1
|
Jiang Y, Glandorff C, Sun M. GSH and Ferroptosis: Side-by-Side Partners in the Fight against Tumors. Antioxidants (Basel) 2024; 13:697. [PMID: 38929136 PMCID: PMC11201279 DOI: 10.3390/antiox13060697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Glutathione (GSH), a prominent antioxidant in organisms, exhibits diverse biological functions and is crucial in safeguarding cells against oxidative harm and upholding a stable redox milieu. The metabolism of GSH is implicated in numerous diseases, particularly in the progression of malignant tumors. Consequently, therapeutic strategies targeting the regulation of GSH synthesis and metabolism to modulate GSH levels represent a promising avenue for future research. This study aimed to elucidate the intricate relationship between GSH metabolism and ferroptosis, highlighting how modulation of GSH metabolism can impact cellular susceptibility to ferroptosis and consequently influence the development of tumors and other diseases. The paper provides a comprehensive overview of the physiological functions of GSH, including its structural characteristics, physicochemical properties, sources, and metabolic pathways, as well as investigate the molecular mechanisms underlying GSH regulation of ferroptosis and potential therapeutic interventions. Unraveling the biological role of GSH holds promise for individuals afflicted with tumors.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Christian Glandorff
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- University Clinic of Hamburg at the HanseMerkur Center of TCM, 20251 Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
2
|
Yang K, Yi T. Tumor cell stemness in gastrointestinal cancer: regulation and targeted therapy. Front Mol Biosci 2024; 10:1297611. [PMID: 38455361 PMCID: PMC10918437 DOI: 10.3389/fmolb.2023.1297611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024] Open
Abstract
The cancer stem cells are a rare group of self-renewable cancer cells capable of the initiation, progression, metastasis and recurrence of tumors, and also a key contributor to the therapeutic resistance. Thus, understanding the molecular mechanism of tumor stemness regulation, especially in the gastrointestinal (GI) cancers, is of great importance for targeting CSC and designing novel therapeutic strategies. This review aims to elucidate current advancements in the understanding of CSC regulation, including CSC biomarkers, signaling pathways, and non-coding RNAs. We will also provide a comprehensive view on how the tumor microenvironment (TME) display an overall tumor-promoting effect, including the recruitment and impact of cancer-associated fibroblasts (CAFs), the establishment of an immunosuppressive milieu, and the induction of angiogenesis and hypoxia. Lastly, this review consolidates mainstream novel therapeutic interventions targeting CSC stemness regulation.
Collapse
Affiliation(s)
- Kangqi Yang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tuo Yi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Hayase E, Ara T, Saito Y, Takahashi S, Yoshioka K, Ohigashi H, Ogasawara R, Yokoyama E, Yamakawa T, Ebata K, Hasegawa Y, Tomizuka K, Teshima T. R-Spondin1 protects gastric stem cells and mitigates gastric GVHD in allogeneic hematopoietic stem cell transplantation. Blood Adv 2024; 8:725-731. [PMID: 38147622 PMCID: PMC10847017 DOI: 10.1182/bloodadvances.2023011034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/27/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023] Open
Abstract
ABSTRACT Graft-versus-host disease (GVHD) is the major obstacle to performing allogeneic hematopoietic cell transplantation (allo-HCT). We and others have shown that intestinal stem cells are targeted in lower gastrointestinal GVHD. A leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5)-expressing gastric stem cells (GSCs) reside at the base of the gastric glands in mice. After experimental allo-HCT, Lgr5+ GSCs significantly decreased. Parietal cells, which underwent continuous renewal by GSCs, were injured in gastric GVHD, leading to failure of gastric acidification and aerobic bacterial overgrowth in the duodenum. Fate-mapping analysis demonstrated that administration of R-Spondin1 (R-Spo1) that binds to Lgr5 for 6 days in naïve mice significantly increased proliferating epithelial cells derived from Lgr5+ GSCs. R-Spo1 administered on days -3 to -1 and from days +1 to +3 of allo-HCT protected GSCs, leading to amelioration of gastric GVHD and restoration of gastric acidification, and suppression of aerobic bacterial overgrowth in the duodenum. In conclusion, Lgr5+ GSCs were targeted by gastric GVHD, resulting in disruption of the gastric homeostasis, whereas R-Spo1 protected Lgr5+ GSCs from GVHD and maintained homeostasis in the stomach.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Takahide Ara
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Yumika Saito
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Shuichiro Takahashi
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Kosuke Yoshioka
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Reiki Ogasawara
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Emi Yokoyama
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Tomohiro Yamakawa
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Ko Ebata
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Yuta Hasegawa
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
4
|
Cheng Q, Hu X, Zhang X, Yang D, Zhao G, Sun L, Jiang M, Yang L, Cai J, Wang B, Zhang M, Han F, Li Y, Nie H. N-glycosylation at N57/100/110 affects CD44s localization, function and stability in hepatocellular carcinoma. Eur J Cell Biol 2023; 102:151360. [PMID: 37703748 DOI: 10.1016/j.ejcb.2023.151360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/23/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
The glycosylation levels of proteins in cancer cells are closely related to cancer invasion and migration. CD44 is a transmembrane glycoprotein that is significantly overexpressed in a variety of tumor cells and has been proven to promote the migration and motility of cancer cells, but the effect of its N-glycosylation modification on CD44 protein function in tumors is less studied. Here, we investigated the effect of six N-glycan chains (N25/57/100/110/120/255) on CD44s localization, function and stability in hepatocarcinoma cells. When the six sites were mutated, we found that CD44s lost its membrane localization in Huh7 and MHCC-97H cells. On this basis, we identified three glycosylation sites on CD44s (N57, N100 and N110) that played key roles in intracellular localization. When N57, N100 and N110 were mutated together, CD44 localized to the cytoplasm, while another three-site mutant (N25/N120/N255) was still anchored to the membrane. In addition, the ability of CD44-N57Q/N100Q/N110Q to promote the metastasis and invasion of Huh7 and 97H cells was weakened compared with that of CD44-N25Q/N120Q/N255Q. Furthermore, CD44-N57Q/N100Q/N110Q accumulated abnormally in the ER, and a high level of the ER stress (ERS) marker BiP was detected at the same time compared with wild-type CD44. When the lysosome inhibitor CQ was added, the content of mutant protein that triggered ERS significantly increased, which indicated that the degradation mode of CD44-N57Q/N100Q/N110Q after ERS was mainly through the lysosomal pathway (ERLAD). The results revealed that the N-glycosylation sites N57, N100 and N110 mutated on CD44s affected its function and degraded it by lysosomes after triggering ERS. These findings provide data for new studies on ER-related degradation, further promote the study of the glycan chain function of CD44 and furnish new ideas for the treatment of liver cancer metastasis.
Collapse
Affiliation(s)
- Qixiang Cheng
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China; Hebei Normal University of Science & Technology, Qinhuangdao, Hebei 066004, China
| | - Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Guiping Zhao
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Liping Sun
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Meiyi Jiang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Bing Wang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Mengmeng Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Fang Han
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
5
|
Zhang Y, Zhang X, Huang X, Tang X, Zhang M, Li Z, Hu X, Zhang M, Wang X, Yan Y. Tumor stemness score to estimate epithelial-to-mesenchymal transition (EMT) and cancer stem cells (CSCs) characterization and to predict the prognosis and immunotherapy response in bladder urothelial carcinoma. Stem Cell Res Ther 2023; 14:15. [PMID: 36721217 PMCID: PMC9890713 DOI: 10.1186/s13287-023-03239-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/13/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND A growing number of investigations have suggested a close link between cancer stem cells (CSCs), epithelial-to-mesenchymal transition (EMT), and the tumor microenvironment (TME). However, the relationships between these physiological processes in bladder urothelial carcinoma (BLCA) remain unclear. METHODS We first explored biomarkers of tumor stemness (TS) by single-cell sequencing analysis. Then, subtypes of bladder urothelial carcinoma (BLCA) were identified using clustering analysis based on TS biomarkers. The TS score was constructed using principal component analysis to quantify tumor stemness in BLCA. Then, meta-analysis was performed to measure the hazard ratio of the TS score in BLCA cohorts. Moreover, we evaluated the clinical value of the TS score for predicting the response to tumor immunotherapy using immunotherapy cohorts. Finally, we built an EMT cell model by treating T24 cells with TGF-β and validated the relationship between the TS score and the EMT process in tumors by real-time quantitative PCR, cell invasion assays, and RNA-seq. In total, 3846 BLCA cells, 6 cell lines, 1627 BLCA samples, and 9858 samples from 32 other types of tumors were included in our study. RESULTS Three TS clusters and two TS-related gene clusters were identified with differential EMT activity status, CSC features, and TME characteristics in BLCA. Then, a TS scoring system was established with 61 TS-related genes to quantify the TS. The prognostic value of the TS score was then confirmed in multiple independent cohorts. A high TS score was associated with high EMT activity, CSC characteristics, high stromal cell content, high TP53 mutation rate, poor prognosis, and high tumor immunotherapy tolerance. The cell line experiment and RNA-seq further validated that our TS score can reflect the EMT and CSC characterization of tumor cells. CONCLUSION Overall, this research provides a better understanding of tumor invasion and metastasis mechanisms through an analysis of TS patterns with different EMT processes and CSC characteristics. The TS score provides an index for EMT and CSC research and helps clinicians develop treatment plans and predict outcomes for patients.
Collapse
Affiliation(s)
- Yanlong Zhang
- Department of Urology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Xuefeng Huang
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaomeng Tang
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Menghan Zhang
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziyi Li
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Min Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Xi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
- Beijing Institute of Infectious Diseases, Beijing, 100015 China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015 China
| | - Yong Yan
- Department of Urology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Yang KT, Yen CC, Chang R, Wang JT, Chen JS. CAST as a Potential Oncogene, Identified by Machine Search, in Gastric Cancer Infiltrated with Macrophages and Associated with Lgr5. Biomolecules 2022; 12:biom12050670. [PMID: 35625600 PMCID: PMC9138541 DOI: 10.3390/biom12050670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/25/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the leading malignant diseases worldwide, especially in Asia. CAST is a potential oncogene in GC carcinogenesis. The character of macrophage infiltration in the GC microenvironment also remains unaddressed. Methods: We first applied machine searching to evaluate gene candidates for GC. CAST expression and pan-cancer surveyance were analyzed using the Human Protein Atlas (HPA) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) database. The protein–protein interaction (PPI) network was downloaded from STRING. We investigated the impact of CAST on clinical prognosis using a Kaplan–Meier plotter. The correlations between CAST and Lgr5 and macrophage infiltration in GC were determined using TIMER 2.0. Finally, GeneMANIA was also used to evaluate the possible functional linkages between genes. Results: After the machine-assisted search, CAST expression was found to significantly influence the overall survival of GC patients. STRING revealed CAST-related proteomic and transcriptomic associations, mainly concerning the CAPN family. Moreover, CAST significantly impacts the prognosis of GC based on the validation of other datasets. Notably, high CAST expression was correlated with worse overall survival in GC patients (hazard ratio = 1.59; log-rank P = 9.4 × 10−8). CAST and Lgr5 expression were both positively correlated with WNT 2 and WNT 2B. Among the GC patients in several datasets, CAST and macrophage infiltration, evaluated together, showed no obvious association with poor clinical overall survival. Conclusions: CAST plays an important role in the clinical prognosis of GC and is associated with WNT 2/WNT 2B/Lgr5. Our study demonstrates that CAST’s influence on overall survival in GC is regulated by macrophage infiltration.
