transplantation. On the downstream signaling mechanism, expression of major components of Wnt/β-catenin pathway β-catenin, AXIN2, and C-MYC, accompanied by β-catenin activity was reduced upon Cripto-1 knockdown. The suppressive effects on stemness properties with Cripto-1 knockdown in vitro and in vivo were partially rescued by forced expression of constitutively active β-catenin. Further elucidation revealed the binding of Cripto-1 to Frizzled-7 (FZD7), low-density lipoprotein receptor-related protein 6 (LRP6) and Dishevelled-3 (DVL3) of the Wnt/β-catenin pathway and stabilized DVL3 protein. Analyses with clinical samples validated Cripto-1 overexpression in HCC tissues, as well as a positive correlation between Cripto-1 and AXIN2 expressions. High Cripto-1 level in tumor was associated with poorer disease-free survival of HCC patients. Taken together, Cripto-1 binds to FZD7/LRP6 and DVL3, stabilizes DVL3 expression and activates the Wnt/β-catenin signaling cascade to confer stemness in HCC. Our study findings substantiated the role of Cripto-1 in determining stemness phenotypes of HCC and mechanistically in modulating the Wnt/β-catenin signaling cascade, one of the most frequently deregulated pathways in liver cancer.
Collapse
|
20
|
Damiati S, Peacock M, Leonhardt S, Damiati L, Baghdadi MA, Becker H, Kodzius R, Schuster B. Embedded Disposable Functionalized Electrochemical Biosensor with a 3D-Printed Flow Cell for Detection of Hepatic Oval Cells (HOCs). Genes (Basel) 2018; 9:E89. [PMID: 29443890 PMCID: PMC5852585 DOI: 10.3390/genes9020089] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 01/06/2023] Open
Abstract
Hepatic oval cells (HOCs) are considered the progeny of the intrahepatic stem cells that are found in a small population in the liver after hepatocyte proliferation is inhibited. Due to their small number, isolation and capture of these cells constitute a challenging task for immunosensor technology. This work describes the development of a 3D-printed continuous flow system and exploits disposable screen-printed electrodes for the rapid detection of HOCs that over-express the OV6 marker on their membrane. Multiwall carbon nanotube (MWCNT) electrodes have a chitosan film that serves as a scaffold for the immobilization of oval cell marker antibodies (anti-OV6-Ab), which enhance the sensitivity of the biomarker and makes the designed sensor specific for oval cells. The developed sensor can be easily embedded into the 3D-printed flow cell to allow cells to be exposed continuously to the functionalized surface. The continuous flow is intended to increase capture of most of the target cells in the specimen. Contact angle measurements were performed to characterize the nature and quality of the modified sensor surface, and electrochemical measurements (cyclic voltammetry (CV) and square wave voltammetry (SWV)) were performed to confirm the efficiency and selectivity of the fabricated sensor to detect HOCs. The proposed method is valuable for capturing rare cells and could provide an effective tool for cancer diagnosis and detection.
Collapse
Affiliation(s)
- Samar Damiati
- Department of Biochemistry, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia.
- Institute for Synthetic Bioarchitecture, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria.
| | | | - Stefan Leonhardt
- Institute of Medical and Polymer Engineering, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Laila Damiati
- Centre for Cell Engineering, University of Glasgow, Glasgow G12 8QQ, UK.
- Department of Biology, Jeddah University, Jeddah 23218, Saudi Arabia.
| | - Mohammed A Baghdadi
- Research Centre, King Faisal Specialist Hospital & Research Centre, Jeddah 21499, Saudi Arabia.
| | | | - Rimantas Kodzius
- Mathematics and Natural Sciences Department, The American University of Iraq, Sulaimani, Sulaymaniyah 46001, Iraq.
- Materials Genome Institute, Shanghai University, Shanghai 200444, China.
- Faculty of Medicine, Ludwig Maximilian University of Munich (LMU), 80539 Munich, Germany.
- Faculty of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Bernhard Schuster
- Institute for Synthetic Bioarchitecture, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria.
| |
Collapse
|
21
|
Yang X, Sun D, Dong C, Tian Y, Gao Z, Wang L. Pinin associates with prognosis of hepatocellular carcinoma through promoting cell proliferation and suppressing glucose deprivation-induced apoptosis. Oncotarget 2018; 7:39694-39704. [PMID: 27175589 PMCID: PMC5129963 DOI: 10.18632/oncotarget.9233] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/24/2016] [Indexed: 01/16/2023] Open
Abstract
The roles of Pinin have been well studied in epithelial cell-cell adhesion and RNA alternative splicing, which suggests its involvement in cancer progression. However, little is known about the association between Pinin expression and hepatocellular carcinoma (HCC) tumorigenesis. In this study we report increased expression of Pinin in HCC tissues and cells. Elevated levels of Pinin closely associates with pathological grade and overall survival of patients with hepatocellular carcinoma. Suppression of Pinin expression via lentivirus mediated shRNA knockdown inhibits HCC cell proliferation, colony formation, cell viability, but promotes glucose deprivation (GD)-induced cell apoptosis. On the contrary, overexpression of Pinin reverses these effects observed in Pinin depleted cells. Meanwhile, overexpression of Pinin attenuates GD initiated poly ADP-ribose polymerase (PARP) cleavage and ERK1/2 dephosphorylation, which can be completely blocked with MEK1/2 inhibitor U0126. Therefore, we conclude that Pinin contributes to HCC progression and resistance to GD-induced apoptosis via maintaining ERK1/2 activation and hence may be a potential therapeutic target in hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Xuejun Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116027, China.,Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Deguang Sun
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Chengyong Dong
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yu Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Zhenming Gao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Liming Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
22
|
Leung CON, Mak WN, Kai AKL, Chan KS, Lee TKW, Ng IOL, Lo RCL. Sox9 confers stemness properties in hepatocellular carcinoma through Frizzled-7 mediated Wnt/β-catenin signaling. Oncotarget 2017; 7:29371-86. [PMID: 27105493 PMCID: PMC5045402 DOI: 10.18632/oncotarget.8835] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/29/2016] [Indexed: 01/01/2023] Open
Abstract
Sox9, an SRY-related HMG box transcription factor, is a progenitor/precursor cell marker of the liver expressed during embryogenesis and following liver injury. In this study, we investigated the role of Sox9 and its molecular mechanism with reference to stemness properties in hepatocellular carcinoma (HCC). Here, we observed upregulation of Sox9 in human HCC tissues compared with the non-tumorous liver counterparts (p < 0.001). Upregulation of Sox9 transcript level was associated with poorer tumor cell differentiation (p = 0.003), venous invasion (p = 0.026), advanced tumor stage (p = 0.044) and shorter overall survival (p = 0.042). Transcript levels of Sox9 and CD24 were positively correlated. Silencing of Sox9 in HCC cells inhibited in vitro cell proliferation and tumorsphere formation, sensitized HCC cells to chemotherapeutic agents, and suppressed in vivo tumorigenicity. In addition, knockdown of Sox9 suppressed HCC cell migration, invasion, and in vivo lung metastasis. Further studies showed that Sox9 endowed stemness features through activation of Wnt/β-catenin signaling, which was confirmed by the partial rescue effect on tumorigenicity and self-renewal upon transfection of active β-catenin in Sox9 knockdown cells. By ChIP and luciferase promoter assays, Frizzled-7 was identified to be the direct transcriptional target of Sox9. In conclusion, Sox9 confers stemness properties of HCC through Frizzled-7 mediated Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Carmen Oi-Ning Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Wing-Nga Mak
