1
|
Zhang L, Gao S, Yuan Q, Fu Y, Yang R. An ensemble learning method combined with multiple feature representation strategies to predict lncRNA subcellular localizations. Comput Biol Chem 2025; 115:108336. [PMID: 39752849 DOI: 10.1016/j.compbiolchem.2024.108336] [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: 09/01/2024] [Revised: 10/26/2024] [Accepted: 12/25/2024] [Indexed: 02/26/2025]
Abstract
Long non-coding RNAs (lncRNAs) are strongly associated with cellular physiological mechanisms and implicated in the numerous diseases. By exploring the subcellular localizations of lncRNAs, we can not only gain crucial insights into the molecular mechanisms of lncRNA-related biological processes but also make valuable contributions towards the diagnosis, prevention, and treatment of various human diseases. However, conventional experimental techniques tend to be laborious and time-intensive. In this context, computational methods are in increased demand. The focus of this paper is the development of an innovative ensemble method that incorporates hybrid features to accurately predict the subcellular localizations of lncRNAs. To address the issue of incomplete reflection of inherent correlation with the intended target using singular source features, the utilization of heterogeneous multi-source features is implemented by introducing information on sequence composition, physicochemical properties, and structure. To address the issue of the imbalance classes in the benchmark dataset, the Synthetic Minority Over-sampling Technique (SMOTE) is employed. Finally, the resulting predictor termed lncSLPre is developed by integrating the outputs of the individual classifiers. Experimental findings suggest that the complementarity of multi-source heterogeneous features improves prediction performance. Additionally, it is demonstrated that the application of SMOTE is effective in mitigating the issue of the imbalanced dataset, while the feature selection approach is critical in eliminating extraneous and redundant features. Compared with existing advanced methods, lncSLPre achieves better performance with an overall accuracy improvement of 13.13%, 2.15%, and 3.23%, respectively, indicating that lncSLPre can effectively predict lncRNA subcellular localizations.
Collapse
Affiliation(s)
- Lina Zhang
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Sizan Gao
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Qinghao Yuan
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Yao Fu
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Runtao Yang
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| |
Collapse
|
2
|
Hu W, Yue Y, Yan R, Guan L, Li M. An ensemble deep learning framework for multi-class LncRNA subcellular localization with innovative encoding strategy. BMC Biol 2025; 23:47. [PMID: 39984880 PMCID: PMC11846348 DOI: 10.1186/s12915-025-02148-4] [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: 05/08/2024] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Long non-coding RNA (LncRNA) play pivotal roles in various cellular processes, and elucidating their subcellular localization can offer crucial insights into their functional significance. Accurate prediction of lncRNA subcellular localization is of paramount importance. Despite numerous computational methods developed for this purpose, existing approaches still encounter challenges stemming from the complexity of data representation and the difficulty in capturing nucleotide distribution information within sequences. RESULTS In this study, we propose a novel deep learning-based model, termed MGBLncLoc, which incorporates a unique multi-class encoding technique known as generalized encoding based on the Distribution Density of Multi-Class Nucleotide Groups (MCD-ND). This encoding approach enables more precise reflection of nucleotide distributions, distinguishing between constant and discriminative regions within sequences, thereby enhancing prediction performance. Additionally, our deep learning model integrates advanced neural network modules, including Multi-Dconv Head Transposed Attention, Gated-Dconv Feed-forward Network, Convolutional Neural Network, and Bidirectional Gated Recurrent Unit, to comprehensively exploit sequence features of lncRNA. CONCLUSIONS Comparative analysis against commonly used sequence feature encoding methods and existing prediction models validates the effectiveness of MGBLncLoc, demonstrating superior performance. This research offers novel insights and effective solutions for predicting lncRNA subcellular localization, thereby providing valuable support for related biological investigations.
Collapse
Affiliation(s)
- Wenxing Hu
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Yan Yue
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Ruomei Yan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Lixin Guan
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China
| | - Mengshan Li
- College of Physics and Electronic Information, Gannan Normal University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
3
|
Ou S, Nie X, Qiu X, Jin X, Wu G, Zhang R, Zhu J. Deciphering the mechanisms of long non-coding RNAs in ferroptosis: insights into its clinical significance in cancer progression and immunology. Cell Death Discov 2025; 11:14. [PMID: 39827195 PMCID: PMC11743196 DOI: 10.1038/s41420-025-02290-6] [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: 09/09/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
A new type of nonapoptotic, iron-dependent cell death induced by lipid peroxidation is known as ferroptosis. Numerous pathological processes, including inflammation and cancer, have been demonstrated to be influenced by changes in the ferroptosis-regulating network. Long non-coding RNAs (LncRNAs) are a group of functional RNA molecules that are not translated into proteins, which can regulate gene expression in various manners. An increasing number of studies have shown that lncRNAs can interfere with the progression of ferroptosis by modulating ferroptosis-related genes directly or indirectly. Despite evidence implicating lncRNAs in cancer and inflammation, studies on their mechanisms and therapeutic potential remain scarce. We investigate the mechanisms of lncRNA-mediated regulation of inflammation and cancer immunity, assessing the feasibility and challenges of lncRNAs as therapeutic targets in these conditions.
Collapse
Affiliation(s)
- Shengming Ou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoya Nie
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiangyu Qiu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Jin
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Geyan Wu
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
4
|
Wang X, Wang Q, Wang H, Cai G, An Y, Liu P, Zhou H, Chen HW, Ji S, Ye J, Wang J. Small protein ERSP encoded by LINC02870 promotes triple negative breast cancer progression via IRE1α/XBP1s activation. Cell Death Differ 2025:10.1038/s41418-025-01443-5. [PMID: 39799200 DOI: 10.1038/s41418-025-01443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Clinical treatment options for triple-negative breast cancer (TNBC) are currently limited to chemotherapy because of a lack of effective therapeutic targets. Recent evidence suggests that long noncoding RNAs (lncRNAs) encode bioactive peptides or proteins, thereby playing noncanonical yet significant roles in regulating cellular processes. However, the potential of lncRNA-translated products in cancer progression remains largely unknown. In this study, we identified a previously undocumented small protein encoded by the lncRNA LINC02870. This protein is localized at the endoplasmic reticulum (ER) and participates in ER stress, thus, we named it the endoplasmic reticulum stress protein (ERSP). ERSP was highly expressed in TNBC tissues, and elevated LINC02870 content was correlated with poor prognosis in TNBC patients. Loss of ERSP inhibited TNBC growth and metastasis both in vitro and in vivo. The pro-oncogenic effects of ERSP could be attributed to its selective activation of the IRE1α/XBP1s branch. ERSP enhances the unfolded protein response (UPR) by interacting with XBP1s, facilitating the nuclear accumulation of XBP1s, thereby promoting the expression of ER stress-related genes. These findings highlight the regulatory role of the lncRNA-encoded protein ERSP in ER stress and suggest that it is a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Xiaolu Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qianqian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Hong Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Guodi Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Yana An
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Huihao Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Shufeng Ji
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510006, China.
| | - Jiantao Ye
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| | - Junjian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
5
|
Liu D, Zhao X, Wang Z, Wang G, Chen Z, Ning S, Feng D, Sun X, Sun R, Yao J, Tian X. A novel protein encoded by circARHGAP12 attenuates DNA damage and apoptosis by regulating MDC1 in intestinal ischemia/reperfusion injury. Int J Biol Macromol 2025; 286:138374. [PMID: 39643171 DOI: 10.1016/j.ijbiomac.2024.138374] [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: 08/12/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Excessive intestinal ischemia/reperfusion (I/R)-induced epithelial cell apoptosis results in damage to the intestinal defense barrier. Circular RNAs (circRNAs) are functional RNA transcripts, and their functions as microRNA (miRNA) sponges and binding proteins have been well characterized. Recent evidence has indicated that some circRNAs encode functional proteins. However, whether protein-encoding circRNAs contribute to intestinal I/R remains undiscovered. Here, we identified a protein-encoding circRNA, circARHGAP12, that can significantly attenuate intestinal I/R-induced cell apoptosis. Moreover, circARHGAP12 can encode a 229-amino-acid (aa) protein, ARHGAP12-229aa. The expression of both circARHGAP12 and ARHGAP12-229aa was downregulated in intestinal I/R injury. Additionally, circARHGAP12 protected against intestinal I/R injury mainly by encoding ARHGAP12-229aa. We performed RNA sequencing (RNA-seq) analyses to identify downstream targets of ARHGAP12-229aa. The results revealed that overexpression of ARHGAP12-229aa led to increased expression of MDC1, a DNA damage-related protein, and that this increase was accompanied by a decrease in intestinal epithelial cell DNA damage and apoptosis. Furthermore, MDC1 silencing weakened the protective effect of ARHGAP12-229aa against intestinal I/R injury. Our findings suggest a novel perspective on circRNA function, providing an innovative therapeutic strategy for intestinal I/R.
Collapse
Affiliation(s)
- Deshun Liu
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.; Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116600, China
| | - Xuzi Zhao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Zhao Chen
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Shili Ning
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Dongcheng Feng
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116600, China
| | - Xin Sun
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Ruimin Sun
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China..
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China..
| |
Collapse
|
6
|
Ren Z, Liu J, Chang X, Yang X, Zhang Y, Zhang X, Zhou S, Liang Q, Xu Z, Yang F, Xiao W. A peptide encoded by LINC00944 suppresses the growth of melanoma cells by diminishing EP400-MYC interaction. Biochem Pharmacol 2025; 231:116652. [PMID: 39586403 DOI: 10.1016/j.bcp.2024.116652] [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: 06/04/2024] [Revised: 09/30/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
The peptides encoded by long noncoding RNAs (lncRNAs) have been shown to participate in cancer pathogenesis. In this study, lncRNA LINC00944 was validated to encode an endogenous 102-amino acid (aa) small peptide (named LINC00944 peptide). Functionally, LINC00944 peptide exerted an anti-growth effect in melanoma cells in vitro. Mechanistically, LINC00944 peptide interacted with the E1A binding protein p400 (EP400)/c-MYC complex. LINC00944 peptide also inhibited c-MYC protein expression. Furthermore, LINC00944 peptide repressed the transcriptional activity of MYC by reducing the EP400-MYC interaction, thereby reducing the levels of fatty acid metabolism- and glucose metabolism-related proteins. Our findings uncovered that LINC00944 peptide might be a promising adjuvant therapeutic agent for melanoma. Implications: This study provided the first evidence that LINC00944-encoded peptide played a critical role in the growth of melanoma cells.
Collapse
Affiliation(s)
- Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Jie Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China; Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Xiyue Chang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China; Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Xuejing Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China; Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Yuke Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China; Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Xinyue Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China; Department of Epidemiology, School of Public Health, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110013, China
| | - Siyu Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Qiushi Liang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Zhijie Xu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Fan Yang
- Department of Dermatology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| | - Wan'an Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
7
|
Shaikh M, Doshi G. Unraveling non-coding RNAs in breast cancer: mechanistic insights and therapeutic potential. Med Oncol 2024; 42:37. [PMID: 39730979 DOI: 10.1007/s12032-024-02589-x] [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: 09/29/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Breast cancer remains a leading global health challenge requiring innovative, therapeutic strategies to improve patient outcomes. This review explores the pivotal roles of non-coding RNAs (ncRNAs), including long non-coding RNA, micro RNA, and circular RNA, in breast cancer biology. We highlight how these molecules regulate critical signaling pathways, influence tumor microenvironments, and contribute to treatment resistance. Our findings underscore the potential of ncRNAs as biomarkers for early diagnosis and as treatment targets for personalized treatment strategies. To pave the way for innovative cancer management approaches, we investigate the complex interactions of ncRNAs and their impact on tumor progression. This comprehensive review enhances our understanding of breast cancer biology while emphasizing the translational significance of ncRNA research in developing effective treatment strategies. Additional research and clinical studies are required to confirm the diagnostic and medicinal value of ncRNAs in breast cancer. Investigating the complex networks of ncRNA interactions and their links to other biological pathways can lead to the discovery of new treatment targets. Furthermore, leveraging advanced technologies, such as machine learning and multi-omics methods, will be critical in improving our understanding of ncRNAs biomarkers and translating these insights into impactful clinical applications.
