1
|
Wei C, Zhang X, Li H, Gu J, Xue F, Xie W, Ji G. STMN1 regulates the stemness of gastric cancer cells by binding to HN1L to activate the STAT3 signaling pathway. Discov Oncol 2025; 16:263. [PMID: 40035993 PMCID: PMC11880488 DOI: 10.1007/s12672-025-01879-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND STMN1 is highly expressed in gastric cancer (GC) tissues and the aim of this study was to investigate the role of STMN1 in GC cell stemness. METHODS Analysis of the expression and correlation of STMN1 and its target genes in GC through bioinformatics. Construction of interference plasmids for STMN1 and transfection into GC cells. Sphere formation assay was conducted to detect stem cell sphere-forming ability. WB analysis was performed to detect the expression of stemness genes CD133, ALDH1, CD44, SOX2, Nanog, and STAT3-related proteins. Additionally, CCK-8 assay and TUNEL staining were used to assess GC cell sensitivity to cisplatin (DDP). Construction of a xenograft mouse model to detect the in vivo tumorigenic ability of GC cells. Immunoprecipitation (IP) experiment was conducted to validate the binding of STMN1 and HN1L in GC cells. Overexpression plasmids of HN1L were used for mechanism validation. RESULTS STMN1 and its target HN1L were highly expressed in GC tissues and cells, and were associated with a poor prognosis in GC. Interfering with STMN1 significantly reduced the self-renewal ability of GC cells, downregulated the expression of CD133, ALDH1, CD44, SOX2, Nanog, p-STAT3 and PD-L1. Interfering with STMN1 increased the sensitivity of GC cells to DDP and promoted apoptosis. IP experiments demonstrate that STMN1 and HN1L combine in GC cells. Overexpression of HN1L significantly reversed the effects of Si-STMN1 on GC cells. In vivo experiments demonstrate that the addition of DDP or interference with STMN1 reduced tumor size and weight, and downregulated the expression of CD133, KI67, HN1L, p-STAT3, and PD-L1 in tumor tissues. The combined use of DPP and Si-STMN1 had a more significant effect. CONCLUSION STMN1 regulates GC cell stemness by binding HN1L to activate the HN1L/STAT3/ PD-L1 signaling pathway.
Collapse
Affiliation(s)
- Chunyang Wei
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Xing Zhang
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Hao Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Jianzhong Gu
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Fei Xue
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Wenna Xie
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Gang Ji
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Me dical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Yun H, Dong F, Wei X, Yan X, Zhang R, Zhang X, Wang Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol Rep 2025; 53:14. [PMID: 39611496 PMCID: PMC11622107 DOI: 10.3892/or.2024.8847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Abstract
Gastric cancer (GC) is characterized by a complex and heterogeneous tumor microenvironment (TME) that significantly influences disease progression and treatment outcomes. The tumor stroma, which is composed of a variety of cell types such as cancer‑associated fibroblasts, immune cells and vascular components, displays significant spatial and temporal diversity. These stromal elements engage in dynamic crosstalk with cancer cells, shaping their proliferative, invasive and metastatic potential. Furthermore, the TME is instrumental in facilitating resistance to traditional chemotherapy, specific treatments and immunotherapy strategies. Understanding the underlying mechanisms by which the GC microenvironment evolves and supports tumor growth and therapeutic resistance is critical for developing effective treatment strategies. The present review explores the latest progress in understanding the intricate interactions between cancer cells and their immediate environment in GC, highlighting the implications for disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Fangde Dong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiaoqin Wei
- Department of Pain, The Second People's Hospital of Baiyin, Baiyin, Gansu 730900, P.R. China
| | - Xinyong Yan
- Department of Proctology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Ronglong Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiuyu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Yulin Wang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| |
Collapse
|
3
|
Razavi Z, Soltani M, Pazoki-Toroudi H, Dabagh M. Microfluidic systems for modeling digestive cancer: a review of recent progress. Biomed Phys Eng Express 2024; 10:052002. [PMID: 39142294 DOI: 10.1088/2057-1976/ad6f15] [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: 03/24/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024]
Abstract
Purpose. This review aims to highlight current improvements in microfluidic devices designed for digestive cancer simulation. The review emphasizes the use of multicellular 3D tissue engineering models to understand the complicated biology of the tumor microenvironment (TME) and cancer progression. The purpose is to develop oncology research and improve digestive cancer patients' lives.Methods. This review analyzes recent research on microfluidic devices for mimicking digestive cancer. It uses tissue-engineered microfluidic devices, notably organs on a chip (OOC), to simulate human organ function in the lab. Cell cultivation on modern three-dimensional hydrogel platforms allows precise geometry, biological components, and physiological qualities. The review analyzes novel methodologies, key findings, and technical progress to explain this field's advances.Results. This study discusses current advances in microfluidic devices for mimicking digestive cancer. Micro physiological systems with multicellular 3D tissue engineering models are emphasized. These systems capture complex biochemical gradients, niche variables, and dynamic cell-cell interactions in the tumor microenvironment (TME). These models reveal stomach cancer biology and progression by duplicating the TME. Recent discoveries and technology advances have improved our understanding of gut cancer biology, as shown in the review.Conclusion. Microfluidic systems play a crucial role in modeling digestive cancer and furthering oncology research. These platforms could transform drug development and treatment by revealing the complex biology of the tumor microenvironment and cancer progression. The review provides a complete summary of recent advances and suggests future research for field professionals. The review's major goal is to further medical research and improve digestive cancer patients' lives.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran
- Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K N Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Centre for Sustainable Business, International Business University, Toronto, Canada
| | | | - Mahsa Dabagh
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, WI 53211, United States of America
| |
Collapse
|
4
|
Wang J, Zhang J, Liu H, Meng L, Gao X, Zhao Y, Wang C, Gao X, Fan A, Cao T, Fan D, Zhao X, Lu Y. N6-methyladenosine reader hnRNPA2B1 recognizes and stabilizes NEAT1 to confer chemoresistance in gastric cancer. Cancer Commun (Lond) 2024; 44:469-490. [PMID: 38512764 PMCID: PMC11024687 DOI: 10.1002/cac2.12534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Chemoresistance is a major cause of treatment failure in gastric cancer (GC). Heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) is an N6-methyladenosine (m6A)-binding protein involved in a variety of cancers. However, whether m6A modification and hnRNPA2B1 play a role in GC chemoresistance is largely unknown. In this study, we aimed to investigate the role of hnRNPA2B1 and the downstream mechanism in GC chemoresistance. METHODS The expression of hnRNPA2B1 among public datasets were analyzed and validated by quantitative PCR (qPCR), Western blotting, immunofluorescence, and immunohistochemical staining. The biological functions of hnRNPA2B1 in GC chemoresistance were investigated both in vitro and in vivo. RNA sequencing, methylated RNA immunoprecipitation, RNA immunoprecipitation, and RNA stability assay were performed to assess the association between hnRNPA2B1 and the binding RNA. The role of hnRNPA2B1 in maintenance of GC stemness was evaluated by bioinformatic analysis, qPCR, Western blotting, immunofluorescence, and sphere formation assays. The expression patterns of hnRNPA2B1 and downstream regulators in GC specimens from patients who received adjuvant chemotherapy were analyzed by RNAscope and multiplex immunohistochemistry. RESULTS Elevated expression of hnRNPA2B1 was found in GC cells and tissues, especially in multidrug-resistant (MDR) GC cell lines. The expression of hnRNPA2B1 was associated with poor outcomes of GC patients, especially in those who received 5-fluorouracil treatment. Silencing hnRNPA2B1 effectively sensitized GC cells to chemotherapy by inhibiting cell proliferation and inducing apoptosis both in vitro and in vivo. Mechanically, hnRNPA2B1 interacted with and stabilized long noncoding RNA NEAT1 in an m6A-dependent manner. Furthermore, hnRNPA2B1 and NEAT1 worked together to enhance the stemness properties of GC cells via Wnt/β-catenin signaling pathway. In clinical specimens from GC patients subjected to chemotherapy, the expression levels of hnRNPA2B1, NEAT1, CD133, and CD44 were markedly elevated in non-responders compared with responders. CONCLUSION Our findings indicated that hnRNPA2B1 interacts with and stabilizes lncRNA NEAT1, which contribute to the maintenance of stemness property via Wnt/β-catenin pathway and exacerbate chemoresistance in GC.
