1
|
Jin J, Qin J, Qi X, Zhang J, Zhang Y. Serum exosomal miRNA contributes to the diagnosis of leptomeningeal carcinomatosis. J Neurooncol 2025; 173:419-428. [PMID: 40080246 DOI: 10.1007/s11060-025-04999-x] [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: 02/14/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
PURPOSE Leptomeningeal carcinomatosis (LC) is a severe complication in the advanced stage of lung adenocarcinoma, with an extremely poor prognosis. Currently, the diagnosis of LC poses challenges. Serum exosomal miRNAs (microRNAs) have been demonstrated to possess potential as viable biomarkers. However, their value in the diagnosis of LC remains unclear. METHODS In this study, serum samples were collected from lung adenocarcinoma patients with LC. The control groups consisted of patients with early-stage and advanced-stage lung adenocarcinoma without LC. Serum exosomes were isolated for high - throughput RNA sequencing to screen for differential miRNAs, and the results were validated in 123 samples. Subsequently, the receiver operating characteristic (ROC) curve was used to evaluate the diagnostic ability of exosomal miRNAs for LC. RESULTS The results of miRNA sequencing revealed seven differentially enriched miRNAs (miRNA-1296-5p, miR-503-5p, miR-499a-5p, miR-374a-5p, miR-3173-5p, miR-370-3p and miR-885-3p). The ddPCR confirmed that the expression levels of miRNA-1296-5p, miR-499a-5p and miR-374a-5p were significantly elevated in LC, while miR-370-3p was significantly decreased (P < 0.05). ROC curve analysis showed that the AUC of the combination of miRNA-1296-5p, miR-499a-5p and miR-370-3p with CEA was 0.803 (P < 0.0001), displaying higher diagnostic power for LC. CONCLUSION This study suggests that the specific expression of miRNA-1296-5p, miR-499a-5p, miR-374a-5p and miR-370-3p in the serum exosomes of LC, which has diagnostic potential. And the combination of miRNA-1296-5p, miR-499a-5p and miR-370-3p with CEA can further enhance this potential.
Collapse
Affiliation(s)
- Jie Jin
- Xiong'an Xuanwu Hospital, Baoding, 070001, PR China.
- Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, 050000, PR China.
| | - Junjuan Qin
- Xiong'an Xuanwu Hospital, Baoding, 070001, PR China
| | - Xuejiao Qi
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
- Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, 050000, PR China
| | - Jiasi Zhang
- Xiong'an Xuanwu Hospital, Baoding, 070001, PR China
| | - YingLu Zhang
- Xiong'an Xuanwu Hospital, Baoding, 070001, PR China
| |
Collapse
|
2
|
Wu F, Zhang T, Mei X, Gao T. lncRNA ZNF667‑AS1 inhibits ovarian cancer progression by interacting with the TNF signaling pathway. Exp Ther Med 2025; 29:100. [PMID: 40165800 PMCID: PMC11955820 DOI: 10.3892/etm.2025.12850] [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: 08/14/2024] [Accepted: 01/14/2025] [Indexed: 04/02/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy and poses a significant public health burden. The present study explored the function and mechanism of long noncoding RNA (lncRNA) ZNF667-AS1 in OC progression. The present study conducted a multifaceted evaluation, including transcriptomic analyses, to examine the expression and prognostic value of lncRNA ZNF667-AS1 in OC via the Cancer Genome Atlas and Genotype-Tissue Expression data. In vitro experiments on OC cell lines were used to investigate the functional effect of ZNF667-AS1 via cell proliferation, migration and invasion assays and RNA sequencing and western blotting were used to explore the implicated molecular pathways. ZNF667-AS1 was significantly underexpressed in OC tissues and cell lines. Its expression levels were positively associated with improved patient prognosis and affected both tumor behavior and tumor microenvironment interactions. Functional analysis confirmed the tumor-suppressive role of ZNF667-AS1 and revealed a marked decrease in proliferation, migration and invasion in ZNF667-AS1-overexpressing cells. Additionally, ZNF667-AS1 was identified as a key regulator in the tumor necrosis factor signaling pathway, which suggests a strong link between ZNF667-AS1 expression and OC progression. The present study identified ZNF667-AS1 as a potential biomarker for OC prognosis and treatment and illustrated its significant regulatory effects on the TNF pathway and its broader implications in cancer pathobiology.