Collapse
Affiliation(s)
- Kuang-Tsu Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802213, Taiwan;
- Division of Family Medicine, Department of Community Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802213, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chia-Chi Yen
- Superintendent’s Office, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 804201, Taiwan;
- Department of Nutrition, Institute of Biomedical Nutrition, Hung-Kuang University, Taichung 433304, Taiwan
- Department of Business Management, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
- Department of Recreation and Sports Management, Tajen University, Pingtung 90741, Taiwan
| | - Jui-Tzu Wang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
| | - Jin-Shuen Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Faculty of Medicine, School of Medicine, National Defense Medicine, Taipei 11490, Taiwan
- Correspondence: ; Tel.: +886-7-342-2121
| |
Collapse
|
7
|
Wilson DJ, DuBois RN. Role of prostaglandin E2 in the progression of gastrointestinal cancer. Cancer Prev Res (Phila) 2022; 15:355-363. [PMID: 35288737 DOI: 10.1158/1940-6207.capr-22-0038] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/01/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022]
Abstract
Chronic inflammation is a well-established risk factor for several diseases, including cancer. It influences tumor cell biology and the type and density of immune cells in the tumor microenvironment (TME), promoting cancer development. While pro-inflammatory cytokines and chemokines modulate cancer development, emerging evidence has shown that prostaglandin E2 (PGE2) is a known mediator connecting chronic inflammation to cancerization. This review highlights recent advances in our understanding of how the elevation of PGE2 production promotes gastrointestinal cancer initiation, progression, invasion, metastasis, and recurrence, including modulation of immune checkpoint signaling and the type and density of immune cells in the tumor/tissue microenvironment.
Collapse
Affiliation(s)
- David Jay Wilson
- Medical University of South Carolina, Greenville, South Carolina, United States
| | - Raymond N DuBois
- Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
8
|
Jing Y, Liang W, Zhang L, Tang J, Huang Z. The Role of Mesenchymal Stem Cells in the Induction of Cancer-Stem Cell Phenotype. Front Oncol 2022; 12:817971. [PMID: 35251985 PMCID: PMC8891610 DOI: 10.3389/fonc.2022.817971] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSCs) modify and form their microenvironment by recruiting and activating specific cell types such as mesenchymal stem cells (MSCs). Tumor-infiltrating MSCs help to establish a suitable tumor microenvironment for the restoration of CSCs and tumor progression. In addition, crosstalk between cancer cells and MSCs in the microenvironment induces a CSC phenotype in cancer cells. Many mechanisms are involved in crosstalk between CSCs/cancer cells and MSCs including cell-cell interaction, secretion of exosomes, and paracrine secretion of several molecules including inflammatory mediators, cytokines, and growth factors. Since this crosstalk may contribute to drug resistance, metastasis, and tumor growth, it is suggested that blockade of the crosstalk between MSCs and CSCs/cancer cells can provide a new avenue to improving the cancer therapeutic tools. In this review, we will discuss the role of MSCs in the induction of cancer stem cell phenotype and the restoration of CSCs. We also discuss targeting the crosstalk between MSCs and CSCs/cancer cells as a therapeutic strategy.
Collapse
Affiliation(s)
- Yuanming Jing
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Junjun Tang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zongliang Huang, ; Junjun Tang ,
| | - Zongliang Huang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zongliang Huang, ; Junjun Tang ,
| |
Collapse
|
9
|
Yamasaki J, Hirata Y, Otsuki Y, Suina K, Saito Y, Masuda K, Okazaki S, Ishimoto T, Saya H, Nagano O. MEK Inhibition Suppresses Metastatic Progression of KRAS-Mutated Gastric Cancer. Cancer Sci 2021; 113:916-925. [PMID: 34931404 PMCID: PMC8898706 DOI: 10.1111/cas.15244] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022] Open
Abstract
Metastatic progression of tumors is driven by genetic alterations and tumor‐stroma interaction. To elucidate the mechanism underlying the oncogene‐induced gastric tumor progression, we have developed an organoid‐based model of gastric cancer from GAstric Neoplasia (GAN) mice, which express Wnt1 and the enzymes COX2 and microsomal prostaglandin E synthase 1 in the stomach. Both p53 knockout (GAN‐p53KO) organoids and KRASG12V‐expressing GAN‐p53KO (GAN‐KP) organoids were generated by genetic manipulation of GAN mouse‐derived tumor (GAN wild‐type [WT]) organoids. In contrast with GAN‐WT and GAN‐p53KO organoids, which manifested Wnt addiction, GAN‐KP organoids showed a Wnt‐independent phenotype and the ability to proliferate without formation of a Wnt‐regulated three‐dimensional epithelial architecture. After transplantation in syngeneic mouse stomach, GAN‐p53KO cells formed only small tumors, whereas GAN‐KP cells gave rise to invasive tumors associated with the development of hypoxia as well as to liver metastasis. Spatial transcriptomics analysis suggested that hypoxia signaling contributes to the metastatic progression of GAN‐KP tumors. In particular, such analysis identified a cluster of stromal cells located at the tumor invasive front that expressed genes related to hypoxia signaling, angiogenesis, and cell migration. These cells were also positive for phosphorylated extracellular signal‐regulated kinase (ERK), suggesting that mitogen‐activated protein kinase (MAPK) signaling promotes development of both tumor and microenvironment. The MEK (MAPK kinase) inhibitor trametinib suppressed the development of GAN‐KP gastric tumors, formation of a hypoxic microenvironment, tumor angiogenesis, and liver metastasis. Our findings therefore establish a rationale for application of trametinib to suppress metastatic progression of KRAS‐mutated gastric cancer.
Collapse
Affiliation(s)
- Juntaro Yamasaki
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Yuki Hirata
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Yuji Otsuki
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Kentaro Suina
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Yoshiyuki Saito
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan
| | - Shogo Okazaki
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Takatsugu Ishimoto
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Osamu Nagano
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
10
|
Wang D, Cabalag CS, Clemons NJ, DuBois RN. Cyclooxygenases and Prostaglandins in Tumor Immunology and Microenvironment of Gastrointestinal Cancer. Gastroenterology 2021; 161:1813-1829. [PMID: 34606846 DOI: 10.1053/j.gastro.2021.09.059] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/03/2021] [Accepted: 09/19/2021] [Indexed: 12/21/2022]
Abstract
Chronic inflammation is a known risk factor for gastrointestinal cancer. The evidence that nonsteroidal anti-inflammatory drugs suppress the incidence, growth, and metastasis of gastrointestinal cancer supports the concept that a nonsteroidal anti-inflammatory drug target, cyclooxygenase, and its downstream bioactive lipid products may provide one of the links between inflammation and cancer. Preclinical studies have demonstrated that the cyclooxygenase-2-prostaglandin E2 pathway can promote gastrointestinal cancer development. Although the role of this pathway in cancer has been investigated extensively for 2 decades, only recent studies have described its effects on host defenses against transformed epithelial cells. Overcoming tumor-immune evasion remains one of the major challenges in cancer immunotherapy. This review summarizes the impacts of the cyclooxygenase-2-prostaglandin E2 pathway on gastrointestinal cancer development. Our focus was to highlight recent advances in our understanding of how this pathway induces tumor immune evasion.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Carlos S Cabalag
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas J Clemons
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Raymond N DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
11
|
Hayashi N, Yamasaki A, Ueda S, Okazaki S, Ohno Y, Tanaka T, Endo Y, Tomioka Y, Masuko K, Masuko T, Sugiura R. Oncogenic transformation of NIH/3T3 cells by the overexpression of L-type amino acid transporter 1, a promising anti-cancer target. Oncotarget 2021; 12:1256-1270. [PMID: 34194623 PMCID: PMC8238248 DOI: 10.18632/oncotarget.27981] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/26/2021] [Indexed: 01/10/2023] Open
Abstract
L-type amino acid transporter 1 (LAT1)/SLC7A5 is the first identified CD98 light chain disulfide linked to the CD98 heavy chain (CD98hc/SLC3A2). LAT1 transports large neutral amino acids, including leucine, which activates mTOR, and is highly expressed in human cancers. We investigated the oncogenicity of human LAT1 introduced to NIH/3T3 cells by retrovirus infection. NIH/3T3 cell lines stably expressing human native (164C) or mutant (164S) LAT1 (naLAT1/3T3 or muLAT1/3T3, respectively) were established. We confirmed that endogenous mouse CD98hc forms a disulfide bond with exogenous human LAT1 in naLAT1/3T3, but not in muLAT1/3T3. Endogenous mouse CD98hc mRNA increased in both naNIH/3T3 and muLAT1/3T3, and a similar amount of exogenous human LAT1 protein was detected in both cell lines. Furthermore, naLAT1/3T3 and muLAT1/3T3 cell lines were evaluated for cell growth-related phenotypes (phosphorylation of ERK, cell-cycle progression) and cell malignancy-related phenotypes (anchorage-independent cell growth, tumor formation in nude mice). naLAT1/3T3 had stronger growth- and malignancy- related phenotypes than NIH/3T3 and muLAT1/3T3, suggesting the oncogenicity of native LAT1 through its interaction with CD98hc. Anti-LAT1 monoclonal antibodies significantly inhibited in vitro cell proliferation and in vivo tumor growth of naLAT1/3T3 cells in nude mice, demonstrating LAT1 to be a promising anti-cancer target.
Collapse
Affiliation(s)
- Natsumi Hayashi
- Laboratory of Molecular Pharmacogenomics, Faculty of Pharmacy, Kindai University, Higashiosaka-Shi, Osaka, Japan.,Cell Biology Laboratory, School of Pharmacy, Kindai University, Osaka, Japan.,Co-first authors.,This laboratory (April, 2000~) was closed at the end of March, 2020, after the mandatory retirement of Takashi Masuko
| | - Akitaka Yamasaki
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Osaka, Japan.,Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-Shi, Miyagi, Japan.,Co-first authors.,This laboratory (April, 2000~) was closed at the end of March, 2020, after the mandatory retirement of Takashi Masuko
| | - Shiho Ueda
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Osaka, Japan
| | - Shogo Okazaki
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba, Japan
| | - Yoshiya Ohno
- Laboratory of Immunobiology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe-Shi, Hyogo, Japan
| | - Toshiyuki Tanaka
- Laboratory of Immunobiology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe-Shi, Hyogo, Japan
| | - Yuichi Endo
- Natural Drug Resources, Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Yoshihisa Tomioka
- Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-Shi, Miyagi, Japan
| | - Kazue Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Osaka, Japan
| | - Takashi Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Osaka, Japan.,Natural Drug Resources, Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Reiko Sugiura
- Laboratory of Molecular Pharmacogenomics, Faculty of Pharmacy, Kindai University, Higashiosaka-Shi, Osaka, Japan
| |
Collapse
|
12
|
Slow-cycling (dormant) cancer cells in therapy resistance, cancer relapse and metastasis. Semin Cancer Biol 2021; 78:90-103. [PMID: 33979674 DOI: 10.1016/j.semcancer.2021.04.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that cancer cell heterogeneity and plasticity constitute major barriers to effective clinical treatments and long-term therapeutic efficacy. Research in the past two decades suggest that virtually all treatment-naive human cancers harbor subsets of cancer cells that possess many of the cardinal features of normal stem cells. Such stem-like cancer cells, operationally defined as cancer stem cells (CSCs), are frequently quiescent and dynamically change and evolve during tumor progression and therapeutic interventions. Intrinsic tumor cell heterogeneity is reflected in a different aspect in that tumors also harbor a population of slow-cycling cells (SCCs) that are not in the proliferative cell cycle and thus are intrinsically refractory to anti-mitotic drugs. In this Perspective, we focus our discussions on SCCs in cancer and on various methodologies that can be employed to enrich and purify SCCs, compare the similarities and differences between SCCs, CSCs and cancer cells undergoing EMT, and present evidence for the involvement of SCCs in surviving anti-neoplastic treatments, mediating tumor relapse, maintaining tumor dormancy and mediating metastatic dissemination. Our discussions make it clear that an in-depth understanding of the biological properties of SCCs in cancer will be instrumental to developing new therapeutic strategies to prevent tumor relapse and distant metastasis.
