1
|
Chen J, Wang Z, Yi M, Yang Y, Tian M, Liu Y, Wang G, Shen H. Regenerative properties of bone marrow mesenchymal stem cell derived exosomes in rotator cuff tears. J Transl Med 2025; 23:47. [PMID: 39800717 PMCID: PMC11727793 DOI: 10.1186/s12967-024-06029-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRCT Rotator cuff injury (RCI), characterized by shoulder pain and restricted mobility, represents a subset of tendon-bone insertion injuries (TBI). In the majority of cases, surgical reconstruction of the affected tendons or ligaments is required to address the damage. However, numerous clinical failures have underscored the suboptimal outcomes associated with such procedures. Further investigations have revealed that these failures are largely attributable to delayed healing at the tendon-bone interface, excessive formation of vascularized scar tissue, and inadequate integration of tendon grafts within bone tunnels. As a result, the healing process of rotator cuff injuries faces significant challenges.Bone marrow-derived mesenchymal stem cell exosomes (BMSC-exos) have emerged as a prominent focus of research within the field of bioengineering, owing to their remarkable potential to regulate cellular proliferation and differentiation, modulate immune responses, and facilitate tissue repair and regeneration following cellular damage. In this review, we explore the anti-inflammatory, angiogenic, anti-scarring, and bone metabolism-modulating effects of BMSC-exos in the context of rotator cuff injury. Additionally, we address the limitations and ongoing challenges within current research, offering insights that could guide the clinical application of BMSC-exos in the treatment of rotator cuff injuries in the future.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zihe Wang
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming Yi
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yi Yang
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mengzhao Tian
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yinqi Liu
- School of Materials and Energy, Southwest University, Southwest University Hospital, Chongqing, China.
| | - Guoyou Wang
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Huarui Shen
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Chen Z, Gao Y, Zhang P, Liu Y, Wei B, Chen L, Xi H. Identification of gastric cancer stem cells with CD44 and Lgr5 double labelling and their initial roles on gastric cancer malignancy and chemotherapy resistance. Cell Biol Toxicol 2024; 41:12. [PMID: 39707072 PMCID: PMC11662044 DOI: 10.1007/s10565-024-09960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Accumulating evidences have indicated that cancer stem cells (CSCs) can initiate tumor progression and cause recurrence after therapy. However, specific markers of gastric CSCs (GCSCs) from different origins have not been comprehensively revealed. Here, we further detected whether cell populations labelled with CD44 and Lgr5, well-recognized stem markers for gastric cancer (GC), can better emphasize cancer initiation, therapeutic resistance and recurrence. Flow cytometry was utilized to sort the CD44 + Lgr5 + and CD44 + Lgr5- cells from GC cell line HGC-27 and primary GC cells. The influences of CD44 and Lgr5 GCSCs on the malignant behaviors and their potential mechanisms was investigated, respectively. In our study, we reported the identification and validation of CD44 + Lgr5 + cells that presented stronger stemness characteristics, as evidenced by increase of sphere forming ability, elevation of stem cell transcriptional activity. Additionally, CD44 + Lgr5 + double positive cells have lower apoptosis, greater chemotherapy resistance, and higher EMT capacity and LC3 density compared with CD44 + Lgr5- cells. Tumor xenograft experiments also verified the faster carcinogenesis of CD44 + Lgr5 + GCSCs. Furthermore, a series of key proteins in the Wnt, Hedgehog, Notch, and TGF-β pathways were elevated in the CD44 + Lgr5 + double positive subpopulation, except for Notch 1 and Smad 1. In conclusion, the binding of CD44 and Lgr5 can serve as a precise GCSCs marker that initiate malignant progression and chemotherapy resistance in GC by activating Wnt, Hedgehog, Notch, TGF-β pathways. Those evidences raise the needs to target both markers simultaneously as a potential approach for the GC treatment.
Collapse
Affiliation(s)
- Zhida Chen
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China
- PLA School of Medicine, Beijing, 100853, China
| | - Yunhe Gao
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China
| | - Pengfei Zhang
- PLA School of Medicine, Beijing, 100853, China
- Department of Oncology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yi Liu
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China
- PLA School of Medicine, Beijing, 100853, China
| | - Bo Wei
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China
| | - Lin Chen
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China.
| | - Hongqing Xi
- Department of General Surgery, First Medical Center of Chinese, PLA General Hospital, Haidian District, No.28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
3
|
Wang MY, Wang XW, Zhao WX, Li Y, Cai ML, Wang KX, Xi XM, Zhao C, Zhou HM, Shao RG, Xia GM, Zhang YF, Zhao WL. Enhanced binding of β-catenin and β-TrCP mediates LMPt's anti-CSCs activity in colorectal cancer. Biochem Pharmacol 2023; 212:115577. [PMID: 37137416 DOI: 10.1016/j.bcp.2023.115577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells with the features of self-renewal, tumor initiation, and insensitivity to common physical and chemical agents, are the key to cancer relapses, metastasis, and resistance. Accessible CSCs inhibitory strategies are primarily based on small molecule drugs, yet toxicity limits their application. Here, we report a liposome loaded with low toxicity and high effectiveness of miriplatin, lipo-miriplatin (LMPt) with high miriplatin loading, and robust stability, exhibiting a superior inhibitory effect on CSCs and non-CSCs. LMPt predominantly inhibits the survival of oxaliplatin-resistant (OXA-resistant) cells composed of CSCs. Furthermore, LMPt directly blocks stemness features of self-renewal, tumor initiation, unlimited proliferation, metastasis, and insensitivity. In mechanistic exploration, RNA sequencing (RNA-seq) revealed that LMPt downregulates the levels of pro-stemness proteins and that the β-catenin-mediated stemness pathway is enriched. Further research shows that either in adherent cells or 3D-spheres, the β-catenin-OCT4/NANOG axis, the vital pathway to maintain stemness, is depressed by LMPt. The consecutive activation of the β-catenin pathway induced by mutant β-catenin (S33Y) and OCT4/NANOG overexpression restores LMPt's anti-CSCs effect, elucidating the key role of the β-catenin-OCT4/NANOG axis. Further studies revealed that the strengthened binding of β-catenin and β-TrCP initiates ubiquitination and degradation of β-catenin induced by LMPt. In addition,the ApcMin/+transgenicmouse model, in which colon tumors are spontaneously formed, demonstrates LMPt's potent anti-non-CSCs activity in vivo.
Collapse
Affiliation(s)
- Meng-Yan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Xiao-Wei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Wen-Xia Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Yang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Mei-Lian Cai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Ke-Xin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Xiao-Ming Xi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Cong Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Hui-Min Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China
| | - Rong-Guang Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| | - Gui-Min Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| | - Ye-Fan Zhang
- Department of Hepatobiliary Surgery/National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Wu-Li Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences/Peking Union Medical College, Tiantanxili #1, Beijing 100050, P.R. China.
| |
Collapse
|
4
|
Pang L, Yan X, Su D, Wu X, Jiang H. Feasibility of olfactomedin 4 as a molecular biomarker for early diagnosis of gastric neoplasia after intestinal metaplasia. Scand J Gastroenterol 2023; 58:133-141. [PMID: 36124708 DOI: 10.1080/00365521.2022.2116992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES This study discusses whether olfactomedin 4 (OLFM4) could be used as a sensitive and specific biomarker in the early diagnosis of gastric cancer (GC) after gastric intestinal metaplasia (GIM). METHODS An integrative analysis combining data derived from the Gene Expression Omnibus (GEO) and cBioPortal databases was performed to investigate the potential molecular biomarker. Immunohistochemistry and quantitative real-time polymerase chain reactions were used to measure the expression of messenger ribonucleic acid (mRNA) and protein by OLFM4. In combination with the gastroscopic findings and the OLFM4 expression in GIM-GC, a predictive model was established. The receiver operator characteristic curve (ROC) was applied to assess the diagnostic value of the model for GIM-GC. RESULTS According to the GEO and cBioPortal databases, OLFM4 was identified as a key gene in the diagnosis of GIM-GC. Higher protein expression of OLFM4 was found in GIM and GIM-GC compared with chronic superficial gastritis (GS) (p < 0.05). The positive expression rate of OLFM4 in paracancerous tissue (GCP) was higher than in GIM (p > 0.05). There was no significant difference between GIM-GC and GCP (p > 0.05). The mRNA expression of OLFM4 was similar to the protein expression, and the positive expression rate was higher in early GIM-GC than in GIM (p < 0.05). CONCLUSION Olfactomedin 4 could be used as a biomarker for the early diagnosis of GIM-GC, and the logistic predictive model could be an effective tool for increasing the early diagnostic rate.