Collapse
Affiliation(s)
- Fangfang Wu
- Department of Obstetrics and Gynecology, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236000, P.R. China
| | - Tianci Zhang
- Anhui Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xiaoyue Mei
- Department of Osteotraumatology of Traditional Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Tianwen Gao
- Department of Pharmacy, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236000, P.R. China
| |
Collapse
|
3
|
Li W, Zhang C, Gao T, Sun Y, Yang H, Liu L, Shi M, Ding L, Zhang C, Deng DYB, Li T. Human umbilical cord mesenchymal stem cells small extracellular vesicles-derived miR-370-3p inhibits cervical precancerous lesions by targeting DHCR24. Stem Cells Transl Med 2025; 14:szae087. [PMID: 39552565 PMCID: PMC11825698 DOI: 10.1093/stcltm/szae087] [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: 05/12/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Cervical cancer is often caused by persistent high-risk human papillomavirus (HPV) infection, causing precancerous lesions. Human umbilical cord mesenchymal stem cells-derived small extracellular vesicles (hucMSC-sEV) exhibit diverse effects on tumors. This study investigates hucMSC-sEV, the impact and mechanisms on HPV-positive cervical precancerous lesion cells to provide new treatment insights. MATERIALS AND METHODS We previously obtained hucMSC and hucMSC-sEV. In vitro experiments evaluated hucMSC-sEV effects on the proliferation and migration of S12 cells (derived from cervical precancerous lesions). Bioinformatics identified key microRNA components, and their impact on S12 cell proliferation and migration was investigated. The target gene of the microRNA component was predicted and confirmed via bioinformatics and dual-luciferase reporter assays. Lentiviral systems overexpressed target gene in S12 cells to examine the effects on microRNA impacts. SH-42 inhibitor was used to investigate target gene treatment potential. Immunohistochemistry assessed target gene expression in cervical precancerous lesions tissue. RESULTS hucMSC-sEV significantly inhibited S12 cell proliferation and migration. Bioinformatics identified miR-370-3p as an effective cargo, which also suppressed S12 cell proliferation and migration. miR-370-3p was confirmed targeting DHCR24 (24-Dehydrocholesterol Reductase). DHCR24 overexpression reversed miR-370-3p's inhibitory effects, while SH-42 counteracted DHCR24 overexpression's promoting effects. Clinical specimen analysis supported these findings, demonstrating a positive correlation between DHCR24 protein expression and cervical precancerous lesions' progression. CONCLUSIONS hucMSC-sEV inhibits S12 cell proliferation and migration, mediated by miR-370-3p targeting DHCR24 to regulate cellular cholesterol content. DHCR24 inhibition reduces the cholesterol level and cell functions, suggesting its potential as a therapeutic target in cervical precancerous lesions.
Collapse
Affiliation(s)
- Weizhao Li
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen 518107, People’s Republic of China
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong 999077, People’s Republic of China
| | - Chi Zhang
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Tianshun Gao
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Yazhou Sun
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Huan Yang
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Lixiang Liu
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Ming Shi
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Lu Ding
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Changlin Zhang
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen 518107, People’s Republic of China
| | - David Y B Deng
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
| | - Tian Li
- Department of Gynecology, Pelvic Floor disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, People’s Republic of China
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen 518107, People’s Republic of China
| |
Collapse
|
4
|
Li X, Zhou F, Niu K, Wang Y, Shi Y, Li Y, Gao X, Zhao W, Chen T, Zhang Y. Emerging discoveries on the role of TRIM14: from diseases to immune regulation. Cell Death Discov 2024; 10:513. [PMID: 39719450 DOI: 10.1038/s41420-024-02276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
TRIM14 is an important member of the TRIM family and is widely expressed in a variety of tissues. Like other members of the TRIM family, TRIM14 is also involved in ubiquitination modifications. TRIM14 was initially reported as an interferon-stimulated gene (ISG). In recent years, many studies have focused on the regulatory role of TRIM14 in signaling pathways such as the PI3K/Akt, NF-κB, and cGAS/STING pathways and revealed its mechanism of action in a variety of pathophysiological processes, and the regulation of TRIM14 has attracted the interest of many researchers as a new direction for the treatment of various diseases. However, there are no reviews on the role of TRIM14 in diseases. In this paper, we will describe the structure of TRIM14, review its role in cancer, cardiovascular disease, cervical spondylosis, inflammation and antiviral immunity, and provide an outlook on future research directions.