Collapse
|
13
|
Yoshida GJ, Saya H. Molecular pathology underlying the robustness of cancer stem cells. Regen Ther 2021; 17:38-50. [PMID: 33869685 PMCID: PMC8024885 DOI: 10.1016/j.reth.2021.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Intratumoral heterogeneity is tightly associated with the failure of anticancer treatment modalities including conventional chemotherapy, radiation therapy, and molecularly targeted therapy. Such heterogeneity is generated in an evolutionary manner not only as a result of genetic alterations but also by the presence of cancer stem cells (CSCs). CSCs are proposed to exist at the top of a tumor cell hierarchy and are undifferentiated tumor cells that manifest enhanced tumorigenic and metastatic potential, self-renewal capacity, and therapeutic resistance. Properties that contribute to the robustness of CSCs include the abilities to withstand redox stress, to rapidly repair damaged DNA, to adapt to a hyperinflammatory or hyponutritious tumor microenvironment, and to expel anticancer drugs by the action of ATP-binding cassette transporters as well as plasticity with regard to the transition between dormant CSC and transit-amplifying progenitor cell phenotypes. In addition, CSCs manifest the phenomenon of metabolic reprogramming, which is essential for maintenance of their self-renewal potential and their ability to adapt to changes in the tumor microenvironment. Elucidation of the molecular underpinnings of these biological features of CSCs is key to the development of novel anticancer therapies. In this review, we highlight the pathological relevance of CSCs in terms of their hallmarks and identification, the properties of their niche—both in primary tumors and at potential sites of metastasis—and their resistance to oxidative stress dependent on system xc (−). Intratumoral heterogeneity driven by CSCs is responsible for therapeutic resistance. CTCs survive in the distant organs and achieve colonization, causing metastasis. E/M hybrid cancer cells due to partial EMT exhibit the highest metastatic potential. The CSC niche regulates stemness in metastatic disease as well as in primary tumor. Activation of system xc(-) by CD44 variant in CSCs is a promising therapeutic target.
Collapse
Key Words
- ABC, ATP-binding cassette
- ALDH, Aldehyde dehydrogenase
- BMP, Bone morphogenetic protein
- CAF, Cancer-associated fibroblast
- CD44 variant
- CD44v, CD44 variant
- CSC, Cancer stem cell
- CTC, Circulating tumor cell
- CagA, Cytotoxin-associated gene A
- Cancer stem cell
- DTC, Disseminated tumor cell
- E/M, Epithelial/mesenchymal
- ECM, Extracellular matrix
- EGF, Epidermal growth factor
- EMT, Epithelial-to-mesenchymal transition
- EpCAM, Epithelial cell adhesion moleculeE
- Epithelial-to-mesenchymal transition (EMT)
- GSC, Glioma stem cell
- GSH, reduced glutathione
- HGF, Hepatocyte growth factor
- HNSCC, Head and neck squamous cell cancer
- IL, Interleukin
- Intratumoral heterogeneity
- MAPK, mitogen-activated protein kinase
- MET, mesenchymal-to-epithelial transition
- NSCLC, non–small cell lung cancer
- Niche
- Nrf2, nuclear factor erythroid 2–related factor 2
- OXPHOS, Oxidative phosphorylation
- Plasticity
- Prrx1, Paired-related homeodomain transcription factor 1
- ROS, Reactive oxygen species
- SRP1, Epithelial splicing regulatory protein 1
- TGF-β, Transforming growth factor–β
Collapse
Affiliation(s)
- Go J Yoshida
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research (IAMR), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Bali P, Lozano-Pope I, Pachow C, Obonyo M. Early detection of tumor cells in bone marrow and peripheral blood in a fast‑progressing gastric cancer model. Int J Oncol 2021; 58:388-396. [PMID: 33469673 PMCID: PMC7864146 DOI: 10.3892/ijo.2021.5171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection is a major risk factor for the development of gastric cancer. The authors previously demonstrated that in mice deficient in myeloid differentiation primary response 88 (Myd88−/−), infection with Helicobacter felis (H. felis) a close relative of H. pylori, subsequently rapidly progressed to neoplasia. The present study examined circulating tumor cells (CTCs) by measuring the expression of cytokeratins, epithelial-to-mesenchymal transition (EMT)-related markers and cancer stem cell (CSC) markers in bone marrow and peripheral blood from Myd88−/− and wild-type (WT) mice. Cytokeratins CK8/18 were detected as early as 4 months post-infection in Myd88−/− mice. By contrast, cytokeratins were not detected in WT mice even after 7 months post-infection. The expression of Mucin-1 (MUC1) was observed in both bone marrow and peripheral blood at different time points, suggesting its role in gastric cancer metastasis. Snail, Twist and ZEB were expressed at different levels in bone marrow and peripheral blood. The expression of these EMT-related markers suggests the manifestation of cancer metastasis in the early stages of disease development. LGR5, CD44 and CD133 were the most prominent CSC markers detected. The detection of CSC and EMT markers along with cytokeratins does reinforce their use as biomarkers for gastric cancer metastasis. This early detection of markers suggests that CTCs leave primary site even before cancer is well established. Thus, cytokeratins, EMT, and CSCs could be used as biomarkers to detect aggressive forms of gastric cancers. This information may prove to be of significance in stratifying patients for treatment prior to the onset of severe disease-related characteristics.
Collapse
Affiliation(s)
- Prerna Bali
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Ivonne Lozano-Pope
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Collin Pachow
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Marygorret Obonyo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| |
Collapse
|
15
|
Abstract
Target molecules of existing anti-cancer therapeutic monoclonal antibodies (mAbs) are divided into 1) receptor-type tyrosine kinases, such as human epidermal growth factor receptor (HER) family, 2) differentiation antigens, such as CD20 (Rituxan target), 3) angiogenesis-related molecules, and 4) immune checkpoint molecules (PD-1, etc.). We have recently reported a novel therapy targeting lymphangiogenesis, but not angiogenesis, using an anti-LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1) mAb. At present, many transporters are not considered to be target molecules for the cancer therapy; however, our study strongly suggested that the inhibition of cancer metabolism by mAbs against amino acid transporters will play a significant role in future cancer therapies. Most anti-cancer therapeutic mAbs bind cell-surface molecules on viable cancer cells: therefore, it is necessary to produce mAbs recognizing epitopes on the extracellular domains of native and non-denatured proteins. We concluded that viable cancer cells or cells transfected with cDNA encoding target proteins are suitable immunogens for the production of anti-cancer therapeutic mAbs. We introduce our efforts to develop seeds for therapeutic mAbs using whole cancer cells and transfectants as the immunogen. As many target candidates in the future are multi-pass membrane proteins, such as 12-pass amino acid transporter proteins belonging to the solute carrier (SLC) family, and their possible immunogenic extracellular regions are small, the production of specific mAbs is highly difficult. In this review, we summarize the successful preparation and characterization of mAbs recognizing the extracellular domain of oncoproteins, including transporters.
Collapse
Affiliation(s)
- Takashi Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University
| |
Collapse
|
16
|
Elgendy SM, Alyammahi SK, Alhamad DW, Abdin SM, Omar HA. Ferroptosis: An emerging approach for targeting cancer stem cells and drug resistance. Crit Rev Oncol Hematol 2020; 155:103095. [PMID: 32927333 DOI: 10.1016/j.critrevonc.2020.103095] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/08/2023] Open
Abstract
Resistance to chemotherapeutic agents remains a major challenge in the fierce battle against cancer. Cancer stem cells (CSCs) are a small population of cells in tumors that possesses the ability to self-renew, initiate tumors, and cause resistance to conventional anticancer agents. Targeting this population of cells was proven as a promising approach to eliminate cancer recurrence and improve the clinical outcome. CSCs are less susceptible to death by classical anticancer agents inducing apoptosis. CSCs can be eradicated by ferroptosis, which is a non-apoptotic-regulated mechanism of cell death. The induction of ferroptosis is an attractive strategy to eliminate tumors due to its ability to selectively target aggressive CSCs. The current review critically explored the crosstalk and regulatory pathways controlling ferroptosis, which can selectively induce CSCs death. In addition, successful chemotherapeutic agents that achieve better therapeutic outcomes through the induction of ferroptosis in CSCs were discussed to highlight their promising clinical impact.
Collapse
Affiliation(s)
- Sara M Elgendy
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Shatha K Alyammahi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Dima W Alhamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Shifaa M Abdin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
17
|
The Prognostic Value of EMT in Glioma and its Role in the Glioma Immune Microenvironment. J Mol Neurosci 2020; 70:1501-1511. [PMID: 32495005 DOI: 10.1007/s12031-020-01583-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/13/2020] [Indexed: 01/06/2023]
Abstract
Diffuse glioma is the deadliest form of brain cancer, and the median survival of grade IV glioma (glioblastoma, GBM) is no more than 2 years even with maximal surgical resection followed by radiotherapy and chemotherapy, which are now the standard of care for GBM. Glioma shares common characteristics with most malignant tumours, such as invasiveness, rapid progression, resistance to various therapies and inevitable recurrence, while it also has its own unique features, such as high aggressiveness and immunotherapy resistance, which can be, respectively, attributed to epithelial-mesenchymal transition (EMT) and the immunosuppressive microenvironment. Here, we calculated the EMT score of glioma using The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA) and the Gene Expression Omnibus (GEO) datasets and validated its prognostic value. Then, we investigated its role in the glioma immune microenvironment, identified the enriched EMT-related immune genes and determined their specific biological functions in glioma. Furthermore, clinical relevance analysis showed the translational value of these EMT-related immune genes. In short, our findings reveal a critical link between EMT and the glioma immune microenvironment and offer important clues for further investigation of the underlying molecular mechanism.
Collapse
|
18
|
Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, Staedel C, Boeuf H, Fernandez S, Barthelemy P, Megraud F, Lehours P, Dubus P, Varon C. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer 2019; 146:2255-2267. [PMID: 31489619 DOI: 10.1002/ijc.32667] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/07/2019] [Indexed: 12/28/2022]
Abstract
Gastric carcinomas (GC) are heterogeneous tumors, composed of a subpopulation of cluster of differentiation-44 (CD44)+ tumorigenic and chemoresistant cancer stem cells (CSC). YAP1 and TAZ oncoproteins (Y/T) interact with TEA domain family member 1 (TEAD) transcription factors to promote cell survival and proliferation in multiple tissues. Their activity and role in GC remain unclear. This work aimed to analyze Y/T-TEAD activity and molecular signature in gastric CSC, and to assess the effect of verteporfin, a Food and Drug Administration-approved drug preventing Y/T-TEAD interaction, on gastric CSC tumorigenic properties. Y/T-TEAD molecular signature was investigated using bioinformatical (KmPlot database), transcriptomic and immunostaining analyses in patient-derived GC and cell lines. Verteporfin effects on Y/T-TEAD transcriptional activity, CSC proliferation and tumorigenic properties were evaluated using in vitro tumorsphere assays and mouse models of patient-derived GC xenografts. High expressions of YAP1, TAZ, TEAD1, TEAD4 and their target genes were associated with low overall survival in nonmetastatic human GC patients (n = 444). This Y/T-TEAD molecular signature was enriched in CD44+ patient-derived GC cells and in cells resistant to conventional chemotherapy. Verteporfin treatment inhibited Y/T-TEAD transcriptional activity, cell proliferation and CD44 expression, and decreased the pool of tumorsphere-forming CD44+ /aldehyde dehydrogenase (ALDH)high gastric CSC. Finally, verteporfin treatment inhibited GC tumor growth in vivo; the residual tumor cells exhibited reduced expressions of CD44 and ALDH1, and more importantly, they were unable to initiate new tumorspheres in vitro. All these data demonstrate that Y/T-TEAD activity controls gastric CSC tumorigenic properties. The repositioning of verteporfin targeting YAP1/TAZ-TEAD activity could be a promising CSC-based strategy for the treatment of GC.