Collapse
Affiliation(s)
- Muqtada Shaikh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India.
| |
Collapse
|
8
|
Zhang S, Xu B, Zhang JL, Liu SH, Xiang X, Liu P. Prognostic role of long noncoding RNA CASC11 in cancer patients: A meta-analysis. Medicine (Baltimore) 2024; 103:e40823. [PMID: 39686470 DOI: 10.1097/md.0000000000040823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Long noncoding RNA (lncRNA) is a significant component of the noncoding genome and refers to RNA molecules that exceed 200 nucleotides in length. It plays a crucial role in both promoting and suppressing cancer by regulating the proliferation, invasion, and metastasis of tumor cells. We have found a correlation between cancer susceptibility candidate 11 (CASC11) experssion and tumorigenesis development prognosis in a number of studies on tumors. Hence, this meta-analysis was conducted to further investigate the effects of CASC11 expression on clinicopathological features and outcome. Furthermore, CASC11 can act both as a therapeutic target and a cancer biomarker. METHOD We conducted a thorough search of PubMed, Embase, Web of Science, and Cochrane Library to identify all eligible studies until September 20, 2023. These studies examined the potential relationship between the expression levels of CASC11 and survival or the range of pathological feature in cancer patients. The impact of CASC11 expression on overall survival (OS) was evaluated using pooled hazard ratios and 95% confidence intervals (CI). The relationship between CASC11 expression and clinicopathological features was assessed through pooled odds ratios and 95% CI. RESULTS A total of 11 studies, involving 660 patients, were examined in this analysis. The results revealed that the overexpression of CASC11 was significantly associated with poor OS (hazard ratios = 2.07, 95%CI = 1.64-2.60) in cancer. Further subgroup analysis demonstrated that the overexpression of CASC11 was consistently linked to poorer OS in diverse types of cancer, including digestive system neoplasm, respiratory neoplasms, and gynecologic tumor. CONCLUSION Overall, this analysis established a strong correlation between CASC11 expression and tumor prognosis, suggesting its potential as a predictive marker for tumor progression in diverse cancer types.
Collapse
Affiliation(s)
- Song Zhang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People's Hospital of Neijiang, Sichuan, China
| | - Bo Xu
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People's Hospital of Neijiang, Sichuan, China
| | - Ji-Ling Zhang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People's Hospital of Neijiang, Sichuan, China
| | - Shun-Hai Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People's Hospital o Neijiang, Sichuan, China
| | - Xin Xiang
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People's Hospital o Neijiang, Sichuan, China
| | - Pan Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People's Hospital o Neijiang, Sichuan, China
| |
Collapse
|
9
|
Luo D, Luo A, Ye G, Li D, Hu S, Zhao H, Peng B. Regulation of a novel circATP8B4/miR-31-5p/nestin ceRNA crosstalk in proliferation, motility, invasion and radiosensitivity of human glioma cells. JOURNAL OF RADIATION RESEARCH 2024; 65:752-764. [PMID: 39287101 PMCID: PMC11630049 DOI: 10.1093/jrr/rrae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/04/2024] [Indexed: 09/19/2024]
Abstract
Deregulation of circular RNAs (circRNAs) is frequent in human glioma. Although circRNA ATPase phospholipid transporting 8B4 (circATP8B4) is highly expressed in glioma, its precise action in glioma development is still not fully understood. The relationship of microRNA (miR)-31-5p and circATP8B4 or nestin (NES) was predicted by bioinformatic analysis and confirmed by RNA pull-down and Dual-luciferase reporter assays. CircATP8B4, miR-31-5p and NES were quantified by qRT-PCR or western blot. Cell functional behaviors were assessed by EdU, wound-healing and transwell invasion assays. Xenograft model experiments were performed to define circATP8B4's activity in vivo. CircATP8B4, a true circular transcript, was upregulated in human glioma. CircATP8B4 downregulation weakened glioma cell growth, motility, and invasion and facilitated radiosensitivity. Mechanistically, circATP8B4 and NES 3'UTR harbored a shared miR-31-5p pairing site, and circATP8B4 involved the post-transcriptional NES regulation by functioning as a competing endogenous RNA (ceRNA). Furthermore, the miR-31-5p/NES axis participated in circATP8B4's activity in glioma cell proliferation, motility, invasion and radiosensitivity. Additionally, circATP8B4 loss diminished tumor growth and enhanced the anticancer effect of radiotherapy in vivo. We have uncovered an uncharacterized ceRNA cascade, circATP8B4/miR-31-5p/NES axis, underlying glioma development and radiosensitivity. Targeting the ceRNA crosstalk may have potential to improve the outcome of glioma patients.
Collapse
Affiliation(s)
- Dongdong Luo
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Aiping Luo
- Department of Radiology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Ganwei Ye
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Dan Li
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Su Hu
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Hailin Zhao
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| | - Biao Peng
- Department of Neurosurgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, No. 78 Hengzhigang Road, Yuexiu District, Guangzhou, 510095, Guangdong, China
| |
Collapse
|
10
|
Xiong Z, Wang Y, Li Z, Li C, Tu C, Li Z. A review on the crosstalk between non-coding RNAs and the cGAS-STING signaling pathway. Int J Biol Macromol 2024; 283:137748. [PMID: 39566795 DOI: 10.1016/j.ijbiomac.2024.137748] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
In the innate immune system, the cyclic GMP-AMP synthase (cGAS)-interferon gene stimulator (STING) pathway activates the type I interferon (IFN) response and the NF-κB pathway by recognizing double-stranded DNAs, the imbalance of which plays a pivotal role in human diseases, including cancer, autoimmune and inflammatory diseases. Non-coding RNAs (ncRNAs) are a diverse group of transcripts that do not code for proteins but regulate various targets and signaling pathways in physiological and pathological processes. Recently, there has been increasing interest in investigating the interplay between the cGAS-STING pathway and ncRNAs. In this review, we provide a concise overview of the cGAS-STING pathway and ncRNAs. Then, we specifically delve into the regulation of the cGAS-STING pathway by long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), the three major classes of ncRNAs, and the influence of the cGAS-STING pathway on the expression of ncRNAs. Furthermore, we introduce the therapeutic applications targeting the cGAS-STING pathway and ncRNA therapy, and propose the utilization of drug delivery systems to deliver ncRNAs that influence the cGAS-STING pathway. Overall, this review highlights the emerging understanding of the intricate relationship between the cGAS-STING pathway and ncRNAs, shedding light on their potential as therapeutic targets in various diseases.
Collapse
Affiliation(s)
- Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yu Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
11
|
Tao Q, Li X, Xia Y, Zheng B, Yan Y, Wang S, Jia L. LINC00261 triggers DNA damage via the miR-23a-3p/CELF2 axis to mitigate the malignant characteristics of 131I-resistant papillary thyroid carcinoma cells. Biochem Biophys Rep 2024; 40:101858. [PMID: 39552712 PMCID: PMC11564912 DOI: 10.1016/j.bbrep.2024.101858] [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: 08/30/2024] [Revised: 10/11/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024] Open
Abstract
Background Long-chain non-coding RNA (LINC00261) in the treatment of papillary thyroid carcinoma (PTC) with 131I is still unknown despite its proven anti-tumour effect in thyroid cancer (TC) and other types of cancer. Methods The database and RT-qPCR were used to analyze the expression level of LINC00261 in PTC and cell lines. PTC cells resistant to 131I (TPC-1/R) were created through ongoing exposure to a lethal dose of 131I, and a subcutaneous xenotransplantation model was developed using PTC mice. Bioinformatics analysis and dual-luciferase assays demonstrated the interaction between LINC00261, miR-23a-3p, and CELF2. RT-qPCR and Western blot were used to detect the expression of LINC00261, miR-23a-3p, and CELF2. Additionally, CCK-8, flow cytometry, immunofluorescence (IF), Western blot, and comet assay were employed to measure cell viability level and DNA damage. Results PTC cell lines exhibited a decrease in the expression of LINC00261. The growth and progression through the S-phase of TPC-1/R cells were suppressed by LINC00261, leading to increased apoptosis and DNA damage. The objective of LINC00261 was to regulate the axis of miR-23a-3p/CELF2. Downregulating LINC00261 enhances the growth and advancement of 131I-resistant cells in the S-phase by activating the miR-23a-3p/CELF2 pathway while suppressing cell death and DNA harm. The miR-23a-3p/CELF2 axis activates DNA damage in 131I-resistant PTC cells by LINC00261. Conclusions LINC00261 activates DNA damage in 131I-resistant PTC cells caused by miR-23a-3p/CELF2 axis, improving the progression of cancer cells of PTC.
Collapse
Affiliation(s)
- Qingyuan Tao
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Xiaojin Li
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Central Laboratory, Kunming, Yunnan, 650021, China
| | - Yanyan Xia
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Bin Zheng
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Yijun Yan
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Songrun Wang
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| | - Li Jia
- The Affiliated Hospital of Yunnan University (Yunnan Second People's Hospital), Nuclear Medicine, Kunming, Yunnan, 650021, China
| |
Collapse
|
12
|
Huang J, Yang P, Pan W, Wu F, Qiu J, Ma Z. The role of polypeptides encoded by ncRNAs in cancer. Gene 2024; 928:148817. [PMID: 39098512 DOI: 10.1016/j.gene.2024.148817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
It was previously thought that ncRNA could not encode polypeptides, but recent reports have challenged this notion. As research into ncRNA progresses, it is increasingly clear that it serves roles beyond traditional mechanisms, playing significant regulatory roles in various diseases, notably cancer, which is responsible for 70% of human deaths. Numerous studies have highlighted the diverse regulatory mechanisms of ncRNA that are pivotal in cancer initiation and progression. The role of ncRNA-encoded polypeptides in cancer regulation has gained prominence. This article explores the newly identified regulatory functions of these polypeptides in three types of ncRNA-lncRNA, pri-miRNA, and circRNA. These polypeptides can interact with proteins, influence signaling pathways, enhance miRNA stability, and regulate cancer progression, malignancy, resistance, and other clinical challenges. Furthermore, we discuss the evolutionary significance of these polypeptides in the transition from RNA to protein, examining their emergence and conservation throughout evolution.
Collapse
Affiliation(s)
- Jiayuan Huang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Ping Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming 650118,China
| | - Wei Pan
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Fan Wu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jianhua Qiu
- Department of Anesthesiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201800, China.
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
13
|
Yin X, Li J, Zhao J, Zheng W, Zhang A, Ma J. Epigenetic modifications involving ncRNAs in digestive system cancers: focus on histone modification. Clin Epigenetics 2024; 16:162. [PMID: 39563475 PMCID: PMC11577885 DOI: 10.1186/s13148-024-01773-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
In recent years, epigenetic modifications have been strongly linked to tumor development, with histone modifications representing a key epigenetic mechanism. In addition, non-coding RNAs (ncRNAs) play a critical role in regulating cancer-related pathways. The abnormal interaction between histone modifications and ncRNAs, both pivotal epigenetic regulators, has been widely observed across various cancer types. Here, we systematically explore the molecular mechanisms through which histone modifications and ncRNAs contribute in the pathogenesis of digestive system cancers, and aberrant ncRNA-mediated histone modifications manipulate various biological behaviors of tumor cells including proliferation, migration, angiogenesis, etc. In addition, we provide new insights into diagnostic, prognostic markers, therapeutic targets and chemoradiation resistance for digestive system cancers from the epigenetic perspective.