Collapse
Affiliation(s)
- Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Feilong Zhou
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kaiyi Niu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yizhu Wang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanlong Shi
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yunxin Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xin Gao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weijie Zhao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianyi Chen
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Tang Y, Ji H, Yan Y, Hu D, Xu M, Xu M, Zhao X, Chen M. Enhancing diabetic foot ulcer healing: Impact of the regulation of the FUS and ILF2 RNA‑binding proteins through negative pressure wound therapy. Int J Mol Med 2024; 54:103. [PMID: 39301661 PMCID: PMC11414528 DOI: 10.3892/ijmm.2024.5427] [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: 05/24/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetic foot ulcer (DFU) is a destructive complication of diabetes. Negative pressure wound therapy (NPWT) promotes DFU wound healing through an undetermined mechanism. In the present study, RNA sequencing was performed on wound granulation tissue from 3 patients with DFU before and after 1 week of NPWT. The fused in sarcoma (FUS) and interleukin enhancer binding factor 2 (ILF2) encoding RNA‑binding proteins (RBPs) were screened from the sequencing data, and wound tissue samples from 24 patients with DFU were validated and analyzed before and after receiving NPWT by reverse transcription‑quantitative PCR, western blotting and immunohistochemistry. In addition, in vitro and in vivo experiments were conducted to determine the effect of the expression of FUS and ILF2 on the function of human epidermal keratinocyte cells (HaCaT cells) and the healing of diabetic skin wounds. The results indicated that NPWT induced the upregulation of 101 genes and the downregulation of 98 genes in DFU wound granulation tissue. After NPWT, the expression of FUS and ILF2 was significantly upregulated (P<0.05). Pearson's correlation coefficient showed that the changes in FUS and ILF2 before and after NPWT were negatively correlated with changes in white blood cells, the neutrophil percentage, C‑reactive protein, tumor necrosis factor‑α, reactive oxygen species, lipid peroxides, matrix metalloproteinase (MMP) 2 and MMP9 (P<0.05), but positively correlated with the anti‑inflammatory factor, IL‑4 (P<0.01). There was also a positive correlation (P<0.05) with the 4‑week ulcer healing rate. Additionally, the knockdown of FUS and ILF2 expression inhibited the proliferation and migration of HaCaT cells, while increasing cell apoptosis. In vivo, the knockdown of FUS and ILF2 significantly reduced the rate of skin wound healing in diabetic mice. The results of the present study therefore provide new insights into the mechanism by which NPWT promotes DFU wound healing. In conclusion, the RBPs, FUS and ILF2, promoted DFU wound healing by regulating the function of keratinocytes and reducing the inflammatory response and oxidative stress.
Collapse
Affiliation(s)
| | | | - Yanyan Yan
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Die Hu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Murong Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Min Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
6
|
Feng Y, Zhang Z, Yang H, Miao F, Li Y, Zhang M, Cao Y, Li M. The lncRNA TPTEP1 suppresses PI3K/AKT signalling and inhibits ovarian cancer progression by interacting with PTBP1. J Cell Mol Med 2024; 28:e70106. [PMID: 39422584 PMCID: PMC11488117 DOI: 10.1111/jcmm.70106] [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: 12/28/2023] [Revised: 07/15/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The expression of the long noncoding RNA (lncRNA) TPTE pseudogene 1 (TPTEP1) is significantly downregulated in ovarian cancer (OC). However, the function and mechanism of the lncRNA TPTEP1 in OC have not been identified. To investigate the expression of the lncRNA TPTEP1, we analysed a publicly available dataset and 20 pairs of OC and normal ovarian samples tissue from the First Affiliated Hospital of Anhui Medical University. Functional assays were used to determine the role of the lncRNA TPTEP1 in OC progression. Furthermore, Western blot, FISH, RNA pull-down, mass spectrometry and RNA immunoprecipitation approaches were used to determine the mechanism by which the lncRNA TPTEP1 affects OC progression. Animal experiments were used to determine the role of the lncRNA TPTEP1 in ovarian tumorigenicity in vivo. The expression of the lncRNA TPTEP1 in OC tissues was significantly lower than that in normal tissues and low expression of the lncRNA TPTEP1 was significantly correlated with advanced FIGO stage and the presence of malignant ascites in OC patients. In vitro and in vivo, regulation of the expression of the lncRNA TPTEP1 caused changes in OC cell proliferation, migration, invasion and apoptosis. Mechanistically, we found that TPTEP1 directly binds to the polypyrimidine tract-binding protein 1 (PTBP1) protein and inhibits PI3K/AKT signalling. The lncRNA TPTEP1 inhibits PI3K/AKT signalling by directly binding PTBP1, possibly indicating the molecular mechanism underlying its biological function. With further research, these findings may aid in the development of clinically useful strategies for the treatment of OC.