Collapse
Affiliation(s)
- Julie Giraud
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Silvia Molina-Castro
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France.,INISA/School of Medicine, University of Costa Rica, San José, Costa Rica
| | - Lornella Seeneevassen
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Elodie Sifré
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Julien Izotte
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Camille Tiffon
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Cathy Staedel
- INSERM U1212, CNRS UMR5320, ARNA Laboratory, University of Bordeaux, Bordeaux, France
| | - Hélène Boeuf
- INSERM U1026 BioTIS, University of Bordeaux, Bordeaux, France
| | - Solène Fernandez
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| | - Philippe Barthelemy
- INSERM U1212, CNRS UMR5320, ARNA Laboratory, University of Bordeaux, Bordeaux, France
| | - Francis Megraud
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Bordeaux, France.,National Reference Center for Campylobacters and Helicobacters, Bordeaux, France
| | - Philippe Lehours
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Bordeaux, France.,National Reference Center for Campylobacters and Helicobacters, Bordeaux, France
| | - Pierre Dubus
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Bordeaux, France
| | - Christine Varon
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux, France
| |
Collapse
|
19
|
Bild A, Teo JL, Kahn M. Enhanced Kat3A/Catenin transcription: a common mechanism of therapeutic resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:917-932. [PMID: 32426696 PMCID: PMC7234864 DOI: 10.20517/cdr.2019.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/04/2019] [Accepted: 06/18/2019] [Indexed: 11/12/2022]
Abstract
Cancers are heterogeneous at the cellular level. Cancer stem cells/tumor initiating cells (CSC/TIC) both initiate tumorigenesis and are responsible for therapeutic resistance and disease relapse. Elimination of CSC/TIC should therefore be able to reverse therapy resistance. In principle, this could be accomplished by either targeting cancer stem cell surface markers or "stemness" pathways. Although the successful therapeutic elimination of "cancer stemness" is a critical goal, it is complex in that it should be achieved without depletion of or increases in somatic mutations in normal tissue stem cell populations. In this perspective, we will discuss the prospects for this goal via pharmacologically targeting differential Kat3 coactivator/Catenin usage, a fundamental transcriptional control mechanism in stem cell biology.
Collapse
Affiliation(s)
- Andrea Bild
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Jia-Ling Teo
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Michael Kahn
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
20
|
Yang D, Wang X, Yuan W, Chen Z. Intake of Anthocyanins and Gastric Cancer Risk: A Comprehensive Meta-Analysis on Cohort and Case-Control Studies. J Nutr Sci Vitaminol (Tokyo) 2019; 65:72-81. [PMID: 30814415 DOI: 10.3177/jnsv.65.72] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This meta-analysis aimed to explore the association between anthocyanins intake and the risk of gastric cancer. All the relative articles have been searched in the online databases, including PubMed, EMBASE, Web of Science, and the Cochrane Library until June 11th, 2018. Risk ratios (RRs) or odds ratio (ORs) and their 95% confidence intervals were calculated and pooled through the STATA 12.0. A total of 6 studies were finally selected in the meta-analysis. No significant association was found between total anthocyanins consumption and gastric cancer risk (RR=0.92, 95%CI: 0.81-1.04). Likewise, there was also no significant evidence of the relationship between anthocyanins intake and gastric cancer in tumor site (cardia: RR=0.90, 95%CI: 0.62-1.31; noncardia: RR=0.86, 95%CI: 0.69-1.07) and gender (men: RR=1.02, 95%CI: 0.73-1.40; women: RR=0.80, 95%CI: 0.52-1.23). The dose-response relationship was also not found in this meta-analysis. The Grades of Recommendations Assessment, Development and Evaluation (GRADE) quality in our study was very low. The results of our meta-analysis suggested the intake of anthocyanins had no association with the risk of gastric cancer and further studies are needed.
Collapse
Affiliation(s)
- DeYi Yang
- Department of General Surgery, Xiangya Hospital, Central South University
| | - Xin Wang
- Department of General Surgery, Shijitan Hospital of Capital Medical University
| | - WeiJie Yuan
- Department of General Surgery, Xiangya Hospital, Central South University
| | - ZiHua Chen
- Department of General Surgery, Xiangya Hospital, Central South University
| |
Collapse
|
21
|
Mutated Rnf43 Aggravates Helicobacter Pylori-Induced Gastric Pathology. Cancers (Basel) 2019; 11:cancers11030372. [PMID: 30884828 PMCID: PMC6468876 DOI: 10.3390/cancers11030372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/10/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
The E3 ubiquitin ligase ring finger protein 43 (RNF43) is frequently mutated in gastric tumors and loss of RNF43 expression was suggested to be one of the key events during the transition from adenoma to gastric carcinoma. Functional studies on RNF43 have shown that it acts as a tumor suppressor by negatively regulating Wnt signaling. Interestingly, we observed that RNF43H292R/H295R mice bearing two point mutations in the ring domain displayed thickening of the mucosa at early age but did not develop neoplasia. In this study, we infected these mice for 6 months with Helicobacter pylori, which has been described as one of the major risk factors for gastric cancer. Mice bearing mutant RNF43H292R/H295R showed higher gastritis scores upon H. pylori infection compared to wild-type mice, accompanied by increased lymphocyte infiltration and Ifng levels. Furthermore, infected Rnf43 mutant mice developed atrophy, hyperplasia and MUC2 expressing metaplasia and displayed higher levels of the gastric stem cell marker CD44 and canonical NF-κB signaling. In summary, our results show that transactivating mutations in the tumor suppressor Rnf43 can worsen H. pylori induced pathology.
Collapse
|
22
|
Go SI, Ko GH, Lee WS, Lee JH, Jeong SH, Lee YJ, Hong SC, Ha WS. The Use of CD44 Variant 9 and Ki-67 Combination Can Predicts Prognosis Better Than Their Single Use in Early Gastric Cancer. Cancer Res Treat 2019; 51:1411-1419. [PMID: 30913874 PMCID: PMC6790844 DOI: 10.4143/crt.2018.663] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/23/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose We previously demonstrated that CD44v9 and Ki-67 played an important role in predicting poor prognosis of early gastric cancer (EGC). However, little is known about combined use of both biomarkers as prognostic biomarker. The present study was performed to investigate the significance of CD44v9 and Ki-67 expression as a combination biomarker for EGC. Materials and Methods With tissue microarray for 158 EGC tissues, we performed immunohistochemical staining for CD44v9 and Ki-67. The whole patients were divided into three groups (group A, CD44v9-negative/Ki-67–low; group B, neither group A or C; and group C, CD44v9-positive/Ki-67–high). Its clinical significance was re-analyzed with adjustment via propensity score matching (PSM). For validation, we performed bootstrap resampling. Results The median follow-up duration was 90.4 months (range, 3.7 to 120.4 months). In the comparison according to CD44v9/Ki-67 expression, the combined use of the two biomarker clearly separated the three groups by 5-year survival rates (5-YSR, 96.3%, 89.8%, and 76.8% in group A, B, and C, respectively; p=0.009). After PSM, 5-YSR were 97.7% and 76.8% in group A+B and group C, respectively (p=0.002). Multivariable analysis demonstrated that group C had independently poor prognosis (hazard ratio, 9.137; 95% confidence interval, 1.187 to 70.366; p=0.034) compared with group A. Bootstrap resampling internally validated this result (p=0.016). Conclusion This study suggests that both positive CD44v9 and high Ki-67 expression are associated with poor prognosis in EGC, and the combined use of these markers provides better prognostic stratification than the single use of them.
Collapse
Affiliation(s)
- Se-Il Go
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Gyung Hyuck Ko
- Department of Pathology, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jeong-Hee Lee
- Department of Pathology, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Sang-Ho Jeong
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Young-Joon Lee
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Soon Chan Hong
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Woo Song Ha
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
23
|
Takaki A, Kawano S, Uchida D, Takahara M, Hiraoka S, Okada H. Paradoxical Roles of Oxidative Stress Response in the Digestive System before and after Carcinogenesis. Cancers (Basel) 2019; 11:cancers11020213. [PMID: 30781816 PMCID: PMC6406746 DOI: 10.3390/cancers11020213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/03/2019] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
Oxidative stress is recognized as a cancer-initiating stress response in the digestive system. It is produced through mitochondrial respiration and induces DNA damage, resulting in cancer cell transformation. However, recent findings indicate that oxidative stress is also a necessary anticancer response for destroying cancer cells. The oxidative stress response has also been reported to be an important step in increasing the anticancer response of newly developed molecular targeted agents. Oxidative stress might therefore be a cancer-initiating response that should be downregulated in the precancerous stage in patients at risk of cancer but an anticancer cell response that should not be downregulated in the postcancerous stage when cancer cells are still present. Many commercial antioxidant agents are marketed as “cancer-eliminating agents” or as products to improve one’s health, so cancer patients often take these antioxidant agents. However, care should be taken to avoid harming the anticancerous oxidative stress response. In this review, we will highlight the paradoxical effects of oxidative stress and antioxidant agents in the digestive system before and after carcinogenesis.
Collapse
Affiliation(s)
- Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Seiji Kawano
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Daisuke Uchida
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Masahiro Takahara
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Sakiko Hiraoka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| |
Collapse
|
24
|
Oliveira LAD, Oshima CTF, Soffner PA, Silva MDS, Lins RR, Malinverni ACDM, Waisberg J. THE CANONICAL WNT PATHWAY IN GASTRIC CARCINOMA. ABCD-ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA 2019; 32:e1414. [PMID: 30624523 PMCID: PMC6323632 DOI: 10.1590/0102-672020180001e1414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/25/2018] [Indexed: 12/29/2022]
Abstract
Background: It is believed that the Wnt pathway is one of the most important signaling
involved in gastric carcinogenesis. Aim: To analyze the protein expression of canonical and non-canonical Wnt pathways
in gastric carcinoma. Method: The immunohistochemistry was performed in 72 specimens of gastric carcinomas
for evaluating the expression of Wnt-5a, FZD5, GSK3β, axin, CK1, ubiquitin,
cyclin D1 and c-myc. Results: There were significant differences for cytoplasm and nucleus ubiquitin for
moderately and well differentiated tumors (p=0.03) and for those of the
intestinal type of the Lauren classification (p=0.03). The absence of c-myc
was related to Lauren’s intestinal tumors (p=0.03). Expression of CK1 in the
cytoplasm was related to compromised margin (p=0.03). Expression of cyclin
D1 protein was more intense in male patients (p=0.03) There was no relation
of the positive or negative expression of the Wnt-5a, FZD5, GSK3 and Axin
with any clinicopathological variables. Conclusion: The canonical WNT pathway is involved in gastric carcinoma.
Collapse
Affiliation(s)
| | - Celina Tizuko Fujiyama Oshima
- Laboratory of Molecular and Experimental Pathology, Department of Pathology, Federal University of São Paulo, UNIFESP/EPM, São Paulo, SP
| | | | - Marcelo de Souza Silva
- Laboratory of Molecular and Experimental Pathology, Department of Pathology, Federal University of São Paulo, UNIFESP/EPM, São Paulo, SP
| | | | | | - Jaques Waisberg
- Interdisciplinar Program in Surgical Sciences.,Department of Surgery, ABC Medical School, Santo André, SP, Brazil
| |
Collapse
|
25
|
Ueda S, Hayashi H, Miyamoto T, Abe S, Hirai K, Matsukura K, Yagi H, Hara Y, Yoshida K, Okazaki S, Tamura M, Abe Y, Agatsuma T, Niwa S, Masuko K, Masuko T. Anti-tumor effects of mAb against L-type amino acid transporter 1 (LAT1) bound to human and monkey LAT1 with dual avidity modes. Cancer Sci 2019; 110:674-685. [PMID: 30548114 PMCID: PMC6361610 DOI: 10.1111/cas.13908] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023] Open
Abstract
l‐Type amino acid transporter 1 (LAT1) disulfide linked to CD98 heavy chain (hc) is highly expressed in most cancer cells, but weakly expressed in normal cells. In the present study, we developed novel anti‐LAT1 mAbs and showed internalization activity, inhibitory effects of amino acid uptake and cell growth and antibody‐dependent cellular cytotoxicity, as well as in vivo antitumor effects in athymic mice. Furthermore, we examined the reactivity of mAbs with LAT1 of Macaca fascicularis to evaluate possible side‐effects of antihuman LAT1 mAbs in clinical trials. Antihuman LAT1 mAbs reacted with ACHN human and MK.P3 macaca kidney‐derived cells, and this reactivity was significantly decreased by siRNAs against LAT1. Macaca LAT1 cDNA was cloned from MK.P3, and only two amino acid differences between human and macaca LAT1 were seen. RH7777 rat hepatoma and HEK293 human embryonic kidney cells expressing macaca LAT1 were established as stable transfectants, and antihuman LAT1 mAbs were equivalently reactive against transfectants expressing human or macaca LAT1. Dual (high and low) avidity modes were detected in transfectants expressing macaca LAT1, MK.P3, ACHN and HCT116 human colon cancer cells, and KA values were increased by anti‐CD98hc mAb, suggesting anti‐LAT1 mAbs detect an epitope on LAT1‐CD98hc complexes on the cell surface. Based on these results, LAT1 may be a promising anticancer target and Macaca fascicularis can be used in preclinical studies with antihuman LAT1 mAbs.