Collapse
Affiliation(s)
- Jiayao Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- The Air Force Hospital of Southern Theater CommandGuangzhouGuangdongP. R. China
| | - Jiehao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- The Air Force Hospital of Southern Theater CommandGuangzhouGuangdongP. R. China
| | - Hao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Lingnan Meng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNanningGuangxiP. R. China
| | - Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Yihan Zhao
- Second Clinical CollegeShaanxi University of Traditional Chinese MedicineXianyangShaanxiP. R. China
| | - Chen Wang
- College of Life SciencesNorthwest UniversityXi'anShaanxiP. R. China
| | - Xiaoliang Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Ahui Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Tianyu Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| |
Collapse
|
5
|
Liu J, Yuan Q, Guo H, Guan H, Hong Z, Shang D. Deciphering drug resistance in gastric cancer: Potential mechanisms and future perspectives. Biomed Pharmacother 2024; 173:116310. [PMID: 38394851 DOI: 10.1016/j.biopha.2024.116310] [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: 11/24/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Gastric cancer (GC) is a malignant tumor that originates from the epithelium of the gastric mucosa. The latest global cancer statistics show that GC ranks fifth in incidence and fourth in mortality among all cancers, posing a serious threat to public health. While early-stage GC is primarily treated through surgery, chemotherapy is the frontline option for advanced cases. Currently, commonly used chemotherapy regimens include FOLFOX (oxaliplatin + leucovorin + 5-fluorouracil) and XELOX (oxaliplatin + capecitabine). However, with the widespread use of chemotherapy, an increasing number of cases of drug resistance have emerged. This article primarily explores the potential mechanisms of chemotherapy resistance in GC patients from five perspectives: cell death, tumor microenvironment, non-coding RNA, epigenetics, and epithelial-mesenchymal transition. Additionally, it proposes feasibility strategies to overcome drug resistance from four angles: cancer stem cells, tumor microenvironment, natural products, and combined therapy. The hope is that this article will provide guidance for researchers in the field and bring hope to more GC patients.
Collapse
Affiliation(s)
- Jiahua Liu
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hewen Guan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Zhijun Hong
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Dong Shang
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
6
|
Xiong JX, Li YT, Tan XY, Chen T, Liu BH, Fu L. Targeting PRSS23 with tipranavir induces gastric cancer stem cell apoptosis and inhibits growth of gastric cancer via the MKK3/p38 MAPK-IL24 pathway. Acta Pharmacol Sin 2024; 45:405-421. [PMID: 37814123 PMCID: PMC10789761 DOI: 10.1038/s41401-023-01165-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
Gastric cancer stem cells (GCSCs) contribute to the refractory features of gastric cancer (GC) and are responsible for metastasis, relapse, and drug resistance. The key factors drive GCSC function and affect the clinical outcome of GC patients remain poorly understood. PRSS23 is a novel serine protease that is significantly up-regulated in several types of cancers and cancer stem cells, and related to tumor progression and drug resistance. In this study, we investigated the role of PRSS23 in GCSCs as well as the mechanism by which PRSS23 regulated the GCSC functions. We demonstrated that PRSS23 was critical for sustaining GCSC survival. By screening a collection of human immunodeficiency virus (HIV) protease inhibitors (PIs), we identified tipranavir as a PRSS23-targeting drug, which effectively killed both GCSC and GC cell lines (its IC50 values were 4.7 and 6.4 μM in GCSC1 cells and GCSC2 cells, respectively). Administration of tipranavir (25 mg·kg-1·d-1, i.p., for 8 days) in GCSC-derived xenograft mice markedly inhibited the growth of subcutaneous GCSC tumors without apparent toxicity. In contrast, combined treatment with 5-FU plus cisplatin did not affect the tumor growth but causing significant weight loss. Furthermore, we revealed that tipranavir induced GCSC cell apoptosis by suppressing PRSS23 expression, releasing MKK3 from the PRSS23/MKK3 complex to activate p38 MAPK, and thereby activating the IL24-mediated Bax/Bak mitochondrial apoptotic pathway. In addition, tipranavir was found to kill other types of cancer cell lines and drug-resistant cell lines. Collectively, this study demonstrates that by targeting both GCSCs and GC cells, tipranavir is a promising anti-cancer drug, and the clinical development of tipranavir or other drugs specifically targeting the PRSS23/MKK3/p38MAPK-IL24 mitochondrial apoptotic pathway may offer an effective approach to combat gastric and other cancers.
