1
|
Ma T, Jin Y, Wang S, Hu H, Wang M, Yan G, Tang Q, Huang R, Wang G. Innovation in CRC Research: Targeting SPACA6P-AS for Progression. J Biochem Mol Toxicol 2024; 38:e70039. [PMID: 39655708 PMCID: PMC11629451 DOI: 10.1002/jbt.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 12/13/2024]
Abstract
The purpose of this research was to provide light on the functional role that the long noncoding RNA SPACA6P-AS plays in the biology of colorectal cancer (CRC). The presence of elevated SPACA6P-AS expression in colorectal cancer tissues is linked to advanced stages of the disease as well as a decreased overall survival rate. It has been demonstrated through knockdown tests conducted on CRC cell lines that SPACA6P-AS stimulates cell growth in both in vitro and in vivo settings. By acting as a competing endogenous RNA, SPACA6P-AS is able to modulate the levels of miR-339-5p and promote the proliferation of colorectal cancer cells by way of the miR-339-5p/FAM167AFAM167A signaling axis. Based on these findings, SPACA6P-AS is a promising candidate for both a prognostic marker and a therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Tianyi Ma
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Yinghu Jin
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Song Wang
- Department of Colorectal Surgery, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hanqing Hu
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Meng Wang
- Department of Colorectal SurgeryZhejiang Cancer Hospital (Affiliated Cancer Hospital of the Chinese Academy of Sciences)HangzhouChina
| | - Guoqing Yan
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Qingchao Tang
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Rui Huang
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Guiyu Wang
- Department of Colorectal Surgerythe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| |
Collapse
|
2
|
Vaghari-Tabari M, Qujeq D, Hashemzadeh MS. Long noncoding RNAs as potential targets for overcoming chemoresistance in upper gastrointestinal cancers. Biomed Pharmacother 2024; 179:117368. [PMID: 39214010 DOI: 10.1016/j.biopha.2024.117368] [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: 05/27/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
In the last decade, researchers have paid much attention to the role of noncoding RNA molecules in human diseases. Among the most important of these molecules are LncRNAs, which are RNA molecules with a length of more than 200 nucleotides. LncRNAs can regulate gene expression through various mechanisms, such as binding to DNA sequences and interacting with miRNAs. Studies have shown that LncRNAs may be valuable therapeutic targets in treating various cancers, including upper-gastrointestinal cancers. Upper gastrointestinal cancers, mainly referring to esophageal and gastric cancers, are among the deadliest gastrointestinal cancers. Despite notable advances, traditional chemotherapy remains a common strategy for treating these cancers. However, chemoresistance poses a significant obstacle to the effective treatment of upper gastrointestinal cancers, resulting in a low survival rate. Chemoresistance arises from various events, such as the enhancement of efflux and detoxification of chemotherapy agents, reduction of drug uptake, alteration of drug targeting, reduction of prodrug activation, strengthening of EMT and stemness, and the attenuation of apoptosis in cancerous cells. Tumor microenvironment also plays an important role in chemoresistance. Interestingly, a series of studies have revealed that LncRNAs can influence important mechanisms associated with some of the aforementioned events and may serve as promising targets for mitigating chemoresistance in upper gastrointestinal cancers. In this review paper, following a concise overview of chemoresistance mechanisms in upper gastrointestinal cancers, we will review the most intriguing findings of these investigations in detail.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | | |
Collapse
|
3
|
Xu P, Li C, Yuan J, Bao Z, Liu W. Predict lncRNA-drug associations based on graph neural network. Front Genet 2024; 15:1388015. [PMID: 38737125 PMCID: PMC11082279 DOI: 10.3389/fgene.2024.1388015] [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: 02/19/2024] [Accepted: 04/05/2024] [Indexed: 05/14/2024] Open
Abstract
LncRNAs are an essential type of non-coding RNAs, which have been reported to be involved in various human pathological conditions. Increasing evidence suggests that drugs can regulate lncRNAs expression, which makes it possible to develop lncRNAs as therapeutic targets. Thus, developing in-silico methods to predict lncRNA-drug associations (LDAs) is a critical step for developing lncRNA-based therapies. In this study, we predict LDAs by using graph convolutional networks (GCN) and graph attention networks (GAT) based on lncRNA and drug similarity networks. Results show that our proposed method achieves good performance (average AUCs > 0.92) on five datasets. In addition, case studies and KEGG functional enrichment analysis further prove that the model can effectively identify novel LDAs. On the whole, this study provides a deep learning-based framework for predicting novel LDAs, which will accelerate the lncRNA-targeted drug development process.