Collapse
Affiliation(s)
- Shiho Ueda
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | | | - Takako Miyamoto
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Shinya Abe
- Laboratory of Biological ProtectionInstitute for Viral Research, Kyoto UniversityKyotoJapan
| | - Kana Hirai
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Kanji Matsukura
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Hideki Yagi
- School of PharmacyInternational University of Health and WelfareOtawaraJapan
| | - Yuta Hara
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Kinji Yoshida
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Shogo Okazaki
- Division of Development and Aging, Research Institute for Biomedical SciencesTokyo University of ScienceChibaJapan
| | - Masakazu Tamura
- Modality Research Laboratories, Biologics DivisionDaiichi Sankyo Co., LtdTokyoJapan
| | - Yuki Abe
- Biologics & Immuno‐Oncology Laboratories, R&D DivisionDaiichi Sankyo Co., LtdTokyoJapan
| | - Toshinori Agatsuma
- Biologics & Immuno‐Oncology Laboratories, R&D DivisionDaiichi Sankyo Co., LtdTokyoJapan
| | | | - Kazue Masuko
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| | - Takashi Masuko
- Cell Biology LaboratorySchool of PharmacyKindai UniversityOsakaJapan
| |
Collapse
|
26
|
Taniguchi D, Saeki H, Nakashima Y, Kudou K, Nakanishi R, Kubo N, Ando K, Oki E, Oda Y, Maehara Y. CD44v9 is associated with epithelial-mesenchymal transition and poor outcomes in esophageal squamous cell carcinoma. Cancer Med 2018; 7:6258-6268. [PMID: 30474922 PMCID: PMC6308082 DOI: 10.1002/cam4.1874] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/05/2018] [Accepted: 10/24/2018] [Indexed: 12/27/2022] Open
Abstract
CD44 serves as a marker of cancer stem cells. Alternative splicing generates the CD44v9 isoform. Cancer stem cells are associated with the epithelial-mesenchymal transition in cancers, although little is known about their role in esophageal squamous cell carcinoma. Here, we aimed to clarify the relationship between CD44v9 expression, the epithelial-mesenchymal transition, and clinicopathological features of patients with esophageal squamous cell carcinoma. CD44v9 levels were higher at the tumor invasive front compared with the center of the tumor and higher in metastatic lymph nodes compared with primary tumors. High levels of CD44v9 at the tumor invasive front were significantly associated with deeper tumor invasion and shorter overall survival and recurrence-free survival. The expression of CD44v9 was increased by treatment with transforming growth factor-β, which induced esophageal squamous cell carcinoma cells to undergo the epithelial-mesenchymal transition. Moreover, inhibition of CD44v9 expression decreased the migration and invasiveness of esophageal squamous cell carcinoma cells. These results indicate that the expression of CD44v9 at the tumor invasive front induced by stemness was strongly associated with the epithelial-mesenchymal transition and poor prognosis of patients with esophageal squamous cell carcinoma. CD44v9 may therefore serve as a novel prognostic biomarker and a potential therapeutic target for esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Daisuke Taniguchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Saeki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichiro Nakashima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Kudou
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhide Kubo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
27
|
High expression level of CD44v8-10 in cancer stem-like cells is associated with poor prognosis in esophageal squamous cell carcinoma patients treated with chemoradiotherapy. Oncotarget 2018; 9:34876-34888. [PMID: 30405881 PMCID: PMC6201859 DOI: 10.18632/oncotarget.26172] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/13/2018] [Indexed: 01/01/2023] Open
Abstract
Background Strong reactive oxygen species (ROS) suppression in cancer stem-like cell components in various solid tumors is associated with therapeutic resistance. In this study, we investigated the influence of CD44v8-10 expression on the overall survival of esophageal squamous cell carcinoma (E-SCC) patients after definitive chemoradiotherapy (dCRT) and on radio-sensitivities of E-SCC cell lines treated with or without sulfasalazine, a CD44v8-10-xCT-GSH axis inhibitor. Methods Seventy-three patients with E-SCC who received dCRT were examined retrospectively. CD44v8-10 expression was analyzed immunohistochemically using paraffin-blocked pre-dCRT biopsy specimens obtained by esophagoscopy and was expressed as a histo-score (H-score). The relationship between the H-score and overall survival was analyzed. From human E-SCC cell lines (T.T, T.Tn, or Kyse-3650), we collected CD44v8-10High and CD44v8-10Low subpopulations using a cell sorter. Water-soluble tetrazolium salt-8 (WST), glutathione-SH (GSH) and ROS assays were performed to compare the effect of sulfasalazine on the radio-sensitivities of these subpopulations in T.Tn and Kyse-3650. Results High CD44v8-10 expression was independently associated with poor prognosis in E-SCC patients treated with dCRT (hazard ratio = 2.906, 95% CI = 1.277–6.611, p = 0.011). In CD44v8-10High cells of each cell line, sulfasalazine decreased cellular GSH levels, resulting in increased radiation-induced ROS and reduced cell viability. In contrast, sulfasalazine had no significant effects in CD44v8-10Low cells. Conclusion High CD44v8-10 expression was an independent prognostic factor in E-SCC patients treated with dCRT. CD44v8-10-xCT-GSH axis inhibition sensitized CD44v8-10High E-SCC cells to ROS-inducing treatments such as radiotherapy. Targeting CD44v8-10-xCT-GSH axis may improve the prognosis of post-dCRT E-SCC patients.
Collapse
|
28
|
Overexpression of CD44 Variant 9: A Novel Cancer Stem Cell Marker in Human Cholangiocarcinoma in Relation to Inflammation. Mediators Inflamm 2018; 2018:4867234. [PMID: 30402042 PMCID: PMC6198546 DOI: 10.1155/2018/4867234] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/29/2018] [Accepted: 09/09/2018] [Indexed: 12/15/2022] Open
Abstract
Various CD44 isoforms are expressed in several cancer stem cells during tumor progression and metastasis. In particular, CD44 variant 9 (CD44v9) is highly expressed in chronic inflammation-induced cancer. We investigated the expression of CD44v9 and assessed whether CD44v9 is a selective biomarker of human cholangiocarcinoma (CCA). The expression profile of CD44v9 was evaluated in human liver fluke Opisthorchis viverrini-related CCA (OV-CCA) tissues, human CCA (independent of OV infection, non-OV-CCA) tissues, and normal liver tissues. CD44v9 overexpression was detected by immunohistochemistry (IHC) in CCA tissues. There was a higher level of CD44v9 expression and IHC score in OV-CCA tissues than in non-OV-CCA tissues, and there was no CD44v9 staining in the bile duct cells of normal liver tissues. In addition, we observed significantly higher expression of inflammation-related markers, such as S100P and COX-2, in OV-CCA tissues compared to that in non-OV and normal liver tissues. Thus, these findings suggest that CD44v9 may be a novel candidate CCA stem cell marker and may be related to inflammation-associated cancer development.
Collapse
|
29
|
Hara Y, Torii R, Ueda S, Kurimoto E, Ueda E, Okura H, Tatano Y, Yagi H, Ohno Y, Tanaka T, Masuko K, Masuko T. Inhibition of tumor formation and metastasis by a monoclonal antibody against lymphatic vessel endothelial hyaluronan receptor 1. Cancer Sci 2018; 109:3171-3182. [PMID: 30058195 PMCID: PMC6172044 DOI: 10.1111/cas.13755] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/21/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
Although cancer metastasis is associated with poor prognosis, the mechanisms of this event, especially via lymphatic vessels, remain unclear. Lymphatic vessel endothelial hyaluronan receptor 1 (LYVE‐1) is expressed on lymphatic vessel endothelium and is considered to be a specific marker of lymphatic vessels, but it is unknown how LYVE‐1 is involved in the growth and metastasis of cancer cells. We produced rat monoclonal antibodies (mAb) recognizing the extracellular domain of mouse LYVE‐1, and investigated the roles of LYVE‐1 in tumor formation and metastasis. The mAb 38M and 64R were selected from hybridoma clones created by cell fusion between spleen cells of rats immunized with RH7777 rat hepatoma cells expressing green fluorescent protein (GFP)‐fused mouse LYVE‐1 proteins and mouse myeloma cells. Two mAb reacted with RH7777 and HEK293F human embryonic kidney cells expressing GFP‐fused mouse LYVE‐1 proteins in a GFP expression‐dependent manner, and each recognized a distinct epitope. On immunohistology, the 38M mAb specifically stained lymphatic vessels in several mouse tissues. In the wound healing assay, the 64R mAb inhibited cell migration of HEK293F cells expressing LYVE‐1 and mouse lymphatic endothelial cells (LEC), as well as tube formation by LEC. Furthermore, this mAb inhibited primary tumor formation and metastasis to lymph nodes in metastatic MDA‐MB‐231 xenograft models. This shows that LYVE‐1 is involved in primary tumor formation and metastasis, and it may be a promising molecular target for cancer therapy.
Collapse
Affiliation(s)
- Yuta Hara
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Ryota Torii
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Shiho Ueda
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Erina Kurimoto
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Eri Ueda
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Hiroshi Okura
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Yutaka Tatano
- Department of Pharmaceuticals, Faculty of Pharmacy, International University of Health and Welfare, Otawara, Tochigi, Japan
| | - Hideki Yagi
- Department of Pharmaceuticals, Faculty of Pharmacy, International University of Health and Welfare, Otawara, Tochigi, Japan
| | - Yoshiya Ohno
- Laboratory of Immunobiology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Toshiyuki Tanaka
- Laboratory of Immunobiology, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Kazue Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| | - Takashi Masuko
- Cell Biology Laboratory, School of Pharmacy, Kindai University, Higashiosaka, Osaka, Japan
| |
Collapse
|
30
|
Wada F, Koga H, Akiba J, Niizeki T, Iwamoto H, Ikezono Y, Nakamura T, Abe M, Masuda A, Sakaue T, Tanaka T, Kakuma T, Yano H, Torimura T. High expression of CD44v9 and xCT in chemoresistant hepatocellular carcinoma: Potential targets by sulfasalazine. Cancer Sci 2018; 109:2801-2810. [PMID: 29981246 PMCID: PMC6125439 DOI: 10.1111/cas.13728] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
CD44v9 is expressed in cancer stem cells (CSC) and stabilizes the glutamate‐cystine transporter xCT on the cytoplasmic membrane, thereby decreasing intracellular levels of reactive oxygen species (ROS). This mechanism confers ROS resistance to CSC and CD44v9‐expressing cancer cells. The aims of the present study were to assess: (i) expression status of CD44v9 and xCT in hepatocellular carcinoma (HCC) tissues, including those derived from patients treated with hepatic arterial infusion chemoembolization (HAIC) therapy with cisplatin (CDDP); and (ii) whether combination of CDDP with sulfasalazine (SASP), an inhibitor of xCT, was more effective on tumor cells than CDDP alone by inducing ROS‐mediated apoptosis. Twenty non‐pretreated HCC tissues and 7 HCC tissues administered HAIC therapy with CDDP before surgical resection were subjected to immunohistochemistry analysis of CD44v9 and xCT expression. Human HCC cell lines HAK‐1A and HAK‐1B were used in this study; the latter was also used for xenograft experiments in nude mice to assess in vivo efficacy of combination treatment. CD44v9 positivity was significantly higher in HAIC‐treated tissues (5/7) than in non‐pretreated tissues (2/30), suggesting the involvement of CD44v9 in the resistance to HAIC. xCT was significantly expressed in poorly differentiated HCC tissues. Combination treatment effectively killed the CD44v9‐harboring HAK‐1B cells through ROS‐mediated apoptosis and significantly decreased xenografted tumor growth. In conclusion, the xCT inhibitor SASP augmented ROS‐mediated apoptosis in CDDP‐treated HCC cells, in which the CD44v9‐xCT system functioned. As CD44v9 is typically expressed in HAIC‐resistant HCC cells, combination treatment with SASP with CDDP may overcome such drug resistance.