Collapse
Affiliation(s)
- Lixing Pang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xin Yan
- Department of Endocrinology, Nanning Second People's Hospital, Nanning, China
| | - Dongxing Su
- Department of Gastroenterology, Nanning Second People's Hospital, Nanning, China
| | - Xianbin Wu
- Department of Gastroenterology, Nanning Second People's Hospital, Nanning, China
| | - Haixing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Li X, Dong H, Chen L, Wang Y, Hao Z, Zhang Y, Jiao Y, Zhao Z, Peng X, Zhan X. Identification of N7-methylguanosine related subtypes and construction of prognostic model in gastric cancer. Front Immunol 2022; 13:984149. [PMID: 36300121 PMCID: PMC9589367 DOI: 10.3389/fimmu.2022.984149] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background N7-methylguanosine (m7G), one of the most common post-transcriptional modifications, can be present in tRNA, mRNA, and miRNA to mediate the progression of various tumors. However, the possible role of m7G in gastric cancer (GC) is still unknown. Materials and Methods In this study, SNVs (single nucleotide variations), CNVs (copy number variations), and methylation of m7G-related genes (m7GRGs) were analyzed. The relationship between them and the expression of m7GRGs and prognosis of GC patients was explored. Based on 13 prognostic-related m7GRGs, 567 GC samples were classified into three subtypes using the ConsensusClusterPlus package. we compared survival status, clinical traits, immune cell infiltration, immune checkpoints, tumor microenvironment (TME), tumor immune dysfunction and exclusion (TIDE), and potential biological pathways among the three subtypes. Then, patients were again grouped into different genetic subtypes based on the DEGs among the three subtypes. In addition, a prognostic m7GRG_Score was constructed using five risk genes applicable to patients of any age, gender and stage. We also assessed tumor mutational burden (TMB), microsatellite instability (MSI), cancer stem cell (CSC) index, sensitivity of antineoplastic drugs, efficacy of anti-PD-1 and anti-CTLA4 immunotherapy between high and low m7GRG_Score groups. Finally, we established a nomogram based on m7GRG_Score and tumor stage to enhance the clinical application of the model. miRNAs and lncRNAs that could regulate expression of risk genes were searched. Results SNVs, CNVs, and methylation of m7GRGs were associated with m7GRGs expression. However, they did not significantly affect the survival of GC patients. Our results also confirmed that patients in subtypes B and C and low m7GRG_Score groups had longer survival time, better clinical stage, more immune cell infiltration, fewer immune escape and dysfunction compared to subtype A and high m7GRG_Score groups. A low m7GRG_score was featured with increased microsatellite instability-high (MSI-H), TMB, and efficacy of immunotherapy. Conclusion The m7GRG_Score model may become a beneficial tool for predicting prognosis and guiding personalized treatment in GC patients. These findings will improve our knowledge of m7G in GC and provide new methods for more effective treatment strategies.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Hao Dong
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ling Chen
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yujie Wang
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yuan Jiao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Zhiyue Zhao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| |
Collapse
|
6
|
Shi Z, Zhu C, Zhang Y, Wang Y, Hou W, Li X, Lu J, Guo X, Xu F, Jiang X, Wang Y, Liu J, Jin M. Deep learning for automatic diagnosis of gastric dysplasia using whole-slide histopathology images in endoscopic specimens. Gastric Cancer 2022; 25:751-760. [PMID: 35394573 DOI: 10.1007/s10120-022-01294-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Distinguishing gastric epithelial regeneration change from dysplasia and histopathological diagnosis of dysplasia is subject to interobserver disagreement in endoscopic specimens. In this study, we developed a method to distinguish gastric epithelial regeneration change from dysplasia and further subclassify dysplasia. Meanwhile, optimized the cross-hospital diagnosis using domain adaption (DA). METHODS 897 whole slide images (WSIs) of endoscopic specimens from two hospitals were divided into training, internal validation, and external validation cohorts. We developed a deep learning (DL) with DA (DLDA) model to classify gastric dysplasia and epithelial regeneration change into three categories: negative for dysplasia (NFD), low-grade dysplasia (LGD), and high-grade dysplasia (HGD)/intramucosal invasion neoplasia (IMN). The diagnosis based on the DLDA model was compared to 12 pathologists using 100 gastric biopsy cases. RESULTS In the internal validation cohort, the diagnostic performance measured by the macro-averaged area under the receiver operating characteristic curve (AUC) was 0.97. In the independent external validation cohort, our DLDA models increased macro-averaged AUC from 0.67 to 0.82. In terms of the NFD and HGD cases, our model's diagnostic sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were significantly higher than junior and senior pathologists. Our model's diagnostic sensitivity, NPV, was higher than specialist pathologists. CONCLUSIONS We demonstrated that our DLDA model could distinguish gastric epithelial regeneration change from dysplasia and further subclassify dysplasia in endoscopic specimens. Meanwhile, achieved significant improvement of diagnosis cross-hospital.
Collapse
Affiliation(s)
- Zhongyue Shi
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Chuang Zhu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yu Zhang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yakun Wang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Weihua Hou
- Department of Pathology, PLA Joint Logistics Support Force 989 Hospital (Formerly, the 152 Central Hospital), Henan, China
| | - Xue Li
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jun Lu
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xinmeng Guo
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Feng Xu
- Department of Breast Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xingran Jiang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ying Wang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jun Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Mulan Jin
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
7
|
Mohammadi A, Khanbabaei H, Zandi F, Ahmadi A, Haftcheshmeh SM, Johnston TP, Sahebkar A. Curcumin: A therapeutic strategy for targeting the Helicobacter pylori-related diseases. Microb Pathog 2022; 166:105552. [DOI: 10.1016/j.micpath.2022.105552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022]
|
8
|
Wang MY, Qiu YH, Cai ML, Zhang CH, Wang XW, Liu H, Chen Y, Zhao WL, Liu JB, Shao RG. Role and molecular mechanism of stem cells in colorectal cancer initiation. J Drug Target 2019; 28:1-10. [PMID: 31244351 DOI: 10.1080/1061186x.2019.1632317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, the rate of colorectal cancer has sharply increased, especially in China, where it ranks second for the number of cancer fatalities. Currently, the treatment of colorectal cancer patients involves the combination of resection surgery and treatment with postoperative anticancer drugs such as 5-FU and oxaliplatin. However, recurrence and metastasis after treatment are still the dominant reasons for the low survival rate. Colorectal cancer stem cells (CSCs) are regarded as the key contributors to tumour recurrence and metastasis due to their resistance to chemotherapy drugs and their extremely high tumourigenicity. Once CSCs overcome chemotherapy treatment, they continue to survive and reinitiate proliferation to form tumours, leading to recurrence. The dominant reason for CSC resistance is that most anticancer drugs are aimed at inhibiting proliferative pathways in cancer cells that differ from those in CSCs. Therefore, studies on the characteristics of CSCs and their intracellular molecular pathways are essential for the exploration of CSC-targeted drugs. In this report, we review recent advances in the research of CSCs and, in particular, review the important intracellular molecular pathways, such as HOXA5-catenin, STRAP-NOTCH and YAP/TAZ, related to the maintenance and differentiation of stem cells to generate a theoretical basis for the exploration of CSC-targeted drugs.
Collapse
Affiliation(s)
- Meng-Yan Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yu-Han Qiu
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Mei-Lian Cai
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Cong-Hui Zhang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Wei Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Liu
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yi- Chen
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing, China
| | - Wu-Li Zhao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jing-Bo Liu
- Department of Urology, Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Rong-Guang Shao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Gao JP, Xu W, Liu WT, Yan M, Zhu ZG. Tumor heterogeneity of gastric cancer: From the perspective of tumor-initiating cell. World J Gastroenterol 2018; 24:2567-2581. [PMID: 29962814 PMCID: PMC6021770 DOI: 10.3748/wjg.v24.i24.2567] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/30/2018] [Accepted: 05/26/2018] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) remains one of the most common and malignant types of cancer due to its rapid progression, distant metastasis, and resistance to conventional chemotherapy, although efforts have been made to understand the underlying mechanism of this resistance and to improve clinical outcome. It is well recognized that tumor heterogeneity, a fundamental feature of malignancy, plays an essential role in the cancer development and chemoresistance. The model of tumor-initiating cell (TIC) has been proposed to explain the genetic, histological, and phenotypical heterogeneity of GC. TIC accounts for a minor subpopulation of tumor cells with key characteristics including high tumorigenicity, maintenance of self-renewal potential, giving rise to both tumorigenic and non-tumorigenic cancer cells, and resistance to chemotherapy. Regarding tumor-initiating cell of GC (GATIC), substantial studies have been performed to (1) identify the putative specific cell markers for purification and functional validation of GATICs; (2) trace the origin of GATICs; and (3) decode the regulatory mechanism of GATICs. Furthermore, recent studies demonstrate the plasticity of GATIC and the interaction between GATIC and its surrounding factors (TIC niche or tumor microenvironment). All these investigations pave the way for the development of GATIC-targeted therapy, which is in the phase of preclinical studies and clinical trials. Here, we interpret the heterogeneity of GC from the perspectives of TIC by reviewing the above-mentioned fundamental and clinical studies of GATICs. Problems encountered during the GATIC investigations and the potential solutions are also discussed.
Collapse
Affiliation(s)
- Jian-Peng Gao
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Wei Xu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Wen-Tao Liu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Min Yan
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Zheng-Gang Zhu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
10
|
Waldum HL, Öberg K, Sørdal ØF, Sandvik AK, Gustafsson BI, Mjønes P, Fossmark R. Not only stem cells, but also mature cells, particularly neuroendocrine cells, may develop into tumours: time for a paradigm shift. Therap Adv Gastroenterol 2018; 11:1756284818775054. [PMID: 29872453 PMCID: PMC5974566 DOI: 10.1177/1756284818775054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 04/03/2018] [Indexed: 02/04/2023] Open
Abstract
Stem cells are considered the origin of neoplasms in general, and malignant tumours in particular, and the stage at which the stem cells stop their differentiation determines the degree of malignancy. However, there is increasing evidence supporting an alternative paradigm. Tumours may develop by dedifferentiation from mature cells able to proliferate. Studies of gastric carcinogenesis demonstrate that mature neuroendocrine (NE) cells upon long-term overstimulation may develop through stages of hyperplasia, dysplasia, and rather benign tumours, into highly malignant carcinomas. Dedifferentiation of cells may change the histological appearance and impede the identification of the cellular origin, as seen with gastric carcinomas, which in many cases are dedifferentiated neuroendocrine tumours. Finding the cell of origin is important to identify risk factors for cancer, prevent tumour development, and tailor treatment. In the present review, we focus not only on gastric tumours, but also evaluate the role of neuroendocrine cells in tumourigenesis in two other foregut-derived organs, the lungs and the pancreas, as well as in the midgut-derived small intestine.