Collapse
Affiliation(s)
- Yifan Feng
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive TractAnhui Medical UniversityHefeiAnhuiChina
- Key Laboratory of Population Health Across Life CycleAnhui Medical UniversityMinistry of Education of the People's Republic of ChinaHefeiAnhuiChina
| | - Zhe Zhang
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive TractAnhui Medical UniversityHefeiAnhuiChina
- Key Laboratory of Population Health Across Life CycleAnhui Medical UniversityMinistry of Education of the People's Republic of ChinaHefeiAnhuiChina
| | - Huijun Yang
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Reproductive Health and GeneticsHefeiAnhuiChina
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial OrgansHefeiAnhuiChina
| | - Fulu Miao
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Reproductive Health and GeneticsHefeiAnhuiChina
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial OrgansHefeiAnhuiChina
| | - Yuyang Li
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Reproductive Health and GeneticsHefeiAnhuiChina
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial OrgansHefeiAnhuiChina
| | - Minmin Zhang
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive TractAnhui Medical UniversityHefeiAnhuiChina
- Key Laboratory of Population Health Across Life CycleAnhui Medical UniversityMinistry of Education of the People's Republic of ChinaHefeiAnhuiChina
| | - Yunxia Cao
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive TractAnhui Medical UniversityHefeiAnhuiChina
- Key Laboratory of Population Health Across Life CycleAnhui Medical UniversityMinistry of Education of the People's Republic of ChinaHefeiAnhuiChina
| | - Min Li
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive TractAnhui Medical UniversityHefeiAnhuiChina
- Key Laboratory of Population Health Across Life CycleAnhui Medical UniversityMinistry of Education of the People's Republic of ChinaHefeiAnhuiChina
| |
Collapse
|
7
|
Lin Y, Zhao W, Pu R, Lv Z, Xie H, Li Y, Zhang Z. Long non‑coding RNAs as diagnostic and prognostic biomarkers for colorectal cancer (Review). Oncol Lett 2024; 28:486. [PMID: 39185489 PMCID: PMC11342420 DOI: 10.3892/ol.2024.14619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the 3rd most common cancer globally and is the 2nd leading cause of cancer-related death. Owing to the lack of specific early symptoms and the limitations of existing early diagnostic methods, most patients with CRC are diagnosed at advanced stages. To overcome these challenges, researchers have increasingly focused on molecular biomarkers, with particular interest in long non-coding RNAs (lncRNAs). These non-protein-coding RNAs, which exceed 200 nucleotides in length, play critical roles in the development and progression of CRC. The stability and detectability of lncRNAs in the circulatory system make them promising candidate biomarkers. The analysis of circulating lncRNAs in peripheral blood represents a potential option for minimally invasive diagnostic tests based on liquid biopsy samples. The present review aimed to evaluate the efficacy of lncRNAs with altered expression levels in peripheral blood as diagnostic markers for CRC. Additionally, the clinical significance of lncRNAs as prognostic markers for this disease were summarized.