Collapse
Affiliation(s)
- Ji-Xian Xiong
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| | - Yu-Ting Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Xiang-Yu Tan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Tie Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Bao-Hua Liu
- Department of Biochemistry, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Zheng L, Chen J, Ye W, Fan Q, Chen H, Yan H. An individualized stemness-related signature to predict prognosis and immunotherapy responses for gastric cancer using single-cell and bulk tissue transcriptomes. Cancer Med 2024; 13:e6908. [PMID: 38168907 PMCID: PMC10807574 DOI: 10.1002/cam4.6908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Currently, many stemness-related signatures have been developed for gastric cancer (GC) to predict prognosis and immunotherapy outcomes. However, due to batch effects, these signatures cannot accurately analyze patients one by one, rendering them impractical in real clinical scenarios. Therefore, we aimed to develop an individualized and clinically applicable signature based on GC stemness. METHODS Malignant epithelial cells from single-cell RNA-Seq data of GC were used to identify stemness-related signature genes based on the CytoTRACE score. Using two bulk tissue datasets as training data, the enrichment scores of the signature genes were applied to classify samples into two subtypes. Then, using the identified subtypes as criteria, we developed an individualized stemness-related signature based on the within-sample relative expression orderings of genes. RESULTS We identified 175 stemness-related signature genes, which exhibited significantly higher AUCell scores in poorly differentiated GCs compared to differentiated GCs. In training datasets, GC samples were classified into two subtypes with significantly different survival times and genomic characteristics. Utilizing the two subtypes, an individualized signature was constructed containing 47 gene pairs. In four independent testing datasets, GC samples classified as high risk exhibited significantly shorter survival times, higher infiltration of M2 macrophages, and lower immune responses compared to low-risk samples. Moreover, the potential therapeutic targets and corresponding drugs were identified for the high-risk group, such as CD248 targeted by ontuxizumab. CONCLUSIONS We developed an individualized stemness-related signature, which can accurately predict the prognosis and efficacy of immunotherapy for each GC sample.
Collapse
Affiliation(s)
- Linyong Zheng
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and EngineeringFujian Medical UniversityFuzhouChina
| | - Jingyan Chen
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and EngineeringFujian Medical UniversityFuzhouChina
| | - Wenhai Ye
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and EngineeringFujian Medical UniversityFuzhouChina
| | - Qi Fan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and EngineeringFujian Medical UniversityFuzhouChina
| | - Haifeng Chen
- Department of Gastrointestinal SurgeryFuzhou Second HospitalFuzhouChina
| | - Haidan Yan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and EngineeringFujian Medical UniversityFuzhouChina
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical SciencesFujian Medical UniversityFuzhouChina
| |
Collapse
|
8
|
Yang Y, Yuan Y, Xia B. RHBDD1 Promotes the Growth and Stemness Characteristics of Gastric Cancer Cells by Activating Wnt/β-catenin Signaling Pathway. Curr Stem Cell Res Ther 2024; 19:1021-1028. [PMID: 37861049 DOI: 10.2174/011574888x259932231010112521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUNDS Gastric cancer (GC) is threatening public health, with at least one million new cases reported each year. Rhomboid domain-containing protein 1 (RHBDD1) has been identified to regulate the proliferation, migration, and metastasis of cancer cells. However, the role of RHBDD1 in GC has not been elucidated. OBJECTS This study aimed to investigate the role of RHBDD1 on the growth, metastasis, and stemness characteristics of GC. METHODS RHBDD1 expression was analyzed from the TCGA databank. qRT-PCR was conducted to evaluate the transcription level of RHBDD1. Western blots were used to evaluate the protein expression of RHBDD1, CD133, CD44, Nanog, β-catenin and c-myc. Colony formation assay and transwell assay were conducted to evaluate the growth and metastasis of NCI-N87 cells, respectively. Sphere-forming assay was performed to study the stemness characteristics. The nude mice xenotransplantation model and immunohistochemistry (IHC) were performed to evaluate the growth of GC in vivo. Results: RHBDD1 expression is elevated in GC cells and clinical tissues. RHBDD1 expression is positively associated with cell proliferation and metastasis of GC cells. RHBDD1 knockdown suppresses the expression of CD133, CD44 and Nanog and attenuates sphere-forming ability. RHBDD1 activates the Wnt/β-catenin pathway via promoting the expression of β-catenin / c-myc and inducing β-catenin translocation into nuclear. RHBDD1 knockdown inhibits the growth of GC in nude mice xenotransplantation model. CONCLUSION RHBDD1 is highly expressed in GC, and its knockdown inhibits the growth, metastasis and stemness characteristics of GC cells through activating the Wnt/β-catenin pathway, suggesting that RHBDD1 has the potential to be a novel therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Yingxue Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical, Chongqing, 400010, China
| | - Yuan Yuan
- Department of Ultrasound, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400010, China
| | - Boning Xia
- Department of Gastrointestinal Anorectal Surgery, The Second Affiliated Hospital of Chongqing Medical, Chongqing, 400010, China
| |
Collapse
|
9
|
Xu Q, Cheng W, Wei J, Ou Y, Xiao X, Jia Y. Synergist for antitumor therapy: Astragalus polysaccharides acting on immune microenvironment. Discov Oncol 2023; 14:179. [PMID: 37741920 PMCID: PMC10517906 DOI: 10.1007/s12672-023-00798-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023] Open
Abstract
Various new treatments are emerging constantly in anti-tumor therapies, including chemotherapy, immunotherapy, and targeted therapy. However, the efficacy is still not satisfactory. Astragalus polysaccharide is an important bioactive component derived from the dry root of Radix astragali. Studies found that astragalus polysaccharides have gained great significance in increasing the sensitivity of anti-tumor treatment, reducing the side effects of anti-tumor treatment, reversing the drug resistance of anti-tumor drugs, etc. In this review, we focused on the role of astragalus polysaccharides in tumor immune microenvironment. We reviewed the immunomodulatory effect of astragalus polysaccharides on macrophages, dendritic cells, natural killer cells, T lymphocytes, and B lymphocytes. We found that astragalus polysaccharides can promote the activities of macrophages, dendritic cells, natural killer cells, T lymphocytes, and B lymphocytes and induce the expression of a variety of cytokines and chemokines. Furthermore, we summarized the clinical applications of astragalus polysaccharides in patients with digestive tract tumors. We summarized the effective mechanism of astragalus polysaccharides on digestive tract tumors, including apoptosis induction, proliferation inhibition, immunoactivity regulation, enhancement of the anticancer effect and chemosensitivity. Therefore, in view of the multiple functions of astragalus polysaccharides in tumor immune microenvironment and its clinical efficacy, the combination of astragalus polysaccharides with antitumor therapy such as immunotherapy may provide new sparks to the bottleneck of current treatment methods.