Collapse
Affiliation(s)
- Fumitaka Wada
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Niizeki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Yu Ikezono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Mitsuhiko Abe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Toshimitsu Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | | | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan.,Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| |
Collapse
|
31
|
Yoshida GJ. Emerging roles of Myc in stem cell biology and novel tumor therapies. J Exp Clin Cancer Res 2018; 37:173. [PMID: 30053872 PMCID: PMC6062976 DOI: 10.1186/s13046-018-0835-y] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/06/2018] [Indexed: 02/08/2023] Open
Abstract
The pathophysiological roles and the therapeutic potentials of Myc family are reviewed in this article. The physiological functions and molecular machineries in stem cells, including embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, are clearly described. The c-Myc/Max complex inhibits the ectopic differentiation of both types of artificial stem cells. Whereas c-Myc plays a fundamental role as a "double-edged sword" promoting both iPS cells generation and malignant transformation, L-Myc contributes to the nuclear reprogramming with the significant down-regulation of differentiation-associated genetic expression. Furthermore, given the therapeutic resistance of neuroendocrine tumors such as small-cell lung cancer and neuroblastoma, the roles of N-Myc in difficult-to-treat tumors are discussed. N-Myc and p53 exhibit the co-localization in the nucleus and alter p53-dependent transcriptional responses which are necessary for DNA repair, anti-apoptosis, and lipid metabolic reprogramming. NCYM protein stabilizes N-Myc, resulting in the stimulation of Oct4 expression, while Oct4 induces both N-Myc and NCYM via direct transcriptional activation of N-Myc, [corrected] thereby leading to the enhanced metastatic potential. Importantly enough, accumulating evidence strongly suggests that c-Myc can be a promising therapeutic target molecule among Myc family in terms of the biological characteristics of cancer stem-like cells (CSCs). The presence of CSCs leads to the intra-tumoral heterogeneity, which is mainly responsible for the therapeutic resistance. Mechanistically, it has been shown that Myc-induced epigenetic reprogramming enhances the CSC phenotypes. In this review article, the author describes two major therapeutic strategies of CSCs by targeting c-Myc; Firstly, Myc-dependent metabolic reprogramming is closely related to CD44 variant-dependent redox stress regulation in CSCs. It has been shown that c-Myc increases NADPH production via enhanced glutaminolysis with a finely-regulated mechanism. Secondly, the dormancy of CSCs due to FBW7-depedent c-Myc degradation pathway is also responsible for the therapeutic resistance to the conventional anti-tumor agents, the action points of which are largely dependent on the operation of the cell cycle. That is why the loss-of-functional mutations of FBW7 gene are expected to trigger "awakening" of dormant CSCs in the niche with c-Myc up-regulation. Collectively, although the further research is warranted to develop the effective anti-tumor therapeutic strategy targeting Myc family, we cancer researchers should always catch up with the current advances in the complex functions of Myc family in highly-malignant and heterogeneous tumor cells to realize the precision medicine.
Collapse
Affiliation(s)
- Go J Yoshida
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
32
|
The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018; 11:64. [PMID: 29747682 PMCID: PMC5946470 DOI: 10.1186/s13045-018-0605-5] [Citation(s) in RCA: 762] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
CD44, a non-kinase transmembrane glycoprotein, is overexpressed in several cell types including cancer stem cells and frequently shows alternative spliced variants that are thought to play a role in cancer development and progression. Hyaluronan, the main ligand for CD44, binds to and activates CD44 resulting in activation of cell signaling pathways that induces cell proliferation, increases cell survival, modulates cytoskeletal changes, and enhances cellular motility. The different functional roles of CD44 standard (CD44s) and specific CD44 variant (CD44v) isoforms are not fully understood. CD44v contain additional peptide motifs that can interact with and sequester growth factors and cytokines at the cell surface thereby functioning as coreceptors to facilitate cell signaling. Moreover, CD44v were expressed in metastasized tumors, whereas switching between CD44v and CD44s may play a role in regulating epithelial to mesenchymal transition (EMT) and in the adaptive plasticity of cancer cells. Here, we review current data on the structural and functional properties of CD44, the known roles for CD44 in tumorigencity, the regulation of CD44 expression, and the potential for targeting CD44 for cancer therapy.
Collapse
|
33
|
Abstract
Chronic inflammation is a risk factor for gastrointestinal cancer and other diseases. Most studies have focused on cytokines and chemokines as mediators connecting chronic inflammation to cancer, whereas the involvement of lipid mediators, including prostanoids, has not been extensively investigated. Prostanoids are among the earliest signaling molecules released in response to inflammation. Multiple lines of evidence suggest that prostanoids are involved in gastrointestinal cancer. In this Review, we discuss how prostanoids impact gastrointestinal cancer development. In particular, we highlight recent advances in our understanding of how prostaglandin E2 induces the immunosuppressive microenvironment in gastrointestinal cancers.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Raymond N DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Research and Division of Gastroenterology, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
34
|
Capillary morphogenesis gene 2 maintains gastric cancer stem-like cell phenotype by activating a Wnt/β-catenin pathway. Oncogene 2018; 37:3953-3966. [PMID: 29662192 PMCID: PMC6053357 DOI: 10.1038/s41388-018-0226-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/29/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
A growing body of evidence shows that the development and progression of gastric cancer (GC) is mainly associated to the presence of gastric cancer stem-like cells (GCSLCs). However, it is unclear how GCSLC population is maintained. This study aimed to explore the role of capillary morphogenesis gene 2 (CMG2) in GCSLC maintenance and the relevance to GC progression. We found that CMG2 was highly expressed in GC tissues and the expression levels were associated with the invasion depth and lymph node metastasis of GC, and inversely correlated with the survival of GC patients. Sorted CMG2High GC cells preferentially clustered in CD44High stem-like cell population, which expressed high levels of stemness-related genes with increased capabilities of self-renewal and tumorigenicity. Depletion of CMG2 gene resulted in reduction of GCSLC population with attenuated stemness and decrease of invasive and metastatic capabilities with subdued epithelial–mesenchymal transition phenotype in GC cells. Mechanistically, CMG2 interacted with LRP6 in GCSLCs to activate a Wnt/β-catenin pathway. Thus, our results demonstrate that CMG2 promotes GC progression by maintaining GCSLCs and can serve as a new prognostic indicator and a target for human GC therapy.
Collapse
|
35
|
Arima K, Komohara Y, Bu L, Tsukamoto M, Itoyama R, Miyake K, Uchihara T, Ogata Y, Nakagawa S, Okabe H, Imai K, Hashimoto D, Chikamoto A, Yamashita YI, Baba H, Ishimoto T. Downregulation of 15-hydroxyprostaglandin dehydrogenase by interleukin-1β from activated macrophages leads to poor prognosis in pancreatic cancer. Cancer Sci 2018; 109:462-470. [PMID: 29224225 PMCID: PMC5797824 DOI: 10.1111/cas.13467] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
Chronic inflammation has a crucial role in cancer development and the progression of various tumors, including pancreatic ductal adenocarcinoma (PDAC). The arachidonate cascade is a major inflammatory pathway that produces several metabolites, such as prostaglandin E2. The enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) degrades prostaglandin and is frequently decreased in several types of cancer; however, the molecular mechanisms of 15-PGDH suppression are unclear. The current study was carried out to elucidate the molecular mechanisms and clinical significance of 15-PGDH suppression in PDAC. Here, we showed that interleukin-1β (IL-1β), a pro-inflammatory cytokine, downregulates 15-PGDH expression in PDAC cells, and that IL-1β expression was inversely correlated with 15-PGDH levels in frozen PDAC tissues. We also found that activated macrophages produced IL-1β and reduced 15-PGDH expression in PDAC cells. Furthermore, the number of CD163-positive tumor-associated macrophages was shown to be inversely correlated with 15-PGDH levels in PDAC cells by immunohistochemical staining of 107 PDAC samples. Finally, we found that low 15-PGDH expression was significantly associated with advanced tumors, presence of lymph node metastasis and nerve invasion, and poor prognosis in PDAC patients. Our results indicate that IL-1β derived from TAMs suppresses 15-PGDH expression in PDAC cells, resulting in poor prognosis of PDAC patients.
Collapse
Affiliation(s)
- Kota Arima
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Luke Bu
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masayo Tsukamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rumi Itoyama
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keisuke Miyake
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoyuki Uchihara
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirohisa Okabe
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
36
|
Jamnongkan W, Thanee M, Yongvanit P, Loilome W, Thanan R, Kimawaha P, Boonmars T, Silakit R, Namwat N, Techasen A. Antifibrotic effect of xanthohumol in combination with praziquantel is associated with altered redox status and reduced iron accumulation during liver fluke-associated cholangiocarcinogenesis. PeerJ 2018; 6:e4281. [PMID: 29375936 PMCID: PMC5784579 DOI: 10.7717/peerj.4281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/29/2017] [Indexed: 12/20/2022] Open
Abstract
Cholangiocarcinoma (CCA) caused by infection of the liver fluke Opisthorchis viverrini, (Ov) is the major public health problem in northeast Thailand. Following Ov infection the subsequent molecular changes can be associated by reactive oxygen species (ROS) induced chronic inflammation, advanced periductal fibrosis, and cholangiocarcinogenesis. Notably, resistance to an activation of cell death in prolonged oxidative stress conditions can occur but some damaged/mutated cells could survive and enable clonal expansion. Our study used a natural product, xanthohumol (XN), which is an anti-oxidant and anti-inflammatory compound, to examine whether it could prevent Ov-associated CCA carcinogenesis. We measured the effect of XN with or without praziquantel (PZ), an anti-helminthic treatment, on DNA damage, redox status change including iron accumulation and periductal fibrosis during CCA genesis induced by administration of Ov and N-dinitrosomethylamine (NDMA) in hamsters. Animals were randomly divided into four groups: group I, Ov infection and NDMA administration (ON); group II, Ov infection and NDMA administration and PZ treatment (ONP); the latter 2 groups were similar to group I and II, but group III received additional XN (XON) and group IV received XN plus PZ (XONP). The results showed that high 8-oxodG (a marker of DNA damage) was observed throughout cholangiocarcinogenesis. Moreover, increased expression of CD44v8-10 (a cell surface in regulation of the ROS defense system), whereas decreased expression of phospho-p38MAPK (a major ROS target), was found during the progression of the bile duct cell transformation. In addition, high accumulation of iron and expression of transferrin receptor-1 (TfR-1) in both malignant bile ducts and inflammatory cells were detected. Furthermore, fibrosis also increased with the highest level being on day 180. On the other hand, the groups of XN with or without PZ supplementations showed an effective reduction in all the markers examined, including fibrosis when compared with the ON group. In particular, the XONP group, in which a significant reduction DNA damage occurred, was also found to have iron accumulation and fibrosis compared to the other groups. Our results show that XN administered in combination with PZ could efficiently prevent CCA development and hence provide potential chemopreventive benefits in Ov-induced cholangiocarcinogenesis.