Collapse
Affiliation(s)
- Helge L. Waldum
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, N-7491, Norway Department of Gastroenterology and Hepatology, St. Olav’s University Hospital, Trondheim, Norway
| | - Kjell Öberg
- Department of Endocrine Oncology Uppsala University and University Hospital, Uppsala, Sweden
| | - Øystein F. Sørdal
- Department of Gastroenterology and Hepatology, St. Olav’s University Hospital, Trondheim, Norway
| | - Arne K. Sandvik
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Gastroenterology and Hepatology, St. Olav’s University Hospital, Trondheim, Norway
| | - Bjørn I. Gustafsson
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Gastroenterology and Hepatology, St. Olav’s University Hospital, Trondheim, Norway
| | - Patricia Mjønes
- epartment of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Pathology, St. Olav’s University Hospital, Trondheim, Norway
| | - Reidar Fossmark
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Gastroenterology and Hepatology, St. Olav’s University Hospital, Trondheim, Norway
| |
Collapse
|
11
|
Cui H, Yang S, Jiang Y, Li C, Zhao Y, Shi Y, Hao Y, Qian F, Tang B, Yu P. The glycosyltransferase ST6Gal-I is enriched in cancer stem-like cells in colorectal carcinoma and contributes to their chemo-resistance. Clin Transl Oncol 2018; 20:1175-1184. [PMID: 29423671 DOI: 10.1007/s12094-018-1840-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/23/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Presence of cancer stem cells (CSCs) contributes to tumor outgrowth, chemo-resistance and relapse in some cancers including colorectal carcinoma (CRC). The current characterization methods of CSCs in CRC only allows enrichment of CSCs but not their purification. Recent reports showed that ST6 beta-galactoside alpha-2,6-sialyltransferase 1 (ST6Gal-I) plays an essential role in protecting tumor cells against harsh environment like oxidative stress and nutrient deprivation. Therefore, whether ST6Gal-I may be highly expressed in CSCs or whether it may enhance resistance of tumor cells to chemotherapy deserves exploration. METHOD ST6Gal-I levels were determined in CRC specimens, compared to paired normal colorectal tissue, and examined in CD133+ vs CD133- CRC cells, and CD44+ vs CD44- CRC cells. ST6Gal-I levels and their association with patient survival were examined. In vivo, 2 CRC cell lines Caco-2 and SW48 were transduced with two lentiviruses, one lentivirus carrying a green fluorescent protein reporter and a luciferase reporter under a cytomegalovirus promoter to allow tracing tumor cells by both fluorescence and luciferase activity, and one lentivirus carrying a nuclear red fluorescent protein under the control of ST6Gal-I promoter to allow separation of ST6Gal-I+ vs ST6Gal-I- CRC cells. Tumor sphere formation, resistance to fluorouracil-induced apoptosis, and frequency of tumor formation after serial adoptive transplantation were done on ST6Gal-I+ vs ST6Gal-I- CRC cells. RESULT ST6Gal-I levels were significantly upregulated in clinically obtained CRC specimens, compared to paired normal colorectal tissue. Poorer patient survival was detected in ST6Gal-I-high CRC, compared to ST6Gal-I-low subjects. Higher levels of ST6Gal-I were detected in CD133+ CRC cells than CD133- CRC cells, and in CD44+ CRC cells than in CD44- CRC cells. Compared to ST6Gal-I- CRC cells, ST6Gal-I+ CRC cells generated significantly more tumor spheres in culture, were more resistant to fluorouracil-induced apoptosis likely through upregulating cell autophagy, and generated tumor more frequently after serial adoptive transplantation. CONCLUSION ST6Gal-I may be highly expressed in the cancer stem-like cells in CRC and enhances cancer cell resistance to chemotherapy.
Collapse
Affiliation(s)
- H Cui
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - S Yang
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - Y Jiang
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - C Li
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - Y Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - Y Shi
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - Y Hao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - F Qian
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China
| | - B Tang
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China.
| | - P Yu
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyanzheng Street, Chongqing, 400038, China.
| |
Collapse
|
12
|
Expression of Cancer Stem Cell Marker CD44 and Its Polymorphisms in Patients with Chronic Gastritis, Precancerous Gastric Lesion, and Gastric Cancer: A Cross-Sectional Multicenter Study in Thailand. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4384823. [PMID: 29445738 PMCID: PMC5763069 DOI: 10.1155/2017/4384823] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/05/2017] [Indexed: 12/13/2022]
Abstract
Here we investigated CD44 protein expression and its polymorphisms in patients with chronic gastritis, precancerous gastric lesions, and gastric cancer; and we evaluated our result with the risk of CD44 protein expression and clinicopathological characteristics. Our results obtained by analyzing 162 gastric cancer patients, 125 chronic gastritis, and 165 precancerous gastric lesions from three study centers in Thailand showed that CD44 expression was significantly higher in patients with precancerous gastric lesions and gastric cancer while patients with chronic gastritis were negative for CD44 staining (p = 0.036). We further observed the significant association of variant genotype; gastric cancer patients carrying AG or GG of CD44 rs187116 had more increased risk of CD44 expression than wild-type (WT) carriers (AG: odds ratio (OR) = 5.67; 95% CI = 1.57-7.23; p = 0.024 and GG: OR = 8.32; 95% CI = 2.94-11.42; p = 0.016), but no significant difference in the risk of CD44 expression due to polymorphism in patients with precancerous gastric lesions. Our results suggested that CD44 expression could be used as a marker for the prediction of gastric cancer development, particularly in patients with precancerous gastric lesions carrying AG or GG, who were selected to surveillance follow-up for gastric cancer prevention.
Collapse
|
13
|
Kim YS, Lee HJ, Park JM, Han YM, Kangwan N, Oh JY, Lee DY, Hahm KB. Targeted molecular ablation of cancer stem cells for curing gastrointestinal cancers. Expert Rev Gastroenterol Hepatol 2017; 11:1059-1070. [PMID: 28707966 DOI: 10.1080/17474124.2017.1356224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abundance of the ATPase-binding cassette (ABC) transporters and deranged self-renewal pathways characterize the presence of cancer stem cells (CSCs) in gastrointestinal cancers (GI cancers), which play crucial roles in tumorigenesis, chemotherapy resistance, tumor recurrence, and cancer metastasis. Therefore, in order to ensure high cure rates, chemoquiescence, CSCs should be ablated. Recent advances in either understanding CSCs or biomarker identification enable scientists to develop techniques for ablating CSCs and clinicians to provide cancer cure, especially in GI cancers characterized by inflammation-driven carcinogenesis. Areas covered: A novel approach to ablate CSCs in GI cancers, including esophageal, gastric, and colon cancers, is introduced along with explored underlying molecular mechanisms. Expert commentary: Though CSC ablation is still in the empirical stages and not in clinical practice, several strategies for ablating CSCs in GI cancers had been published, proton-pump inhibitors (PPIs) that regulate the membrane-bound ABC transporters, which underlie drug resistance; chloroquine (CQ) that inhibits autophagy, which is responsible for tumor survival; Hedgehog/Wnt/Notch inhibitors that influence the underlying stem-cell growth, and some natural products including Korean red ginseng, cancer-preventive kimchi, Artemisia extract, EGCG from green tea, and walnut extracts.
Collapse
Affiliation(s)
- Yong Seok Kim
- a Department of Biochemistry and Molecular Biology , Hanyang University College of Medicine , Seoul , Korea
| | - Ho Jae Lee
- b Department of Biochemistry , Gachon University College of Medicine , Incheon , Korea
| | - Jong-Min Park
- c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea
| | - Young-Min Han
- c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea
| | - Napapan Kangwan
- d Division of Physiology, School of Medical Sciences , University of Phayao , Phayao , Thailand
| | | | | | - Ki Baik Hahm
- a Department of Biochemistry and Molecular Biology , Hanyang University College of Medicine , Seoul , Korea.,c CHA Cancer Prevention Research Center , CHA University , Seongnam , Korea.,f Digestive Disease Center , CHA University Bundang Medical Center , Seongnam , Korea
| |
Collapse
|
14
|
Lee DH, Lee SY, Oh SC. Hedgehog signaling pathway as a potential target in the treatment of advanced gastric cancer. Tumour Biol 2017. [DOI: 10.1177/1010428317692266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Dae-Hee Lee
- Division of Brain Korea 21 Program for Biomedicine Science, College of Medicine, Korea University, Seoul, Republic of Korea
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Suk-young Lee
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Sang Cheul Oh
- Division of Brain Korea 21 Program for Biomedicine Science, College of Medicine, Korea University, Seoul, Republic of Korea
- Division of Oncology and Hematology, Department of Internal Medicine, College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
15
|
Zeng JF, Ma XQ, Wang LP, Wang W. MicroRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer. World J Gastroenterol 2017; 23:2337-2345. [PMID: 28428713 PMCID: PMC5385400 DOI: 10.3748/wjg.v23.i13.2337] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/09/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To determine the potential roles of CD4 and microRNA (miR)-145 in gastric cancer.
METHODS The levels of CD44 and miR-145 were determined in gastric cancer cells. Quantitative real-time polymerase chain reaction was used to measure to the level of CD44 mRNA. A luciferase reporter assay and western blotting were performed to examine the effect of miR-145 on CD44 expression. Tumor sphere and MTT assays were carried out to evaluate the self-renewal and chemo-resistance properties of gastric cancer cells.
RESULTS The expression of CD44 was greatly increased and miR-145 was decreased in gastric cancer cells that were highly enriched in cancer stem cells (CSCs). The results demonstrated that miR-145 regulated CD44 by targeting directly the CD44 3’-untranslated region (3’-UTR). In gastric cancer cells, overexpression of miR-145 repressed the activity of the CD44 3’-UTR, and disruption of miR-145/CD44 3’-UTR interactions abrogated the silencing effects. In addition, miR-145 inhibition stimulated CD44 3’-UTR activity and disruption of miR-145/CD44 3’-UTR interactions abrogated this stimulatory effect. Enforced CD44 expression greatly increased tumor sphere formation and chemo-resistance in gastric cancer cells. Furthermore, the inhibition of CSCs and the chemo-sensitivity of gastric cancer cells treated with miR-145 were significantly abrogated by overexpression of CD44.