Collapse
Affiliation(s)
- Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Chuchu Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Zhenshen Bao
- College of Information Engineering, Taizhou University, Taizhou, Jiangsu, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Zhang J, Wu L, Wang C, Xie X, Han Y. Research Progress of Long Non-Coding RNA in Tumor Drug Resistance: A New Paradigm. Drug Des Devel Ther 2024; 18:1385-1398. [PMID: 38689609 PMCID: PMC11060174 DOI: 10.2147/dddt.s448707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
In the past few decades, chemotherapy has been one of the most effective cancer treatment options. Drug resistance is currently one of the greatest obstacles to effective cancer treatment. Even though drug resistance mechanisms have been extensively investigated, they have not been fully elucidated. Recent genome-wide investigations have revealed the existence of a substantial quantity of long non-coding RNAs (lncRNAs) transcribed from the human genome, which actively participate in numerous biological processes, such as transcription, splicing, epigenetics, the cell cycle, cell differentiation, development, pluripotency, immune microenvironment. The abnormal expression of lncRNA is considered a contributing factor to the drug resistance. Furthermore, drug resistance may be influenced by genetic and epigenetic variations, as well as individual differences in patient treatment response, attributable to polymorphisms in metabolic enzyme genes. This review focuses on the mechanism of lncRNAs resistance to target drugs in the study of tumors with high mortality, aiming to establish a theoretical foundation for targeted therapy.
Collapse
Affiliation(s)
- Jing Zhang
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi’an, Shaanxi, People’s Republic of China
| | - Le Wu
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi’an, Shaanxi, People’s Republic of China
| | - Chenchen Wang
- Department of Critical Care Medicine, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, People’s Republic of China
| | - Xin Xie
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi’an, Shaanxi, People’s Republic of China
| | - Yuying Han
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi’an, Shaanxi, People’s Republic of China
- Department of Critical Care Medicine, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, People’s Republic of China
- Science and Education Department, Xi’an No. 5 Hospital, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
5
|
Lee J, Kim EA, Kang J, Chae YS, Park HY, Kang B, Lee SJ, Lee IH, Park JY, Park NJY, Jung JH. Long non-coding RNA SOX2OT in tamoxifen-resistant breast cancer. BMC Mol Cell Biol 2024; 25:12. [PMID: 38649821 PMCID: PMC11036730 DOI: 10.1186/s12860-024-00510-y] [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: 10/31/2023] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
Hormone receptor (HR)-positive breast cancer can become aggressive after developing hormone-treatment resistance. This study elucidated the role of long non-coding RNA (lncRNA) SOX2OT in tamoxifen-resistant (TAMR) breast cancer and its potential interplay with the tumor microenvironment (TME). TAMR breast cancer cell lines TAMR-V and TAMR-H were compared with the luminal type A cell line (MCF-7). LncRNA expression was assessed via next-generation sequencing, RNA extraction, lncRNA profiling, and quantitative RT-qPCR. SOX2OT overexpression effects on cell proliferation, migration, and invasion were evaluated using various assays. SOX2OT was consistently downregulated in TAMR cell lines and TAMR breast cancer tissue. Overexpression of SOX2OT in TAMR cells increased cell proliferation and cell invasion. However, SOX2OT overexpression did not significantly alter SOX2 levels, suggesting an independent interaction within TAMR cells. Kaplan-Meier plot analysis revealed an inverse relationship between SOX2OT expression and prognosis in luminal A and B breast cancers. Our findings highlight the potential role of SOX2OT in TAMR breast cancer progression. The downregulation of SOX2OT in TAMR breast cancer indicates its involvement in resistance mechanisms. Further studies should explore the intricate interactions between SOX2OT, SOX2, and TME in breast cancer subtypes.