Collapse
Affiliation(s)
- Wassana Jamnongkan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Malinee Thanee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Raynoo Thanan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Phongsaran Kimawaha
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Tidarat Boonmars
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.,Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Runglawan Silakit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Anchalee Techasen
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
37
|
Shibata W, Sue S, Tsumura S, Ishii Y, Sato T, Kameta E, Sugimori M, Yamada H, Kaneko H, Sasaki T, Ishii T, Tamura T, Kondo M, Maeda S. Helicobacter-induced gastric inflammation alters the properties of gastric tissue stem/progenitor cells. BMC Gastroenterol 2017; 17:145. [PMID: 29212456 PMCID: PMC5719643 DOI: 10.1186/s12876-017-0706-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 11/24/2017] [Indexed: 12/13/2022] Open
Abstract
Background Although Helicobacter-induced gastric inflammation is the major predisposing factor for gastric carcinogenesis, the precise mechanism by which chronic gastritis causes gastric cancer remains unclear. Intestinal and spasmolytic polypeptide-expressing metaplasia (SPEM) is considered as precancerous lesions, changes in epithelial tissue stem/progenitor cells after chronic inflammation has not been clarified yet. In this study, we utilized three-dimensional gastric epithelial cell culture systems that could form organoids, mimicking gastric epithelial layer, and characterized the changes in epithelial cells after chronic Helicobacter felis infection. Methods We used three mice model; 1) long-term H. felis infection, 2) H. felis eradication, and 3) MNU chemical carcinogenesis model. We performed cRNA microarray analysis after organoid culture, and analyzed the effects of chronic gastric inflammation on tissue stem cells, by the size of organoid, mRNA expression profile and immunohistochemical analysis. Results The number of organoids cultured from gastric epithelial cells was significantly higher in organoids isolated from H. felis-infected mice compared with those from uninfected gastric mucosa. Based on the mRNA expression profile, we found that possible stem cell markers such as Cd44, Dclk1, and genes associated with the intestinal phenotype, such as Villin, were increased in organoids isolated from H. felis-infected mucosa compared with the control. The upregulation of these genes were cancelled after H. felis eradication. In a xenograft model, tumors were generated only from organoids cultured from carcinogen-treated gastric mucosa, not from H. felis infected mucosa or control organoids. Conclusions Our results suggested that, as a possible mechanism of gastric carcinogenesis, chronic inflammation induced by H. felis infection increased the number of tissue stem/progenitor cells and the expression of stem cell markers. These findings suggest that chronic inflammation may alter the direction of differentiation toward undifferentiated state and that drawbacks may enable cells to redifferentiate to intestinal metaplasia or neoplasia. Electronic supplementary material The online version of this article (10.1186/s12876-017-0706-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wataru Shibata
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Division of Translational Research, Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Soichiro Sue
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sachiko Tsumura
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yasuaki Ishii
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Sato
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Eri Kameta
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Makoto Sugimori
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroaki Yamada
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroaki Kaneko
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Sasaki
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiro Ishii
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshihide Tamura
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaaki Kondo
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
38
|
Suzuki Y, Kitahara S, Suematsu T, Oshima M, Sato Y. Requisite role of vasohibin-2 in spontaneous gastric cancer formation and accumulation of cancer-associated fibroblasts. Cancer Sci 2017; 108:2342-2351. [PMID: 28960674 PMCID: PMC5715352 DOI: 10.1111/cas.13411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/30/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022] Open
Abstract
The vasohibin (VASH) family consists of two genes, VASH1 and VASH2. VASH1 is mainly expressed in vascular endothelial cells and suppresses angiogenesis in an autocrine manner, whereas VASH2 is mainly expressed in cancer cells and exhibits pro‐angiogenic activity. Employing adenomatous polyposis coli gene mutant mice, we recently reported on the role of Vash2 in the spontaneous formation of intestinal tumors. In this study, we used K19‐Wnt1/C2mE (Gan) mice and examined the role of Vash2 in spontaneous gastric cancer formation. Gan mice spontaneously develop gastric tumors by activation of Wnt and prostaglandin E2 signaling pathways in gastric mucosa after 30 weeks of age. Expression of Vash2 mRNA was significantly increased in gastric tumor tissues compared with normal stomach tissues. When Gan mice were crossed with the Vash2‐deficient (Vash2LacZ/LacZ) strain, gastric cancer formation was significantly suppressed in Vash2LacZ/LacZGan mice. Normal composition of gastric mucosa was partially maintained in Vash2LacZ/LacZGan mice. Knockout of Vash2 caused minimal reduction of tumor angiogenesis but a significant decrease in cancer‐associated fibroblasts (CAF) in tumor stroma. DNA microarray analysis and real‐time RT‐PCR showed that mRNA levels of epiregulin (Ereg) and interleukin‐11 (Il11) were significantly downregulated in gastric tumors of Vash2LacZ/LacZGan mice. Furthermore, conditioned medium of gastric cancer cells stimulated migration of and α‐smooth muscle actin expression in fibroblasts, whereas conditioned medium of VASH2 knockdown cells attenuated these effects in vitro. These results suggest that VASH2 plays an important role in gastric tumor progression via the accumulation of CAF accompanying upregulation of EREG and IL‐11 expression.
Collapse
Affiliation(s)
- Yasuhiro Suzuki
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Shuji Kitahara
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Takuya Suematsu
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yasufumi Sato
- Department of Vascular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
39
|
Fu DJ, Miller AD, Southard TL, Flesken-Nikitin A, Ellenson LH, Nikitin AY. Stem Cell Pathology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:71-92. [PMID: 29059010 DOI: 10.1146/annurev-pathol-020117-043935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rapid advances in stem cell biology and regenerative medicine have opened new opportunities for better understanding disease pathogenesis and the development of new diagnostic, prognostic, and treatment approaches. Many stem cell niches are well defined anatomically, thereby allowing their routine pathological evaluation during disease initiation and progression. Evaluation of the consequences of genetic manipulations in stem cells and investigation of the roles of stem cells in regenerative medicine and pathogenesis of various diseases such as cancer require significant expertise in pathology for accurate interpretation of novel findings. Therefore, there is an urgent need for developing stem cell pathology as a discipline to facilitate stem cell research and regenerative medicine. This review provides examples of anatomically defined niches suitable for evaluation by diagnostic pathologists, describes neoplastic lesions associated with them, and discusses further directions of stem cell pathology.
Collapse
Affiliation(s)
- Dah-Jiun Fu
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Andrew D Miller
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Teresa L Southard
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Andrea Flesken-Nikitin
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Lora H Ellenson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| |
Collapse
|
40
|
|
41
|
Jiao Y, Wang Y, Guo S, Wang G. Glutathione peroxidases as oncotargets. Oncotarget 2017; 8:80093-80102. [PMID: 29108391 PMCID: PMC5668124 DOI: 10.18632/oncotarget.20278] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is a disturbance in the equilibrium among free radicals, reactive oxygen species, and endogenous antioxidant defense mechanisms. Oxidative stress is a result of imbalance between the production of reactive oxygen and the biological system's ability to detoxify the reactive intermediates or to repair the resulting damage. Mounting evidence has implicated oxidative stress in various physiological and pathological processes, including DNA damage, proliferation, cell adhesion, and survival of cancer cells. Glutathione peroxidases (GPxs) (EC 1.11.1.9) are an enzyme family with peroxidase activity whose main biological roles are to protect organisms from oxidative damage by reducing lipid hydroperoxides as well as free hydrogen peroxide. Currently, 8 sub-members of GPxs have been identified in humans, all capable of reducing H2O2 and soluble fatty acid hydroperoxides. A large number of publications has demonstrated that GPxs have significant roles in different stages of carcinogenesis. In this review, we will update recent progress in the study of the roles of GPxs in cancer. Better mechanistic understanding of GPxs will potentially contribute to the development and advancement of improved cancer treatment models.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Stomatology, PLA Army General Hospital, Beijing, P.R. China
| | - Yirong Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, P.R. China
| | - Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
42
|
Kim YM, Gang EJ, Kahn M. CBP/Catenin antagonists: Targeting LSCs' Achilles heel. Exp Hematol 2017; 52:1-11. [PMID: 28479420 PMCID: PMC5526056 DOI: 10.1016/j.exphem.2017.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/07/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs), including leukemia stem cells (LSCs), exhibit self-renewal capacity and differentiation potential and have the capacity to maintain or renew and propagate a tumor/leukemia. The initial isolation of CSCs/LSCs was in adult myelogenous leukemia, although more recently, the existence of CSCs in a wide variety of other cancers has been reported. CSCs, in general, and LSCs, specifically with respect to this review, are responsible for initiation of disease, therapeutic resistance and ultimately disease relapse. One key focus in cancer research over the past decade has been the development of therapies that safely eliminate the LSC/CSC population. One major obstacle to this goal is the identification of key mechanisms that distinguish LSCs from normal endogenous hematopoietic stem cells. An additional daunting feature that has recently come to light with advances in next-generation sequencing and single-cell sequencing is the heterogeneity within leukemias/tumors, with multiple combinations of mutations, gain and loss of function of genes, and so on being capable of driving disease, even within the CSC/LSC population. The focus of this review/perspective is on our work in identifying and validating, in both chronic myelogenous leukemia and acute lymphoblastic leukemia, a safe and efficacious mechanism to target an evolutionarily conserved signaling nexus, which constitutes a common "Achilles heel" for LSCs/CSCs, using small molecule-specific CBP/catenin antagonists.
Collapse
Affiliation(s)
- Yong-Mi Kim
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Blood and Bone Marrow Transplantation, University of Southern California, Los Angeles, CA
| | - Eun-Ji Gang
- Children's Hospital Los Angeles, Department of Pediatrics, Division of Blood and Bone Marrow Transplantation, University of Southern California, Los Angeles, CA
| | - Michael Kahn
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA; Department of Molecular Pharmacology and Toxicology, University of Southern California, Los Angeles, CA; Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.
| |
Collapse
|
43
|
Jafari N, Abediankenari S. MicroRNA-34 dysregulation in gastric cancer and gastric cancer stem cell. Tumour Biol 2017; 39:1010428317701652. [PMID: 28468587 DOI: 10.1177/1010428317701652] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a major cause of cancer mortality worldwide, with a low survival rate for patients with advanced forms of the disease. Over the recent decades, the investigation of the pathophysiological mechanisms of tumourigenesis has opened promising avenues to understand some of the complexities of cancer treatment. However, tumour regeneration and metastasis impose great difficulty for gastric cancer cure. In recent years, cancer stem cells - a small subset of tumour cells in many cancers - have become a major focus of cancer research. Cancer stem cells are capable of self-renewal and are known to be responsible for tumour initiation, metastasis, therapy resistance and cancer recurrence. Recent studies have revealed the key role of microRNAs - small noncoding RNAs regulating gene expression - in these processes. MicroRNAs play crucial roles in the regulation of a wide range of biological processes in a post-transcriptional manner, though their expression is dysregulated in most malignancies, including gastric cancer. In this article, we review the consequences of aberrant expression of microRNA-34 in cancer and cancer stem cells, with a specific focus on the miR-34 dysregulation in gastric cancer and gastric cancer stem cells. We address the critical effects of the aberrant expression of miR-34 and its target genes in maintaining cancer stem cell properties. Information collection and discussion about the advancements in gastric cancer stem cells and microRNAs can be useful for providing novel insights into patient treatment.