CONCLUSION miR-145 targeting of CD44 plays critical roles in the regulation of tumor growth and chemo-resistance in gastric cancer.
Collapse
|
16
|
Rivas-Ortiz CI, Lopez-Vidal Y, Arredondo-Hernandez LJR, Castillo-Rojas G. Genetic Alterations in Gastric Cancer Associated with Helicobacter pylori Infection. Front Med (Lausanne) 2017; 4:47. [PMID: 28512631 PMCID: PMC5411440 DOI: 10.3389/fmed.2017.00047] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer is a world health problem and depicts the fourth leading mortality cause from malignancy in Mexico. Causation of gastric cancer is not only due to the combined effects of environmental factors and genetic variants. Recent molecular studies have transgressed a number of genes involved in gastric carcinogenesis. The aim of this review is to understand the recent basics of gene expression in the development of the process of gastric carcinogenesis. Genetic variants, polymorphisms, desoxyribonucleic acid methylation, and genes involved in mediating inflammation have been associated with the development of gastric carcinogenesis. Recently, these genes (interleukin 10, Il-17, mucin 1, β-catenin, CDX1, SMAD4, SERPINE1, hypoxia-inducible factor 1 subunit alpha, GSK3β, CDH17, matrix metalloproteinase 7, RUNX3, RASSF1A, TFF1, HAI-2, and COX-2) have been studied in association with oncogenic activation or inactivation of tumor suppressor genes. All these mechanisms have been investigated to elucidate the process of gastric carcinogenesis, as well as their potential use as biomarkers and/or molecular targets to treatment of disease.
Collapse
Affiliation(s)
- Claudia I. Rivas-Ortiz
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Yolanda Lopez-Vidal
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Gonzalo Castillo-Rojas
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- *Correspondence: Gonzalo Castillo-Rojas,
| |
Collapse
|
17
|
Nanovesicle-mediated systemic delivery of microRNA-34a for CD44 overexpressing gastric cancer stem cell therapy. Biomaterials 2016; 105:12-24. [DOI: 10.1016/j.biomaterials.2016.07.036] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/24/2016] [Accepted: 07/29/2016] [Indexed: 12/22/2022]
|
18
|
Błogowski W, Zuba-Surma E, Sałata D, Budkowska M, Dołęgowska B, Starzyńska T. Peripheral trafficking of bone-marrow-derived stem cells in patients with different types of gastric neoplasms. Oncoimmunology 2015; 5:e1099798. [PMID: 27141380 DOI: 10.1080/2162402x.2015.1099798] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
Recently, there has been a growing interest in the importance of stem cells (SCs) in the development/progression of gastric neoplasms. In this study, we performed a comprehensive analysis of different populations of bone-marrow-derived stem cells (BMSCs) in patients with various types of gastric malignancies, including gastric cancer, gastrointestinal stromal tumors (GISTs), neuroendocrine neoplasms (NENs), and lymphomas. We found significantly lower numbers of circulating Lin-/CD45 +/ CD133 + hematopoietic stem/progenitor cells (HSPCs), and intensified peripheral trafficking of both Lin-/CD45-/CXCR4+/CD34+/CD133+ very small embryonic/epiblast-like stem cells (VSELs) and CD105 + /STRO-1 +/ CD45- mesenchymal SCs (MSCs) in patients with gastric cancer, but not in those with other types of gastric neoplasms. No significant differences in the absolute numbers of circulating CD34 +/ KDR +/ CD31 +/ CD45- endothelial progenitor cells (EPCs) were observed between the groups. This abnormal balance in the peripheral trafficking of BMSCs in patients with gastric cancer was neither associated with clinical stage of the disease nor with systemic levels of stromal-derived factor-1 (SDF-1), as these were comparable to the values observed in control individuals. Interestingly, the absolute numbers of circulating BMSCs correlated with the concentrations of complement cascade-derived anaphylatoxins/molecules (mainly C5b-9/membrane attack complex-MAC) and sphingosine-1-phosphate (S1P). In summary, our translational study revealed that abnormal peripheral trafficking of BMSCs occurs in patients with gastric cancer, but not in those with other types of gastric neoplasms. Further, our findings indicate that highlighted complement cascade-derived molecules and S1P, but not SDF-1, are significant players associated with this phenomenon.
Collapse
Affiliation(s)
- Wojciech Błogowski
- Department of Internal Medicine, University of Zielona Góra, Zielona Góra, Poland; Department of Gastroenterology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Daria Sałata
- Department of Medical Analytics, Pomeranian Medical University in Szczecin , Szczecin, Poland
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University in Szczecin , Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Medical Analytics, Pomeranian Medical University in Szczecin , Szczecin, Poland
| | - Teresa Starzyńska
- Department of Gastroenterology, Pomeranian Medical University in Szczecin , Szczecin, Poland
| |
Collapse
|
19
|
Abstract
Stomach cancer remains, stubbornly, highly prevalent in East Asia. Still, stomach cancer has few biomarkers by which it can be predicted. Helicobacter pylori infection, a known carcinogen of stomach cancer, usually goes undetected prior to cancer diagnosis, due to the poor mucosal environments that its related gastric atrophy causes. We propose, herein, an endoscopic-biopsy-based cancer-predicting DNA methylation marker. We semi-quantitatively examined the methylation-variable sites near the CpG-island margins by radioisotope-labeling methylation-specific polymerase chain reaction in association with H. pylori, which increases age-related over-methylation in CpG islands of gastric mucosa. These age-related methylation patterns of the transitional-CpG sites are proposed as useful surrogate markers for stomach cancer. It would be helpful for setting the optimal screening interval for high-risk subjects as well as for estimating the prognosis and the predictability for recurrence of early gastric cancer in patients having undergone endoscopic submucosal dissection. New screening-interval guidelines for gastric cancer should be suggested considering individual risk based on age, severity of atrophy, H. pylori status, and DNA methylation pattern.
Collapse
Affiliation(s)
- Jung-Hwan Oh
- Departments of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hoon Jung
- Departments of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Jin Hong
- Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mun-Gan Rhyu
- Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
20
|
Metabolic alteration--Overcoming therapy resistance in gastric cancer via PGK-1 inhibition in a combined therapy with standard chemotherapeutics. Int J Surg 2015; 22:92-8. [PMID: 26298000 DOI: 10.1016/j.ijsu.2015.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/15/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES It can be assumed that PGK1 is involved in metastatic spread of gastric carcinomas. Furthermore PGK1 has a proven influence on the characteristics of tumor stem cells. The presence of malignant stem cells, regarding treatment resistance and recurrence, is of considerable importance. We hypothesized that inhibition of PGK1 makes these cells more sensitive to chemotherapeutic agents and therefore mediates an overcome of the existing therapy resistance. METHODS All investigations were performed with human gastric adenocarcinoma cell lines. Small hairpin RNA knockdown of PGK1 via adenovirus-shPGK1 was used for PGK1-inhibition. Chemotherapeutic agents were 5-FU and mitomycin. FACS, qRT-PCR, and xCELLigence were performed. RESULTS Using the medium-sole-control indicating the highest cell viability and Triton indicating the lowest, mitomycin and 5-FU alone showed a significant decrease in cell viability. The treatment with AdvshPGK1 alone already showed a better decrease. The simultaneous application of chemotherapeutics and adenovirus showed the strongest effect and is comparable to the effect of Triton. CONCLUSIONS We showed a significant decrease in cell viability after the simultaneous application of chemotherapeutics and adenovirus. These results suggest that PGK1-inhibition is able to increase the vulnerability of gastric cancer cells and tumor stem cells to overcome the chemotherapeutic therapy resistance.
Collapse
|
21
|
Levi E, Sochacki P, Khoury N, Patel BB, Majumdar APN. Cancer stem cells in Helicobacter pylori infection and aging: Implications for gastric carcinogenesis. World J Gastrointest Pathophysiol 2014; 5:366-372. [PMID: 25133037 PMCID: PMC4133534 DOI: 10.4291/wjgp.v5.i3.366] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 04/04/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To demonstrated the combined effects of aging and carcinogen treatment on cancer stem/stem-like cells (CSCs) of gastric mucosa in an animal model.
METHODS: In this study we investigated the effects of aging and Helicobacter pylori (H. pylori) inflammation as a model for inflammation induced carcinogenesis in human and rat gastric mucosa samples. In aging studies, we compared 4-mo old (young) with 22 mo (aged) old Fischer-344 rats. For human studies, gastric biopsies and resection specimens representing normal mucosa or different stages of H. pylori gastritis and gastric adenocarcinomas were used for determining the expression of stem cell markers CD166, ALDH1 and LGR5. In addition we performed immunofluorescent double labeling for B-catenin and Lgr5 in both rat and human gastric tissues to examine the status of Wnt signaling in these cells.
RESULTS: CSC markers ALDH1, LGR5, and CD166 were expressed in very low levels in normal human gastric mucosa or young rat gastric mucosa. In contrast, level of expression for all three markers significantly increased in H. pylori gastritis and gastric adenocarcinomas as well as in normal gastric mucosa in aged rats. We also observed cytoplasmic B-catenin staining in both aged rat and human H. pylori inflamed gastric mucosa, which were found to be colocalized with Lgr5 immunoreactive cells. The increased number of ALDH1, CD166 and LGR5 positive cells in H. pylori gastritis indicates that increased number of stem-like cells in gastric mucosa is an early event, and may constitute an important step in the progression to neoplasia.