Collapse
Affiliation(s)
- Qian Xu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wen Cheng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jinrui Wei
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yan Ou
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xian Xiao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingjie Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
10
|
Akrami H, Shamsdin SA, Nikmanesh Y, Fattahi M. Effect of Mir-4270 Inhibitor and Mimic on Viability and Stemness in Gastric Cancer Stem-Like Cells Derived from MKN-45 Cell Line. IRANIAN BIOMEDICAL JOURNAL 2023; 27:100-7. [PMID: 37070617 PMCID: PMC10314761 DOI: 10.61186/ibj.3851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2023]
Abstract
Background MicroRNAs (miRNAs) are significant regulatory factors in stem cell proliferation, and change in miRNA expression influences the cancer stem cell viability and gene expression. Herein, we evaluated the effect of the hsa-miR-4270 inhibitor and its mimic on the expression of stem cell markers in gastric cancer (GC) stem-like cells. Methods GC stem-like cells were isolated from the MKN-45 cell line by a non-adherent surface system. The cells were confirmed by differentiation assays using dexamethasone and insulin as adipogenesis-inducing agents and also Staurosporine as a neural-inducing agent. Isolated GC stem-like cells were treated with different concentrations (0, 15, 20, 25, 30, 40, 50, and 60 nM) of hsa-miR-4270 inhibitor and its mimic. The quantity of cell viability was determined by trypan blue method. Transcription of the stem cell marker genes, including CD44, OCT3/4, SOX2, Nanog, and KLF4, was evaluated by real-time RT-PCR. Results The results showed that GC stem-like cells were differentiated into both adipose cells using dexamethasone and insulin and neural cells by Staurosporine. Treatment of GC stem-like cells with hsa-miR-4270 inhibitor decreased cell viability and downregulated OCT3/4, CD44, and Nanog to 86%, 79%, and 91% respectively. Also, SOX2 and KLF4 were overexpressed to 8.1- and 1.94-folds, respectively. However, hsa-miR-4270 mimic had opposite effects on the cell viability and gene expression of the stem cell markers. Conclusion The effect of hsa-miR-4270 inhibitor and its mimic on the expression of the stem cell markers in GCSCs indicated that hsa-miR-4270 stimulates the stemness property of GCSCs, likely through stimulating the development of gastric stem cells.
Collapse
Affiliation(s)
- Hassan Akrami
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | |
Collapse
|
11
|
Li X, Li L, Wu J. The members of the miR-148/152 family inhibit cancer stem cell-like properties in gastric cancer via negative regulation of ITGA5. J Transl Med 2023; 21:105. [PMID: 36765401 PMCID: PMC9912648 DOI: 10.1186/s12967-023-03894-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND The role of microRNA (miRNA) in modulating the function of cancer stem cells through diverse signaling pathway has been evidenced. We here identified a role of microRNA (miRNA) family, specifically miR-148/152, in gastric cancer and delineated its functional effects on gastric cancer stem cells. METHODS Bioinformatics analysis was conducted to analyze expression of integrin α5 (ITGA5) which was verified through expression determination in clinical tissue samples. Next, the upstream regulatory factors of ITGA5 were determined. CD44+EpCAM (high) cells sorted from AGS cells subjected to gain-of-function experiments, followed by evaluation of their capacity of colony formation, generation of tumorosphere, cell migration and viability in vitro and xenograft tumor formation in vivo. RESULTS ITGA5 was elevated in gastric cancer tissues and confirmed as a target gene of the miR-148/152 family members. The miR-148/152 family members were downregulated in gastric cancer tissues and cells. Decreased expression of miR-148/152 family members was also detected in gastric cancer stem cells. However, the raised expression led to reduced colony formation, tumorosphere, cell migration, cell viability, and drug resistance of CD44+EpCAM (high) AGS cells in vitro, and tumorigenesis in vitro. ITGA5 overexpression reversed the effect of the miR-148/152 family members. CONCLUSIONS This study demonstrates that the miR-148/152 family members may prevent gastric cancer stem cell-like properties by targeting ITGA5, which can serve as an appealing target for gastric cancer treatment.
Collapse
Affiliation(s)
- Xiaoying Li
- grid.412644.10000 0004 5909 0696Department of Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 People’s Republic of China
| | - Lin Li
- grid.412644.10000 0004 5909 0696Department of Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032 People’s Republic of China
| | - Jiangying Wu
- Department of Intervention, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, People's Republic of China.
| |
Collapse
|
12
|
Yang Y, Meng WJ, Wang ZQ. The origin of gastric cancer stem cells and their effects on gastric cancer: Novel therapeutic targets for gastric cancer. Front Oncol 2022; 12:960539. [PMID: 36185219 PMCID: PMC9520244 DOI: 10.3389/fonc.2022.960539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent malignancies and the most common causes of cancer-related mortality worldwide. Furthermore, the prognosis of advanced GC remains poor even after surgery combined with chemoradiotherapy. As a small group of cells with unlimited differentiation and self-renewal ability in GC, accumulating evidence shows that GC stem cells (GCSCs) are closely associated with the refractory characteristics of GC, such as drug resistance, recurrence, and metastasis. With the extensive development of research on GCSCs, GCSCs seem to be promising therapeutic targets for GC. However, the relationship between GCSCs and GC is profound and intricate, and its mechanism of action is still under exploration. In this review, we elaborate on the source and key concepts of GCSCs, systematically summarize the role of GCSCs in GC and their underlying mechanisms. Finally, we review the latest information available on the treatment of GC by targeting GCSCs. Thus, this article may provide a theoretical basis for the future development of the novel targets based on GCSCs for the treatment of GC.
Collapse
|
13
|
Islas JF, Quiroz-Reyes AG, Delgado-Gonzalez P, Franco-Villarreal H, Delgado-Gallegos JL, Garza-Treviño EN, Gonzalez-Villarreal CA. Cancer Stem Cells in Tumor Microenvironment of Adenocarcinoma of the Stomach, Colon, and Rectum. Cancers (Basel) 2022; 14:3948. [PMID: 36010940 PMCID: PMC9405851 DOI: 10.3390/cancers14163948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Gastrointestinal adenocarcinomas are one of the world's deadliest cancers. Cancer stem cells and the tissue microenvironment are highly regulated by cell and molecular mechanisms. Cancer stem cells are essential for maintenance and progression and are associated with resistance to conventional treatments. This article reviews the current knowledge of the role of the microenvironment during the primary establishment of gastrointestinal adenocarcinomas in the stomach, colon, and rectum and its relationship with cancer stem cells. We also describe novel developments in cancer therapeutics, such as targeted therapy, and discuss the advantages and disadvantages of different treatments for improving gastrointestinal cancer prognosis.
Collapse
Affiliation(s)
- Jose Francisco Islas
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Adriana G. Quiroz-Reyes
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Paulina Delgado-Gonzalez
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | | - Juan Luis Delgado-Gallegos
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Elsa N. Garza-Treviño
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | |
Collapse
|
14
|
Abstract
Like most solid tumours, the microenvironment of epithelial-derived gastric adenocarcinoma (GAC) consists of a variety of stromal cell types, including fibroblasts, and neuronal, endothelial and immune cells. In this article, we review the role of the immune microenvironment in the progression of chronic inflammation to GAC, primarily the immune microenvironment driven by the gram-negative bacterial species Helicobacter pylori. The infection-driven nature of most GACs has renewed awareness of the immune microenvironment and its effect on tumour development and progression. About 75-90% of GACs are associated with prior H. pylori infection and 5-10% with Epstein-Barr virus infection. Although 50% of the world's population is infected with H. pylori, only 1-3% will progress to GAC, with progression the result of a combination of the H. pylori strain, host susceptibility and composition of the chronic inflammatory response. Other environmental risk factors include exposure to a high-salt diet and nitrates. Genetically, chromosome instability occurs in ~50% of GACs and 21% of GACs are microsatellite instability-high tumours. Here, we review the timeline and pathogenesis of the events triggered by H. pylori that can create an immunosuppressive microenvironment by modulating the host's innate and adaptive immune responses, and subsequently favour GAC development.