Collapse
Affiliation(s)
- Jeeyeon Lee
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Eun-Ae Kim
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jieun Kang
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Yee Soo Chae
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Ho Yong Park
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Byeongju Kang
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Soo Jung Lee
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - In Hee Lee
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Ji-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Nora Jee-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea
| | - Jin Hyang Jung
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
- Kyungpook National University Chilgok Hospital, Hoguk-ro 807, Buk-gu, 41404, Daegu, Republic of Korea.
| |
Collapse
|
6
|
Chuang YT, Shiau JP, Tang JY, Farooqi AA, Chang FR, Tsai YH, Yen CY, Chang HW. Connection of Cancer Exosomal LncRNAs, Sponging miRNAs, and Exosomal Processing and Their Potential Modulation by Natural Products. Cancers (Basel) 2023; 15:cancers15082215. [PMID: 37190145 DOI: 10.3390/cancers15082215] [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: 03/20/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Cancerous exosomes contain diverse biomolecules that regulate cancer progression. Modulating exosome biogenesis with clinical drugs has become an effective strategy for cancer therapy. Suppressing exosomal processing (assembly and secretion) may block exosomal function to reduce the proliferation of cancer cells. However, the information on natural products that modulate cancer exosomes lacks systemic organization, particularly for exosomal long noncoding RNAs (lncRNAs). There is a gap in the connection between exosomal lncRNAs and exosomal processing. This review introduces the database (LncTarD) to explore the potential of exosomal lncRNAs and their sponging miRNAs. The names of sponging miRNAs were transferred to the database (miRDB) for the target prediction of exosomal processing genes. Moreover, the impacts of lncRNAs, sponging miRNAs, and exosomal processing on the tumor microenvironment (TME) and natural-product-modulating anticancer effects were then retrieved and organized. This review sheds light on the functions of exosomal lncRNAs, sponging miRNAs, and exosomal processing in anticancer processes. It also provides future directions for the application of natural products when regulating cancerous exosomal lncRNAs.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Hong Tsai
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Hsueh-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
7
|
Tan S, Yang Y, Yang W, Han Y, Huang L, Yang R, Hu Z, Tao Y, Liu L, Li Y, Oyang L, Lin J, Peng Q, Jiang X, Xu X, Xia L, Peng M, Wu N, Tang Y, Cao D, Liao Q, Zhou Y. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J Exp Clin Cancer Res 2023; 42:59. [PMID: 36899389 PMCID: PMC9999652 DOI: 10.1186/s13046-023-02634-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of cancer. As nutrients are scarce in the tumor microenvironment (TME), tumor cells adopt multiple metabolic adaptations to meet their growth requirements. Metabolic reprogramming is not only present in tumor cells, but exosomal cargos mediates intercellular communication between tumor cells and non-tumor cells in the TME, inducing metabolic remodeling to create an outpost of microvascular enrichment and immune escape. Here, we highlight the composition and characteristics of TME, meanwhile summarize the components of exosomal cargos and their corresponding sorting mode. Functionally, these exosomal cargos-mediated metabolic reprogramming improves the "soil" for tumor growth and metastasis. Moreover, we discuss the abnormal tumor metabolism targeted by exosomal cargos and its potential antitumor therapy. In conclusion, this review updates the current role of exosomal cargos in TME metabolic reprogramming and enriches the future application scenarios of exosomes.
Collapse
Affiliation(s)
- Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wenjuan Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Lisheng Huang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Ruiqian Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Zifan Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Yi Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Lin Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yun Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Deliang Cao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
8
|
Zhao F, Tian H, Wang Y, Zhang J, Liu F, Fu L. LINC01004-SPI1 axis-activated SIGLEC9 in tumor-associated macrophages induces radioresistance and the formation of immunosuppressive tumor microenvironment in esophageal squamous cell carcinoma. Cancer Immunol Immunother 2023; 72:1835-1851. [PMID: 36688997 DOI: 10.1007/s00262-022-03364-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/27/2022] [Indexed: 01/24/2023]
Abstract
Radioresistance and immunosuppression remain the major obstacles in the anti-cancer treatments. This work studies the functions of sialic acid binding Ig like lectin 9 (SIGLEC9) and its related molecules in radioresistance and immunosuppression in esophageal squamous cell carcinoma (ESCC). The single-cell analysis showed that SIGLEC9 was mainly expressed on tumor-associated macrophages (TAMs). Monocytes-derived macrophages were co-cultured with ESCC cells and subjected to radiotherapy. High or low doses of radiotherapy induced SIGLEC9 upregulation and M2 polarization of TAMs. Artificial inhibition of SIGLEC9 in TAMs suppressed the radioresistance and immunosuppressive tumor microenvironment (TME) in the co-cultured ESCC cells. Upstream molecules of SIGLEC9 were predicted via bioinformatics. LINC01004 recruited Spi-1 proto-oncogene (SPI1) in nucleus of TAMs to induce transcriptional activation of SIGLEC9. SIGLEC9 interacted with mucin 1 (MUC1). MUC1 overexpression in ESCCs induced M2 skewing of TAMs, enhanced radioresistance and immunosuppression, and promoted nuclear translocation of β-catenin to suppress radiotherapy-induced ferroptosis of ESCC cells. These effects were blocked upon SIGLEC9 suppression. In vitro results were reproduced in the animal models with xenograft tumors. Taken together, this study demonstrates that the LINC01004-SPI1 axis-activated SIGLEC9 in TAMs induces radioresistance and the formation of immunosuppressive TME in ESCC.