CONCLUSION: Our observation of the age-related increase in cancer stem/stem-like cells in the gastric mucosa may explain the increased incidence of gastric cancer during aging. Combination of aging and H. pylori infection may have additive effects in progression to neoplasia.
Collapse
|
22
|
Yang D, Wang H, Zhang J, Li C, Lu Z, Liu J, Lin C, Li G, Qian H. In vitro characterization of stem cell-like properties of drug-resistant colon cancer subline. Oncol Res 2014; 21:51-7. [PMID: 24330852 DOI: 10.3727/096504013x13793555706768] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The objective of this study was to investigate the stem cell-like properties of drug-resistant colon cancer cells. Oxaliplatin was used to induce the drug-resistant subline of HCT116(p53+/+) cell line. The stem cell-like characteristics of the drug-resistant subline were assayed for the proliferation capacity, cell cycle, adhesion, invasion, multiple drug resistance, and clone sphere formation capacity. The expression of ABCG2 (ATP-binding cassette superfamily G member 2) and "stemness" indicators SOX2 (SRY-related HMG box-containing transcription factor-2) and OCT4 (octamer-binding transcription factor 4) was determined by Western blot. We established the HCT116(p53+/+)-oxaliplatin subline (HCT116(p53+/+)OXA), which was resistant to oxaliplatin with a resistance index (RI) of 3.03 ± 0.14. The HCT116(p53+/+)OXA was also resistant to Taxol, showing lower proliferation, higher adhesion and invasion ability, greater proportion of G0/G1 phase, and higher sphere-forming capacity than its parental cells. SOX2, OCT4, and ABCG2 were expressed at higher levels in drug-resistant cells than in their parental cells. We verified that the HCT116(p53+/+)OXA was enriched with cancer stem cell properties and provided an ideal cell model for drug-resistance study.
Collapse
Affiliation(s)
- Dong Yang
- State Key Laboratory of Molecular Oncology, Cancer Institute/Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Cao L, Hu X, Zhang J, Liang P, Zhang Y. CD44(+) CD324(-) expression and prognosis in gastric cancer patients. J Surg Oncol 2014; 110:727-33. [PMID: 24910454 DOI: 10.1002/jso.23690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/14/2014] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the frequency of cancer stem cell marker CD44 combined CD324 expression in gastric cancer specimens and association of the expression with survival of gastric cancer patients. METHODS A total of 203 patients with primary gastric cancer who underwent curative gastric resection with lymphadenectomy were studied. The expression of CD44 and CD324 was analyzed by immunohistochemistry and the association of CD44 and CD324 expression analyzed by clinicopathological characteristics. Survival of the patients was evaluated. RESULTS CD44(+) CD324(-) tumors showed strong correlation with positive lymph node metastasis. Patients with CD44(-) CD324(+) had a significantly better prognosis (54.8%) than that with CD44(+) CD324(-) (24.5%). A multivariate Cox proportional hazards model using those variables associated with survival in our study (depth of invasion, tumor size, lymph node metastasis, gastrectomy, CD44 expression, CD324 expression, CD44 and CD324 expression) revealed that CD44(+) CD324(-) expression (P = 0.005) was one of significant independent prognostic indicator, whereas CD44 expression (P = 0.081), CD324 expression (P = 0.068) was not. CONCLUSION CD44(+) CD324(-) expression was one of significant independent prognostic survival factors for patients with gastric cancer.
Collapse
Affiliation(s)
- Liang Cao
- Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | | | | | | | | |
Collapse
|
24
|
Kalamohan K, Periasamy J, Bhaskar Rao D, Barnabas GD, Ponnaiyan S, Ganesan K. Transcriptional coexpression network reveals the involvement of varying stem cell features with different dysregulations in different gastric cancer subtypes. Mol Oncol 2014; 8:1306-25. [PMID: 24917244 DOI: 10.1016/j.molonc.2014.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 03/22/2014] [Accepted: 04/14/2014] [Indexed: 01/29/2023] Open
Abstract
Despite the advancements in the cancer therapeutics, gastric cancer ranks as the second most common cancers with high global mortality rate. Integrative functional genomic investigation is a powerful approach to understand the major dysregulations and to identify the potential targets toward the development of targeted therapeutics for various cancers. Intestinal and diffuse type gastric tumors remain the major subtypes and the molecular determinants and drivers of these distinct subtypes remain unidentified. In this investigation, by exploring the network of gene coexpression association in gastric tumors, mRNA expressions of 20,318 genes across 200 gastric tumors were categorized into 21 modules. The genes and the hub genes of the modules show gastric cancer subtype specific expression. The expression patterns of the modules were correlated with intestinal and diffuse subtypes as well as with the differentiation status of gastric tumors. Among these, G1 module has been identified as a major driving force of diffuse type gastric tumors with the features of (i) enriched mesenchymal, mesenchymal stem cell like, and mesenchymal derived multiple lineages, (ii) elevated OCT1 mediated transcription, (iii) involvement of Notch activation, and (iv) reduced polycomb mediated epigenetic repression. G13 module has been identified as key factor in intestinal type gastric tumors and found to have the characteristic features of (i) involvement of embryonic stem cell like properties, (ii) Wnt, MYC and E2F mediated transcription programs, and (iii) involvement of polycomb mediated repression. Thus the differential transcription programs, differential epigenetic regulation and varying stem cell features involved in two major subtypes of gastric cancer were delineated by exploring the gene coexpression network. The identified subtype specific dysregulations could be optimally employed in developing subtype specific therapeutic targeting strategies for gastric cancer.
Collapse
Affiliation(s)
- Kalaivani Kalamohan
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India
| | - Jayaprakash Periasamy
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India
| | - Divya Bhaskar Rao
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India
| | - Georgina D Barnabas
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India
| | - Srigayatri Ponnaiyan
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India
| | - Kumaresan Ganesan
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, India.
| |
Collapse
|
25
|
Yu L, Yang L, An W, Su X. Anticancer Bioactive Peptide-3 Inhibits Human Gastric Cancer Growth by Suppressing Gastric Cancer Stem Cells. J Cell Biochem 2014; 115:697-711. [DOI: 10.1002/jcb.24711] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 11/05/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Lan Yu
- Department of Cell Biology; Capital Medical University; No. 10 You An Men Wai Street Fengtai District Beijing 100069 China
| | - Ling Yang
- Clinical Medical Research Center of the Affiliated Hospital; Inner Mongolia Medical University; No. 1 Tongdao North Street Huimin District Hohhot 010050 China
| | - Wei An
- Department of Cell Biology; Capital Medical University; No. 10 You An Men Wai Street Fengtai District Beijing 100069 China
| | - Xiulan Su
- Department of Cell Biology; Capital Medical University; No. 10 You An Men Wai Street Fengtai District Beijing 100069 China
- Clinical Medical Research Center of the Affiliated Hospital; Inner Mongolia Medical University; No. 1 Tongdao North Street Huimin District Hohhot 010050 China
| |
Collapse
|
26
|
Choi JE, Bae JS, Lee JH, Jang KY, Chung MJ, Moon WS. Musashi-1 expression and clinicopathological significance in young gastric cancer patients: a matched case-control study. Int J Oncol 2014; 44:1185-92. [PMID: 24452324 DOI: 10.3892/ijo.2014.2263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/29/2013] [Indexed: 11/05/2022] Open
Abstract
Musashi-1 (Msi-1) is proposed to be a marker of progenitor cells in the human gastric mucosa. We examined Msi-1 expression and localization in surgical specimens of gastric cancer in young patients using immunohistochemistry and tested associations of Msi-1 expression with clinicopathological features. Patients (n=611) with gastric cancer who underwent radical gastrectomy were included in the present study. To minimize confounding effects, we matched 26 gastric cancer patients 30 years of age or younger (age ≤30) with 26 patients 60 years of age or older (age ≥60). The groups were matched by gender, tumor histological type and tumor stage. Gastric cancer in the younger patients was significantly associated with female gender and with diffuse histological type, compared with 585 gastric cancer patients older than 31 years. Msi-1 expression was more frequently upregulated in gastric cancer in young patients than in patients older than 60 years. Msi-1 expression was significantly associated with diffuse histological type, depth of tumor invasion, lymph node metastasis and tumor stage in the 26 young patients with gastric cancer. Univariate Kaplan-Meier survival analysis identified Msi-1 expression as a significantly negative factor in the survival of young gastric cancer patients. However, Msi-1 expression was not significantly associated with survival in the 26 matched older patients. According to mucin phenotype, the gastric foveolar type predominated in Msi-1-positive gastric cancers. Our principal findings included a significantly higher level of Msi-1 expression in younger gastric cancer patients compared to older ones, and a probable association of tumor Msi-1 expression in young gastric cancer patients with more aggressive tumor type.
Collapse
Affiliation(s)
- Ji Eun Choi
- Department of Pathology, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Ju Hyung Lee
- Preventive Medicine, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University, Medical School, Research Institute of Clinical Medicine of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju 561-756, Republic of Korea
| |
Collapse
|
27
|
Stojnev S, Krstic M, Ristic-Petrovic A, Stefanovic V, Hattori T. Gastric cancer stem cells: therapeutic targets. Gastric Cancer 2014; 17:13-25. [PMID: 23563919 DOI: 10.1007/s10120-013-0254-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 03/15/2013] [Indexed: 02/07/2023]
Abstract
During the past decade, a growing body of evidence has implied that cancer stem cells (CSCs) play an important role in the development of gastric cancer (GC). The notion that CSCs give rise to GC and may be responsible for invasion, metastasis, and resistance to treatment has profound implications for anti-cancer therapy. Recent major advances in the rapidly evolving field of CSCs have opened novel exciting opportunities for developing CSC-targeted therapies. Discovery of specific markers and signaling pathways in gastric CSCs (GCSCs), with the perfecting of technologies for identification, isolation, and validation of CSCs, may provide the basis for a revolutionary cancer treatment approach based on the eradication of GCSCs. Emerging therapeutic tools based on specific properties and functions of CSCs, including activation of self-renewal signaling pathways, differences in gene expression profiles, and increased activity of telomerase or chemoresistance mechanisms, are developing in parallel with advances in nanotechnology and bioengineering. The addition of GCSC-targeted therapies to current oncological protocols and their complementary application may be the key to successfully fighting GC.