Collapse
|
15
|
Saito S, Ku CC, Wuputra K, Pan JB, Lin CS, Lin YC, Wu DC, Yokoyama KK. Biomarkers of Cancer Stem Cells for Experimental Research and Clinical Application. J Pers Med 2022; 12:715. [PMID: 35629138 PMCID: PMC9147761 DOI: 10.3390/jpm12050715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
The use of biomarkers in cancer diagnosis, therapy, and prognosis has been highly effective over several decades. Studies of biomarkers in cancer patients pre- and post-treatment and during cancer progression have helped identify cancer stem cells (CSCs) and their related microenvironments. These analyses are critical for the therapeutic application of drugs and the efficient targeting and prevention of cancer progression, as well as the investigation of the mechanism of the cancer development. Biomarkers that characterize CSCs have thus been identified and correlated to diagnosis, therapy, and prognosis. However, CSCs demonstrate elevated levels of plasticity, which alters their functional phenotype and appearance by interacting with their microenvironments, in response to chemotherapy and radiotherapeutics. In turn, these changes induce different metabolic adaptations of CSCs. This article provides a review of the most frequently used CSCs and stem cell markers.
Collapse
Affiliation(s)
- Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita 329-1571, Japan
- Horus Co., Ltd., Nakano, Tokyo 164-0001, Japan
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Jia-Bin Pan
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.); (J.-B.P.); (C.-S.L.)
| | - Ying-Chu Lin
- School of Dentistry, Department of Dentistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Deng-Chyang Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.); (J.-B.P.); (C.-S.L.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| |
Collapse
|
16
|
Aldehyde Dehydrogenase 1 in Gastric Cancer. JOURNAL OF ONCOLOGY 2022; 2022:5734549. [PMID: 35310914 PMCID: PMC8926486 DOI: 10.1155/2022/5734549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/01/2022] [Accepted: 02/03/2022] [Indexed: 01/09/2023]
Abstract
Gastric cancer (GC) is a disease that threatens human health. It is thus crucial to clarify the mechanisms involved in GC development and discover diagnostic biomarkers and therapeutics. As a cancer stem cell marker, aldehyde dehydrogenase 1 (ALDH1) is involved in the development, progression, and treatment of GC. This review evaluated the prognostic value of ALDH1 and explored its mechanism of action in GC. Importantly, ALDH1 is an informative biomarker in clinical practice as it has specific relationships with indicators, such as metastasis and overall survival. Additionally, ALDH1 interacts with genes and exhibits properties that mimic stem cell characteristics amongst other mechanisms employed in the occurrence and progression of GC. Our results, therefore, provide evidence of possible clinical utility of ALDH1 as a GC therapeutic target.
Collapse
|
17
|
Farahani DB, Akrami H, Moradi B, Mehdizadeh K, Fattahi MR. The Effect of hsa-miR-451b Knockdown on Biological Functions of Gastric Cancer Stem-Like Cells. Biochem Genet 2021; 59:1203-1224. [PMID: 33725258 DOI: 10.1007/s10528-021-10057-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/02/2021] [Indexed: 01/15/2023]
Abstract
Numerous researches have extensively studied factors such as microRNAs that lead to cancer. Thus, the current study's purpose is to investigate the biological consequences of hsa-miR-451b inhibition on the properties and functions of gastric cancer stem-like cells. First, gastric cancer stem-like cells were transfected by hsa-miR-451b inhibitor then we used real-time RT-PCR to evaluate its effect on the expression of hsa-miR-451b and two of its direct target genes, Stemness markers such as KLF4, SOX2, CD44, OCT3/4 and NANOG genes and finally Akt, PI3K, Bcl-2, Bax, CASP3 and PCNA genes involved in apoptosis. Here, we conducted a DNA Laddering assay to investigate apoptosis. The level of the MMP-2 and -9 Activities and Migration were examined by Zymography and Transwell invasion assay. HUVEC cells were used to investigate angiogenesis. The outcomes revealed that the level of the MMP-2 and -9 Activities, migration and angiogenesis decreased, but apoptosis was induced by inhibiting hsa-miR-451b. Evaluating KREMEN1 and CASK expression showed that the former increased, and the latter dropped under hsa-miR-451b inhibition. Also, upregulation of the KLF4 and SOX2 and downregulation of the CD44, OCT3/4, and NANOG decreased Self-renewal ability of gastric cancer stem cells under hsa-miR-451b inhibition. Even, under hsa-miR-451b inhibition, downregulation of Akt, PI3K, Bcl-2 and PCNA as well as upregulation of Bax and CASP3 revealed a movement towards apoptosis in MKN-45 stem-like cells. In summary, hsa-miR-451b is an oncomir in the carcinogenesis of gastric cancer stem-like cells and may be suggested as an appropriate therapeutic target for future gastric cancer treatment.
Collapse
Affiliation(s)
| | - Hassan Akrami
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, 71935-1311, Shiraz, Iran.
| | - Behrouz Moradi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Kiumars Mehdizadeh
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Mohammad Reza Fattahi
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, 71935-1311, Shiraz, Iran
| |
Collapse
|
18
|
Yasuda T, Ishimoto T, Baba H. Conflicting metabolic alterations in cancer stem cells and regulation by the stromal niche. Regen Ther 2021; 17:8-12. [PMID: 33598509 PMCID: PMC7851492 DOI: 10.1016/j.reth.2021.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies have revealed that cancer stem cells (CSCs) undergo metabolic alterations that differentiate them from non-CSCs. Inhibition of specific metabolic pathways in CSCs has been conducted to eliminate the CSC population in many types of cancer. However, there is conflicting evidence about whether CSCs depend on glycolysis or mitochondrial oxidative phosphorylation (OXPHOS) to maintain their stem cell properties. This review summarizes the latest knowledge regarding CSC-specific metabolic alterations and offers recent evidence that the surrounding microenvironments may play an important role in the maintenance of CSC properties.