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Alan Ka-Lun Kai
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Kwan-Shuen Chan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Terence Kin-Wah Lee
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Regina Cheuk-Lam Lo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,State Key Laboratory for Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| |
Collapse
|
23
|
Cui CP, Wong CCL, Kai AKL, Ho DWH, Lau EYT, Tsui YM, Chan LK, Cheung TT, Chok KSH, Chan ACY, Lo RCL, Lee JMF, Lee TKW, Ng IOL. SENP1 promotes hypoxia-induced cancer stemness by HIF-1α deSUMOylation and SENP1/HIF-1α positive feedback loop. Gut 2017; 66:2149-2159. [PMID: 28258134 PMCID: PMC5749365 DOI: 10.1136/gutjnl-2016-313264] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/25/2017] [Accepted: 02/08/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVE We investigated the effect and mechanism of hypoxic microenvironment and hypoxia-inducible factors (HIFs) on hepatocellular carcinoma (HCC) cancer stemness. DESIGN HCC cancer stemness was analysed by self-renewal ability, chemoresistance, expression of stemness-related genes and cancer stem cell (CSC) marker-positive cell population. Specific small ubiquitin-like modifier (SUMO) proteases 1 (SENP1) mRNA level was examined with quantitative PCR in human paired HCCs. Immunoprecipitation was used to examine the binding of proteins and chromatin immunoprecipitation assay to detect the binding of HIFs with hypoxia response element sequence. In vivo characterisation was performed in immunocompromised mice and stem cell frequency was analysed. RESULTS We showed that hypoxia enhanced the stemness of HCC cells and hepatocarcinogenesis through enhancing HIF-1α deSUMOylation by SENP1 and increasing stabilisation and transcriptional activity of HIF-1α. Furthermore, we demonstrated that SENP1 is a direct target of HIF-1/2α and a previously unrecognised positive feedback loop exists between SENP1 and HIF-1α. CONCLUSIONS Taken together, our findings suggest the significance of this positive feedback loop between HIF-1α and SENP1 in contributing to the increased cancer stemness in HCC and hepatocarcinogenesis under hypoxia. Drugs that specifically target SENP1 may offer a potential novel therapeutic approach for HCC.
Collapse
Affiliation(s)
- Chun-Ping Cui
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Carmen Chak-Lui Wong
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Alan Ka-Lun Kai
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Eunice Yuen-Ting Lau
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Yu-Man Tsui
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Lo-Kong Chan
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Tan-To Cheung
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Kenneth Siu-Ho Chok
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Albert C Y Chan
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Regina Cheuk-Lam Lo
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Joyce Man-Fong Lee
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Terence Kin-Wah Lee
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Irene Oi Lin Ng
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
- State Key Laboratory for Liver Research, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| |
Collapse
|
24
|
Wei Y, Wang Y, Gong J, Rao L, Wu Z, Nie T, Shi D, Zhang L. High expression of MAGE-A9 contributes to stemness and malignancy of human hepatocellular carcinoma. Int J Oncol 2017; 52:219-230. [PMID: 29138811 DOI: 10.3892/ijo.2017.4198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/23/2017] [Indexed: 01/30/2023] Open
Abstract
MAGE-A9, a well-characterized cancer testis antigen (CTA), belongs to a member of melanoma antigen gene (MAGE) family. In human malignancies, aberrant expression of MAGE genes correlated with poor clinical prognosis, increased tumor growth, metastases, and enrichment in stem cell populations of certain cancers. Cancer stem cells (CSCs) have been proposed to contribute to the major malignant phenotypes of liver cancer, including recurrence, metastasis and chemoresistance. However, expression and potential role of MAGE-A9 in liver cancer stem cells (LCSCs) still remain unclear. In the present study, we first analyzed the expression profiling of MAGE family genes in EpCAM+ and EpCAM- human hepatocellular carcinoma (HCC), based on public Gene Expression Omnibus (GEO) database. Among these examined MAGE members, MAGE-A9 is the only one with significantly higher expression in EpCAM+ HCC specimens as compared with EpCAM- HCC. Quantitative PCR analysis further confirmed that MAGE-A9 expression significantly elevated in a subtype of HCC patients that had features of hepatic stem/progenitor cells with high-level expression of EpCAM and α-fetoprotein (AFP). Moreover, MAGE-A9 displayed remarkably enriched expression in EpCAM+ HCC cells that were sorted by fluorescence-activated cell sorting and cultured HCC cell spheroids with characteristics of stem/progenitor cells. Functional experiments further revealed that MAGE-A9 overexpression promoted cell proliferation, colony formation, migration, chemoresistance, and tumorigenicity in the context of EpCAM+ HCC cells, whereas MAGE-A9 knockdown significantly inhibited anchorage-dependent and spheroid colony formation and in vivo tumorigenicity. Collectively, these data demonstrate that MAGE-A9 functions as an important regulator of LCSCs, and MAGE-A9 may serve as a potential therapeutic target against HCC stem/progenitor cells.
Collapse
Affiliation(s)
- Youping Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Yanqin Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Jing Gong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Lihua Rao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Zhiwei Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Teng Nie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Dongling Shi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| | - Liming Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330003, P.R. China
| |
Collapse
|
25
|
Flores-Téllez TNJ, Villa-Treviño S, Piña-Vázquez C. Road to stemness in hepatocellular carcinoma. World J Gastroenterol 2017; 23:6750-6776. [PMID: 29085221 PMCID: PMC5645611 DOI: 10.3748/wjg.v23.i37.6750] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/27/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
Carcinogenic process has been proposed to relay on the capacity to induce local tissue damage and proliferative repair. Liver has a great regeneration capacity and currently, most studies point towards the dominant role of hepatocytes in regeneration at all levels of liver damage. The most frequent liver cancer is hepatocellular carcinoma (HCC). Historical findings originally led to the idea that the cell of origin of HCC might be a progenitor cell. However, current linage tracing studies put the progenitor hypothesis of HCC origin into question. In agreement with their dominant role in liver regeneration, mature hepatocytes are emerging as the cell of origin of HCC, although, the specific hepatocyte subpopulation of origin is yet to be determined. The relationship between the cancer cell of origin (CCO) and cancer-propagating cells, known as hepatic cancer stem cell (HCSC) is unknown. It has been challenging to identify the definitive phenotypic marker of HCSC, probably due to the existence of different cancer stem cells (CSC) subpopulations with different functions within HCC. There is a dynamic interconversion among different CSCs, and between CSC and non-CSCs. Because of that, CSC-state is currently defined as a description of a highly adaptable and dynamic intrinsic property of tumor cells, instead of a static subpopulation of a tumor. Altered conditions could trigger the gain of stemness, some of them include: EMT-MET, epigenetics, microenvironment and selective stimulus such as chemotherapy. This CSC heterogeneity and dynamism makes them out reach from therapeutic protocols directed to a single target. A further avenue of research in this line will be to uncover mechanisms that trigger this interconversion of cell populations within tumors and target it.