Collapse
Affiliation(s)
- Slavica Stojnev
- Faculty of Medicine, Institute of Pathology, University of Nis, Zorana Djindjica Blvd 81, 18000, Nis, Serbia,
| | | | | | | | | |
Collapse
|
28
|
Kuang RG, Kuang Y, Luo QF, Zhou CJ, Ji R, Wang JW. Expression and significance of Musashi-1 in gastric cancer and precancerous lesions. World J Gastroenterol 2013; 19:6637-6644. [PMID: 24151393 PMCID: PMC3801380 DOI: 10.3748/wjg.v19.i39.6637] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate expression of stem cell marker Musashi-1 (Msi-1) in relationship to tumorigenesis and progression of intestinal-type gastric cancer (GC).
METHODS: Endoscopic biopsy specimens and surgical specimens were obtained, including 54 cases of intestinal-type GC, 41 high-grade intraepithelial neoplasia, 57 low-grade intraepithelial neoplasia, 31 intestinal metaplasia, and 36 normal gastric mucosa. Specimens were fixed in 10% paraformaldehyde, conventionally dehydrated, embedded in paraffin, and sliced in 4-μm-thick serial sections. Two-step immunohistochemical staining was used to detect Msi-1 and proliferating cell nuclear antigen (PCNA) expression. Correlation analysis was conducted between Msi-1 and PCNA expression. The relationship between Msi-1 expression and clinicopathological parameters of GC was analyzed statistically.
RESULTS: There were significant differences in Msi-1 and PCNA expression in different pathological tissues (χ2 = 15.37, P < 0.01; χ2 = 115.36, P < 0.01). Msi-1 and PCNA-positive cells were restricted to the isthmus of normal gastric glands. Expression levels of Msi-1 and PCNA in intestinal metaplasia were significantly higher than in normal mucosa (U = 392.0, P < 0.05; U = 40.50, P < 0.01), whereas there was no significant difference compared to low or high-grade intraepithelial neoplasia. Msi-1 and PCNA expression in intestinal-type GC was higher than in high-grade intraepithelial neoplasia (U = 798.0, P < 0.05; U = 688.0, P < 0.01). There was a significantly positive correlation between Msi-1 and PCNA expression (rs = 0.20, P < 0.01). Msi-1 expression in GC tissues was correlated with their lymph node metastasis and tumor node metastasis stage (χ2 = 12.62, P < 0.01; χ2 = 11.24, P < 0.05), but not with depth of invasion and the presence of distant metastasis.
CONCLUSION: Msi-1-positive cells may play a key role in the early events of gastric carcinogenesis and may be involved in invasion and metastasis of GC.
Collapse
|
29
|
Canovas-Jorda D, Louisse J, Pistollato F, Zagoura D, Bremer S. Regenerative toxicology: the role of stem cells in the development of chronic toxicities. Expert Opin Drug Metab Toxicol 2013; 10:39-50. [PMID: 24102433 DOI: 10.1517/17425255.2013.844228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Human stem cell lines and their derivatives, as alternatives to the use of animal cells or cancer cell lines, have been widely discussed as cellular models in predictive toxicology. However, the role of stem cells in the development of long-term toxicities and carcinogenesis has not received great attention so far, despite growing evidence indicating the relationship of stem cell damage to adverse effects later in life. However, testing this in vitro is a scientific/technical challenge in particular due to the complex interplay of factors existing under physiological conditions. Current major research programs in stem cell toxicity are not aiming to demonstrate that stem cells can be targeted by toxicants. Therefore, this knowledge gap needs to be addressed in additional research activities developing technical solutions and defining appropriate experimental designs. AREAS COVERED The current review describes selected examples of the role of stem cells in the development of long-term toxicities in the brain, heart or liver and in the development of cancer. EXPERT OPINION The presented examples illustrate the need to analyze the contribution of stem cells to chronic toxicity in order to make a final conclusion whether stem cell toxicities are an underestimated risk in mechanism-based safety assessments. This requires the development of predictive in vitro models allowing the assessment of adverse effects to stem cells on chronic toxicity and carcinogenicity.
Collapse
Affiliation(s)
- David Canovas-Jorda
- Institute for Health and Consumer Protection, DG Joint Research Centre (JRC), European Commission, Systems Toxicology Unit , Via E. Fermi 2749, TP 580, 21027 Ispra (VA) , Italy +39 0332 785914 ; +39 0332 785336 ;
| | | | | | | | | |
Collapse
|
30
|
Abstract
Gastric cancer (GC) remains one of the most common cancers worldwide. Its prevalence is still on the rise in the developing countries due to the ageing population. The cancer stem cell (CSC) theory provides a new insight into the interpretation of tumor initiation, aggressive growth, recurrence, and metastasis of cancer, as well as the development of new strategies for cancer treatment. This review will focus on the progress of biomarkers and signaling pathways of CSCs, the complex crosstalk networks between the microenvironment and CSCs, and the development of therapeutic approaches against CSCs, predominantly focusing on GC.
Collapse
|
31
|
β -Elemene-Attenuated Tumor Angiogenesis by Targeting Notch-1 in Gastric Cancer Stem-Like Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:268468. [PMID: 23710217 PMCID: PMC3655606 DOI: 10.1155/2013/268468] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 03/15/2013] [Accepted: 03/23/2013] [Indexed: 12/22/2022]
Abstract
Emerging evidence suggests that cancer stem cells are involved in tumor angiogenesis. The Notch signaling pathway is one of the most important regulators of these processes. β-Elemene, a naturally occurring compound extracted from Curcumae Radix, has been used as an antitumor drug for various cancers in China. However, its underlying mechanism in the treatment of gastric cancer remains largely unknown. Here, we report that CD44+ gastric cancer stem-like cells (GCSCs) showed enhanced proliferation capacity compared to their CD44− counterparts, and this proliferation was accompanied by the high expression of Notch-1 (in vitro). These cells were also more superior in spheroid colony formation (in vitro) and tumorigenicity (in vivo) and positively associated with microvessel density (in vivo). β-Elemene was demonstrated to effectively inhibit the viability of GCSCs in a dose-dependent manner, most likely by suppressing Notch-1 (in vitro). β-Elemene also contributed to growth suppression and attenuated the angiogenesis capacity of these cells (in vivo) most likely by interfering with the expression of Notch-1 but not with Dll4. Our findings indicated that GCSCs play an important role in tumor angiogenesis, and Notch-1 is one of the most likely mediators involved in these processes. β-Elemene was effective at attenuating angiogenesis by targeting the GCSCs, which could be regarded as a potential mechanism for its efficacy in gastric cancer management in the future.
Collapse
|
32
|
Zhang JH, Feng YG, Gao GP, Tao YY, Zhang XQ, Zhang HM, Jiao JX. Effects of CagA +Helicobacter pylori infection on expression of HIF-2α and ABCG2 in human gastric cancer cell line SGC7901 under normoxia and hypoxia conditions. Shijie Huaren Xiaohua Zazhi 2013; 21:293-299. [DOI: 10.11569/wcjd.v21.i4.293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the effect of CagA+Helicobacter pylori (H. pylori) on the expression of HIF-2α and ABCG2 in human gastric cancer cell line SGC7901 under normoxia and hypoxia conditions to investigate whether H. pylori infection and the tumor microenvironment have a synergistic effect in the initiation and development of gastric cancer.
METHODS: Gastric mucosal biopsy specimens collected by endoscopy were cultured under microaerophilic conditions and H. pylori isolates were identified. CagA+H. pylori strains were confirmed by PCR. Gastric cancer cell line SGC7901 was co-cultured with a CagA+H. pylori strain for 48 h under either normoxia or hypoxia condition (cells were divided into a normoxia control group, a hypoxia control group, a normoxia plus CagA+H. pylori group, and a hypoxia plus CagA+H. pylori group). Immunocytochemistry was used to detect the expression of HIF-2α and ABCG2 proteins, and RT-PCR was used to detect the expression of ABCG2 mRNA.
RESULTS: Immunocytochemistry results showed that HIF-2α and ABCG2 proteins were expressed at low levels under normoxia, while both hypoxia and CagA+H. pylori could significantly induce the expression of HIF-2α and ABCG2 proteins compared to the normoxia control group (all P < 0.01). Compared to the hypoxia control group and normoxia plus CagA+H. pylori group, the expression of HIF-2α and ABCG2 proteins was further elevated in the hypoxia plus CagA+H. pylori group (all P < 0.01). There was a positive correlation between the expression of HIF-2α and that of ABCG2 (r = 0.976, P < 0.05). Similar results were obtained for ABCG2 mRNA expression by RT-PCR.
CONCLUSION: CagA+H. pylori can stimulate the expression of HIF-2α and ABCG2 in gastric cancer cells under normoxia condition, and their expression can be further up-regulated under hypoxia condition. CagA+H. pylori and hypoxia have a synergistic effect on the expression of HIF-2α and ABCG2, suggesting that CagA+H. pylori and hypoxia may play an important role in inducing gastric cancer cell de-differentiation and chemotherapy resistance.