Collapse
Affiliation(s)
- Fen Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, Shandong, People's Republic of China
| | - Hui Tian
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yungang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Jianbo Zhang
- Departments of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Fang Liu
- Department of Imaging, Shandong Medical College, Jinan, 250002, Shandong, People's Republic of China
| | - Lei Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
9
|
Zhang LJ, Chen F, Liang XR, Ponnusamy M, Qin H, Lin ZJ. Crosstalk among long non-coding RNA, tumor-associated macrophages and small extracellular vesicles in tumorigenesis and dissemination. Front Oncol 2022; 12:1008856. [PMID: 36263199 PMCID: PMC9574020 DOI: 10.3389/fonc.2022.1008856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Long noncoding RNAs (lncRNAs), which lack protein-coding ability, can regulate cancer cell growth, proliferation, invasion, and metastasis. Tumor-associated macrophages (TAMs) are key components of the tumor microenvironment that have a significant impact on cancer progression. Small extracellular vesicles (sEV) are crucial mediators of intercellular communications. Cancer cell and macrophage-derived sEV can carry lncRNAs that influence the onset and progression of cancer. Dysregulation of lncRNAs, TAMs, and sEV is widely observed in tumors which makes them valuable targets for cancer immunotherapy. In this review, we summarize current updates on the interactions among sEV, lncRNAs, and TAMs in tumors and provide new perspectives on cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Li-jie Zhang
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Feng Chen
- Department of General Surgery, Weifang Traditional Chinese Hospital, Weifang, China
| | - Xiao-ru Liang
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | | | - Hao Qin
- Department of Public Health, Weifang Medical University, Weifang, China
| | - Zhi-juan Lin
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
- *Correspondence: Zhi-juan Lin,
| |
Collapse
|
10
|
Najafi S, Khatami SH, Khorsand M, Jamali Z, Shabaninejad Z, Moazamfard M, Majidpoor J, Aghaei Zarch SM, Movahedpour A. Long non-coding RNAs (lncRNAs); roles in tumorigenesis and potentials as biomarkers in cancer diagnosis. Exp Cell Res 2022; 418:113294. [PMID: 35870535 DOI: 10.1016/j.yexcr.2022.113294] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 12/15/2022]
Abstract
New research has indicated that long non-coding RNAs (lncRNAs) play critical roles in a broad range of biological processes, including the pathogenesis of many complex human diseases, including cancer. The detailed regulation mechanisms of many lncRNAs in cancer initiation and progression have yet to be discovered, even though a few of lncRNAs' functions in cancer have been characterized. In the present study, we summarize recent advances in the mechanisms and functions of lncRNAs in cancer. We focused on the roles of newly-identified lncRNAs as oncogenes and tumor suppressors, as well as the potential pathways these molecules could play. The paper also discusses their potential uses as biomarkers for the diagnosis and prognosis of cancer.