Collapse
Key Words
- ALDH, aldehyde dehydrogenase
- ATP, adenosine triphosphate
- CD44v, CD44 variant isoform
- CSCs
- CSCs, cancer stem cells
- EMT, epithelial–mesenchymal transition
- EVs, extracellular vesicles
- FAO, fatty acid oxidation
- FBP1, fructose-1,6-biphosphatase 1
- GLUT1, glucose transporter 1
- GP6, glucose-6-phosphate
- Glycolysis
- HCC, hepatocellular carcinoma
- HIF1a, hypoxia inducible factor 1a
- IMP2, insulin-like growth factor 2
- IncRNAs, long noncoding RNAs
- LSCs, leukemia stem cells
- Mitochondrial OXPHOS
- NRF2, nuclear factor erythroid 2–related factor 2
- OXPHOS, oxidative phosphorylation
- PDK1, pyruvate dehydrogenase kinase 1
- PPP, pentose phosphate pathway
- ROS
- ROS, reactive oxygen species
- SOD2, superoxide dismutase 2
- Stromal niche
- TCA, tricarboxylic acid
- TICs, tumor initiating stem-like cells
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Tadahito Yasuda
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Lin T, Peng W, Mai P, Zhang E, Peng L. Human Gastric Cancer Stem Cell (GCSC) Markers Are Prognostic Factors Correlated With Immune Infiltration of Gastric Cancer. Front Mol Biosci 2021; 8:626966. [PMID: 34113647 PMCID: PMC8185345 DOI: 10.3389/fmolb.2021.626966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
The prognosis of patients with gastric cancer (GC) is still unsatisfying. Numerous markers of gastric cancer stem cells (GCSCs) have been identified and were thought to be related to cancer aggressiveness. However, the roles of GCSC markers in GC patients’ prognosis and immune infiltration remain unknown. Expression of GCSC markers was analyzed using Oncomine and Gene Expression Profiling Interactive Analysis (GEPIA). Their associations with clinicopathological parameters were analyzed using UALCAN and LinkedOmics. Alternations and protein expression of GCSC markers were analyzed by cBioPortal and the Human Protein Atlas databases, respectively. The prognostic significance of GCSC markers was evaluated using Kaplan-Meier plotter. Correlations between the expression of GCSC markers and immune infiltration along with biomarkers of tumor-infiltrating immune cells (TIICs) were assessed combined Tumor Immune Estimation Resource and GEPIA. GeneMANIA was used to discover the interactive genes of GCSC markers, and enrichment analysis was performed using Database for Annotation, Visualization, and Integrated Discovery server. We identified six GCSC markers significantly up-expressed in GC, compared with normal stomach tissues. Among them, the overexpression of ICAM1, THY1, and CXCR4 significantly indicated adverse, while EPCAM indicated beneficial clinicopathological features of GC patients. The up-regulation of CXCR4 showed unfavorable prognostic significance, whereas EPCAM and TFRC showed the opposite. The six GCSC markers were all correlated with the infiltration and activation of distinct TIICs. Especially, ICAM1, THY1, and CXCR4 showed strongly positive correlations with tumor-associated macrophages. Besides, chemokine, Toll-like receptor, NF-kappa B, and HIF-1 signaling pathways might be involved in the regulation of GCSC markers on cancer development. This study proposed that GCSC markers might be promising targets of GC treatment to weaken cancer stem-like properties and strengthen anticancer immunity.
Collapse
Affiliation(s)
- Tong Lin
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wenya Peng
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Peipei Mai
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - E Zhang
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lisheng Peng
- Department of Science and Education, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
20
|
Shiozaki A, Katsurahara K, Kudou M, Shimizu H, Kosuga T, Ito H, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, Okamoto K, Otsuji E. Amlodipine and Verapamil, Voltage-Gated Ca 2+ Channel Inhibitors, Suppressed the Growth of Gastric Cancer Stem Cells. Ann Surg Oncol 2021; 28:5400-5411. [PMID: 33566246 DOI: 10.1245/s10434-021-09645-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/06/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND The membrane transporters activated in cancer stem cells (CSCs) are the target of novel cancer therapies for gastric cancer. The present study investigated ion channel expression profiles in gastric CSCs (GCSCs). METHODS Cells strongly expressing CD44 were separated from MKN74 cells, a human gastric cancer cell line, by fluorescence-activated cell sorting (FACS), and GCSCs were identified based on tumorsphere formation. Gene expression profiles in GCSCs were examined by a microarray analysis. RESULTS Among MKN74 cells, CD44 messenger RNA levels were higher in CSCs than in non-CSCs. These CSCs also exhibited resistance to cisplatin. The microarray analysis revealed that the expression of several genes related to voltage-gated Ca2+ channels (VGCCs), including CACNA2D1 and CACNB4, was upregulated. The cytotoxicities of the CACNA2D1 inhibitor amlodipine and the CACNB4 inhibitor verapamil were greater at lower concentrations in CSCs than in non-CSCs, and markedly reduced tumorsphere numbers. Tumor volumes were significantly smaller in a xenograft nude mouse model treated with amlodipine or verapamil in combination with cisplatin than in that treated with cisplatin alone. CONCLUSIONS The present results indicate that VGCCs play a role in maintaining CSCs, and demonstrated the potential of their specific inhibitors, amlodipine and verapamil, as targeted therapeutic agents against gastric cancer.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Keita Katsurahara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ito
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
21
|
Tumor microenvironment characterization in stage IV gastric cancer. Biosci Rep 2021; 41:227294. [PMID: 33416081 PMCID: PMC7796192 DOI: 10.1042/bsr20201248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is remarkably affected by the immune environment of the principal tumor. Nonetheless, the immune environment's clinical relevance in stage IV gastric cancer (GC) is largely unknown. The gene expression profiles of 403 stage IV GC patients in the three cohorts: GEO (Gene Expression Omnibus, GSE84437 (n=292) and GSE62254 (n=77), and TCGA (The Cancer Genome Atlas, n=34) were used in the present study. Using four publicly available stage IV GC expression datasets, 29 immune signatures were expression profiled, and on this basis, we classified stage IV GC. The classification was conducted using the hierarchical clustering method. Three stage IV GC subtypes L, M, and H were identified representing low, medium, and high immunity, respectively. Immune correlation analysis of these three types revealed that Immune H exhibited a better prognostic outcome as well as a higher immune score compared with other subtypes. There was a noticeable difference in the three subgroups of HLA genes. Further, on comparing with other subtypes, CD86, CD80, CD274, CTLA4, PDCD1, and PDCD1LG2 had higher expression in the Immunity H subtype. In stage IV GC, potentially positive associations between immune and pathway activities were displayed, due to the enrichment of pathways including TNF signaling, Th-17 cell differentiation, and JAK-STAT signaling pathways in Immunity H vs Immunity L subtypes. External cohorts from TCGA cohort ratified these results. The identification of stage IV GC subtypes has potential clinical implications in stage IV GC treatment.