Collapse
Affiliation(s)
- Yuning Lin
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Wenzhen Zhao
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Ruonan Pu
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Zhenyi Lv
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Hongyan Xie
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Ying Li
- Department of Ultrasonography, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhongying Zhang
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| |
Collapse
|
8
|
Yang T, Li Y, Zheng Z, Qu P, Shao Z, Wang J, Ding N, Wang W. Comprehensive analysis of lncRNA-mediated ceRNA network in renal cell carcinoma based on GEO database. Medicine (Baltimore) 2024; 103:e39424. [PMID: 39213211 PMCID: PMC11365686 DOI: 10.1097/md.0000000000039424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Renal cell carcinoma (RCC) ranks among the leading causes of cancer-related mortality. Despite extensive research, the precise etiology and progression of RCC remain incompletely elucidated. Long noncoding RNA (lncRNA) has been identified as competitive endogenous RNA (ceRNA) capable of binding to microRNA (miRNA) sites, thereby modulating the expression of messenger RNAs (mRNA) and target genes. This regulatory network is known to exert a pivotal influence on cancer initiation and progression. However, the specific role and functional significance of the lncRNA-miRNA-mRNA ceRNA network in RCC remain poorly understood. The RCC transcriptome data was obtained from the gene expression omnibus database. The identification of differentially expressed long noncoding RNAs (DElncRNAs), differentially expressed miRNAs, and differentially expressed mRNAs (DEmRNAs) between RCC and corresponding paracancer tissues was performed using the "Limma" package in R 4.3.1 software. We employed a weighted gene co-expression network analysis to identify the key DElncRNAs that are most relevant to RCC. Subsequently, we utilized the encyclopedia of RNA interactomes database to predict the interactions between these DElncRNAs and miRNAs, and the miRDB database to predict the interactions between miRNAs and mRNAs. Therefore, key DElncRNAs were obtained to verify the expression of their related genes in the The Cancer Genome Atlas database and to analyze the prognosis. The construction of RCC-specific lncRNA-miRNA-mRNA ceRNA network was carried out using Cytoscape 3.7.0. A total of 286 DElncRNAs, 56 differentially expressed miRNAs, and 2065 DEmRNAs were identified in RCC. Seven key DElncRNAs (GAS6 antisense RNA 1, myocardial infarction associated transcript, long intergenic nonprotein coding RNA 921, MMP25 antisense RNA 1, Chromosome 22 Open Reading Frame 34, MIR34A host gene, MIR4435-2 host gene) were identified using weighted gene co-expression network analysis and encyclopedia of RNA interactomes databases. Subsequently, a network diagram comprising 217 nodes and 463 edges was constructed based on these key DElncRNAs. The functional analysis of DEmRNAs in the ceRNA network was conducted using Kyoto Encyclopedia of Genes and Genomes and gene ontology. We constructed RCC-specific ceRNA networks and identified the crucial lncRNAs associated with RCC using bioinformatics analysis, which will help us further understand the pathogenesis of this disease.
Collapse
Affiliation(s)
- Tianci Yang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Yixuan Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Zhouhang Zheng
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Pei Qu
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Zhiang Shao
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Nan Ding
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Wei Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
9
|
Song J, Xie D, Wei X, Liu B, Yao F, Ye W. A cuproptosis-related lncRNAs signature predicts prognosis and reveals pivotal interactions between immune cells in colon cancer. Heliyon 2024; 10:e34586. [PMID: 39114018 PMCID: PMC11305305 DOI: 10.1016/j.heliyon.2024.e34586] [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: 09/17/2023] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Copper-mediated cell death presents distinct pathways from established apoptosis processes, suggesting alternative therapeutic approaches for colon cancer. Our research aims to develop a predictive framework utilizing long-noncoding RNAs (lncRNAs) related to cuproptosis to predict colon cancer outcomes while examining immune interactions and intercellular signaling. We obtained colon cancer-related human mRNA expression profiles and clinical information from the Cancer Genome Atlas repository. To isolate lncRNAs involved in cuproptosis, we applied Cox proportional hazards modeling alongside the least absolute shrinkage and selection operator technique. We elucidated the underlying mechanisms by examining the tumor mutational burden, the extent of immune cell penetration, and intercellular communication dynamics. Based on the model, drugs were predicted and validated with cytological experiments. A 13 lncRNA-cuproptosis-associated risk model was constructed. Two colon cancer cell lines were used to validate the predicted representative mRNAs with high correlation coefficients with copper-induced cell death. Survival enhancement in the low-risk cohort was evidenced by the trends in Kaplan-Meier survival estimates. Analysis of immune cell infiltration suggested that survival was induced by the increased infiltration of naïve CD4+ T cells and a reduction of M2 macrophages within the low-risk faction. Decreased infiltration of naïve B cells, resting NK cells, and M0 macrophages was significantly associated with better overall survival. Combined single-cell analysis suggested that CCL5-ACKR1, CCL2-ACKR1, and CCL5-CCR1 pathways play key roles in mediating intercellular dialogues among immune constituents within the neoplastic microhabitat. We identified three drugs with a high sensitivity in the high-risk group. In summary, this discovery establishes the possibility of using 13 cuproptosis-associated lncRNAs as a risk model to assess the prognosis, unravel the immune mechanisms and cell communication, and improve treatment options, which may provide a new idea for treating colon cancer.