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Khorsand
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyed Mohsen Aghaei Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
11
|
Zhao F, Dong Z, Li Y, Liu S, Guo P, Zhang D, Li S. Comprehensive Analysis of Molecular Clusters and Prognostic Signature Based on m7G-related LncRNAs in Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:893186. [PMID: 35912250 PMCID: PMC9329704 DOI: 10.3389/fonc.2022.893186] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/24/2022] [Indexed: 01/19/2023] Open
Abstract
N7-Methylguanosine (m7G) and long non-coding RNAs (lncRNAs) have been widely reported to play an important role in cancer. However, there is little known about the relationship between m7G-related lncRNAs and esophageal squamous cell carcinoma (ESCC). In this study, we aimed to find new potential biomarkers and construct an m7G-related lncRNA prognostic signature for ESCC. Three molecular clusters were identified by consensus clustering of 963 m7G-related lncRNAs, of which cluster B is preferentially related to poorer prognosis, higher immune and stromal scores, higher mRNA levels of immune checkpoints, and higher immune infiltrate level. We constructed a robust and effective m7G-related lncRNA prognostic signature (m7G-LPS, including 7 m7G-related prognostic lncRNAs) and demonstrated its prognostic value and predictive ability in the GEO and TCGA cohorts. The risk score was able to serve as an independent risk factor for patients with ESCC and showed better prediction than the traditional clinical risk factors. The immune score, stromal score, the infiltration level of immune cells and expression of immune checkpoints were significantly higher in the high-risk subgroup compared to the low-risk subgroup. The establishment of nomogram further improved the performance of m7G-LPS and facilitated its clinical application. Finally, we used GTEx RNA-seq data and qRT-PCR experiments to verify the expression levels of 7 m7G-related lncRNAs. To a certain degree, m7G-lncRNAs can be used as prognostic markers and therapeutic targets for ESCC patients.
Collapse
Affiliation(s)
- Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Zefang Dong
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yishuai Li
- Department of Thoracic Surgery, Hebei Chest Hospital, Shijiazhuang, China
| | - Shiquan Liu
- Department of Thoracic Surgery, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Pengfei Guo
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Dengfeng Zhang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- School of Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Shujun Li, , orcid.org/0000-0001-5959-3160
| |
Collapse
|
12
|
Krstic J, Deutsch A, Fuchs J, Gauster M, Gorsek Sparovec T, Hiden U, Krappinger JC, Moser G, Pansy K, Szmyra M, Gold D, Feichtinger J, Huppertz B. (Dis)similarities between the Decidual and Tumor Microenvironment. Biomedicines 2022; 10:1065. [PMID: 35625802 PMCID: PMC9138511 DOI: 10.3390/biomedicines10051065] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 02/05/2023] Open
Abstract
Placenta-specific trophoblast and tumor cells exhibit many common characteristics. Trophoblast cells invade maternal tissues while being tolerated by the maternal immune system. Similarly, tumor cells can invade surrounding tissues and escape the immune system. Importantly, both trophoblast and tumor cells are supported by an abetting microenvironment, which influences invasion, angiogenesis, and immune tolerance/evasion, among others. However, in contrast to tumor cells, the metabolic, proliferative, migrative, and invasive states of trophoblast cells are under tight regulatory control. In this review, we provide an overview of similarities and dissimilarities in regulatory processes that drive trophoblast and tumor cell fate, particularly focusing on the role of the abetting microenvironments.
Collapse
Affiliation(s)
- Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Alexander Deutsch
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
- Division of Biophysics, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Tina Gorsek Sparovec
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julian Christopher Krappinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Katrin Pansy
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Marta Szmyra
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Daniela Gold
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| |
Collapse
|
13
|
Wu Z, Ju Q. Non-Coding RNAs Implicated in the Tumor Microenvironment of Colorectal Cancer: Roles, Mechanisms and Clinical Study. Front Oncol 2022; 12:888276. [PMID: 35574420 PMCID: PMC9096125 DOI: 10.3389/fonc.2022.888276] [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: 03/02/2022] [Accepted: 04/04/2022] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors. The morbidity and mortality rates have been increasing all over the world. It is critical to elucidate the mechanism of CRC occurrence and development. However, tumor microenvironment (TME) includes immune cells, fibroblasts, endothelial cells, cytokines, chemokines and other components that affect the progression of CRC and patients' prognosis. Non-coding RNAs (ncRNAs) including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs) without protein-coding ability have been shown to engage in tumor microenvironment-mediated angiogenesis and metastasis. Therefore, clarifying the mechanism of ncRNAs regulating the microenvironment is very important to develop the therapeutic target of CRC and improve the survival time of patients. This review focuses on the role and mechanism of ncRNAs in the CRC microenvironment and puts forward possible clinical treatment strategies.
Collapse
Affiliation(s)
| | - Qiang Ju
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|