Collapse
Affiliation(s)
- Xiaodi Yin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Jingyi Li
- Intensive Care Medicine, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jiahui Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Weihan Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Aohua Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China
| | - Jun Ma
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, #2 Jingba Road, Zhengzhou, 450014, China.
| |
Collapse
|
14
|
Fernando A, Liyanage C, Srinivasan S, Panchadsaram J, Rothnagel JA, Clements J, Batra J. Iroquois homeobox 4 (IRX4) derived micropeptide promotes prostate cancer progression and chemoresistance through Wnt signalling dysregulation. COMMUNICATIONS MEDICINE 2024; 4:224. [PMID: 39487222 PMCID: PMC11530646 DOI: 10.1038/s43856-024-00613-9] [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: 09/16/2023] [Accepted: 09/17/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a commonly diagnosed cancer. Genome-wide association studies have implicated Iroquois homeobox 4 (IRX4) in PCa susceptibility, yet its functional roles remain unclear. We discovered a 78-amino acid micropeptide (miPEP, IRX4_PEP1), encoded from the alternative start site within the IRX4 gene. The miPEPs, encoded through short open reading frames (sORFs) have emerged as regulators of diverse biological processes. However, the significance of miPEPs in prostate tumorigenesis and therapy response remains unexplored to date. Here, we demonstrated the unique role of IRX4_PEP1 in PCa. METHODS The role of IRX4_PEP1 was evaluated in PCa in vitro via functional assays and comprehensive pathway analysis. The interacting partners of IRX4_PEP1 were identified using an immunoprecipitation assay, and the impact of IRX4_PEP1 on PCa stem cells was assessed through a stem cell enrichment assay. Additionally, the expression of IRX4_PEP1 was evaluated in PCa patient samples for its potential diagnostic and prognostic significance. RESULTS Here we show IRX4_PEP1 promotes PCa cell proliferation, migration, and invasion by interacting with heterogeneous nuclear ribonucleoprotein K (HNRPK). Notably, IRX4_PEP1 dysregulates Wnt signalling by interacting with Catenin beta 1 (β catenin; CTNB1), elevating PCa stemness markers, and fostering docetaxel resistance. Clinically, IRX4_PEP1 expression is elevated in PCa tissues and correlates positively with disease aggressiveness. CTNNB1, HNRNPK levels, and ssGSEA enrichment score of WNT/CTNB1 signalling correlate positively with IRX4_PEP1 in PCa tissues. CONCLUSIONS These findings highlight IRX4_PEP1 role in PCa stemness and chemoresistance, suggesting it as a therapeutic target and potential diagnostic marker.
Collapse
Affiliation(s)
- Achala Fernando
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Chamikara Liyanage
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Janaththani Panchadsaram
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Joseph A Rothnagel
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia Campus, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.
- The Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Australia.
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
15
|
Poliseno L, Lanza M, Pandolfi PP. Coding, or non-coding, that is the question. Cell Res 2024; 34:609-629. [PMID: 39054345 PMCID: PMC11369213 DOI: 10.1038/s41422-024-00975-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/30/2024] [Indexed: 07/27/2024] Open
Abstract
The advent of high-throughput sequencing uncovered that our genome is pervasively transcribed into RNAs that are seemingly not translated into proteins. It was also found that non-coding RNA transcripts outnumber canonical protein-coding genes. This mindboggling discovery prompted a surge in non-coding RNA research that started unraveling the functional relevance of these new genetic units, shaking the classic definition of "gene". While the non-coding RNA revolution was still taking place, polysome/ribosome profiling and mass spectrometry analyses revealed that peptides can be translated from non-canonical open reading frames. Therefore, it is becoming evident that the coding vs non-coding dichotomy is way blurrier than anticipated. In this review, we focus on several examples in which the binary classification of coding vs non-coding genes is outdated, since the same bifunctional gene expresses both coding and non-coding products. We discuss the implications of this intricate usage of transcripts in terms of molecular mechanisms of gene expression and biological outputs, which are often concordant, but can also surprisingly be discordant. Finally, we discuss the methodological caveats that are associated with the study of bifunctional genes, and we highlight the opportunities and challenges of therapeutic exploitation of this intricacy towards the development of anticancer therapies.
Collapse
Affiliation(s)
- Laura Poliseno
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa, Italy.
- Institute of Clinical Physiology, CNR, Pisa, Italy.
| | - Martina Lanza
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa, Italy
- Institute of Clinical Physiology, CNR, Pisa, Italy
- University of Siena, Siena, Italy
| | - Pier Paolo Pandolfi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Torino, Italy.
- Renown Institute for Cancer, Nevada System of Higher Education, Reno, NV, USA.
| |
Collapse
|
16
|
Zhang B, Li Z, Ye G, Hu K. Biologic activity and treatment resistance to gastrointestinal cancer: the role of circular RNA in autophagy regulation. Front Oncol 2024; 14:1393670. [PMID: 39281375 PMCID: PMC11392687 DOI: 10.3389/fonc.2024.1393670] [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: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Circular RNAs (circRNAs) lack the 5'-end methylated guanine cap structure and 3' polyadenylate tail structure, classifying it as a non-coding RNA. With the extensive investigation of circRNA, its role in regulating cell death has garnered significant attention in recent years, establishing it as a recognized participant in cancer's biological processes. Autophagy, an essential pathway in programmed cell death (PCD), involves the formation of autophagosomes using lysosomes to degrade cellular contents under the regulation of various autophagy-related (ATG) genes. Numerous studies have demonstrated that circRNA can modulate the biological activity of cancer cells by influencing the autophagy pathway, exhibiting a dualistic role in suppressing or promoting carcinogenesis. In this review, we comprehensively analyze how autophagy-related circRNA impacts the progression of gastrointestinal cancer (GIC). Additionally, we discuss drug resistance phenomena associated with autophagy regulation in GIC. This review offers valuable insights into exploring potential biological targets for prognosis and treatment strategies related to GIC.
Collapse
Affiliation(s)
- Bo Zhang
- Health Science Center, Ningbo University, Ningbo, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zhe Li
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Guoliang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Kefeng Hu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Zhang J, Lu H, Jiang Y, Ma Y, Deng L. ncRNA Coding Potential Prediction Using BiLSTM and Transformer Encoder-Based Model. J Chem Inf Model 2024; 64:6712-6722. [PMID: 39120528 DOI: 10.1021/acs.jcim.4c01097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Many noncoding RNAs (ncRNAs) have been identified, and many of them play vital roles in various biological processes, including gene expression regulation, epigenetic regulation, transcription, and control. Recently, a few observations revealed that ncRNAs are translated into functional peptides. Moreover, many computational methods have been developed to predict the coding potential of these transcripts, which contributes to a deeper investigation of their functions. However, most of these are used to distinguish ncRNAs and mRNAs. It is important to develop a highly accurate computational tool for identifying the coding potential of ncRNAs, thereby contributing to the discovery of novel peptides. In this Article, we propose a novel BiLSTM And Transformer encoder-based model (nBAT) with intrinsic features encoded for ncRNA coding potential prediction. In nBAT, we introduce a learnable position encoding mechanism to better obtain the embeddings of the ncRNA sequence. Moreover, we extract 43 intrinsic features from different perspectives and encode these features into the Transformer encoder by calculating their distances. Our performance comparisons show that nBAT achieves a superior performance than the state-of-the-art methods for coding potential prediction on different datasets. We also apply the method to new ncRNAs for identifying the coding potential, and the results further indicate the competitive performance of nBAT. We expect the method can be exploited as a useful tool for high-throughput coding potential prediction for ncRNAs.
Collapse
Affiliation(s)
- Jingpu Zhang
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan 467000, China
| | - Hao Lu
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan 467000, China
| | - Ying Jiang
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Yuanyuan Ma
- School of Computer Engineering, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| |
Collapse
|
18
|
Han B, Cheng D, Luo H, Li J, Wu J, Jia X, Xu M, Sun P, Cheng S. Peptidomic analysis reveals novel peptide PDLC promotes cell proliferation in hepatocellular carcinoma via Ras/Raf/MEK/ERK pathway. Sci Rep 2024; 14:18757. [PMID: 39138279 PMCID: PMC11322383 DOI: 10.1038/s41598-024-69789-3] [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: 09/22/2023] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Hepatocellular carcinoma (HCC) still presents poor prognosis with low overall survival rates and limited therapeutic options available. Recently, attention has been drawn to peptidomic analysis, an emerging field of proteomics for the exploration of new potential peptide drugs for the treatment of various diseases. However, research on the potential function of HCC peptides is lacking. Here, we analyzed the peptide spectrum in HCC tissues using peptidomic techniques and explored the potentially beneficial peptides involved in HCC. Changes in peptide profiles in HCC were examined using liquid chromatography-mass spectrometry (LC-MS/MS). Analyze the physicochemical properties and function of differently expressed peptides using bioinformatics. The effect of candidate functional peptides on HCC cell growth and migration was evaluated using the CCK-8, colony formation, and transwell assays. Transcriptome sequencing analysis and western blot were employed to delve into the mode of action of potential peptide on HCC. Peptidomic analysis of HCC tissue yielded a total of 8683 peptides, of which 452 exhibited up-regulation and 362 showed down-regulation. The peptides that were differentially expressed, according to bioinformatic analysis, were closely linked to carbon metabolism and the mitochondrial inner membrane. The peptide functional validation identified a novel peptide, PDLC (peptide derived from liver cancer), which was found to dramatically boost HCC cell proliferation through the Ras/Raf/MEK/ERK signaling cascade. Our research defined the peptide's properties and pattern of expression in HCC and identified a novel peptide, PDLC, with a function in encouraging HCC progression, offering an entirely new potential therapeutic target the disease.
Collapse
Affiliation(s)
- Bo Han
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daqing Cheng
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huizhao Luo
- Rehabilitation Department, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jutang Li
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaoxiang Wu
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing Jia
- Department of Urology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Xu
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Sheng Cheng
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Yang Y, Huang K, Jiang H, Wang S, Xu X, Liu Y, Liu Q, Wei M, Li Z. Unveiling the role of circRBBP7 in myoblast proliferation and differentiation: A novel regulator of muscle development. FASEB J 2024; 38:e23808. [PMID: 38994637 DOI: 10.1096/fj.202302599rr] [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: 12/18/2023] [Revised: 06/13/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
Muscle development is a multistep process regulated by diverse gene networks, and circRNAs are considered novel regulators mediating myogenesis. Here, we systematically analyzed the role and underlying regulatory mechanisms of circRBBP7 in myoblast proliferation and differentiation. Results showed that circRBBP7 has a typical circular structure and encodes a 13 -kDa protein. By performing circRBBP7 overexpression and RNA interference, we found that the function of circRBBP7 was positively correlated with the proliferation and differentiation of myoblasts. Using RNA sequencing, we identified 1633 and 532 differentially expressed genes (DEGs) during myoblast proliferation or differentiation, respectively. The DEGs were found mainly enriched in cell cycle- and skeletal muscle development-related pathways, such as the MDM2/p53 and PI3K-Akt signaling pathways. Further co-IP and IF co-localization analysis revealed that VEGFR-1 is a target of circRBBP7 in myoblasts. qRT-PCR and WB analysis further confirmed the positive correlation between VEGFR-1 and circRBBP7. Moreover, we found that in vivo transfection of circRBBP7 into injured muscle tissues significantly promoted the regeneration and repair of myofibers in mice. Therefore, we speculate that circRBBP7 may affect the activity of MDM2 by targeting VEGFR-1, altering the expression of muscle development-related genes by mediating p53 degradation, and ultimately promoting myoblast development and muscle regeneration. This study provides essential evidence that circRBBP7 can serve as a potential target for myogenesis regulation and a reference for the application of circRBBP7 in cattle genetic breeding and muscle injury treatment.