Collapse
|
33
|
Gomceli I, Demiriz B, Tez M. Gastric carcinogenesis. World J Gastroenterol 2012; 18:5164-70. [PMID: 23066309 PMCID: PMC3468847 DOI: 10.3748/wjg.v18.i37.5164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/30/2012] [Accepted: 05/05/2012] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the second most common cancer worldwide and the second most common cause of cancer-related deaths. Despite complete resection of gastric cancer and lymph node dissection, as well as improvements in chemotherapy and radiotherapy, there are still 700 000 gastric cancer-related deaths per year worldwide and more than 80% of patients with advanced gastric cancer die of the disease or recurrent disease within 1 year after diagnosis. None of the treatment modalities we have been applying today can influence the overall survival rates: at present, the overall 5-year relative survival rate for gastric cancer is about 28%. Cellular metaplasia due to chronic inflammation, injury and repair are the most documented processes for neoplasia. It appears that chronic inflammation stimulates tumor development and plays a critical role in initiating, sustaining and advancing tumor growth. It is also evident that not all inflammation is tumorigenic. Additional mutations can be acquired, and this leads to the cancer cell gaining a further growth advantage and acquiring a more malignant phenotype. Intestinalization of gastric units, which is called “intestinal metaplasia”; phenotypic antralization of fundic units, which is called “spasmolytic polypeptide-expressing metaplasia”; and the development directly from the stem/progenitor cell zone are three pathways that have been described for gastric carcinogenesis. Also, an important factor for the development of gastrointestinal cancers is peritumoral stroma. However, the initiating cellular event in gastric metaplasia is still controversial. Understanding gastric carcinogenesis and its precursor lesions has been under intense investigation, and our paper attempts to highlight recent progress in this field of cancer research.
Collapse
|
34
|
Ni X, Long J, Cen P, Chen L, Yang J, Li M. Pancreatic cancer tumour initiating cells: the molecular regulation and therapeutic values. J Cell Mol Med 2012; 16:988-94. [PMID: 22050663 PMCID: PMC3298733 DOI: 10.1111/j.1582-4934.2011.01478.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is an aggressive solid tumour characterized by its local invasion, early metastasis and resistance to standard chemotherapy or radiation therapy. Tumour initiating cells (TICs) are not only capable of self-renewal and differentiation, but also play an important role in multi-drug resistance, and thus become a popular topic in cancer research especially in pancreatic cancer. In this review, we summarize the current progress of TICs in tumourigenesis, various newly identified surface markers of pancreatic TICs, and the signalling pathways such as epithelial-mesenchymal transition, sonic hedgehog and Notch that regulate TICs. We also discuss the role which microRNA plays in TICs as well as its application in TIC-targeted therapy along with other approaches.
Collapse
Affiliation(s)
- Xiaoling Ni
- The Vivian L Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
35
|
Bauer L, Langer R, Becker K, Hapfelmeier A, Ott K, Novotny A, Höfler H, Keller G. Expression profiling of stem cell-related genes in neoadjuvant-treated gastric cancer: a NOTCH2, GSK3B and β-catenin gene signature predicts survival. PLoS One 2012; 7:e44566. [PMID: 22970250 PMCID: PMC3438181 DOI: 10.1371/journal.pone.0044566] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 08/08/2012] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cell (CSC) based gene expression signatures are associated with prognosis in various tumour types and CSCs are suggested to be particularly drug resistant. The aim of our study was first, to determine the prognostic significance of CSC-related gene expression in residual tumour cells of neoadjuvant-treated gastric cancer (GC) patients. Second, we wished to examine, whether expression alterations between pre- and post-therapeutic tumour samples exist, consistent with an enrichment of drug resistant tumour cells. The expression of 44 genes was analysed in 63 formalin-fixed, paraffin embedded tumour specimens with partial tumour regression (10-50% residual tumour) after neoadjuvant chemotherapy by quantitative real time PCR low-density arrays. A signature of combined GSK3B(high), β-catenin (CTNNB1)(high) and NOTCH2(low) expression was strongly correlated with better patient survival (p<0.001). A prognostic relevance of these genes was also found analysing publically available gene expression data. The expression of 9 genes was compared between pre-therapeutic biopsies and post-therapeutic resected specimens. A significant post-therapeutic increase in NOTCH2, LGR5 and POU5F1 expression was found in tumours with different tumour regression grades. No significant alterations were observed for GSK3B and CTNNB1. Immunohistochemical analysis demonstrated a chemotherapy-associated increase in the intensity of NOTCH2 staining, but not in the percentage of NOTCH2. Taken together, the GSK3B, CTNNB1 and NOTCH2 expression signature is a novel, promising prognostic parameter for GC. The results of the differential expression analysis indicate a prominent role for NOTCH2 and chemotherapy resistance in GC, which seems to be related to an effect of the drugs on NOTCH2 expression rather than to an enrichment of NOTCH2 expressing tumour cells.
Collapse
Affiliation(s)
- Lukas Bauer
- Institute of Pathology, Technische Universität München, München, Germany
| | - Rupert Langer
- Institute of Pathology, Technische Universität München, München, Germany
| | - Karen Becker
- Institute of Pathology, Technische Universität München, München, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Statistics and Epidemiology, Technische Universität München, München, Germany
| | - Katja Ott
- Department of Surgery, Universität Heidelberg, Heidelberg, Germany
| | - Alexander Novotny
- Department of Surgery, Technische Universität München, München, Germany
| | - Heinz Höfler
- Institute of Pathology, Technische Universität München, München, Germany
- Institute of Pathology, Helmholtz-Zentrum München, Neuherberg, Germany
| | - Gisela Keller
- Institute of Pathology, Technische Universität München, München, Germany
- * E-mail:
| |
Collapse
|
36
|
Taborda AG, Prolla JC. Alimentary factors in the development of gastric intestinal metaplasia in functional dyspeptic patients. ARQUIVOS DE GASTROENTEROLOGIA 2012; 49:208-13. [DOI: 10.1590/s0004-28032012000300008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Indexed: 12/26/2022]
Abstract
CONTEXT: Intestinal metaplasia of the stomach is a lesion in which metaplasia of gastric epithelial cells occurs for an intestinal phenotype. Gastric intestinal metaplasia is a lesion associated with an increase in the risk of gastric carcinoma development. Epidemiologic studies indicate a relation between dietary habits and stomach cancer development, some habits increasing the risk for it, and others have a protective effect, suggesting that antioxidants, such as vitamins A, C, and E, decrease the risk of this type of cancer. The relationship of these alimentary factors and intestinal metaplasia is unknown. METHODS: It is a case-control, observational study in which 320 patients with functional dyspepsia, divided in two groups, were assessed. The case I group (individuals with intestinal metaplasia) had their dietary pattern compared to that of the control group, constituted of individuals similar to those in the case group but without intestinal metaplasia, through a food frequency questionnaire. RESULTS: The analysis of the dietary pattern of functional dyspeptic patients with intestinal metaplasia, and its comparison with those without intestinal metaplasia, showed a higher frequency of canned and smoked foods consumption in the first group and, on the other hand, a higher consumption of fruits and vegetables in patients without intestinal metaplasia. No effect of salt consumption was detected. CONCLUSIONS: The results obtained in this study suggest changes in the diet, with a decrease in the consumption of smoked and canned foods, and an increase in the consumption of fruits and vegetables, can lead to a diminution of gastric intestinal metaplasia cases.
Collapse
|
37
|
Yu QM, Wang XB, Luo J, Wang S, Fang XH, Yu JL, Ling ZQ. CDH1 methylation in preoperative peritoneal washes is an independent prognostic factor for gastric cancer. J Surg Oncol 2012; 106:765-71. [PMID: 22514028 PMCID: PMC3495294 DOI: 10.1002/jso.23116] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 03/15/2012] [Indexed: 02/06/2023]
Abstract
Background and Objectives To investigate the clinical value of CDH1 methylation in preoperative peritoneal washes (PPW) from gastric cancer patients. Methods CDH1 methylation was detected by real-time methylation specific-PCR in tumor tissues and corresponding PPW from 92 gastric cancer patients, gastric mucosa from 40 chronic gastritis patients and 48 normal persons. Results CDH1 methylation was found in 75 of 92 (81.5%) gastric cancer tissues, which significantly correlated with size, growth pattern, differentiation, lymphatic invasion, venous invasion, invasion depth, lymph node metastasis, distant metastasis, and TNM stage of tumor (all P < 0.05), but its relationship to age, gender, tumor site, and H. pylori infection was not found (all P > 0.05). The percentage of CDH1 methylation in PPW was 48.9%, of which the Aζ value of ROC curve was 0.8 compared to that in gastric cancer tissues. Kaplan–Meier analysis showed that there was a significant difference in disease-free survival (DFS) between the patients with or without methylated CDH1 in their PPW (χ2 = 109.64, P < 0.000). Cox regression analysis revealed CDH1 methylation in PPW was an independent risk factor for gastric cancer patients, with a remarkable decrease in DFS after postoperative 30 months. Conclusions Methylated CDH1 in PPW predicts poor prognosis for gastric cancer patients. J. Surg. Oncol. 2012; 106:765–771. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qi-Ming Yu
- Zhejiang Cancer Research Institute, Zhejiang Province Cancer Hospital, Zhejiang Cancer Center, Banshanqiao District, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Liu GM, Zhou C, Xie C, Yang Z, Lv NH. Recent advances in research of gastric cancer stem cells. Shijie Huaren Xiaohua Zazhi 2012; 20:574-579. [DOI: 10.11569/wcjd.v20.i7.574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a common malignancy of the digestive tract that has a high mortality and seriously affects people's health. At present, the pathogenesis of gastric cancer is still unclear. According to the cancer stem cell theory, cancer stem cells are malignant cells with the characteristics of normal stem cells, probably formed by the mutation of normal stem cells. Tumor stem cells have been identified in a variety of solid tumors. Recent studies have shown that the location of gastric cancer is consistent with the settlement area of stem cells, indicating that gastric cancer may be a kind of stem cells disease. In this article we will review the existence, origin, identification and separation of cancer stem cells.