Collapse
Affiliation(s)
- Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
44
|
Chen Z, Soutto M, Rahman B, Fazili MW, Peng D, Blanca Piazuelo M, Chen H, Kay Washington M, Shyr Y, El-Rifai W. Integrated expression analysis identifies transcription networks in mouse and human gastric neoplasia. Genes Chromosomes Cancer 2017; 56:535-547. [PMID: 28281307 DOI: 10.1002/gcc.22456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide. The Tff1 knockout (KO) mouse model develops gastric lesions that include low-grade dysplasia (LGD), high-grade dysplasia (HGD), and adenocarcinomas. In this study, we used Affymetrix microarrays gene expression platforms for analysis of molecular signatures in the mouse stomach [Tff1-KO (LGD) and Tff1 wild-type (normal)] and human gastric cancer tissues and their adjacent normal tissue samples. Combined integrated bioinformatics analysis of mouse and human datasets indicated that 172 genes were consistently deregulated in both human gastric cancer samples and Tff1-KO LGD lesions (P < .05). Using Ingenuity pathway analysis, these genes mapped to important transcription networks that include MYC, STAT3, β-catenin, RELA, NFATC2, HIF1A, and ETS1 in both human and mouse. Further analysis demonstrated activation of FOXM1 and inhibition of TP53 transcription networks in human gastric cancers but not in Tff1-KO LGD lesions. Using real-time RT-PCR, we validated the deregulated expression of several genes (VCAM1, BGN, CLDN2, COL1A1, COL1A2, COL3A1, EpCAM, IFITM1, MMP9, MMP12, MMP14, PDGFRB, PLAU, and TIMP1) that map to altered transcription networks in both mouse and human gastric neoplasia. Our study demonstrates significant similarities in deregulated transcription networks in human gastric cancer and gastric tumorigenesis in the Tff1-KO mouse model. The data also suggest that activation of MYC, STAT3, RELA, and β-catenin transcription networks could be an early molecular step in gastric carcinogenesis.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Mohammed Soutto
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Bushra Rahman
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Muhammad W Fazili
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Maria Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Gastroenterology, Hepatology, & Nutrition, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Heidi Chen
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Yu Shyr
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Wael El-Rifai
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| |
Collapse
|
45
|
Yamakawa Y, Kusuhara M, Terashima M, Kinugasa Y, Sugino T, Abe M, Mochizuki T, Hatakeyama K, Kami K, Yamaguchi K. CD44 variant 9 expression as a predictor for gastric cancer recurrence: immunohistochemical and metabolomic analysis of surgically resected tissues. Biomed Res 2017; 38:41-52. [PMID: 28239031 DOI: 10.2220/biomedres.38.41] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
CD44 variant 9 (CD44v9) and the heavy chain of 4F2 cell-surface antigen (CD98hc) appear important for regulation of reactive oxygen species defence and tumor growth in gastric cancer. This study examined the roles of CD44v9 and CD98hc as markers of gastric cancer recurrence, and investigated associations with energy metabolism. We applied capillary electrophoresis time-of-flight mass spectrometry to metabolome profiling of gastric cancer specimens from 103 patients who underwent resection with no residual tumor or microscopic residual tumor, and compared metabolite levels to immunohistochemical staining for CD44v9 and CD98hc. Positive expression rates were 40.7% for CD44v9 and 42.7% for CD98hc. Various tumor characteristics were significantly associated with CD44v9 expression. Five-year recurrence-free survival rate was significantly lower for CD44v9-positive tumors (39.1%) than for CD44v9-negative tumors (73.5%; P < 0.0001), but no significant differences in recurrence-free survival were seen according to CD98hc expression. Uni- and multivariate analyses identified positive CD44v9 expression as an independent predictor of poorer recurrence-free survival. Metabolome analysis of 110 metabolites found that levels of glutathione disulfide were significantly lower and reduced glutathione (GSH)/ glutathione disulfide (GSSG) ratio was significantly higher in CD44v9-positive tumors than in CD44v9-negative tumors, suggesting that CD44v9 may enhance pentose phosphate pathway flux and maintain GSH levels in cancer cells.
Collapse
Affiliation(s)
- Yushi Yamakawa
- Division of Colon and Rectal Surgery, Shizuoka Cancer Center
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol 2017; 10:67. [PMID: 28279189 PMCID: PMC5345270 DOI: 10.1186/s13045-017-0436-9] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/02/2017] [Indexed: 02/07/2023] Open
Abstract
The 2016 Nobel Prize in Physiology or Medicine was awarded to the researcher that discovered autophagy, which is an evolutionally conserved catabolic process which degrades cytoplasmic constituents and organelles in the lysosome. Autophagy plays a crucial role in both normal tissue homeostasis and tumor development and is necessary for cancer cells to adapt efficiently to an unfavorable tumor microenvironment characterized by hypo-nutrient conditions. This protein degradation process leads to amino acid recycling, which provides sufficient amino acid substrates for cellular survival and proliferation. Autophagy is constitutively activated in cancer cells due to the deregulation of PI3K/Akt/mTOR signaling pathway, which enables them to adapt to hypo-nutrient microenvironment and exhibit the robust proliferation at the pre-metastatic niche. That is why just the activation of autophagy with mTOR inhibitor often fails in vain. In contrast, disturbance of autophagy–lysosome flux leads to endoplasmic reticulum (ER) stress and an unfolded protein response (UPR), which finally leads to increased apoptotic cell death in the tumor tissue. Accumulating evidence suggests that autophagy has a close relationship with programmed cell death, while uncontrolled autophagy itself often induces autophagic cell death in tumor cells. Autophagic cell death was originally defined as cell death accompanied by large-scale autophagic vacuolization of the cytoplasm. However, autophagy is a “double-edged sword” for cancer cells as it can either promote or suppress the survival and proliferation in the tumor microenvironment. Furthermore, several studies of drug re-positioning suggest that “conventional” agents used to treat diseases other than cancer can have antitumor therapeutic effects by activating/suppressing autophagy. Because of ever increasing failure rates and high cost associated with anticancer drug development, this therapeutic development strategy has attracted increasing attention because the safety profiles of these medicines are well known. Antimalarial agents such as artemisinin and disease-modifying antirheumatic drug (DMARD) are the typical examples of drug re-positioning which affect the autophagy regulation for the therapeutic use. This review article focuses on recent advances in some of the novel therapeutic strategies that target autophagy with a view to treating/preventing malignant neoplasms.
Collapse
Affiliation(s)
- Go J Yoshida
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan. .,Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan.
| |
Collapse
|
47
|
Qian HR, Yang Y. Functional role of autophagy in gastric cancer. Oncotarget 2017; 7:17641-51. [PMID: 26910278 PMCID: PMC4951239 DOI: 10.18632/oncotarget.7508] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/06/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a highly regulated catabolic pathway responsible for the degradation of long-lived proteins and damaged intracellular organelles. Perturbations in autophagy are found in gastric cancer. In host gastric cells, autophagy can be induced by Helicobacter pylori (or H. pylori) infection, which is associated with the oncogenesis of gastric cancer. In gastric cancer cells, autophagy has both pro-survival and pro-death functions in determining cell fate. Besides, autophagy modulates gastric cancer metastasis by affecting a wide range of pathological events, including extracellular matrix (ECM) degradation, epithelial-to-mesenchymal transition (EMT), tumor angiogenesis, and tumor microenvironment. In addition, some of the autophagy-related proteins, such as Beclin 1, microtubule-associated protein 1 light chain 3 (MAP1-LC3), and p62/sequestosome 1 (SQSTM1) have certain prognostic values for gastric cancer. In this article, we review the recent studies regarding the functional role of autophagy in gastric cancer.
Collapse
Affiliation(s)
- Hao-ran Qian
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yi Yang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
48
|
Wang W, Li M, Zhang Z, Cui C, Zhou J, Yin L, Lv H. Design, synthesis and evaluation of multi-functional tLyP-1-hyaluronic acid-paclitaxel conjugate endowed with broad anticancer scope. Carbohydr Polym 2017; 156:97-107. [DOI: 10.1016/j.carbpol.2016.08.100] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 02/08/2023]
|
49
|
Choi YJ, Kim N, Lee HS, Park SM, Park JH, Yoon H, Shin CM, Park YS, Kim JW, Lee DH. Expression of Leucine-rich Repeat-containing G-protein Coupled Receptor 5 and CD44: Potential Implications for Gastric Cancer Stem Cell Marker. J Cancer Prev 2016; 21:279-287. [PMID: 28053963 PMCID: PMC5207613 DOI: 10.15430/jcp.2016.21.4.279] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The human leucine-rich repeat-containing G-protein coupled receptor (LGR) 5 and CD44 are one of the candidates for the marker of gastric cancer stem cells. We compared the expressions of two genes among control, dysplasia and cancer groups. METHODS We compared the mRNA expression of LGR5, CD44 and CD44v8-10 and immunohistochemistry (IHC) of LGR5 and CD44 in gastric antral mucosa of 45 controls, 36 patients with gastric dysplasia, and 39 patients with early gastric cancer. Additionally, IHC of LGR5 in gastric body mucosa was analyzed. Normal mucosa adjacent to dysplastic or cancer lesions was used for the quantitative real-time-PCR and IHC. RESULTS Immunoreactivity of LGR5 in base of antral mucosa was higher in non-cancerous tissues of cancer than those of control (P = 0.006), whereas the expression of LGR5 mRNA was not different among the three groups. Immunostaining of LGR5 was much stronger in the antrum than in the body of stomach (P < 0.001). Although there was no difference in antral immunointensity of LGR5 according to the severity of intestinal metaplasia, stronger immunostaining was found in the body with an aggravation of intestinal metaplasia (P trend < 0.001). The expression of CD44v8-10 mRNA was higher in cancer patients than control subjects and patients with dysplasia (P = 0.018 and 0.009) while the expression of CD44 mRNA was higher in the control groups than the others. CONCLUSIONS IHC of LGR5 in crypt base and CD44 may be used for gastric CSC markers. LGR5 expression may be associated with the developing of corporal intestinal metaplasia. The expression of CD44v8-10 mRNA would be more suitable for gastric cancer stem cell marker than CD44 or LGR5 mRNA.
Collapse
Affiliation(s)
- Yoon Jin Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seon Mee Park
- Department of Internal Medicine, Chungbuk National University College of Medicine and Medical Research Institute, Cheongju, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin-Wook Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
50
|
Prognostic impact of CD44-positive cancer stem-like cells at the invasive front of gastric cancer. Br J Cancer 2016; 116:186-194. [PMID: 27931044 PMCID: PMC5243989 DOI: 10.1038/bjc.2016.401] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/04/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
Background: The invasive tumour front may provide prognostic information. We examined the relationship between the presence of cancer stem cells (CSCs) at the invasive tumour front and prognosis in gastric cancer (GC). Methods: CD44 is a CSC marker; accordingly, CD44 standard (CD44s), CD44 variant-6 (CD44v6), and CD44 variant-9 (CD44v9) expression were examined in 123 resected primary GCs and the clinical significance of CSCs at the invasive tumour front was analysed. Results: Thirteen (10.6%), 79 (64.2%), and 47 (38.2%) GCs were CD44s-, CD44v6-, and CD44v9-positive, respectively. Patients with CD44-positive expression at the invasive tumour front had significantly poorer disease-specific survival than those with negative expression (CD44s: P<0.00001, CD44v6: P=0.013, CD44v9: P=0.0002). CD44s expression at the invasive tumour front was an independent prognostic factor in resectable GC patients (hazard ratio=3.13; 95% confidence interval, 1.09–9.01; P=0.035) and was significantly associated with peritoneal (P<0.001), lymphatic (P<0.001), and haematogenous recurrences (P=0.008). In addition, the number of CD44 isoforms expressed in cancer cells at the invasive tumour front was associated with patient prognosis. No conventional clinicopathological factors were independently associated with CD44 expression at the invasive tumour front. Conclusions: CD44-positive cancer stem-like cells at the invasive tumour front indicate poor survival and can be a unique biological prognostic factor for GC.
Collapse
|