Collapse
Affiliation(s)
- Yufeng Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Guangxi Agricultural Vocational University, Nanning, China
| | - Kongwei Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Shuwan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaoxian Xu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yang Liu
- Guangxi Zhuang Autonomous Region Center for Analysis and Test Research, Nanning, China
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Mingsong Wei
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Guangxi Agricultural Vocational University, Nanning, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
20
|
Ma H, Weng F, Tong X, Li H, Yao Y, Yuan J. LncRNA TRPM2-AS promotes endometrial carcinoma progression and angiogenesis via targeting miR-497-5p/SPP1 axis. Cell Mol Biol Lett 2024; 29:93. [PMID: 38956502 PMCID: PMC11218065 DOI: 10.1186/s11658-024-00612-7] [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: 01/30/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Anti-angiogenic therapy has become one of the effective treatment methods for tumors. Long noncoding RNAs (lncRNAs) are emerging as important regulators of tumorigenesis and angiogenesis in EC. However, the underlying mechanisms of lncRNA TRPM2-AS in EC are still not clear. METHODS We screened the differently expressed lncRNAs that were highly associated with poor prognosis and angiogenesis of EC by bioinformatics analysis, and constructed a ceRNA network based on the prognostic lncRNAs. The subcellular localization of TRPM2-AS was determined by fluorescence in situ hybridization (FISH) and nuclear cytoplasmic fractionation assay. CCK-8, EdU, transwell, western blot, qRT-PCR and endothelial tube formation assay were used to evaluate the effects of TRPM2-AS on the proliferation, invasion, migration of EC cells and angiogenesis. The targeted microRNA (miRNA) of TRPM2-AS was predicted by bioinformatic methods. The interaction between TRPM2-AS and miR497-5p, miR497-5p and SPP1 were analyzed by RNA immunoprecipitation and dual-luciferase reporter assay. A subcutaneous tumor model was used to explore TRPM2-AS's function in vivo. CIBERSORT was used to analyze the correlation between TRPM2-AS and immune cell immersion in EC. RESULTS We found that the expression of TRPM2-AS and SPP1 was aberrantly upregulated, while miR-497-5p expression was significantly downregulated in EC tissues and cells. TRPM2-AS was closely correlated with the angiogenesis and poor prognosis in EC patients. Mechanistically, TRPM2-AS could sponge miR-497-5p to release SPP1, thus promoting the proliferation, invasion and migration of EC cells and angiogenesis of HUVECs. Knockdown of TRPM2-AS in xenograft mouse model inhibited tumor proliferation and angiogenesis in vivo. In addition, TRPM2-AS plays a vital role in regulating the tumor immune microenvironment of EC, overexpression of TRPM2-AS in EC cells stimulated the polarization of M2 macrophages and angiogenesis through secreting SPP1 enriched exosomes. CONCLUSION The depletion of TRPM2-AS inhibits the oncogenicity of EC by targeting the miR-497-5p/SPP1 axis. This study offers a better understanding of TRPM2-AS's role in regulating angiogenesis and provides a novel target for EC treatment.
Collapse
Affiliation(s)
- Hanbo Ma
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Fengyun Weng
- Department of Obstetrics and Gynecology, Shangyu People's Hospital of Shaoxing, Zhejiang, 312300, China
| | - Xiaowen Tong
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huaifang Li
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yinan Yao
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Jiangjing Yuan
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| |
Collapse
|
21
|
Yu J, Zhang Y, Xue Y, Pei H, Li B. Emerging roles of long noncoding RNAs in enzymes related intracellular metabolic pathways in cancer biology. Biomed Pharmacother 2024; 176:116831. [PMID: 38824835 DOI: 10.1016/j.biopha.2024.116831] [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: 02/03/2024] [Revised: 05/13/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Metabolic reprogramming plays critical roles in the development and progression of tumor by providing cancer cells with a sufficient supply of nutrients and other factors needed for fast-proliferating. Emerging evidence indicates that long noncoding RNAs (lncRNAs) are involved in the initiation of metastasis via regulating the metabolic reprogramming in various cancers. In this paper, we aim to summarize that lncRNAs could participate in intracellular nutrient metabolism including glucose, amino acid, lipid, and nucleotide, regardless of whether lncRNAs have tumor-promoting or tumor-suppressor function. Meanwhile, modulation of lncRNAs in glucose metabolic enzymes in glycolysis, pentose phosphate pathway and tricarboxylic acid cycle (TCA) in cancer is reviewed. We also discuss therapeutic strategies targeted at interfering with enzyme activity to decrease the utilization of glucoses, amino acid, nucleotide acid and lipid in tumor cells. This review focuses on our current understanding of lncRNAs participating in cancer cell metabolic reprogramming, paving the way for further investigation into the combination of such approaches with existing anti-cancer therapies.
Collapse
Affiliation(s)
- Jing Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, Suzhou 215123, China; Department of clinical laboratory Center, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yue Zhang
- School of Clinical Medicine, Medical College of Soochow University, Suzhou 215123, China
| | - Yaqi Xue
- Department of Clinical Nutrition, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Bingyan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
22
|
Liu SC. Comprehensive analysis of clinical and biological value of ING family genes in liver cancer. World J Gastrointest Oncol 2024; 16:2580-2597. [DOI: 10.4251/wjgo.v16.i6.2580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/17/2024] [Accepted: 04/22/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Liver cancer (LIHC) is a malignant tumor that occurs in the liver and has a high mortality in cancer. The ING family genes were identified as tumor suppressor genes. Dysregulated expression of these genes can lead to cell cycle arrest, senescence and/or apoptosis. ING family genes are promising targets for anticancer therapy. However, their role in LIHC is still not well understood.
AIM To have a better understanding of the important roles of ING family members in LIHC.
METHODS A series of bioinformatics approaches (including gene expression analysis, genetic alteration analysis, survival analysis, immune infiltration analysis, prediction of upstream microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) of ING1, and ING1-related gene functional enrichment analysis) was applied to study the expression profile, clinical relationship, prognostic significance and immune infiltration of ING in LIHC. The relationship between ING family genes expression and tumor associated immune checkpoints was investigated in LIHC. The molecular mechanism of ING1 mediated hepatocarcinogenesis was preliminarily discussed.
RESULTS mRNA/protein expression of different ING family genes in LIHC was analyzed in different databases, showing that ING family genes were highly expressed in LIHC. In 47 samples from 366 LIHC patients, the ING family genes were altered at a rate of 13%. By comprehensively analyzing the expression, clinical pathological parameters and prognostic value of ING family genes, ING1/5 was identified. ING1/5 was related to poor prognosis of LIHC, suggesting that they may play key roles in LIHC tumorigenesis and progression. One of the target miRNAs of ING1 was identified as hsa-miR-214-3p. Two upstream lncRNAs of hsa-miR-214-3p, U91328.1, and HCG17, were identified. At the same time, we found that the expression of ING family genes was correlated with immune cell infiltration and immune checkpoint genes.
CONCLUSION This study lays a foundation for further research on the potential mechanism and clinical value of ING family genes in the treatment and prognosis of LIHC.
Collapse
Affiliation(s)
- Shi-Cai Liu
- School of Medical Information, Wannan Medical College, Wuhu 241002, Anhui Province, China
| |
Collapse
|
23
|
Liu SC. Comprehensive analysis of clinical and biological value of ING family genes in liver cancer. World J Gastrointest Oncol 2024; 16:2592-2609. [PMID: 38994155 PMCID: PMC11236222 DOI: 10.4251/wjgo.v16.i6.2592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/17/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Liver cancer (LIHC) is a malignant tumor that occurs in the liver and has a high mortality in cancer. The ING family genes were identified as tumor suppressor genes. Dysregulated expression of these genes can lead to cell cycle arrest, senescence and/or apoptosis. ING family genes are promising targets for anticancer therapy. However, their role in LIHC is still not well understood. AIM To have a better understanding of the important roles of ING family members in LIHC. METHODS A series of bioinformatics approaches (including gene expression analysis, genetic alteration analysis, survival analysis, immune infiltration analysis, prediction of upstream microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) of ING1, and ING1-related gene functional enrichment analysis) was applied to study the expression profile, clinical relationship, prognostic significance and immune infiltration of ING in LIHC. The relationship between ING family genes expression and tumor associated immune checkpoints was investigated in LIHC. The molecular mechanism of ING1 mediated hepatocarcinogenesis was preliminarily discussed. RESULTS mRNA/protein expression of different ING family genes in LIHC was analyzed in different databases, showing that ING family genes were highly expressed in LIHC. In 47 samples from 366 LIHC patients, the ING family genes were altered at a rate of 13%. By comprehensively analyzing the expression, clinical pathological parameters and prognostic value of ING family genes, ING1/5 was identified. ING1/5 was related to poor prognosis of LIHC, suggesting that they may play key roles in LIHC tumorigenesis and progression. One of the target miRNAs of ING1 was identified as hsa-miR-214-3p. Two upstream lncRNAs of hsa-miR-214-3p, U91328.1, and HCG17, were identified. At the same time, we found that the expression of ING family genes was correlated with immune cell infiltration and immune checkpoint genes. CONCLUSION This study lays a foundation for further research on the potential mechanism and clinical value of ING family genes in the treatment and prognosis of LIHC.
Collapse
Affiliation(s)
- Shi-Cai Liu
- School of Medical Information, Wannan Medical College, Wuhu 241002, Anhui Province, China
| |
Collapse
|
24
|
Wen K, Chen X, Gu J, Chen Z, Wang Z. Beyond traditional translation: ncRNA derived peptides as modulators of tumor behaviors. J Biomed Sci 2024; 31:63. [PMID: 38877495 PMCID: PMC11177406 DOI: 10.1186/s12929-024-01047-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/24/2024] [Indexed: 06/16/2024] Open
Abstract
Within the intricate tapestry of molecular research, noncoding RNAs (ncRNAs) were historically overshadowed by a pervasive presumption of their inability to encode proteins or peptides. However, groundbreaking revelations have challenged this notion, unveiling select ncRNAs that surprisingly encode peptides specifically those nearing a succinct 100 amino acids. At the forefront of this epiphany stand lncRNAs and circRNAs, distinctively characterized by their embedded small open reading frames (sORFs). Increasing evidence has revealed different functions and mechanisms of peptides/proteins encoded by ncRNAs in cancer, including promotion or inhibition of cancer cell proliferation, cellular metabolism (glucose metabolism and lipid metabolism), and promotion or concerted metastasis of cancer cells. The discoveries not only accentuate the depth of ncRNA functionality but also open novel avenues for oncological research and therapeutic innovations. The main difficulties in the study of these ncRNA-derived peptides hinge crucially on precise peptide detection and sORFs identification. Here, we illuminate cutting-edge methodologies, essential instrumentation, and dedicated databases tailored for unearthing sORFs and peptides. In addition, we also conclude the potential of clinical applications in cancer therapy.
Collapse
Affiliation(s)
- Kang Wen
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, P.R. China
| | - Xin Chen
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, P.R. China
| | - Jingyao Gu
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, P.R. China
| | - Zhenyao Chen
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P.R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhaoxia Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, P.R. China.
| |
Collapse
|
25
|
Li Z, Yan H, Wang B, Wang H, Chen A, Zhu T, Liu J, Yu G, Kang M. High methylation of the same promoter of lncRNA ZNF582-AS1/ ZNF582 promotes malignant progression of esophageal cancer. Epigenomics 2024; 16:733-752. [PMID: 38869483 PMCID: PMC11318746 DOI: 10.1080/17501911.2024.2342229] [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: 11/13/2023] [Accepted: 04/09/2024] [Indexed: 06/14/2024] Open
Abstract
Aim: This study aimed to investigate the functions of ZNF582-AS1 and ZNF582 in esophageal cancer (EC). Materials & methods: Bioinformatics analysis, qRT-PCR and western blot were used to analyze the expression levels. Biological functions were evaluated using cell-counting kit 8, colony formation, Transwell assays and flow cytometry. FISH was used to detect subcellular localization, and methylation-specific PCR determined gene methylation levels. Animal experiments validated the impact on tumor progression. Results: ZNF582-AS1 and ZNF582 were highly methylated and downregulated in EC. Overexpression of ZNF582-AS1 up-regulated the expression of ZNF582, thereby inhibiting EC cell viability and metastasis, promoting apoptosis and inhibiting tumor growth. Conclusion: Low expression of ZNF582-AS1/ZNF582 mediated by DNA hypermethylation facilitates the malignant progression of EC.