Collapse
Affiliation(s)
- Teresita NJ Flores-Téllez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| |
Collapse
|
26
|
Scarabel L, Perrone F, Garziera M, Farra R, Grassi M, Musiani F, Russo Spena C, Salis B, De Stefano L, Toffoli G, Rizzolio F, Tonon F, Abrami M, Chiarappa G, Pozzato G, Forte G, Grassi G, Dapas B. Strategies to optimize siRNA delivery to hepatocellular carcinoma cells. Expert Opin Drug Deliv 2017; 14:797-810. [PMID: 28266887 DOI: 10.1080/17425247.2017.1292247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/03/2017] [Indexed: 02/08/2023]
Abstract
hepatocellular carcinoma (hcc) is the predominant form of primary liver cancer and the second leading cause of cancer-associated mortality worldwide. available therapies for hcc have limited efficacy due to often late diagnosis and the general resistance of hcc to anti-cancer agents; therefore, the development of novel therapeutics is urgently required. small-interfering rna (sirna) molecules are short, double-stranded rnas that specifically recognize and bind the mrna of a target gene to inhibit gene expression. despite the great therapeutic potential of sirnas towards many human tumors including hcc, their use is limited by suboptimal delivery. Areas covered: In this review, we outline the current data regarding the therapeutic potential of siRNAs in HCC and describe the development of effective siRNA delivery systems. We detail the key problems associated with siRNA delivery and discuss the possible solutions. Finally, we provide examples of the various siRNA delivery strategies that have been employed in animal models of HCC and in human patients enrolled in clinical trials. Expert opinion: Despite the existing difficulties in siRNA delivery for HCC, the increasing scientific attention and breakthrough studies in this field is facilitating the design of novel and efficient technical solutions that may soon find practical applications.
Collapse
Affiliation(s)
- Lucia Scarabel
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Francesca Perrone
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Marica Garziera
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Rossella Farra
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Mario Grassi
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Francesco Musiani
- d Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Concetta Russo Spena
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Barbara Salis
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Lucia De Stefano
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Flavio Rizzolio
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Federica Tonon
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Michela Abrami
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Gianluca Chiarappa
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Gabriele Pozzato
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Giancarlo Forte
- f Center for Translational Medicine, International Clinical Research Center , St. Anne's University Hospital , Brno , Czech Republic
| | - Gabriele Grassi
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Barbara Dapas
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| |
Collapse
|
27
|
Tong Y, Yang H, Xu X, Ruan J, Liang M, Wu J, Luo B. Effect of a hypoxic microenvironment after radiofrequency ablation on residual hepatocellular cell migration and invasion. Cancer Sci 2017; 108:753-762. [PMID: 28182306 PMCID: PMC5406608 DOI: 10.1111/cas.13191] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/27/2017] [Accepted: 02/04/2017] [Indexed: 12/26/2022] Open
Abstract
Clinical observations have shown that the boundary of tumor ablation is often less than safe border and that the use of radiofrequency ablation (RFA) in the treatment of hepatocellular carcinoma (HCC) may probably accelerate its recurrence and metastasis. RFA can cause the formation of a transition zone between normal liver tissues and necrotic coagulation, where blood stagnation and thrombosis expose residual cancer cells to a hypoxic microenvironment. As the blocked vessels are slowly reperfused, the oxygen supply is gradually restored. Here, HCC cells underwent heat treatment and were cultured under hypoxic conditions to mimic the aforementioned situation, and morphological changes were observed in the surviving cells. Compared with their parental cells, hypoxic HCC cells showed changes that include enhanced invasive, metastatic, and chemoresistant abilities as well as mesenchymal characteristics. There was also a higher percentage of stem-like cells. However, either improving the hypoxic microenvironment or silencing hypoxia inducible factor (HIF)-1α signaling significantly reduced the invasive, metastatic, and chemoresistant potential and reversed the epithelial-mesenchymal transition to varying degrees. Together, these results indicated that a sustained hypoxic microenvironment after RFA may exert a negative impact on the prognosis of HCC patients, and minimizing exposure to a hypoxic microenvironment and targeting HIF-1α signaling might be effective strategies for patients who experience insufficient RFA therapy.
Collapse
Affiliation(s)
- Yuyang Tong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.,Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haiyun Yang
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaolin Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.,Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingliang Ruan
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Liang
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiayi Wu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Lee JH, Hur W, Hong SW, Kim JH, Kim SM, Lee EB, Yoon SK. ELK3 promotes the migration and invasion of liver cancer stem cells by targeting HIF-1α. Oncol Rep 2016; 37:813-822. [PMID: 27959451 DOI: 10.3892/or.2016.5293] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/25/2016] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common solid cancer and the third most common cause of cancer-related mortality. HCC develops via a multistep process associated with genetic aberrations that facilitate HCC invasion and migration and promote metastasis. A growing body of evidence indicates that cancer stem cells (CSCs) are responsible for tumorigenesis, cancer cell invasion and metastasis. Despite the extremely small proportion of cancer cells represented by this subpopulation of HCC cells, CSCs play a key role in cancer metastasis and poor prognosis. ELK3 (Net/SAP-2/Erp) is a transcription factor that is activated by the Ras/extracellular signal-regulated kinase (ERK) signaling pathway. It plays several important roles in various physiological processes, including cell migration, invasion, wound healing, angiogenesis and tumorigenesis. In the present study, we investigated the role of ELK3 in cancer cell invasion and metastasis in CD133+/CD44+ liver cancer stem cells (LCSCs). We isolated LCSCs expressing CD133 and CD44 from Huh7 HCC cells and evaluated their metastatic potential using invasion and migration assays. We found that CD133+/CD44+ cells had increased metastatic potential compared with non-CD133+/CD44+ cells. We also demonstrated that ELK3 expression was upregulated in CD133+/CD44+ cells and that this aberration enhanced cell migration and invasion. In addition, we identified the molecular mechanism by which ELK3 promotes cancer cell migration and invasion. We found that silencing of ELK3 expression in CD133+/CD44+ LCSCs attenuated their metastatic potential by modulating the expression of heat shock-induced factor-1α (HIF-1α). Collectively, the results of the present study demonstrated that ELK3 overexpression promoted metastasis in CD133+/CD44+ cells by regulating HIF-1α expression and that silencing of ELK3 expression attenuated the metastatic potential of CD133+/CD44+ LCSCs. In conclusion, modulation of ELK3 expression may represent a novel therapeutic strategy for preventing HCC metastasis and invasion.