Collapse
|
22
|
Robinson K, Atherton JC. The Spectrum of Helicobacter-Mediated Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 16:123-144. [PMID: 33197219 DOI: 10.1146/annurev-pathol-032520-024949] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Helicobacter pylori is the leading cause of peptic ulcer disease. The infection has been implicated in more than 75% of duodenal ulcer cases and 17% of gastric ulcer cases. H. pylori has been classified as a human carcinogen, since it is the main cause of distal gastric adenocarcinoma and B cell mucosa-associated lymphoid tissue lymphoma. Evidence also links H. pylori with extragastric conditions including iron deficiency anemia, idiopathic thrombocytopenic purpura, and vitamin B12 deficiency. Studies indicate that H. pylori may be protective against other conditions of the gastrointestinal tract (e.g., reflux esophagitis and related pathologies) and elsewhere in the body (e.g., asthma). The infection is asymptomatic in the vast majority of cases; more serious outcomes occur in only 10-15% of infected individuals. Despite extensive research over the past 3 decades, there is no effective vaccine, and the circumstances leading to disease development remain unclear. In addition, there is now a growing prevalence of antimicrobial resistance in H. pylori. This review discusses these important issues.
Collapse
Affiliation(s)
- Karen Robinson
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham NG7 2RD United Kingdom;
| | - John C Atherton
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham NG7 2RD United Kingdom;
| |
Collapse
|
23
|
Kim DH, Lee S, Kang HG, Park HW, Lee HW, Kim D, Yoem DH, Ahn JH, Ha E, You WK, Lee SH, Kim SJ, Chun KH. Synergistic antitumor activity of a DLL4/VEGF bispecific therapeutic antibody in combination with irinotecan in gastric cancer. BMB Rep 2020. [PMID: 32580836 PMCID: PMC7607148 DOI: 10.5483/bmbrep.2020.53.10.103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Notch signaling has been identified as a critical pathway in gastric cancer (GC) progression and metastasis, and inhibition of Delta-like ligand 4 (DLL4), a Notch ligand, is suggested as a potent therapeutic approach for GC. Expression of both DLL4 and vascular endothelial growth factor receptor 2 (VEGFR2) was similar in the malignant tissues of GC patients. We focused on vascular endothelial growth factor (VEGF), a known angiogenesis regulator and activator of DLL4. Here, we used ABL001, a DLL4/VEGF bispecific therapeutic antibody, and investigated its therapeutic effect in GC. Treatment with human DLL4 therapeutic antibody (anti-hDLL4) or ABL001 slightly reduced GC cell growth in monolayer culture; however, they significantly inhibited cell growth in 3D-culture, suggesting a reduction in the cancer stem cell population. Treatment with anti-hDLL4 or ABL001 also decreased GC cell migration and invasion. Moreover, the combined treatment of irinotecan with anti-hDLL4 or ABL001 showed synergistic antitumor activity. Both combination treatments further reduced cell growth in 3D-culture as well as cell invasion. Interestingly, the combination treatment of ABL001 with irinotecan synergistically reduced the GC burden in both xenograft and orthotopic mouse models. Collectively, DLL4 inhibition significantly decreased cell motility and stem-like phenotype and the combination treatment of DLL4/VEGF bispecific therapeutic antibody with irinotecan synergistically reduced the GC burden in mouse models. Our data suggest that ABL001 potentially represents a potent agent in GC therapy. Further biochemical and pre-clinical studies are needed for its application in the clinic.
Collapse
Affiliation(s)
- Da-Hyun Kim
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Goyang 10408, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Goyang 10408, Korea
| | - Seul Lee
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Goyang 10408, Korea
| | - Hyeok Gu Kang
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Goyang 10408, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Goyang 10408, Korea
| | - Hyun-Woo Park
- Department of Biochemistry, College of Life Science, Yonsei University, Seoul 03722, Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science, Yonsei University, Seoul 03722, Korea
| | - Dongin Kim
- R&D center, ABL Bio Inc., Seongnam 13488, Korea
| | | | | | - Eunsin Ha
- R&D center, ABL Bio Inc., Seongnam 13488, Korea
- National OncoVenture, National Cancer Center, Goyang 10408, Korea
| | | | | | - Seok-Jun Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Goyang 10408, Korea
- Brain Korea 21 Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju 61452, Korea
- Corresponding authors. Seok-Jun Kim, Tel: +82-62-230-6664; Fax: +82-62-234-4326; E-mail: ; Kyung-Hee Chun, Tel: +82-2-2228-1699; Fax: +82-2-312-5041; E-mail:
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Goyang 10408, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Goyang 10408, Korea
- Corresponding authors. Seok-Jun Kim, Tel: +82-62-230-6664; Fax: +82-62-234-4326; E-mail: ; Kyung-Hee Chun, Tel: +82-2-2228-1699; Fax: +82-2-312-5041; E-mail:
| |
Collapse
|
24
|
Sun L, Huang C, Zhu M, Guo S, Gao Q, Wang Q, Chen B, Li R, Zhao Y, Wang M, Chen Z, Shen B, Zhu W. Gastric cancer mesenchymal stem cells regulate PD-L1-CTCF enhancing cancer stem cell-like properties and tumorigenesis. Am J Cancer Res 2020; 10:11950-11962. [PMID: 33204322 PMCID: PMC7667687 DOI: 10.7150/thno.49717] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: Mesenchymal stem cells (MSCs) have been the focus of many studies because of their abilities to modulate immune responses, angiogenesis, and promote tumor growth and metastasis. Our previous work showed that gastric cancer MSCs (GCMSCs) promoted immune escape by secreting of IL-8, which induced programmed cell death ligand 1 (PD-L1) expression in GC cells. Mounting evidence has revealed that PD-L1 expression is related to intrinsic tumor cell properties. Here, we investigated whether GCMSCs maintained a pool of cancer stem cells (CSCs) through PD-L1 signaling and the specific underlying molecular mechanism. Methods: Stem cell surface markers, aldehyde dehydrogenase (ALDH) activity, migration and sphere formation abilities were tested to evaluate the stemness of GC cells. PD-L1-expressing lentivirus and PD-L1 specific siRNA were used to analyze the effects of PD-L1 on GC cells stemness. Annexin V/PI double staining was used to assess apoptosis of GC cells induced by chemotherapy. Co-Immunoprecipitation (Co-IP) and Mass spectrometry were employed to determine the PD-L1 binding partner in GC cells. PD-L1Negative and PD-L1Positive cells were sorted by flow cytometry and used for limiting dilution assays to verify the effect of PD-L1 on tumorigenic ability in GC cells. Results: The results showed that GCMSCs enhanced the CSC-like properties of GC cells through PD-L1, which led to the resistance of GC cells to chemotherapy. PD-L1 associated with CTCF to contribute to the stemness and self-renewal of GC cells. In vivo, PD-L1Positive GC cells had greater stemness potential and tumorigenicity than PD-L1Negative GC cells. The results also indicated that GC cells were heterogeneous, and that PD-L1 in GC cells had different reactivity to GCMSCs. Conclusions: Overall, our data indicated that GCMSCs enriched CSC-like cells in GC cells, which gives a new insight into the mechanism of GCMSCs prompting GC progression and provides a potential combined therapeutic target.