Collapse
Affiliation(s)
- Zhupeng Li
- Second Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Hefang Yan
- Department of Endocrinology, Rheumatology & immunology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Bin Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Haiyong Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Aixia Chen
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Ting Zhu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Jianjiang Liu
- Department of Radiotherapy, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Mingqiang Kang
- Second Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| |
Collapse
|
26
|
Zhang Y, Lu Y, Wang N, Yang Y, Hao F, Fei X, Chen Y, Wang J. Alternative splicing-related long noncoding RNA ANRIL facilitates hepatocellular carcinoma by targeting the miR-199a-5p/SRSF1 axis and impacting Anillin. Mol Carcinog 2024; 63:1064-1078. [PMID: 38411272 DOI: 10.1002/mc.23709] [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: 10/07/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by aberrant alternative splicing (AS), which plays an important part in the pathological process of this disease. However, available reports about genes and mechanisms involved in AS process are limited. Our previous research has identified ANRIL as a long noncoding RNA related to the AS process of HCC. Here, we investigated the exact effect and the mechanism of ANRIL on HCC progress. The ANRIL expression profile was validated using the real-time quantitative polymerase chain reaction assay. The western blot analysis and IHC assay were conducted on candidate targets, including SRSF1 and Anillin. The clinicopathological features of 97 patients were collected and analyzed. Loss-of and gain-of-function experiments were conducted. The dual-luciferase reporter assay was applied to verify the interaction between ANRIL, miR-199a-5p, and SRSF1. Anomalous upregulation of ANRIL in HCC was observed, correlating with worse clinicopathological features of HCC. HCC cell proliferation, mobility, tumorigenesis, and metastasis were impaired by depleting ANRIL. We found that ANRIL acts as a sponger of miRNA-199a-5p, resulting in an elevated level of its target protein SRSF1. The phenotypes induced by ANRIL/miR-199a-5p/SRSF1 alteration are associated with Anillin, a validated HCC promoter. ANRIL is an AS-related lncRNA promoting HCC progress by modulating the miR-199a-5p/SRSF1 axis. The downstream effector of this axis in the development of HCC is Anillin.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yiquan Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Nan Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yuchen Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fengjie Hao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongjun Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
27
|
Zhang L, Wang Y, Gao J, Zhou X, Huang M, Wang X, He Z. Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review). Oncol Lett 2024; 27:255. [PMID: 38646493 PMCID: PMC11027111 DOI: 10.3892/ol.2024.14388] [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: 11/04/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Esophageal cancer (EC) is a common form of malignant tumor in the digestive system that is classified into two types: Esophageal squamous cell carcinomas (ESCC) and esophageal adenocarcinoma. ESCC is known for its early onset of symptoms, which can be difficult to identify, as well as its rapid progression and tendency to develop drug resistance to chemotherapy and radiotherapy. These factors contribute to the high incidence of disease and low cure rate. Therefore, a diagnostic biomarker and therapeutic target need to be identified for ESCC. Non-coding RNAs (ncRNAs) are a class of molecules that are transcribed from DNA but do not encode proteins. Initially, ncRNAs were considered to be non-functional segments generated during transcription. However, with advancements in high-throughput sequencing technologies in recent years, ncRNAs have been associated with poor prognosis, drug resistance and progression of ESCC. The present study provides a comprehensive overview of the biogenesis, characteristics and functions of ncRNAs, particularly focusing on microRNA, long ncRNAs and circular RNAs. Furthermore, the ncRNAs that could potentially be used as diagnostic biomarkers and therapeutic targets for ESCC are summarized to highlight their application value and prospects in ESCC.
Collapse
Affiliation(s)
- Longze Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yanyang Wang
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianmei Gao
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xue Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
28
|
Du B, Zhang Z, Jia L, Zhang H, Zhang S, Wang H, Cheng Z. Micropeptide AF127577.4-ORF hidden in a lncRNA diminishes glioblastoma cell proliferation via the modulation of ERK2/METTL3 interaction. Sci Rep 2024; 14:12090. [PMID: 38802444 PMCID: PMC11130299 DOI: 10.1038/s41598-024-62710-y] [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: 01/26/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024] Open
Abstract
Micropeptides hidden in long non-coding RNAs (lncRNAs) have been uncovered to program various cell-biological changes associated with malignant transformation-glioblastoma (GBM) cascade. Here, we identified and characterized a novel hidden micropeptide implicated in GBM. We screened potential candidate lncRNAs by establishing a workflow involving ribosome-bound lncRNAs, publicly available MS/MS data, and prognosis-related lncRNAs. Micropeptide expression was detected by western blot (WB), immunofluorescence (IF), and immunohistochemistry (IHC). Cell proliferation rate was assessed by calcein/PI staining and EdU assay. Proteins interacted with the micropeptide were analyzed by proteomics after co-immunoprecipitation (Co-IP). We discovered that lncRNA AF127577.4 indeed encoded an endogenous micropeptide, named AF127577.4-ORF. AF127577.4-ORF was associated with GBM clinical grade. In vitro, AF127577.4-ORF could suppress GBM cell proliferation. Moreover, AF127577.4-ORF reduced m6A methylation level of GBM cells. Mechanistically, AF127577.4-ORF diminished ERK2 interaction with m6A reader methyltransferase like 3 (METTL3) and downregulated phosphorylated ERK (p-ERK) level. The ERK inhibitor reduced p-ERK level and downregulated METTL3 protein expression. AF127577.4-ORF weakened the stability of METTL3 protein by ERK. Also, AF127577.4-ORF suppressed GBM cell proliferation via METTL3. Our study identifies a novel micropeptide AF127577.4-ORF hidden in a lncRNA, with a potent anti-proliferating function in GBM by diminishing METTL3 protein stability by reducing the ERK2/METTL3 interaction. This micropeptide may be beneficial for development of therapeutic strategies against GBM.
Collapse
Affiliation(s)
- Baoshun Du
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, 453003, Henan, People's Republic of China
| | - Zheying Zhang
- Department of Pathology, Xinxiang Medical University, No. 601 Jinsui Avenue, Xinxiang, 453003, Henan, People's Republic of China.
| | - Linlin Jia
- Department of Critical Care Medicine, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, 450053, Henan, People's Republic of China
| | - Huan Zhang
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, 453003, Henan, People's Republic of China
| | - Shuai Zhang
- Department of Pathology, Xinxiang Medical University, No. 601 Jinsui Avenue, Xinxiang, 453003, Henan, People's Republic of China
| | - Haijun Wang
- Department of Pathology, Xinxiang Medical University, No. 601 Jinsui Avenue, Xinxiang, 453003, Henan, People's Republic of China
| | - Zhenguo Cheng
- Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, 453003, Henan, People's Republic of China
| |
Collapse
|
29
|
Zheng B, Zhou B, Ye D, Wang Y, Zheng W, Wang X, Liu D, Qian F, Zhou X, Yan T, Li Y, Fang L. LINC01572 promotes triple-negative breast cancer progression through EIF4A3-mediated β-catenin mRNA nuclear exportation. ENVIRONMENTAL TOXICOLOGY 2024; 39:3026-3039. [PMID: 38317508 DOI: 10.1002/tox.24171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 02/07/2024]
Abstract
Long noncoding RNAs have been reported to be involved in the development of breast cancer. LINC01572 was previously reported to promote the development of various tumors. However, the potential biological function of LINC01572 in breast cancer remains largely unknown. R language was used to perform bioinformatic analysis of The Cancer Genome Atlas data. The expression level of RNAs was examined by RT-qPCR. The effect of knocking down or overexpression LINC01572 in triple-negative breast cancer (TNBC) cell lines was evaluated by detecting cell proliferation, migrant action. RNA immunoprecipitation assay and RNA pull-down assay were performed to explore the regulatory relationship between LINC01572, EIF4A3, and β-catenin. Bioinformatics analysis identifies LINC01572 as an oncogene of breast cancer. LINC01572 is over-expressed in TNBC tissues and cell lines, correlated with poor clinical prognosis in BC patients. Cell function studies confirmed that LINC01572 facilitated the proliferation and migration of TNBC cells in both vivo and vitro. Mechanistically, β-catenin mRNA and EIF4A3 combine spatially to form a complex, LINC01572 helps transport this complex from the nucleus to the cytoplasm, thereby facilitating the translation of β-catenin. Our findings confirm that LINC01572 acts as a tumor promoter and may act as a biomarker in TNBC. In addition, novel molecular regulatory relationships involving LINC01572/EIF4A3/β-catenin are critical to the development of TNBC, which led to a new understanding of the mechanisms of TNBC progression and shows a new target for precision treatment for TNBC.
Collapse
Affiliation(s)
- Bowen Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Baian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Danrong Ye
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yuying Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Wenfang Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xuehui Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Diya Liu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fengyuan Qian
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xiqian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tao Yan
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
- Medical College, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Yating Li
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
- Medical College, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
30
|
Shi S, Tang X, Liu H. Disulfidptosis-Related lncRNA for the Establishment of Novel Prognostic Signature and Therapeutic Response Prediction to Endometrial Cancer. Reprod Sci 2024; 31:811-822. [PMID: 37880552 DOI: 10.1007/s43032-023-01382-x] [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: 07/23/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
Disulfidptosis, a newly discovered cellular death mechanism initiated by disulfide stress, features elevated expression of SLC7A11 and restricted glucose availability, rendering it a possible therapeutic target for cancer. Endometrial cancer of the uterine corpus (ECUC) ranks among prevalent gynecological malignancies. Long non-coding RNAs (lncRNAs) have been implicated in ECUC's metabolic pathways, invasive capabilities, and metastatic processes. Yet, the prognostic implications of Disulfidptosis-Linked lncRNAs (DLLs) in ECUC remain ambiguous. Transcriptome and clinical datasets related to ECUC were sourced from The Cancer Genome Atlas (TCGA), while genes linked with disulfidptosis were identified from existing literature. A panel of ten DLLs was discerned through least absolute shrinkage and selection operator (LASSO) coupled with Cox regression methods to formulate and validate risk prognostic models. We engineered a nomogram for ECUC patient prognosis forecasting and further examined the model via gene set enrichment analysis (GSEA), principal component analysis (PCA), gene set analysis (GSA), immune profiling, and sensitivity to antineoplastic agents. Prognostic models employing a set of ten DLLs (including AC005034.2, AC020765.2, AL158071.4, AL161663.2, AP000787.1, CR392039.3, EMSLR, SEC24B-AS1, Z69733.1, Z94721.3) were established. Based on median risk values, patient samples were stratified into high- and low-risk cohorts, revealing notable differences in survival across both training and validation datasets. The risk scores, when amalgamated with clinical variables, acted as standalone predictors of prognosis. GSEA findings indicated that the high-risk category predominantly aligned with pathways like extracellular matrix interactions and cell adhesion molecules, suggesting a likely association with metastatic potential. Concurrently, we scrutinized disparities in immune function and tumor mutational burden across risk categories and identified anticancer drugs with likely efficacy. In summary, a set of ten DLLs proved useful in forecasting patient outcomes and holds potential for informing targeted therapeutic approaches in ECUC.
Collapse
Affiliation(s)
- Shanping Shi
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Xiaojian Tang
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
31
|
Han X, Chen L, Sun P, Wang X, Zhao Q, Liao L, Lou D, Zhou N, Wang Y. A novel lncRNA-hidden polypeptide regulates malignant phenotypes and pemetrexed sensitivity in A549 pulmonary adenocarcinoma cells. Amino Acids 2024; 56:15. [PMID: 38351332 PMCID: PMC10864564 DOI: 10.1007/s00726-023-03361-7] [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: 09/22/2023] [Accepted: 12/20/2023] [Indexed: 02/16/2024]
Abstract
The advance of high-throughput sequencing enhances the discovery of short ORFs embedded in long non-coding RNAs (lncRNAs). Here, we uncovered the production and biological activity of lncRNA-hidden polypeptides in lung adenocarcinoma (LUAD). In the present study, bioinformatics was used to screen the lncRNA-hidden polypeptides in LUAD. Analysis of protein expression was done by western blot or immunofluorescence assay. The functions of the polypeptide were determined by detecting its effects on cell viability, proliferation, migration, invasion, and pemetrexed (PEM) sensitivity. The protein interactors of the polypeptide were analyzed by mass spectrometry after Co-immunoprecipitation (Co-IP) assay. The results showed that the lncRNA LINC00954 was confirmed to encode a novel polypeptide LINC00954-ORF. The polypeptide had tumor-suppressor features in A549 cells by repressing cell growth, motility and invasion. Moreover, the polypeptide enhanced PEM sensitivity and suppressed growth in A549/PEM cells. The protein interactors of this polypeptide had close correlations with RNA processing, amide metabolic process, translation, RNA binding, RNA transport, and DNA replication. As a conclusion, the LINC00954-ORF polypeptide embedded in lncRNA LINC00954 possesses tumor-suppressor features in A549 and PEM-resistant A549 cells and sensitizes PEM-resistant A549 cells to PEM, providing evidence that the LINC00954-ORF polypeptide is a potential anti-cancer agent in LUAD.