Collapse
Affiliation(s)
- Joon Ho Lee
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Wonhee Hur
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Woo Hong
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jung-Hee Kim
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Min Kim
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun Byul Lee
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
29
|
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) is a multistep process involving the progressive accumulation of molecular alterations pinpointing different molecular and cellular events. The next-generation sequencing technology is facilitating the global and systematic evaluation of molecular landscapes in HCC. There is emerging evidence supporting the importance of cancer metabolism and tumor microenvironment in providing a favorable and supportive niche to expedite HCC development. Moreover, recent studies have identified distinct surface markers of cancer stem cell (CSC) in HCC, and they also put forward the profound involvement of altered signaling pathways and epigenetic modifications in CSCs, in addition to the concomitant drug resistance and metastasis. Taken together, multiple key genetic and non-genetic factors, as well as liver CSCs, result in the development and progression of HCC.
Collapse
Affiliation(s)
- Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Hong Kong, SAR, China,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, SAR, China
| | - Regina Cheuk-Lam Lo
- Department of Pathology, The University of Hong Kong, Hong Kong, SAR, China,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, SAR, China
| | - Lo-Kong Chan
- Department of Pathology, The University of Hong Kong, Hong Kong, SAR, China,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, SAR, China
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, SAR, China,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, SAR, China,*Irene O. L. Ng, MD, PhD, Department of Pathology and State Key Laboratory for Liver Research, The University of Hong Kong, Room 127B, University Pathology Building, Department of Pathology, The University of Hong Kong, Queen Mary, Hospital, Pokfulam, Hong Kong, SAR (China), Tel. +852 2255 3967, E-Mail
| |
Collapse
|
30
|
The Progress and Prospects of Putative Biomarkers for Liver Cancer Stem Cells in Hepatocellular Carcinoma. Stem Cells Int 2016; 2016:7614971. [PMID: 27610139 PMCID: PMC5005617 DOI: 10.1155/2016/7614971] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/06/2016] [Accepted: 07/04/2016] [Indexed: 01/30/2023] Open
Abstract
Accumulating evidence suggests that hepatocellular carcinoma (HCC) is organized by liver cancer stem cells (LCSCs), which are a subset of cells with “stem-like” characteristics. Identification of the LCSCs is a fundamental and important problem in HCC research. LCSCs have been investigated by various stem cell biomarkers. There is still lack of consensus regarding the existence of a “global” marker for LCSCs in HCC. In this review article, we summarize the progress and prospects of putative biomarkers for LCSCs in the past decades, which is essential to develop future therapies targeting CSCs and to predict prognosis and curative effect of these therapies.
Collapse
|
31
|
Li HK, Mai RT, Huang HD, Chou CH, Chang YA, Chang YW, You LR, Chen CM, Lee YHW. DDX3 Represses Stemness by Epigenetically Modulating Tumor-suppressive miRNAs in Hepatocellular Carcinoma. Sci Rep 2016; 6:28637. [PMID: 27344963 PMCID: PMC4921922 DOI: 10.1038/srep28637] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022] Open
Abstract
Studies indicate that the presence of cancer stem cells (CSCs) is responsible for poor prognosis of hepatocellular carcinoma (HCC) patients. In this study, the functional role of DDX3 in regulation of hepatic CSCs was investigated. Our results demonstrated that reduced DDX3 expression was not only inversely associated with tumor grade, but also predicted poor prognosis of HCC patients. Knockdown of DDX3 in HCC cell line HepG2 induced stemness gene signature followed by occurrence of self-renewal, chemoreisistance, EMT, migration as well as CSC expansion, and most importantly, DDX3 knockdown promotes tumorigenesis. Moreover, we found positive correlations between DDX3 level and expressions of tumor-suppressive miR-200b, miR-200c, miR-122 and miR-145, but not miR-10b and miR-519a, implying their involvement in DDX3 knockdown-induced CSC phenotypes. In addition, DDX3 reduction promoted up-regulation of DNA methyltransferase 3A (DNMT3A), while neither DNMT3B nor DNMT1 expression was affected. Enriched DNMT3A binding along with hypermethylation on promoters of these tumor-suppressive miRNAs reflected their transcriptional repressions in DDX3-knockdown cells. Furthermore, individual restoration of these tumor-suppressive miRNAs represses DDX3 knockdown-induced CSC phenotypes. In conclusion, our study suggested that DDX3 prevents generation of CSCs through epigenetically regulating a subset of tumor-suppressive miRNAs expressions, which strengthens tumor suppressor role of DDX3 in HCC.
Collapse
Affiliation(s)
- Hao-Kang Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ru-Tsun Mai
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Institute of Bioinformatics and Systems Biology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chih-Hung Chou
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Institute of Bioinformatics and Systems Biology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yi-An Chang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Yao-Wen Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Ming Chen
- Department of Life Sciences and Institute of Genome Sciences, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yan-Hwa Wu Lee
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
32
|
CXCL3 contributes to CD133(+) CSCs maintenance and forms a positive feedback regulation loop with CD133 in HCC via Erk1/2 phosphorylation. Sci Rep 2016; 6:27426. [PMID: 27255419 PMCID: PMC4891684 DOI: 10.1038/srep27426] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/09/2016] [Indexed: 12/25/2022] Open
Abstract
Although the chemotactic cytokine CXCL3 is thought to play an important role in tumor initiation and invasion, little is known about its function in hepatocellular carcinoma (HCC). In our previous study, we found that Ikaros inhibited CD133 expression via the MAPK pathway in HCC. Here, we showed that Ikaros may indirectly down-regulate CXCL3 expression in HCC cells, which leads to better outcomes in patients with CD133+ cancer stem cell (CSC) populations. CD133 overexpression induced CXCL3 expression, and silencing of CD133 down-regulated CXCL3 in HCC cells. Knockdown of CXCL3 inhibited CD133+ HCC CSCs’ self-renewal and tumorigenesis. The serum CXCL3 level was higher in HCC patients’ samples than that in healthy individual. HCC patients with higher CXCL3 expression displayed a poor prognosis, and a high level of CXCL3 was significantly associated with vascular invasion and tumor capsule formation. Exogenous CXCL3 induced Erk1/2 and ETS1 phosphorylation and promoted CD133 expression, indicating a positive feedback loop between CXCL3 and CD133 gene expression in HCC cells via Erk1/2 activation. Together, our findings indicated that CXCL3 might be a potent therapeutic target for HCC.
Collapse
|
33
|
The Therapeutic Targets of miRNA in Hepatic Cancer Stem Cells. Stem Cells Int 2016; 2016:1065230. [PMID: 27118975 PMCID: PMC4826947 DOI: 10.1155/2016/1065230] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide malignancy and the third leading cause of cancer death in patients. Several studies demonstrated that hepatic cancer stem cells (HCSCs), also called tumor-initiating cells, are involved in regulation of HCC initiation, tumor progression, metastasis development, and drug resistance. Despite the extensive research, the underlying mechanisms by which HCSCs are regulated remain still unclear. MicroRNAs (miRNAs) are able to regulate a lot of biological processes such as self-renewal and pluripotency of HCSCs, representing a new promising strategy for treatment of HCC chemotherapy-resistant tumors. In this review, we synthesize the latest findings on therapeutic regulation of HCSCs by miRNAs, in order to highlight the perspective of novel miRNA-based anticancer therapies for HCC treatment.