Collapse
Affiliation(s)
- Jingru Song
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Dong Xie
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xia Wei
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Binbin Liu
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Fang Yao
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| | - Wei Ye
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, Zhejiang, China
| |
Collapse
|
10
|
Lin Y, Zhao W, Lv Z, Xie H, Li Y, Zhang Z. The functions and mechanisms of long non-coding RNA in colorectal cancer. Front Oncol 2024; 14:1419972. [PMID: 39026978 PMCID: PMC11254705 DOI: 10.3389/fonc.2024.1419972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
CRC poses a significant challenge in the global health domain, with a high number of deaths attributed to this disease annually. If CRC is detected only in its advanced stages, the difficulty of treatment increases significantly. Therefore, biomarkers for the early detection of CRC play a crucial role in improving patient outcomes and increasing survival rates. The development of a reliable biomarker for early detection of CRC is particularly important for timely diagnosis and treatment. However, current methods for CRC detection, such as endoscopic examination, blood, and stool tests, have certain limitations and often only detect cases in the late stages. To overcome these constraints, researchers have turned their attention to molecular biomarkers, which are considered a promising approach to improving CRC detection. Non-invasive methods using biomarkers such as mRNA, circulating cell-free DNA, microRNA, LncRNA, and proteins can provide more reliable diagnostic information. These biomarkers can be found in blood, tissue, stool, and volatile organic compounds. Identifying molecular biomarkers with high sensitivity and specificity for the early and safe, economic, and easily measurable detection of CRC remains a significant challenge for researchers.
Collapse
Affiliation(s)
- Yuning Lin
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Wenzhen Zhao
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Zhenyi Lv
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Hongyan Xie
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Ying Li
- Ultrasonography Department, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhongying Zhang
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| |
Collapse
|
11
|
Mahdavi Niyaki Z, Salehzadeh A, Peymani M, Zaefizadeh M. Exploring the Therapeutic Potential of Fe 3O 4@Glu-Oleuropein Nanoparticles in Targeting KRAS Pathway-Regulating lncRNAs in Colorectal Cancer Cells. Biol Trace Elem Res 2024; 202:3073-3085. [PMID: 37792268 DOI: 10.1007/s12011-023-03892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
Cancer, the leading cause of death worldwide, has witnessed significant advancements in treatment through targeted therapies. Among the proto-oncogenes prevalent in human cancers, KRAS stands out, and recent research has focused on long noncoding RNAs (lncRNAs) as regulators of miRNAs targeting the KRAS oncogene. This study specifically explores lncRNAs involved in the KRAS pathway in colorectal cancer (CRC). To investigate this, researchers employed iron oxide nanoparticles coated with glucose and conjugated with Oleuropein (Fe3O4@Glu-Oleuropein NPs) to evaluate their impact on candidate lncRNAs associated with KRAS pathway deregulation. The study utilized TCGA data to identify genes affected by KRAS mutation and lncRNAs linked to KRAS in CRC. Enrichr and MsigDB databases helped identify relevant pathways. Genes with a correlation coefficient above 0.5 and a P-value less than 0.01 with candidate lncRNAs were selected. MTT and flow cytometry assays determined the anti-proliferative and apoptotic effects of Fe3O4@Glu-Oleuropein NPs on CRC cells (SW480) and normal cells (HEK293). The findings showed that increased expression of FEZF1-AS1, GAS6-AS1, and LINC00920 correlated with mutated KRAS, and co-expressed genes were significantly involved in hypoxia, KRAS signaling, DNA repair, and IL-2/STAT5 signaling pathways. Fe3O4@Glu-Oleuropein NPs exhibited higher toxicity toward cancer cells, with IC50 values of 92 μg/ml for SW480 and 281 μg/ml for HEK293. Flow cytometry analysis revealed a substantial increase in necrotic and apoptotic cells when treated with Fe3O4@Glu-Oleuropein, along with down-regulation of GAS6-AS1, LINC00920, and FEZF1-AS1 lncRNAs in treated cells. In conclusion, this study highlights the therapeutic potential of Fe3O4@Glu-Oleuropein on colon cancer cells in vitro. The identification of lncRNAs involved in the KRAS pathway provides insights into the underlying mechanisms and offers avenues for further research in targeted cancer therapies.