Collapse
Affiliation(s)
- Xiaobing Han
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China.
| | - Liangxin Chen
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Peng Sun
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Xiuqing Wang
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Qian Zhao
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Lingfeng Liao
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Dejin Lou
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Nan Zhou
- Department of Oncology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China
| | - Yujun Wang
- Department of Gastroenterology, Xinyang Central Hospital, No. 1 Siyi Road, Shihe District, Xinyang, 464000, Henan, China.
| |
Collapse
|
32
|
Su D, Ding C, Qiu J, Yang G, Wang R, Liu Y, Tao J, Luo W, Weng G, Zhang T. Ribosome profiling: a powerful tool in oncological research. Biomark Res 2024; 12:11. [PMID: 38273337 PMCID: PMC10809610 DOI: 10.1186/s40364-024-00562-4] [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: 08/19/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
Neoplastic cells need to adapt their gene expression pattern to survive in an ever-changing or unfavorable tumor microenvironment. Protein synthesis (or mRNA translation), an essential part of gene expression, is dysregulated in cancer. The emergence of distinct translatomic technologies has revolutionized oncological studies to elucidate translational regulatory mechanisms. Ribosome profiling can provide adequate information on diverse aspects of translation by aiding in quantitatively analyzing the intensity of translating ribosome-protected fragments. Here, we review the primary currently used translatomics techniques and highlight their advantages and disadvantages as tools for translatomics studies. Subsequently, we clarified the areas in which ribosome profiling could be applied to better understand translational control. Finally, we summarized the latest advances in cancer studies using ribosome profiling to highlight the extensive application of this powerful and promising translatomic tool.
Collapse
Affiliation(s)
- Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Chen Ding
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Gang Yang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R. China
| | - Guihu Weng
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R. China.
| |
Collapse
|
33
|
Yang Z, Zhang S, Xiong J, Xia T, Zhu R, Miao M, Li K, Chen W, Zhang L, You Y, You B. The m 6A demethylases FTO and ALKBH5 aggravate the malignant progression of nasopharyngeal carcinoma by coregulating ARHGAP35. Cell Death Discov 2024; 10:43. [PMID: 38263362 PMCID: PMC10806234 DOI: 10.1038/s41420-024-01810-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
N6-methyladenosine (m6A) is an RNA modification that can be removed by demethylases [fat mass and obesity-associated protein (FTO) and AlkB homolog 5 (ALKBH5)], which regulate gene expression and cell function. We show that m6A levels and m6A demethylase levels are altered in nasopharyngeal carcinoma (NPC) tissues vs. normal tissues. High FTO and ALKBH5 predict a poor prognosis in NPC patients. Silencing FTO and ALKBH5 inhibited the malignant behavior of patient-derived NPC cells in a short time. However, as time progressed, the inhibitory effect of FTO or ALKBH5 was weakened, and the cosilencing of FTO and ALKBH5 maintained a better inhibitory effect. Combined transcriptome and m6A-seq analysis revealed a downstream target gene that was jointly regulated by FTO and ALKBH5 in NPC, and ARHGAP35 was chosen to do further study. The synergistic silencing of FTO and ALKBH5 increased the methylation level on the mRNA CDS of a new transcription factor (ARHGAP35) and positively regulate the protein coding capacity and mRNA stability of ARHGAP35, thus leading to increased expression of ARHGAP35 and inhibition of the malignant phenotype of tumor cells. Our study revealed that the growth and metastasis of NPC can be stably inhibited through synergistic silencing of the demethylases FTO and ALKBH5, which play a positive role in the treatment of NPC by regulating the downstream transcript ARHGAP35 and increasing its m6A level.
Collapse
Affiliation(s)
- Zhiyuan Yang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Siyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Jiayan Xiong
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Tian Xia
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Rui Zhu
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Mengyu Miao
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Keying Li
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Wenyue Chen
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Lin Zhang
- Haimen People's Hospital, Nantong, China
| | - Yiwen You
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Bo You
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| |
Collapse
|
34
|
Zhong S, Xu H, Wang D, Yang S, Li H, Zhang H, Feng J, Zhou S. circNFIB decreases synthesis of arachidonic acid and inhibits breast tumor growth and metastasis. Eur J Pharmacol 2024; 963:176221. [PMID: 38128869 DOI: 10.1016/j.ejphar.2023.176221] [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: 08/31/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023]
Abstract
We identified circNFIB (hsa_circ_0086376) as a down-regulated circRNA in breast cancer but its effect is unclear. We aimed to explore the roles of circNFIB in breast cancer. The expression levels of circNFIB in breast cancer tissues and cells were detected. Both in vitro and in vivo experiments were used to assess the effects and mechanisms of circNFIB. circNFIB was down-regulated in 29 breast cancer tissues compared to adjacent normal tissues. circNFIB is a highly conserved circRNA and mainly located in cytoplasm of breast cancer cells. In vitro experiments showed that overexpression of circNFIB inhibited proliferation and invasion of breast cancer cells, whereas knockdown of circNFIB induced proliferation and invasion. Animal experiments indicated that circNFIB inhibited tumor growth and metastasis in vivo. Bioinformatics analysis showed that circNFIB contained an open reading frame (ORF) spanning its spliced junction, an internal ribosome entry site (IRES) and a N6-methyladenosine (m6A) site, suggesting circNFIB had the potential to encode a 56 amino acid (aa) protein, which was then confirmed by experiments. Metabonomics analysis results indicated that circNFIB may inhibit synthesis of arachidonic acid (AA) by regulating phospholipase. EIF4A3 and U2AF65 may regulate circNFIB expression by binding to the flanking sequence of circNFIB. In conclusion, circNFIB is a down-regulated circRNA in breast cancer tissues and encodes a 56 aa protein. circNFIB down-regulates AA in breast cancer cells, thus decreasing AA metabolites. Based on reported evidences of AA metabolites on cancer, we speculated that circNFIB may inhibit breast tumor growth and metastasis partly by inhibiting AA.
Collapse
Affiliation(s)
- Shanliang Zhong
- Center of Clinical Laboratory Science, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
| | - Hanzi Xu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
| | - Dandan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Sujin Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Huixin Li
- Department of Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| | - Heda Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Jifeng Feng
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, China.
| | - Siying Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215168, China.
| |
Collapse
|
35
|
Liu S, Jiao B, Zhao H, Liang X, Jin F, Liu X, Hu J. LncRNAs-circRNAs as Rising Epigenetic Binary Superstars in Regulating Lipid Metabolic Reprogramming of Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303570. [PMID: 37939296 PMCID: PMC10767464 DOI: 10.1002/advs.202303570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/28/2023] [Indexed: 11/10/2023]
Abstract
As one of novel hallmarks of cancer, lipid metabolic reprogramming has recently been becoming fascinating and widely studied. Lipid metabolic reprogramming in cancer is shown to support carcinogenesis, progression, distal metastasis, and chemotherapy resistance by generating ATP, biosynthesizing macromolecules, and maintaining appropriate redox status. Notably, increasing evidence confirms that lipid metabolic reprogramming is under the control of dysregulated non-coding RNAs in cancer, especially lncRNAs and circRNAs. This review highlights the present research findings on the aberrantly expressed lncRNAs and circRNAs involved in the lipid metabolic reprogramming of cancer. Emphasis is placed on their regulatory targets in lipid metabolic reprogramming and associated mechanisms, including the clinical relevance in cancer through lipid metabolism modulation. Such insights will be pivotal in identifying new theranostic targets and treatment strategies for cancer patients afflicted with lipid metabolic reprogramming.
Collapse
Affiliation(s)
- Shanshan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationCancer Center, First HospitalJilin UniversityChangchun130021China
- Hematology DepartmentFirst HospitalJilin UniversityChangchun130021China
| | - Benzheng Jiao
- NHC Key Laboratory of Radiobiology (Jilin University)School of Public HealthJilin UniversityChangchun130021China
- Nuclear Medicine DepartmentFirst HospitalJilin UniversityChangchun130021China
| | - Hongguang Zhao
- Nuclear Medicine DepartmentFirst HospitalJilin UniversityChangchun130021China
| | - Xinyue Liang
- Hematology DepartmentFirst HospitalJilin UniversityChangchun130021China
| | - Fengyan Jin
- Hematology DepartmentFirst HospitalJilin UniversityChangchun130021China
| | - Xiaodong Liu
- NHC Key Laboratory of Radiobiology (Jilin University)School of Public HealthJilin UniversityChangchun130021China
- Radiation Medicine Department, School of Public Health and ManagementWenzhou Medical UniversityWenzhou325035China
| | - Ji‐Fan Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationCancer Center, First HospitalJilin UniversityChangchun130021China
- Palo Alto Veterans Institute for ResearchStanford University Medical SchoolPalo AltoCA94304USA
| |
Collapse
|
36
|
Liang J, Cao H, Wang G, Wei L, Dong L, Chen G, Li Q, Zhu D. TMPOP2 Inhibition Suppresses Pancreatic Cancer Cell Migration and Development by Repressing The JNK/STAT3 Pathway. Adv Biol (Weinh) 2023; 7:e2300113. [PMID: 37469237 DOI: 10.1002/adbi.202300113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/18/2023] [Indexed: 07/21/2023]
Abstract
Pancreatic cancer is a malignancy with a poor prognosis and high mortality. The lincRNA TMPOP2 is highly expressed in gynecological cancers and may exhibit tumor-promoting functions. However, the function of TMPOP2 in pancreatic cancer is limited. TMPOP2 expression in pancreatic cancer and adjacent tissues is analyzed from The Cancer Genome Atlas (TCGA) and GTEx database. It shows the high expression of TMPOP2 in pancreatic cancer tissues. Similar results are observed in resected pancreatic adenocarcinoma tumors and adjacent tissues from 20 patients and the relative cell lines. When the pancreatic cell lines are transfected with si-TMPOP2, it shows that TMPOP2 downregulation inhibits the cells migration and EMT. Furthermore, the potential mechanism is explored by detecting the expression of c-Jun N-terminal kinase (JNK), phosphorylated JNK, signal transducer and activator of transcription 3 (STAT3), and phosphorylated STAT3. It suggests that TMPOP2 knockdown inactivates JNK and STAT3 phosphorylation. When a JNK activator (anisomycin) is added to the cells with si-NC or si-TMPOP2, it can partially reverse the migration and EMT inhibition of the cells with inhibited TMPOP2. TMPOP2 inhibition suppresses the migration and EMT of pancreatic cancer by repressing the JNK/STAT3 pathway. Thus, this may be a novel target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Jun Liang
- Department of Oncology Medicine Center, Shanghai East Hospital, School of Medicine,Tongji University, Shanghai, 200092, China
| | - Huangming Cao
- Department of Oncology, East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai, 200123, China
| | - Guangxue Wang
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Liang Wei
- Department of Neurosurgery, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lin Dong
- Department of Cardiothoracic Surgery, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Guohan Chen
- Department of Cardiothoracic Surgery, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qinchuan Li
- Department of Cardiothoracic Surgery, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Dongyi Zhu
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai, 200123, China
| |
Collapse
|
37
|
Deng J, Xu W, Jie Y, Chong Y. Subcellular localization and relevant mechanisms of human cancer-related micropeptides. FASEB J 2023; 37:e23270. [PMID: 37994683 DOI: 10.1096/fj.202301019rr] [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: 05/21/2023] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 11/24/2023]
Abstract
Rapid advances in high-quality sequencing and bioinformatics have invalidated the argument that noncoding RNAs (ncRNAs) are junk transcripts that do not encode proteins. Increasing evidence suggests that small open reading frames (sORFs) in ncRNAs can encode micropeptides and polypeptides within 100 amino acids in length. Several micropeptides have been characterized and proven to have various functions in human physiology and pathology, particularly in cancer. The present review mainly highlights the latest studies on ncRNA-encoded micropeptides in different cancers and categorizes them based on their subcellular localization, thereby providing a theoretical basis for micropeptide applications in the early diagnosis and prognosis of cancer and as therapeutic targets. However, considering the inherent characteristics of micropeptides and the limitations of the assay technology methods, more detailed information is warranted.