Collapse
|
34
|
Zheng LY, Wu L, Lu J, Zou DJ, Huang Q. Expression of Phosphorylated AMP-Activated Protein Kinase Predicts Response to Transarterial Chemoembolization in Postoperative Cases of Hepatocellular Carcinoma. Medicine (Baltimore) 2016; 95:e2908. [PMID: 26986101 PMCID: PMC4839882 DOI: 10.1097/md.0000000000002908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies in the world. Transcatheter arterial chemoembolization (TACE) was commonly used for HCC patients postoperatively. However, the survival benefits of adjuvant TACE were controversial due to the extensive heterogeneity of HCC. Hence, there is a critical need to explore potential biomarkers that can predict the clinical response to TACE. The AMP-activated protein kinase (AMPK) is a highly conserved heterotrimeric serine/threonine kinase that plays a central role in linking metabolism and cancer development. In this study, we aimed at evaluating the association of pAMPKα (Thr172) status with clinical outcomes in HCC patients treated with or without postoperative adjuvant TACE.pAMPKα (Thr172) expression was assessed using immunohistochemical analysis in a cohort of 378 Chinese HCC patients who had undergone tumor resection. Kaplan-Meier analysis and multivariate Cox proportional hazards models were used to study the impact on clinical outcomes.High pAMPKα (Thr172) expression was associated with improved disease-free and overall survival and was an independent prognostic factor for overall survival by multivariate analysis. Furthermore, low pAMPKα (Thr172) expression level was correlated with high percentage of OV6 tumor-initiating cells (T-ICs) in HCC specimens.To our knowledge, it can be demonstrated for the first time that pAMPKα (Thr172) status is associated with response to postoperative adjuvant TACE. High pAMPKα (Thr172) level in HCC may serve as a positive predictor of survival in HCC patients undergoing TACE.
Collapse
Affiliation(s)
- Long-Yi Zheng
- From the Department of Endocrinology, Changhai Hospital (L-YZ, JL, D-JZ, QH), and The First Department of Interventional Radiology, Eastern Hepato-biliary Surgery Hospital, Second Military Medical University (LW), Shanghai, China
| | | | | | | | | |
Collapse
|
35
|
Yang F, Lv LZ, Cai QC, Jiang Y. Potential roles of EZH2, Bmi-1 and miR-203 in cell proliferation and invasion in hepatocellular carcinoma cell line Hep3B. World J Gastroenterol 2015; 21:13268-13276. [PMID: 26715809 PMCID: PMC4679758 DOI: 10.3748/wjg.v21.i47.13268] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 05/03/2015] [Accepted: 07/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the potential roles of enhancer of zeste homolog2 (EZH2), Bmi-1 and miR-203 in cell proliferation and invasion in hepatocellular carcinoma (HCC) cell line Hep3B.
METHODS: A total of 73 patients who underwent surgical resection at Fuzong Clinical Medical College of Fujian Medical University were enrolled in this study. Hep3B cells were cultivated in RPMI 1640 medium supplemented with 10% fetal bovine serum at 37 °C. Vectors that containing cDNA of the EZH2 gene or miR-203 targeted shRNA plasmid were constructed, and then transfected into Hep3B cells. The mRNA expression of miR-203, EZH2, and Bmi-1 was analyzed using quantitative real-time polymerase chain reaction analysis, and the protein levels of EZH2 and Bmi-1 were detected by Western blot analysis. Effect of EZH2 or miR-203 on cell proliferation was observed by methyl thiazolyl tetrazolium assay, and cell apoptosis was assessed using flow cytometry. Besides, effect of EZH2 or miR-203 on tumor cell invasion was detected using Transwell assay.
RESULTS: The mRNA levels of EZH2 and Bmi-1 in HCC tissues and in Hep3B cells were significantly higher compared with those in normal samples (P < 0.01), while miR-203 level was significantly lower in HCC tissues (P < 0.01). Hep3B cells transfected with EZH2-shRNA or miR-203-shRNA showed lower expression levels of EZH2 and Bmi-1 (P < 0.05). Compared with controls, Hep3B cells transfected with EZH2-shRNA had relative slow cell proliferation, indicating that low expression of EZH2 and Bmi-1 and overexpression of miR-203 could inhibit Hep3B cell proliferation (P < 0.05). The average apoptosis rate of Hep3B cells transfected with EZH2-shRNA vector was about 18.631%, while that of Hep3B cells transfected with shRNA vector was about 5.33%, suggesting that EZH2 was down-regulated by transfecting with EZH2-shRNA, and the down-regulated EZH2 contributed to the cell apoptosis. Low expression of EZH2 and Bmi-1 and overexpression of miR-203 could reduce Hep3B cell invasion (P < 0.05).
CONCLUSION: Our study suggests that EZH2 and Bmi-1 are up-regulated while miR-203 is down-regulated in Hep3B cells. MiR-203 may contribute to the metastasis and enhance apoptosis of HCC cells by regulating EZH2 and Bmi-1. Our study may provide a theoretical basis for metastasis of HCC and targeted therapy of HCC.
Collapse
|
36
|
Ganapathy V, Moghe PV, Roth CM. Targeting tumor metastases: Drug delivery mechanisms and technologies. J Control Release 2015; 219:215-223. [PMID: 26409123 PMCID: PMC4745901 DOI: 10.1016/j.jconrel.2015.09.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Primary sites of tumor are the focal triggers of cancers, yet it is the subsequent metastasis events that cause the majority of the morbidity and mortality. Metastatic tumor cells exhibit a phenotype that differs from that of the parent cells, as they represent a resistant, invasive subpopulation of the original tumor, may have acquired additional genetic or epigenetic alterations under exposure to prior chemotherapeutic or radiotherapeutic treatments, and reside in a microenvironment differing from that of its origin. This combination of resistant phenotype and distal location make tracking and treating metastases particularly challenging. In this review, we highlight some of the unique biological traits of metastasis, which in turn, inspire emerging strategies for targeted imaging of metastasized tumors and metastasis-directed delivery of therapeutics.