Collapse
Affiliation(s)
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
12
|
Zhu Z, Li M, Weng J, Li S, Guo T, Guo Y, Xu Y. LncRNA GAS6-AS1 contributes to 5-fluorouracil resistance in colorectal cancer by facilitating the binding of PCBP1 with MCM3. Cancer Lett 2024; 589:216828. [PMID: 38521199 DOI: 10.1016/j.canlet.2024.216828] [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: 02/05/2024] [Revised: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
5-Fluorouracil (5-FU) resistance has always been a formidable obstacle in the adjuvant treatment of advanced colorectal cancer (CRC). In recent years, long non-coding RNAs have emerged as key regulators in various pathophysiological processes including 5-FU resistance. TRG is a postoperative pathological score of the chemotherapy effectiveness for CRC, of which TRG 0-1 is classified as chemotherapy sensitivity and TRG 3 as chemotherapy resistance. Here, RNA-seq combined with weighted gene correlation network analysis confirmed the close association of GAS6-AS1 with TRG. GAS6-AS1 expression was positively correlated with advanced clinicopathological features and poor prognosis in CRC. GAS6-AS1 increased the 50% inhibiting concentration of 5-FU, enhanced cell proliferation and accelerated G1/S transition, both with and without 5-FU, both in vitro and in vivo. Mechanistically, GAS6-AS1 enhanced the stability of MCM3 mRNA by recruiting PCBP1, consequently increasing MCM3 expression. Furthermore, PCBP1 and MCM3 counteracted the effects of GAS6-AS1 on 5-FU resistance. Notably, the PDX model indicated that combining chemotherapeutic drugs with GAS6-AS1 knockdown yielded superior outcomes in vivo. Together, our findings elucidate that GAS6-AS1 directly binds to PCBP1, enhancing MCM3 expression and thereby promoting 5-FU resistance. GAS6-AS1 may serve as a robust biomarker and potential therapeutic target for combination therapy in CRC.
Collapse
Affiliation(s)
- Zhonglin Zhu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Minghan Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Junyong Weng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Shanbao Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Tianan Guo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Yang Guo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
13
|
Fan S, Zhou L, Zhang W, Wang D, Tang D. Role of imbalanced gut microbiota in promoting CRC metastasis: from theory to clinical application. Cell Commun Signal 2024; 22:232. [PMID: 38637851 PMCID: PMC11025274 DOI: 10.1186/s12964-024-01615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
Metastasis poses a major challenge in colorectal cancer (CRC) treatment and remains a primary cause of mortality among patients with CRC. Recent investigations have elucidated the involvement of disrupted gut microbiota homeostasis in various facets of CRC metastasis, exerting a pivotal influence in shaping the metastatic microenvironment, triggering epithelial-mesenchymal transition (EMT), and so on. Moreover, therapeutic interventions targeting the gut microbiota demonstrate promise in enhancing the efficacy of conventional treatments for metastatic CRC (mCRC), presenting novel avenues for mCRC clinical management. Grounded in the "seed and soil" hypothesis, this review consolidates insights into the mechanisms by which imbalanced gut microbiota promotes mCRC and highlights recent strides in leveraging gut microbiota modulation for the clinical prevention and treatment of mCRC. Emphasis is placed on the considerable potential of manipulating gut microbiota within clinical settings for managing mCRC.
Collapse
Affiliation(s)
- Shiying Fan
- Clinical Medical College, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Lujia Zhou
- Clinical Medical College, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, 400030, Chongqing, P. R. China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, 225000, Yangzhou, P. R. China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, 225000, Yangzhou, P. R. China.
| |
Collapse
|
14
|
Xie Y, Zhang Z, Lai D, Liang J, Zhao Z, Lu W, Ke J, Lin W, He H. Lymph node metastasis-related lncRNA GAS6-AS1 facilitates the progression of esophageal squamous cell carcinoma. J Gastrointest Oncol 2023; 14:2293-2308. [PMID: 38196547 PMCID: PMC10772685 DOI: 10.21037/jgo-23-798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024] Open
Abstract
Background Lymph node metastasis is the main type of metastasis in esophageal squamous cell carcinoma (ESCC), especially when the primary tumor invasion depth reaches above the adventitia layer (T3 stage), the incidence of lymph node metastasis increases sharply. Abnormal expression of long non-coding RNAs (lncRNAs) has been confirmed in ESCC, but there are still many unknown connections between lncRNAs and lymph node metastasis. Methods We used transcriptome sequencing (RNA-seq) to analyze 10 pairs of ESCC tissues with primary tumor stage T3 and their paired normal epithelium. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to further verify the sequencing results, and survival curve analysis, logistic regression analysis, and receiver operating characteristic (ROC) curve analysis were used to investigate its clinical application value. We investigated the growth and metastasis effects of lncRNA GAS6-AS1 on ESCC cell lines TE-1 and KYSE410 in vitro and in vivo. Other functional experiments included cell apoptosis and cell cycle experiments. Results Based on our RNA-seq data, lncRNA GAS6-AS1 is highly expressed in ESCC tissues, especially in cancer tissues with lymph node metastasis. The qRT-PCR experiment analysis showed that high expression of GAS6-AS1 was related to poor tumor differentiation and tumor stage. Logistic regression analysis showed that it was an independent risk factor for lymph node metastasis, and ROC analysis validated that it could predict lymph node metastasis. Further survival analysis suggested that high expression of GAS6-AS1 was associated with patients' poor prognosis. In vitro experiments, knocking down GAS6-AS1 inhibited the growth and metastasis of ESCC cells and inhibited tumor growth in vivo. In addition, knocking down GAS6-AS1 can inhibit cell cycle and promote cell apoptosis. Conclusions Our results revealed that lncRNA GAS6-AS1 obtained from RNA-seq can be used as an independent risk factor for ESCC lymph node metastasis and an effective biomarker to predict, and that it was related to the growth and metastasis of ESCC. It may represent a new biomarker to aid in the assessment of the lymph node metastasis of ESCC.