Collapse
Affiliation(s)
- Jing Deng
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenli Xu
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yusheng Jie
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yutian Chong
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Mohsen JJ, Martel AA, Slavoff SA. Microproteins-Discovery, structure, and function. Proteomics 2023; 23:e2100211. [PMID: 37603371 PMCID: PMC10841188 DOI: 10.1002/pmic.202100211] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023]
Abstract
Advances in proteogenomic technologies have revealed hundreds to thousands of translated small open reading frames (sORFs) that encode microproteins in genomes across evolutionary space. While many microproteins have now been shown to play critical roles in biology and human disease, a majority of recently identified microproteins have little or no experimental evidence regarding their functionality. Computational tools have some limitations for analysis of short, poorly conserved microprotein sequences, so additional approaches are needed to determine the role of each member of this recently discovered polypeptide class. A currently underexplored avenue in the study of microproteins is structure prediction and determination, which delivers a depth of functional information. In this review, we provide a brief overview of microprotein discovery methods, then examine examples of microprotein structures (and, conversely, intrinsic disorder) that have been experimentally determined using crystallography, cryo-electron microscopy, and NMR, which provide insight into their molecular functions and mechanisms. Additionally, we discuss examples of predicted microprotein structures that have provided insight or context regarding their function. Analysis of microprotein structure at the angstrom level, and confirmation of predicted structures, therefore, has potential to identify translated microproteins that are of biological importance and to provide molecular mechanism for their in vivo roles.
Collapse
Affiliation(s)
- Jessica J. Mohsen
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
| | - Alina A. Martel
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
| | - Sarah A. Slavoff
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| |
Collapse
|
39
|
Du J, Su Y, Gao J, Tai Y. The expression and function of long noncoding RNAs in hepatocellular carcinoma. CANCER INNOVATION 2023; 2:488-499. [PMID: 38125766 PMCID: PMC10730004 DOI: 10.1002/cai2.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 12/23/2023]
Abstract
With the deepening of the genome project study, attention on noncoding RNAs is increasing. Long noncoding RNAs (lncRNAs) have become a new research hotspot. A growing number of studies have revealed that lncRNAs are involved in tumorigenesis and tumor suppressor pathways. Aberrant expressions of lncRNAs have been found in a variety of human tumors including hepatocellular carcinoma (HCC). In this review, we provide a brief introduction to lncRNA and highlight recent research on the functions and clinical significance of lncRNAs in HCC.
Collapse
Affiliation(s)
- Jingli Du
- Senior Department of TuberculosisThe 8th Medical Center of PLA General HospitalBeijingChina
| | - Yue Su
- Senior Department of TuberculosisThe 8th Medical Center of PLA General HospitalBeijingChina
| | - Jianzhi Gao
- Department of OncologyZhuozhou Hospital, ZhuozhouHebeiChina
| | - Yanhong Tai
- Department of PathologyThe 5th Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
40
|
Zhan Y, Liu Y, Yang R, Chen Q, Teng F, Huang Y, Jiang X, Wang Y, Yu B, Zhang D, Bao L, Liu X, Huang J. CircPTEN suppresses human clear cell renal carcinoma progression and resistance to mTOR inhibitors by targeting epigenetic modification. Drug Resist Updat 2023; 71:101003. [PMID: 37866104 DOI: 10.1016/j.drup.2023.101003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 10/24/2023]
Abstract
Renal cell carcinoma (RCC) is known to be the most commonly diagnosed kidney cancer. Clear cell RCC (ccRCC) represents approximately 85 % of diagnosed RCC cases. Targeted therapeutics, such as multi-targeted tyrosine kinase inhibitors (TKI) and mTOR inhibitors, are widely used in ccRCC therapy. However, patients treated with mTOR and TKI inhibitors easily acquire drug resistance, making the therapy less effective. Here, we demonstrated that circPTEN inhibits the expression of its parental gene PTEN by reducing methylation of the PTEN promotor and inhibits GLUT1 expression by reducing m6A methylation of GLUT1, which suppresses ccRCC progression and resistance to mTOR inhibitors.
Collapse
Affiliation(s)
- Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Third Affiliated hospital of Navy Medical University, 225 Changhai Road, Yangpu District, Shanghai, China
| | - Yang Liu
- Department of Orthopedics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Road, Pudong District, Shanghai 200127, China
| | - Rui Yang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Qiong Chen
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Pudong District, Shanghai 200127, China
| | - Fei Teng
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Yueying Huang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Third Affiliated hospital of Navy Medical University, 225 Changhai Road, Yangpu District, Shanghai, China
| | - Xin Jiang
- Department of Anesthesiology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Yueming Wang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Pudong District, Shanghai 200127, China
| | - Bin Yu
- WisGen Biosciences Inc., No.400 Fucheng Road, Baiyang Street, Qiantang District, Hangzhou 310000, Zhejiang, China
| | - Ding Zhang
- The Medical Department, 3D Medicines Inc., 118 Furonghua Road, Pudong District, Shanghai 201114, China
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Third Affiliated hospital of Navy Medical University, 225 Changhai Road, Yangpu District, Shanghai, China.
| | - Xinli Liu
- Department of Digestive Oncology, Liaoning Cancer Hospital & Institute, Dalian University of Technology, 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China.
| | - Jiwei Huang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Pudong District, Shanghai 200127, China.
| |
Collapse
|
41
|
Wang M, Liu Z, Fang X, Cong X, Hu Y. The emerging role of m 6A modification of non-coding RNA in gastrointestinal cancers: a comprehensive review. Front Cell Dev Biol 2023; 11:1264552. [PMID: 37965577 PMCID: PMC10642577 DOI: 10.3389/fcell.2023.1264552] [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: 07/21/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Gastrointestinal (GI) cancer is a series of malignant tumors with a high incidence globally. Although approaches for tumor diagnosis and therapy have advanced substantially, the mechanisms underlying the occurrence and progression of GI cancer are still unclear. Increasing evidence supports an important role for N6-methyladenosine (m6A) modification in many biological processes, including cancer-related processes via splicing, export, degradation, and translation of mRNAs. Under distinct cancer contexts, m6A regulators have different expression patterns and can regulate or be regulated by mRNAs and non-coding RNAs, especially long non-coding RNAs. The roles of m6A in cancer development have attracted increasing attention in epigenetics research. In this review, we synthesize progress in our understanding of m6A and its roles in GI cancer, especially esophageal, gastric, and colorectal cancers. Furthermore, we clarify the mechanism by which m6A contributes to GI cancer, providing a basis for the development of diagnostic, prognostic, and therapeutic targets.
Collapse
Affiliation(s)
- Meiqi Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- Department of Biobank, the China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- Department of Biobank, the China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Zhang J, Mei Z, Yao W, Zhao C, Wu S, Ouyang J. SIX1 induced HULC modulates neuropathic pain and Schwann cell oxidative stress after sciatic nerve injury. Gene 2023; 882:147655. [PMID: 37479098 DOI: 10.1016/j.gene.2023.147655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/02/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Neuropathic pain is a severe and debilitating condition caused by damage to the peripheral nerve or central nervous system. Although several mechanisms have been identified, the underlying pathophysiology of neuropathic pain is still not fully understood. Unfortunately, few effective therapies are available for this condition. Therefore, there is an urgent need to investigate the underlying mechanisms of neuropathic pain to develop more effective treatments. Long non-coding RNAs (lncRNAs) have recently gained attention due to their potential to modulate protein expression through various mechanisms. LncRNAs have been implicated in many diseases, including neuropathic pain. This study aimed to identify a novel lncRNA involved in neuropathic pain progression. The lncRNA microarray analysis showed that lncRNA Upregulated in Liver Cancer (HULC) was significantly upregulated in spinal cord tissue of sciatic nerve injury (SNI) rats. Further experiments confirmed that HULC promoted neuropathic pain progression and aggravated H2O2-induced Schwann cell injury. Mechanistically, Sine Oculis Homeobox 1 (SIX1) regulated the transcriptional expression of HULC, and both SIX1 and HULC were involved in neuropathic pain and Schwann cell injury. The results of our research indicate the existence of a previously unknown SIX1/HULC axis that plays a significant role in the development and progression of neuropathic pain, shedding light on the complex mechanisms that underlie this debilitating condition. These findings offer novel insights into the molecular pathways involved in neuropathic pain. This study underscores the potential of targeting lncRNAs as a viable approach to alleviate the suffering of patients with neuropathic pain.
Collapse
Affiliation(s)
- Jinyuan Zhang
- Department of Anatomy, Southern Medical University, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangzhou, China; The Second Department of Orthopedics Surgery (Department of Spinal Surgery), Zhongshan People's Hospital, Zhongshan, China.
| | - Zhi Mei
- The Second Department of Orthopedics Surgery (Department of Spinal Surgery), Zhongshan People's Hospital, Zhongshan, China
| | - Wanxiang Yao
- The Second Department of Orthopedics Surgery (Department of Spinal Surgery), Zhongshan People's Hospital, Zhongshan, China
| | - Chenyi Zhao
- The Second Department of Orthopedics Surgery (Department of Spinal Surgery), Zhongshan People's Hospital, Zhongshan, China
| | - Shutong Wu
- Department of Anatomy, Southern Medical University, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangzhou, China
| | - Jun Ouyang
- Department of Anatomy, Southern Medical University, Guangdong Provincial Key Laboratory of Medical Biomechanics, Guangzhou, China.
| |
Collapse
|
43
|
Vashisht I, Dhaka N, Jain R, Sood A, Sharma N, Sharma MK, Sharma R. Non-coding RNAs-mediated environmental surveillance determines male fertility in plants. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 203:108030. [PMID: 37708711 DOI: 10.1016/j.plaphy.2023.108030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023]
Abstract
Plants are continuously exposed to environmental stresses leading to significant yield losses. With the changing climatic conditions, the intensity and duration of these stresses are expected to increase, posing a severe threat to crop productivity worldwide. Male gametogenesis is one of the most sensitive developmental stages. Exposure to environmental stresses during this stage leads to male sterility and yield loss. Elucidating the underlying molecular mechanism of environment-affected male sterility is essential to address this challenge. High-throughput RNA sequencing studies, loss-of-function phenotypes of sRNA biogenesis genes and functional genomics studies with non-coding RNAs have started to unveil the roles of small RNAs, long non-coding RNAs and the complex regulatory interactions between them in regulating male fertility under different growth regimes. Here, we discuss the current understanding of the non-coding RNA-mediated environmental stress surveillance and regulation of male fertility in plants. The candidate ncRNAs emerging from these studies can be leveraged to generate environment-sensitive male sterile lines for hybrid breeding or mitigate the impact of climate change on male fertility, as the situation demands.