Collapse
Affiliation(s)
- Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA.
| |
Collapse
|
37
|
A Schiff base derivative for effective treatment of diethylnitrosamine-induced liver cancer in vivo. Anticancer Drugs 2015; 26:555-64. [PMID: 25714251 DOI: 10.1097/cad.0000000000000221] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Hepatocellular carcinoma is one of the most prevalent cancers, with a high morbidity rate, even in developed countries. In the present study, the curative effect of the Schiff base (SB) heterodinuclear copper(II)Mn(II) complex on diethylnitrosamine (DEN)-induced liver carcinoma was investigated. Hepatocarcinoma was initiated by an injection of DEN and promoted by phenobarbital (0.05%) in the diet. In addition, the potential nephrotoxicity of SB was evaluated in a cisplatin-induced nephrotoxicity model. Rats were administered the SB complex (1 and 2 mg/kg body weight/day) for 24 weeks, and cancer progression was investigated by macroscopic, histopathological, and western blot examinations. The administration of SB decreased the incidence and the number of hepatic nodules in a dose-dependent manner by regulating inflammation response and the apoptotic pathway. Western blot analyses from the livers of rats treated with SB after DEN induction showed significantly enhanced Bax and caspase-3 levels, with a marked decrease in the levels of Bcl-2, NF-κB p65 and cyclooxygenase (COX)-2. Results from the nephrotoxicity study showed that, whereas cisplatin increased serum urea nitrogen and creatinine levels, no increase in serum biochemical parameters was detected in SB-treated animals. Moreover, protein levels of NF-E2-related factor-2 (Nrf2) and heme oxygenase-1 were lower, whereas nuclear factor-κB (NF-κB p65) and activator protein-1 levels were higher in the kidneys of cisplatin-treated animals compared with that of the SB groups. Therefore, the SB complex could be an alternative chemotherapeutic option for liver cancer treatment once its safety in clinical applications has been examined.
Collapse
|
38
|
Ouyang Y, Liu K, Hao M, Zheng R, Zhang C, Wu Y, Zhang X, Li N, Zheng J, Chen D. Radiofrequency ablation-increased CXCL10 is associated with earlier recurrence of hepatocellular carcinoma by promoting stemness. Tumour Biol 2015; 37:3697-704. [PMID: 26462839 DOI: 10.1007/s13277-015-4035-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/02/2015] [Indexed: 12/12/2022] Open
Abstract
Radiofrequency ablation (RFA) represents a valuable choice in hepatocellular carcinoma (HCC); however, local recurrence of HCC is common after RFA. Here, 20 primary HCC patients treated by RFA were enrolled. Before (termed 0d) and after RFA treatment for 1 and 7 days (termed 1d and 7d, respectively), plasma and noncancerous tissue were collected. ELISA assay showed that plasma C-X-C motif chemokine 10 (CXCL10) was increased in ten patients (type I patients) but decreased in the other 10 patients (type II patients). The mean interval for HCC recurrence in type I patients was less than the mean interval in type II patients. Interestingly, a significant negative correlation between interval for HCC recurrence and fold change of plasma CXCL10 (1d/0d or 7d/0d) was identified, suggesting that RFA-induced CXCL10 is associated with earlier HCC recurrence. Immunofluorescence assay showed that the receptor of CXCL10, chemokine (C-X-C motif) receptor 3 (CXCR3), was significantly increased in type I, but not type II, patients after RFA. In vitro assay demonstrated that CXCL10 stimulus increased the rate of CD133(+) cancer stem cells (CSCs) in HepG2 cells by binding to CXCR3 and then inducing c-Myc expression. Many studies have reported that induction of CD133(+) CSCs contributes to HCC recurrence. Thus, CXCL10-increased CD133(+) CSCs by activating CXCR3/c-Myc pathway might accelerate HCC recurrence after RFA. These data might have potential implications for HCC therapy.
Collapse
Affiliation(s)
- Yabo Ouyang
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Kai Liu
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Meijun Hao
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China
| | - Rongling Zheng
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Chunmiao Zhang
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Yanning Wu
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Xiaofeng Zhang
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Ning Li
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.,Beijing Institute of Hepatology, Beijing, 100069, China
| | - Jiasheng Zheng
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China.
| | - Dexi Chen
- Beijing You'an Hospital, Capital Medical University, No. 8 XiTouTiao, YouAnMenWai Street, FengTai District, Beijing, 100069, People's Republic of China. .,Beijing Institute of Hepatology, Beijing, 100069, China.
| |
Collapse
|
39
|
Pan QZ, Pan K, Wang QJ, Weng DS, Zhao JJ, Zheng HX, Zhang XF, Jiang SS, Lv L, Tang Y, Li YQ, He J, Liu Q, Chen CL, Zhang HX, Xia JC. Annexin A3 as a potential target for immunotherapy of liver cancer stem-like cells. Stem Cells 2015; 33:354-66. [PMID: 25267273 DOI: 10.1002/stem.1850] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/04/2014] [Indexed: 12/14/2022]
Abstract
Cancer stem-like cells/cancer-initiating cells (CSCs/CICs) are considered to represent a small population of cancer cells that is resistant to conventional cancer treatments and responsible for tumor recurrence and metastasis. The aim of this study was to establish CSC/CIC-targeting immunotherapy. In this study, we found that Annexin A3 (ANXA3) was preferentially expressed in CSCs/CICs derived from hepatocellular carcinoma (HCC) cells compared to non-CSCs/CICs. In HCC samples, high levels of ANXA3 correlated with expansion of CD133(+) tumor cells representing CSCs/CICs in HCC; the combination of high levels of ANXA3 and CD133 was associated with progression of HCC. Overexpression of ANXA3 increased the proportion of CD133(+) cells, enhancing their tumorigenicity. On the contrary, knockdown of ANXA3 decreased CD133(+) cells and inhibited tumorigenicity. The mechanistic study revealed that ANXA3-mediated maintenance of HCC CSCs/CICs activity was likely involved with the HIF1A/Notch pathway. Using ANXA3 as a target, ANXA3-transfected dendritic cells could induce more functionally active T cells and these effector T cells could superiorly kill CD133(+) HCC CSCs/CICs in vitro and in vivo. Taken together, our findings suggest that ANXA3 plays a role in HCC CSC/CIC maintenance, and that ANXA3 may represent a potential CSC/CIC-specific therapeutic target for improving the treatment of HCC.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li Y, Liu D, Zong Y, Qi J, Li B, Liu K, Xiao H. Developmental Stage-Specific Hepatocytes Induce Maturation of HepG2 Cells by Rebuilding the Regulatory Circuit. Mol Med 2015; 21:285-95. [PMID: 25879626 DOI: 10.2119/molmed.2014.00173] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 04/14/2015] [Indexed: 12/19/2022] Open
Abstract
On the basis of their characteristics, we presume that developmental stage-specific hepatocytes should have the ability to induce maturation of hepatoma cells. A regulatory circuit formed by hepatocyte nuclear factor (HNF)-4α, HNF-1α, HNF-6 and the upstream stimulatory factor (USF-1) play a key role in the maturation of embryonic hepatocytes; however, it is unclear whether the regulatory circuit mediates the embryonic induction of hepatoma cell maturation. In this study, 12.5-d to 15.5-d mouse embryonic hepatocytes or their medium were used to coculture or treat HepG2 cells, and the induced maturation was evaluated in vitro and in vivo. In the induced HepG2 cells, the components of the regulatory circuit were detected, their cross-regulation was evaluated and HNF-4α RNA interference was performed. We found that 13.5-d to 14.5-d embryonic hepatocytes could induce HepG2 cell maturation, demonstrated by morphological changes, increased maturation markers and decreased c-Myc and α-fetoprotein (AFP) in vitro. The majority of HepG2 tumors were eliminated by 13.5-d embryonic induction in vivo. All components of the regulatory circuit were upregulated and the binding of HNF-4α, HNF-1α, HNF-6 and USF-1 to their target sites was promoted to rebuild the regulatory circuit in the induced HepG2 cells. Moreover, RNA interference targeting HNF-4α, which is the core of the regulatory circuit, attenuated the induced maturation of HepG2 cells with downregulation of the regulatory circuit. These results revealed that developmental stage-specific hepatocytes could induce the maturation of HepG2 cells by rebuilding the regulatory circuit.