Collapse
|
39
|
Adamczyk B, Tharmalingam T, Rudd PM. Glycans as cancer biomarkers. Biochim Biophys Acta Gen Subj 2011; 1820:1347-53. [PMID: 22178561 DOI: 10.1016/j.bbagen.2011.12.001] [Citation(s) in RCA: 364] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/11/2011] [Accepted: 12/01/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Non-invasive biomarkers, such as those from serum, are ideal for disease prognosis, staging and monitoring. In the past decade, our understanding of the importance of glycosylation changes with disease has evolved. SCOPE OF REVIEW We describe potential biomarkers derived from serum glycoproteins for liver, pancreatic, prostate, ovarian, breast, lung and stomach cancers. Methods for glycan analysis have progressed and newly developed high-throughput platform technologies have enabled the analysis of large cohorts of samples in an efficient manner. We also describe this evolution and trends to follow in the future. MAJOR CONCLUSIONS Many convincing examples of aberrant glycans associated with cancer have come about from glycosylation analyses. Most studies have been carried out to identify changes in serum glycan profiles or through the isolation and identification of glycoproteins that contain these irregular glycan structures. In a majority of cancers the fucosylation and sialylation expression are found to be significantly modified. Therefore, these aberrations in glycan structures can be utilized as targets to improve existing cancer biomarkers. GENERAL SIGNIFICANCE The ability to distinguish differences in the glycosylation of proteins between cancer and control patients emphasizes glycobiology as a promising field for potential biomarker identification. Furthermore, the high-throughput and reproducible nature of the chromatography platform have highlighted extensive applications in biomarker discovery and allowed integration of glycomics with other -omics fields, such as proteomics and genomics, making systems glycobiology a reality. This article is part of a Special Issue entitled Glycoproteomics.
Collapse
Affiliation(s)
- Barbara Adamczyk
- Dublin-Oxford Glycobiology Laboratory, NIBRT-The National Institute for Bioprocessing Research and Training, Fosters Avenue, Mount Merrion, Blackrock, Co. Dublin, Ireland
| | | | | |
Collapse
|
40
|
Gao ZL, Zhang C, Sheng FY, Jin LW. Intestinal metaplasia, intraepithelial neoplasia and gastric carcinogenesis. Shijie Huaren Xiaohua Zazhi 2011; 19:1981-1984. [DOI: 10.11569/wcjd.v19.i19.1981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a common disease that greatly endangers people's health. The mechanism of gastric carcinogenesis is still unknown. Gastric carcinogenesis is a long-term multistep process, during which a series of precancerous lesions develop sequentially. Intraepithelial dysplasia and intestinal metaplasia are two types of gastric precancerous lesions. Because of high malignancy and unclear etiology of gastric cancer, there are some difficulties in carrying out successful primary prevention. Here, we give an overview of the definitions and classification of gastric intraepithelial neoplasia and intestinal metaplasia, and elaborate the relationship among Helicobacter pylori infection, atrophic gastritis, intestinal metaplasia and gastric carcinoma.
Collapse
|
41
|
Chen G, Wang QR, Qiu H, Ke SD, Xiang Y, Hu SM. Relationship of cellular drug resistance to proportion of side population cells and expression of Mdr1 and ERCC1 in human gastric cell line SGC7901/OXA. Shijie Huaren Xiaohua Zazhi 2011; 19:1219-1224. [DOI: 10.11569/wcjd.v19.i12.1219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the mechanism of acquired drug resistance in human gastric cancer cell line SGC7901/OXA.
METHODS: SGC7901/OXA cell line was obtained by exposure of SGC7901 cells to increasing doses of oxaliplatin (OXA). The parental cell line SGC7901 was used as a control. The reduced rate of cell growth, half maximal inhibitory concentration (IC50), and resistance index (RI) were measured by MTT assay and compared between the two groups of cells. The expression of multidrug resistance 1 (Mdr1) mRNA and excision repair cross-complementing group 1 (ERCC1) protein was detected by RT-PCR and Western blot, respectively. The proportion of side population (SP) cells was determined by flow cytometry.
RESULTS: The reduced rate of cell growth significantly decreased and IC50 increased (38.23 μmol/L vs 7.12 μmol/L) in SGC7901/OXA cells compared to parental SGC7901 cells. The RI was 5.37 in SGC7901/OXA cells. There was no significant difference in the relative expression level of Mdr1 mRNA (0.468 ± 0.147 vs 0.427 ± 0.136, P = 0.079) between the two groups. ERCC1 protein level was significantly higher in SGC7901/OXA cells than in SGC7901 cells. The proportion of SP cells was also significantly higher in SGC7901/OXA cells than in SGC7901 cells (6.1% vs 1.8%, P < 0.05).
CONCLUSION: SGC7901/OXA cells were more significantly resistant to OXA than SGC7901. The mechanism of acquired resistance of SGC7901/OXA cells may be associated with changes in ERCC1 protein expression and proportion of SP cells.
Collapse
|
42
|
Bones J, Byrne JC, O'Donoghue N, McManus C, Scaife C, Boissin H, Nastase A, Rudd PM. Glycomic and glycoproteomic analysis of serum from patients with stomach cancer reveals potential markers arising from host defense response mechanisms. J Proteome Res 2011; 10:1246-65. [PMID: 21142185 DOI: 10.1021/pr101036b] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the reduced incidence of gastric cancer in the developed world, a diagnosis of stomach carcinoma still carries a poor prognosis due to the asymptomatic nature of the disease in the early stages, subsequent advanced stage diagnosis, and a low 5 year survival rate. Endoscopy remains the primary standard for diagnosis of stomach carcinoma and the current marker, carbohydrate antigen 19-9 (CA19-9) lacks the levels of sensitivity and specificity required in order to make it clinically useful for diagnostic monitoring. Therefore, there is a current need for additional markers to improve the diagnostic accuracy for the early stages of stomach cancer. Together, glycomic, proteomic, and glycoproteomic analyses of serum have the potential to identify such probable markers. A discovery study is reported here using preoperative serum from 80 stomach cancer patients, 10 patients bearing benign stomach disease, and 20 matched controls. Glycomic analysis of the total and immunoaffinity depleted serum revealed statistically significant increases in the levels of sialyl Lewis X epitopes (SLe(X)) present on triantennary glycans accompanied by increased levels of core fucosylated agalactosyl biantennary glycans present on IgG (referred to as the IgG G0 glycoform) which are associated with increasing disease pathogenesis. Protein expression analysis using 2D-DiGE returned a number of differentially expressed protein candidates in the depleted serum, many of which were shown to carry triantennary SLe(X) during subsequent glycomic investigations. Biological pathway analysis of the experimental data returned complement activation and acute phase response signaling as the most significantly altered pathways in the stomach cancer patient serum. Upon the basis of these findings, it is suggested that increased expression of IgG G0 and complement activation are a host response to the presence of the stomach tumor while the increased expression of SLe(X) and acute phase response proteins is a result of pro-inflammatory cytokine signaling, including IL-6, during carcinogenesis. The approach presented herein provides an insight into the underlying mechanisms of disease and the resulting changes in the glycome and glycoproteome offer promise as potential markers for diagnosis and prognostic monitoring in stomach cancer.
Collapse
Affiliation(s)
- Jonathan Bones
- NIBRT Dublin-Oxford Glycobiology Laboratory, The National Institute for Bioprocessing Research and Training, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cancer stem cells in pancreatic cancer. Cancers (Basel) 2010; 2:1629-41. [PMID: 24281178 PMCID: PMC3837327 DOI: 10.3390/cancers2031629] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 07/29/2010] [Accepted: 08/18/2010] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is an aggressive malignant solid tumor well-known by early metastasis, local invasion, resistance to standard chemo- and radiotherapy and poor prognosis. Increasing evidence indicates that pancreatic cancer is initiated and propagated by cancer stem cells (CSCs). Here we review the current research results regarding CSCs in pancreatic cancer and discuss the different markers identifying pancreatic CSCs. This review will focus on metastasis, microRNA regulation and anti-CSC therapy in pancreatic cancer.
Collapse
|
44
|
Wang B, Xia YJ, Yan W, Xia LM, Liu M, Tian DA. Regulation of side population cells by TGF-β1-induced epithelial-mesenchymal transition in hepatocellular carcinoma cell lines SMMC-7721 and HepG2. Shijie Huaren Xiaohua Zazhi 2010; 18:2416-2421. [DOI: 10.11569/wcjd.v18.i23.2416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the influence of transforming growth factor-β1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) on side population cells in SMMC-7721.
METHODS: The percentages of SP cells in SMMC-7721 and HepG2 cells were analyzed by flow cytometry. TGF-β1 was added to the cultured SMMC7721 and HepG2 cells to induce EMT in vitro. Seventy-two hours later, cell morphological changes were observed, the protein expression of E-cadherin was detected by Western blot and immunofluorescence, and the changes in the percentages of SP cells were analyzed by flow cytometry.
RESULTS: An experimental system for the selection of SP cells from hepatocellular carcinoma cells was successfully established. There were 4.3% of SP cells in SMMC-7721 cells, and the expression of ABCG2 (ATP-binding cassette superfamily G member 2) was up-regulated in these SP cells. No SP cells were detected in HepG2 cells. After exposure to TGF-β1 for 72 h, SMMC-7721 cells showed significant morphological changes and reduced E-cadherin expression and SP cell percentage.
CONCLUSION: TGF-β1 is able to induce EMT and reduce the percentage of side population cells in hepatocellular carcinoma cell lines, suggesting that EMT may promote the invasion and metastasis of carcinoma cells by suppressing the proliferation of cancer stem cells.
Collapse
|