Collapse
|
25
|
Chen X, Zhang D, Jiang F, Shen Y, Li X, Hu X, Wei P, Shen X. Prognostic Prediction Using a Stemness Index-Related Signature in a Cohort of Gastric Cancer. Front Mol Biosci 2020; 7:570702. [PMID: 33134315 PMCID: PMC7504590 DOI: 10.3389/fmolb.2020.570702] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/14/2020] [Indexed: 12/24/2022] Open
Abstract
Background With characteristic self-renewal and multipotent differentiation, cancer stem cells (CSCs) have a crucial influence on the metastasis, relapse and drug resistance of gastric cancer (GC). However, the genes that participates in the stemness of GC stem cells have not been identified. Methods The mRNA expression-based stemness index (mRNAsi) was analyzed with differential expressions in GC. The weighted gene co-expression network analysis (WGCNA) was utilized to build a co-expression network targeting differentially expressed genes (DEG) and discover mRNAsi-related modules and genes. We assessed the association between the key genes at both the transcription and protein level. Gene Expression Omnibus (GEO) database was used to validate the expression levels of the key genes. The risk model was established according to the least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Furthermore, we determined the prognostic value of the model by employing Kaplan-Meier (KM) plus multivariate Cox analysis. Results GC tissues exhibited a substantially higher mRNAsi relative to the healthy non-tumor tissues. Based on WGCNA, 17 key genes (ARHGAP11A, BUB1, BUB1B, C1orf112, CENPF, KIF14, KIF15, KIF18B, KIF4A, NCAPH, PLK4, RACGAP1, RAD54L, SGO2, TPX2, TTK, and XRCC2) were identified. These key genes were clearly overexpressed in GC and validated in the GEO database. The protein-protein interaction (PPI) network as assessed by STRING indicated that the key genes were tightly connected. After LASSO analysis, a nine-gene risk model (BUB1B, NCAPH, KIF15, RAD54L, KIF18B, KIF4A, TTK, SGO2, C1orf112) was constructed. The overall survival in the high-risk group was relatively poor. The area under curve (AUC) of risk score was higher compared to that of clinicopathological characteristics. According to the multivariate Cox analysis, the nine-gene risk model was a predictor of disease outcomes in GC patients (HR, 7.606; 95% CI, 3.037-19.051; P < 0.001). We constructed a prognostic nomogram with well-fitted calibration curve based on risk score and clinical data. Conclusion The 17 mRNAsi-related key genes identified in this study could be potential treatment targets in GC treatment, considering that they can inhibit the stemness properties. The nine-gene risk model can be employed to predict the disease outcomes of the patients.
Collapse
Affiliation(s)
- Xiaowei Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Dawei Zhang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Fei Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Yan Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Xin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xueju Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Pingmin Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
26
|
Molecular Bases of Mechanisms Accounting for Drug Resistance in Gastric Adenocarcinoma. Cancers (Basel) 2020; 12:cancers12082116. [PMID: 32751679 PMCID: PMC7463778 DOI: 10.3390/cancers12082116] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric adenocarcinoma (GAC) is the most common histological type of gastric cancer, the fifth according to the frequency and the third among the deadliest cancers. GAC high mortality is due to a combination of factors, such as silent evolution, late clinical presentation, underlying genetic heterogeneity, and effective mechanisms of chemoresistance (MOCs) that make the available antitumor drugs scarcely useful. MOCs include reduced drug uptake (MOC-1a), enhanced drug efflux (MOC-1b), low proportion of active agents in tumor cells due to impaired pro-drug activation or active drug inactivation (MOC-2), changes in molecular targets sensitive to anticancer drugs (MOC-3), enhanced ability of cancer cells to repair drug-induced DNA damage (MOC-4), decreased function of pro-apoptotic factors versus up-regulation of anti-apoptotic genes (MOC-5), changes in tumor cell microenvironment altering the response to anticancer agents (MOC-6), and phenotypic transformations, including epithelial-mesenchymal transition (EMT) and the appearance of stemness characteristics (MOC-7). This review summarizes updated information regarding the molecular bases accounting for these mechanisms and their impact on the lack of clinical response to the pharmacological treatment currently used in GAC. This knowledge is required to identify novel biomarkers to predict treatment failure and druggable targets, and to develop sensitizing strategies to overcome drug refractoriness in GAC.
Collapse
|
27
|
Pajuelo-Lozano N, Alcalá S, Sainz B, Perona R, Sanchez-Perez I. Targeting MAD2 modulates stemness and tumorigenesis in human Gastric Cancer cell lines. Am J Cancer Res 2020; 10:9601-9618. [PMID: 32863948 PMCID: PMC7449921 DOI: 10.7150/thno.49270] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Gastric cancer (GC) is a solid tumor that contains subpopulations of cancer stem cells (CSCs), which are considered drivers of tumor initiation and metastasis; responsible for therapeutic resistance; and promoters of tumor relapse. The balance between symmetric and asymmetric division is crucial for stem cell maintenance. The objective of this study is to evaluate the role of MAD2, a key protein for proper mitotic checkpoint activity, in the tumorigenesis of GC. Methods: Gastric cancer stem cells (GCSCs) were obtained from MKN45, SNU638 and ST2957 cell lines. Pluripotency and stemness markers were evaluated by RT-qPCR and autofluorescence and membrane markers by flow cytometry. Relevant signal transduction pathways were studied by WB. We analysed cell cycle progression, migration and invasion after modulation of MAD2 activity or protein expression levels in these in vitro models. In vivo assays were performed in a nude mouse subcutaneous xenograft model. Results: We found that NANOG, CXCR4 and autofluorescence are common and consistent markers for the GCSCs analysed, with other markers showing more variability. The three main signalling pathways (Wnt/β-catenin; Hedgehog and Notch) were activated in GCSCs. Downregulation of MAD2 in MKN45CSCs decreased the expression of markers CXCR4, CD133, CD90, LGR5 and VIM, without affecting cell cycle profile or therapy resistance. Moreover, migration, invasion and tumor growth were clearly reduced, and accordingly, we found that metalloprotease expression decreased. These results were accompanied by a reduction in the levels of transcription factors related with epithelial-to-mesenchymal transition. Conclusions: We can conclude that MAD2 is important for GCSCs stemness and its downregulation in MKN45CSCs plays a central role in GC tumorigenesis, likely through CXCR4-SNAI2-MMP1. Thus, its potential use in the clinical setting should be studied as its functions appear to extend beyond mitosis.
Collapse
|