Collapse
Affiliation(s)
- Yanning Li
- Department of Molecular Biology, Hebei Key Laboratory of Laboratory Animal, Hebei Medical University, Shijiazhuang, China
| | - Demei Liu
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Yanhong Zong
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Jinsheng Qi
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Bin Li
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Kun Liu
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Hui Xiao
- Department of Biochemistry, Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
41
|
Huang H, Hu M, Li P, Lu C, Li M. Mir-152 inhibits cell proliferation and colony formation of CD133(+) liver cancer stem cells by targeting KIT. Tumour Biol 2015; 36:921-928. [PMID: 25311946 DOI: 10.1007/s13277-014-2719-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/07/2014] [Indexed: 02/07/2023] Open
Abstract
miR152 is involved in diverse biological functions and development of disease. This study investigates the role of mir-152 in cell proliferation and colony formation of liver cancer stem cells. We show that exogenous overexpression of mir-152 suppresses cell proliferation and colony formation in CD133(+) hep3B cells. We also show that KIT is a direct target of miR-152 and miR-152 downregulates protein expression of KIT by directly binding to 3' untranslated region of KIT. Downregulation of KIT by specific siRNAs inhibits proliferation and colony formation of CD133(+) hep3B cells, which is similar to inhibitory effects of miR-152. Moreover, exogenous expression of KIT compromises inhibitory effects of miR-152 on cell proliferation and colony formation. Our findings suggest that mir-152 inhibits cell proliferation and colony formation of CD133(+) hep3B cells by targeting KIT.
Collapse
Affiliation(s)
- Haili Huang
- Clinical Research Center, the Affiliated Hospital of Guangdong Medical College, 524001, Zhanjiang, China
| | | | | | | | | |
Collapse
|
42
|
Phenotypic screening in cancer drug discovery - past, present and future. Nat Rev Drug Discov 2014; 13:588-602. [PMID: 25033736 DOI: 10.1038/nrd4366] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been a resurgence of interest in the use of phenotypic screens in drug discovery as an alternative to target-focused approaches. Given that oncology is currently the most active therapeutic area, and also one in which target-focused approaches have been particularly prominent in the past two decades, we investigated the contribution of phenotypic assays to oncology drug discovery by analysing the origins of all new small-molecule cancer drugs approved by the US Food and Drug Administration (FDA) over the past 15 years and those currently in clinical development. Although the majority of these drugs originated from target-based discovery, we identified a significant number whose discovery depended on phenotypic screening approaches. We postulate that the contribution of phenotypic screening to cancer drug discovery has been hampered by a reliance on 'classical' nonspecific drug effects such as cytotoxicity and mitotic arrest, exacerbated by a paucity of mechanistically defined cellular models for therapeutically translatable cancer phenotypes. However, technical and biological advances that enable such mechanistically informed phenotypic models have the potential to empower phenotypic drug discovery in oncology.
Collapse
|
43
|
Taniguchi K, Karin M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin Immunol 2014; 26:54-74. [PMID: 24552665 DOI: 10.1016/j.smim.2014.01.001] [Citation(s) in RCA: 519] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/06/2014] [Indexed: 11/17/2022]
Abstract
Inflammatory responses play pivotal roles in cancer development, including tumor initiation, promotion, progression, and metastasis. Cytokines are now recognized as important mediators linking inflammation and cancer, and are therefore potential therapeutic and preventive targets as well as prognostic factors. The interleukin (IL)-6 family of cytokines, especially IL-6 and IL-11, is highly up-regulated in many cancers and considered as one of the most important cytokine families during tumorigenesis and metastasis. This review discusses molecular mechanisms linking the IL-6 cytokine family to solid malignancies and their treatment.
Collapse
Affiliation(s)
- Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; UC San Diego Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Ding J, Wang H. Multiple interactive factors in hepatocarcinogenesis. Cancer Lett 2013; 346:17-23. [PMID: 24374016 DOI: 10.1016/j.canlet.2013.12.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/08/2013] [Accepted: 12/10/2013] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer and the third most frequent cause of cancer mortality globally. Each year there are approximately 630,000 new cases of HCC in the world and more than half of the new cases occur in China. Major risk factors of HCC include HBV or HCV infection, alcoholic liver disease, and nonalcoholic fatty liver disease. Most of these risk factors lead to chronic hepatitis and cirrhosis, which is present in 80-90% of HCC patients. Hepatocarcinogenesis has been regarded as a multi-stage process involving multiple genetic or environmental factors. Interaction and cross-regulation of distinct factors synergistically contributes to HCC occurrence. A comprehensive knowledge on the multiple factors and their interaction in hepatocarcinogenesis is necessary to improve the effectiveness of HCC intervention. In this review, we will focus on the recent progress made in understanding the mechanisms of hepatocarcinogenesis and discuss some potential issues or challenges in this area.
Collapse
Affiliation(s)
- Jin Ding
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China.
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China.
| |
Collapse
|
45
|
Wilson GS, Hu Z, Duan W, Tian A, Wang XM, McLeod D, Lam V, George J, Qiao L. Efficacy of using cancer stem cell markers in isolating and characterizing liver cancer stem cells. Stem Cells Dev 2013; 22:2655-2664. [PMID: 23638793 PMCID: PMC3780293 DOI: 10.1089/scd.2012.0703] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/02/2013] [Indexed: 12/23/2022] Open
Abstract
Recent evidence suggests that a subset of hepatocellular carcinomas (HCCs) are derived from liver cancer stem cells (LCSCs). In order to isolate and characterize LCSCs, reliable markers that are specific to these cells are required. We evaluated the efficacy of a range of cancer stem cell (CSC) markers in isolating and characterizing LCSCs. We show that the most widely used CSC markers are not specific to LCSCs. By western analysis, protein expression of the common markers showed no significant difference between HCC tumor tissues and adjacent non-cancerous liver. Further, isolation of LCSCs from common HCC cell lines using FACScan and microbeads showed no consistent marker expression pattern. We also show that LCSCs have unique subtypes. Immunohistochemistry of HCC tissues showed that different HCCs express unique combinations of LCSC markers. Quantitative real-time polymerase chain reaction analysis showed that LCSCs isolated using different markers in the same HCC phenotype had different expression profiles. Likewise, LCSCs isolated from different HCC phenotypes with the same marker also had unique expression profiles and displayed varying resistance profiles to Sorafenib. Thus, using a range of commonly used CSC markers in HCCs and cell lines, we demonstrate that currently available markers are not specific for LCSCs. LCSCs have unique subtypes that express distinctive combinations of LCSC markers and altered drug resistance profiles, making their identification problematic.