Collapse
Affiliation(s)
- Ira Vashisht
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Namrata Dhaka
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, 123031, India
| | - Rubi Jain
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Akanksha Sood
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Niharika Sharma
- NSW Department of Primary Industries, Orange Agricultural Institute, Orange, NSW, 2800, Australia
| | - Manoj K Sharma
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rita Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan, 333031, India.
| |
Collapse
|
44
|
Zhu Y, Li J, Zhang P, Peng B, Li C, Ming Y, Liu H. Berberine protects hepatocyte from hypoxia/reoxygenation-induced injury through inhibiting circDNTTIP2. PeerJ 2023; 11:e16080. [PMID: 37780378 PMCID: PMC10538280 DOI: 10.7717/peerj.16080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Background During hepatic ischemia-reperfusion injury, the excessive release of inflammatory cytokines can activate the intracellular signal transduction cascade to induce hepatocyte injury. Apoptosis is an important way of cell death after I/R injury. Berberine, a common quaternary ammonium alkaloid, has anti-inflammatory, anti-oxidative stress, and anti-apoptotic effects. An increasing number of studies have revealed the importance of non-coding RNAs, including microRNA, long non-coding RNAs and circular RNAs (circRNAs), as regulators of the effects of berberine. Purpose In this study, we investigated the mechanism of berberine against liver ischemia-reperfusion injury in vitro. Study Design and Methods In this study, hypoxia-reoxygenation (H/R)-treated L02 cells were pretreated with berberine to study the role and mechanism of berberine in resisting hepatic ischemia-reperfusion injury. Results The results show that berberine pre-treatment increased the cell viability of H/R-challenged cells, reduced H/R-induced apoptosis and ROS production, reversed H/R-increased on IL-6, IL-1β, TNF-α, and H/R-decreased IL-10 expression. Mechanically, berberine protect hepatocyte from H/R injury, at least partially, through circDNTTIP2. In addition, circDNTTIP2 can bind to the TATA box of caspase3 promoter, thereby promoting caspase 3-related cell apoptosis and the release of inflammatory cytokines. Conclusion This study found that berberine has a protective effect on H/R-induced hepatocyte damage by inhibiting a novel circRNA, circDNTTIP2. This study provides potential treatment strategies and treatment targets for liver ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yi Zhu
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Junhui Li
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Pengpeng Zhang
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Bo Peng
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Cai Li
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Hong Liu
- The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| |
Collapse
|
45
|
Zhai S, Lin J, Ji Y, Zhang R, Zhang Z, Cao Y, Liu Y, Tang X, Liu J, Liu P, Lin J, Li F, Li H, Shi Y, Fu D, Deng X, Shen B. A microprotein N1DARP encoded by LINC00261 promotes Notch1 intracellular domain (N1ICD) degradation via disrupting USP10-N1ICD interaction to inhibit chemoresistance in Notch1-hyperactivated pancreatic cancer. Cell Discov 2023; 9:95. [PMID: 37714834 PMCID: PMC10504324 DOI: 10.1038/s41421-023-00592-6] [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: 01/18/2023] [Accepted: 07/28/2023] [Indexed: 09/17/2023] Open
Abstract
The extensively activated Notch signaling pathway in pancreatic cancer cells is important in carcinogenesis, chemoresistance, and recurrence. Targeting this pathway is a promising therapeutic strategy for pancreatic cancer; however, few successful approaches have been reported, and currently used molecular inhibitors of this pathway exhibit limited clinical benefits. In this study, we identified a previously uncharacterized microprotein, Notch1 degradation-associated regulatory polypeptide (N1DARP), encoded by LINC00261. N1DARP knockout accelerated tumor progression and enhanced stem cell properties in pancreatic cancer organoids and LSL-Kras, LSL-Trp53, and Pdx1-Cre (KPC) mice. Mechanistically, N1DARP suppressed canonical and non-canonical Notch1 pathways by competitively disrupting the interaction between N1ICD and ubiquitin-specific peptidase 10 (USP10), thereby promoting K11- and K48-linked polyubiquitination of N1ICD. To evaluate the therapeutic potential of N1DARP, we designed a cell-penetrating stapled peptide, SAH-mAH2-5, with a helical structure similar to that of N1DARP that confers remarkable physicochemical stability. SAH-mAH2-5 interacted with and promoted the proteasome-mediated degradation of N1ICD. SAH-mAH2-5 injection provided substantial therapeutic benefits with limited off-target and systemic adverse effects in Notch1-activated pancreatic cancer models. Taken together, these findings confirm that N1DARP acts as a tumor suppressor and chemosensitizer by regulating USP10-Notch1 oncogenic signaling, and suggest a promising therapeutic strategy targeting the N1DARP-N1ICD interaction in Notch1-activated pancreatic cancer.
Collapse
Affiliation(s)
- Shuyu Zhai
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiewei Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuchen Ji
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ronghao Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zehui Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yizhi Cao
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yang Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaomei Tang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jia Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pengyi Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiayu Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fanlu Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hongzhe Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yusheng Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
46
|
Zhu P, Liu B, Fan Z. Noncoding RNAs in tumorigenesis and tumor therapy. FUNDAMENTAL RESEARCH 2023; 3:692-706. [PMID: 38933287 PMCID: PMC11197782 DOI: 10.1016/j.fmre.2023.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/26/2023] [Accepted: 05/07/2023] [Indexed: 06/28/2024] Open
Abstract
Tumorigenesis is a complicated process in which numerous modulators are involved in different ways. Previous studies have focused primarily on tumor-associated protein-coding genes such as oncogenes and tumor suppressor genes, as well as their associated oncogenic pathways. However, noncoding RNAs (ncRNAs), rising stars in diverse physiological and pathological processes, have recently emerged as additional modulators in tumorigenesis. In this review, we focus on two typical kinds of ncRNAs: long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs). We describe the molecular patterns of ncRNAs and focus on the roles of ncRNAs in cancer stem cells (CSCs), tumor cells, and tumor environmental cells. CSCs are a small subset of tumor cells and are generally considered to be cells that initiate tumorigenesis, and dozens of ncRNAs have been defined as critical modulators in CSC maintenance and oncogenesis. Moreover, ncRNAs are widely involved in oncogenetic processes, including sustaining proliferation, resisting cell death, genome instability, metabolic disorders, immune escape and metastasis. We also discuss the potential applications of ncRNAs in tumor diagnosis and therapy. The progress in ncRNA research greatly improves our understanding of ncRNAs in oncogenesis and provides new potential targets for future tumor therapy.
Collapse
Affiliation(s)
- Pingping Zhu
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Benyu Liu
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zusen Fan
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
47
|
Palcau AC, Brandi R, Mehterov NH, Botti C, Blandino G, Pulito C. Exploiting Long Non-Coding RNAs and Circular RNAs as Pharmacological Targets in Triple-Negative Breast Cancer Treatment. Cancers (Basel) 2023; 15:4181. [PMID: 37627209 PMCID: PMC10453179 DOI: 10.3390/cancers15164181] [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: 07/04/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer is one of the most frequent causes of cancer death among women worldwide. In particular, triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype because it is characterized by the absence of molecular targets, thus making it an orphan type of malignancy. The discovery of new molecular druggable targets is mandatory to improve treatment success. In that context, non-coding RNAs represent an opportunity for modulation of cancer. They are RNA molecules with apparently no protein coding potential, which have been already demonstrated to play pivotal roles within cells, being involved in different processes, such as proliferation, cell cycle regulation, apoptosis, migration, and diseases, including cancer. Accordingly, they could be used as targets for future TNBC personalized therapy. Moreover, the peculiar characteristics of non-coding RNAs make them reliable biomarkers to monitor cancer treatment, thus, to monitor recurrence or chemoresistance, which are the most challenging aspects in TNBC. In the present review, we focused on the oncogenic or oncosuppressor role of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) mostly involved in TNBC, highlighting their mode of action and depicting their potential role as a biomarker and/or as targets of new non-coding RNA-based therapeutics.
Collapse
Affiliation(s)
- Alina Catalina Palcau
- Translational Oncology Research Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.P.); (R.B.); (G.B.)
| | - Renata Brandi
- Translational Oncology Research Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.P.); (R.B.); (G.B.)
| | - Nikolay Hristov Mehterov
- Department of Medical Biology, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria;
- Research Institute, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Claudio Botti
- Breast Surgery Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Giovanni Blandino
- Translational Oncology Research Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.P.); (R.B.); (G.B.)
| | - Claudio Pulito
- Translational Oncology Research Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.C.P.); (R.B.); (G.B.)
| |
Collapse
|
48
|
Qi K, Dou Y, Zhang Z, Wei Y, Song C, Qiao R, Li X, Yang F, Wang K, Li X, Han X. Expression Profile and Regulatory Properties of m6A-Modified circRNAs in the Longissimus Dorsi of Queshan Black and Large White Pigs. Animals (Basel) 2023; 13:2190. [PMID: 37443988 DOI: 10.3390/ani13132190] [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: 05/17/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
It is well known that N6-methyladenosine (m6A) is the most abundant modification in linear RNA molecules, but many circRNA molecules have now been found to have a wide range of m6A modification sites as well. However, there are few relevant studies and information on the expression profile and functional regulatory properties of m6A-modified circRNAs (m6A-circRNAs) in longissimus dorsi. In this study, a total of 12 putative m6A-circRNAs were identified and characterized in the longissimus dorsi of Queshan Black and Large White pigs-8 of them were significantly more expressed in the longissimus dorsi of Queshan Black than in Large White pigs, while the other 4 were the opposite. These 12 putative m6A-circRNAs were also found to act as miRNA sponge molecules to regulate fat deposition by constructing the ceRNA regulatory network. Enrichment analysis also revealed that the 12 m6A-circRNAs parent genes and their adsorbed miRNA target genes were widely involved in fat deposition and cell proliferation and differentiation-related pathways, such as the HIF-1 signaling pathway, the pentose phosphate pathway, the MAPK signaling pathway, the glycosphingolipid biosynthesis-lacto and neolacto series, and the TNF signaling pathway, suggesting that the analyzed m6A-circRNAs may be largely involved in the formation of pork quality. These results provide new information to study the regulatory properties of m6A-circRNAs in the formation of pork quality.
Collapse
Affiliation(s)
- Kunlong Qi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yaqing Dou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhe Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yilin Wei
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Chenglei Song
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ruimin Qiao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiuling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Feng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Kejun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xinjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xuelei Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
49
|
Xu X, Peng Q, Jiang X, Tan S, Yang Y, Yang W, Han Y, Chen Y, Oyang L, Lin J, Xia L, Peng M, Wu N, Tang Y, Li J, Liao Q, Zhou Y. Metabolic reprogramming and epigenetic modifications in cancer: from the impacts and mechanisms to the treatment potential. Exp Mol Med 2023; 55:1357-1370. [PMID: 37394582 PMCID: PMC10394076 DOI: 10.1038/s12276-023-01020-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 07/04/2023] Open
Abstract
Metabolic reprogramming and epigenetic modifications are hallmarks of cancer cells. In cancer cells, metabolic pathway activity varies during tumorigenesis and cancer progression, indicating regulated metabolic plasticity. Metabolic changes are often closely related to epigenetic changes, such as alterations in the expression or activity of epigenetically modified enzymes, which may exert a direct or an indirect influence on cellular metabolism. Therefore, exploring the mechanisms underlying epigenetic modifications regulating the reprogramming of tumor cell metabolism is important for further understanding tumor pathogenesis. Here, we mainly focus on the latest studies on epigenetic modifications related to cancer cell metabolism regulations, including changes in glucose, lipid and amino acid metabolism in the cancer context, and then emphasize the mechanisms related to tumor cell epigenetic modifications. Specifically, we discuss the role played by DNA methylation, chromatin remodeling, noncoding RNAs and histone lactylation in tumor growth and progression. Finally, we summarize the prospects of potential cancer therapeutic strategies based on metabolic reprogramming and epigenetic changes in tumor cells.
Collapse
Affiliation(s)
- 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
- University of South China, Hengyang, 421001, 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
| | - 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
| | - Yuyu Chen
- 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
| | - 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
| | - Jinyun 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.
| | - 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
|
50
|
Deng L, Jiang Y, Hu X, Zheng R, Huang Z, Zhang J. ABLNCPP: Attention Mechanism-Based Bidirectional Long Short-Term Memory for Noncoding RNA Coding Potential Prediction. J Chem Inf Model 2023; 63:3955-3966. [PMID: 37294848 DOI: 10.1021/acs.jcim.3c00366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
With the continuous development of ribosome profiling, sequencing technology, and proteomics, evidence is mounting that noncoding RNA (ncRNA) may be a novel source of peptides or proteins. These peptides and proteins play crucial roles in inhibiting tumor progression and interfering with cancer metabolism and other essential physiological processes. Therefore, identifying ncRNAs with coding potential is vital to ncRNA functional research. However, existing studies perform well in classifying ncRNAs and mRNAs, and no research has been explicitly raised to distinguish whether ncRNA transcripts have coding potential. For this reason, we propose an attention mechanism-based bidirectional LSTM network called ABLNCPP to assess the coding possibility of ncRNA sequences. Considering the sequential information loss in previous methods, we introduce a novel nonoverlapping trinucleotide embedding (NOLTE) method for ncRNAs to obtain embeddings containing sequential features. The extensive evaluations show that ABLNCPP outperforms other state-of-the-art models. In general, ABLNCPP overcomes the bottleneck of ncRNA coding potential prediction and is expected to provide valuable contributions to cancer discovery and treatment in the future. The source code and data sets are freely available at https://github.com/YinggggJ/ABLNCPP.
Collapse
Affiliation(s)
- Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Ying Jiang
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Xiaowen Hu
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Rongtao Zheng
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Zhijian Huang
- School of Computer Science and Engineering, Central South University, Changsha 410018, China
| | - Jingpu Zhang
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan 467000, China
| |
Collapse
|