Collapse
Affiliation(s)
- George S. Wilson
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney and Westmead Hospital, Westmead, Australia
| | - Zenan Hu
- Division of Gastroenterology and Hepatology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Australia
| | - Aiping Tian
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney and Westmead Hospital, Westmead, Australia
- Division of Gastroenterology and Hepatology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xin M. Wang
- Westmead Millennium Institute for Medical Research, Flow Cytometry Centre, University of Sydney, Westmead, Australia
| | - Duncan McLeod
- Department of Tissue Pathology and Diagnostic Oncology, Institute for Clinical Pathology and Medical Research, Westmead Hospital, Westmead, Australia
| | - Vincent Lam
- Department of Surgery, The University of Sydney, Westmead Hospital, Westmead, Australia
| | - Jacob George
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney and Westmead Hospital, Westmead, Australia
| | - Liang Qiao
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney and Westmead Hospital, Westmead, Australia
| |
Collapse
|
46
|
Callegari E, Elamin BK, Sabbioni S, Gramantieri L, Negrini M. Role of microRNAs in hepatocellular carcinoma: a clinical perspective. Onco Targets Ther 2013; 6:1167-78. [PMID: 24039437 PMCID: PMC3770717 DOI: 10.2147/ott.s36161] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly tumors, and current treatments for the disease are often ineffective. The discovery of the involvement of microRNAs (miRNAs) in hepatocarcinogenesis represents an important area of investigation for the development of their clinical applications. These molecules may act as oncogenes or tumor suppressors by directly or indirectly controlling the expression of key proteins involved in cancer-associated pathways. On the clinical side, because of their tumor-specific expression and stability in tissues and in the circulation, miRNAs have been proposed as novel diagnostic tools for classification and prognostic stratification of HCC. In recent years, the therapeutic potential of miRNAs has been demonstrated in various preclinical studies. Anti-miRNA oligonucleotides and miRNA mimics have been found to have antitumor activity. Moreover, by exploiting tumor-specific expression of miRNA, efforts have been aimed at improving targeting of tumor cells by replicative oncolytic viruses while sparing normal cells. These areas are expected to be explored further in the upcoming years to assess the clinical value of miRNA-based approaches in HCC and cancer in general.
Collapse
Affiliation(s)
- Elisa Callegari
- Dipartimento di Morfologia, Chirurgia e Medicina Sperimentale, Università di Ferrara, Ferrara, Italy
| | | | | | | | | |
Collapse
|
47
|
Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med 2013; 19:998-1004. [PMID: 23921754 DOI: 10.1038/nm.3267] [Citation(s) in RCA: 504] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 05/22/2013] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (PSCs) are a leading candidate for cell-based therapies because of their capacity for unlimited self renewal and pluripotent differentiation. These advances have recently culminated in the first-in-human PSC clinical trials by Geron, Advanced Cell Technology and the Kobe Center for Developmental Biology for the treatment of spinal cord injury and macular degeneration. Despite their therapeutic promise, a crucial hurdle for the clinical implementation of human PSCs is their potential to form tumors in vivo. In this Perspective, we present an overview of the mechanisms underlying the tumorigenic risk of human PSC-based therapies and discuss current advances in addressing these challenges.
Collapse
Affiliation(s)
- Andrew S Lee
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Medicine, Division of Cardiology Stanford University School of Medicine, Stanford, California 94305, USA
| | - Chad Tang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Mahendra S Rao
- National Center for Regenerative Medicine, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C Wu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Medicine, Division of Cardiology Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
48
|
Lo RCL, Ng IOL. Hepatic progenitor cells: their role and functional significance in the new classification of primary liver cancers. Liver Cancer 2013; 2:84-92. [PMID: 24159600 PMCID: PMC3740719 DOI: 10.1159/000343844] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Hepatic progenitor cells (HPCs) are bipotential cells residing in normal liver. Their proliferation is observed in reactive conditions of the liver and in primary liver cancers. The observation that some hepatocellular carcinomas (HCCs) express a biliary-like immunophenotype has led to the identification of HPCs in HCC. Accumulating evidence suggests that HPCs play a role as the cell of origin in a variety of primary liver cancers. This has led to the development of revolutionary concepts in hepatocarcinogenesis. In this article, the role and significance of HPCs in HCC, including its classification, are summarized and discussed.
Collapse
Affiliation(s)
| | - Irene Oi-lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, SAR, China,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, SAR, China,*Departm ent of Pathology and State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong, SAR (China), Tel +852 2255 3967, E-Mail
| |
Collapse
|
49
|
Uehara T, Ainslie GR, Kutanzi K, Pogribny IP, Muskhelishvili L, Izawa T, Yamate J, Kosyk O, Shymonyak S, Bradford BU, Boorman GA, Bataller R, Rusyn I. Molecular mechanisms of fibrosis-associated promotion of liver carcinogenesis. Toxicol Sci 2013; 132:53-63. [PMID: 23288052 DOI: 10.1093/toxsci/kfs342] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) mostly develops in patients with advanced fibrosis; however, the mechanisms of interaction between a genotoxic insult and fibrogenesis are not well understood. This study tested a hypothesis that fibrosis promotes HCC via a mechanism that involves activation of liver stem cells. First, B6C3F1 mice were administered diethylnitrosamine (DEN; single ip injection of 1mg/kg at 14 days of age). Second, carbon tetrachloride (CCl(4); 0.2ml/kg, 2/week ip starting at 8 weeks of age) was administered for 9 or 14 weeks to develop advanced liver fibrosis. In animals treated with DEN as neonates, presence of liver fibrosis led to more than doubling (to 100%) of the liver tumor incidence as early as 5 months of age. This effect was associated with activation of cells with progenitor features in noncancerous liver tissue, including markers of replicative senescence (p16), oncofetal transformation (Afp, H19, and Bex1), and increased "stemness" (Prom1 and Epcam). In contrast, the dose of DEN used did not modify the extent of liver inflammation, fibrogenesis, oxidative stress, proliferation, or apoptosis induced by subchronic CCl(4) administration. This study demonstrates the potential role of liver stem-like cells in the mechanisms of chemical-induced, fibrosis-promoted HCC. We posit that the combination of genotoxic and fibrogenic insults is a sensible approach to model liver carcinogenesis in experimental animals. These results may contribute to identification of cirrhotic patients predisposed to HCC by analyzing the expression of hepatic progenitor cell markers in the noncancerous liver tissue.
Collapse
Affiliation(s)
- Takeki Uehara
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|