Collapse
Affiliation(s)
- Yujie Xie
- Department of Thoracic Surgery, Gaozhou People’s Hospital, Maoming, China
| | - Zhanfei Zhang
- Department of Cardiothoracic Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Dongmei Lai
- Department of Oncology, Gaozhou People’s Hospital, Maoming, China
| | - Jin Liang
- Department of Cardiothoracic Surgery, Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Zhengang Zhao
- Department of Cardiothoracic Surgery, Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Weicheng Lu
- Department of Thoracic Surgery, Maoming People’s Hospital, Maoming, China
| | - Junli Ke
- Department of Cardiothoracic Surgery, Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Wanli Lin
- Department of Thoracic Surgery, Gaozhou People’s Hospital, Maoming, China
| | - Haiquan He
- Department of Thoracic Surgery, Gaozhou People’s Hospital, Maoming, China
| |
Collapse
|
15
|
Wu J, Lu X, Yu J, Li P, Yu X. LINC02253 promote the malignant phenotype of Colon adenocarcinoma cells by up-regulating WWP1-mediated SMAD3 ubiquitination. Mol Cell Probes 2023; 72:101928. [PMID: 37597669 DOI: 10.1016/j.mcp.2023.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
OBJECTIVES Colon adenocarcinoma (COAD) represents a type of common malignant tumor originating in the digestive tract. Long non-coding RNAs (lncRNAs) have been identified to engage in regulating the initiation and development of COAD. LncRNA LINC02253 has been reported abnormal expressed in COAD, but the underlying mechanism has not been discussed so far. This study aimed to determine the role and the molecular biology mechanism of LINC02253 in COAD progression and unearthed its specific molecular mechanism. MATERIALS AND RESULTS RT-qPCR and Western blot assays were conducted to detect gene expression. Function assays were performed to evaluate the effect of gene expression on COAD cell phenotype. Mechanism analyses were done to verify the association among genes after bioinformatics analysis. The obtained data revealed that LINC02253 demonstrated a high expression in COAD tissues and cells. This gene served as an oncogene, permitting to stimulate proliferation and suppress apoptosis of COAD cells. Mechanically, it was found that LINC02253 recruited FUS to stabilize WWP1 mRNA and WWP1 could mediate SMAD3 ubiquitination, thereby promoting the malignant phenotype formation of COAD cells. CONCLUSIONS LINC02253 was uncovered to exert an oncogenic role, enhancing the proliferation of COAD cells and repressing the cell apoptosis by recruiting FUS and encouraging WWP1-mediated SMAD3 ubiquitination.
Collapse
Affiliation(s)
- Jinfeng Wu
- Department of Gastroenterology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, China
| | - Xianhong Lu
- Department of Gastroenterology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, China
| | - Jinzhong Yu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine, Shanghai, 200120, China
| | - Pan Li
- Institute of Ultrasound Imaging Engineering, Chongqing Medical University, Chongqing, 400000, China
| | - Xiqiu Yu
- Department of Gastroenterology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, China.
| |
Collapse
|
16
|
Zhang Y, Zhao L, Bi Y, Zhao J, Gao C, Si X, Dai H, Asmamaw MD, Zhang Q, Chen W, Liu H. The role of lncRNAs and exosomal lncRNAs in cancer metastasis. Biomed Pharmacother 2023; 165:115207. [PMID: 37499455 DOI: 10.1016/j.biopha.2023.115207] [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: 06/09/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
Tumor metastasis is the main reason for cancer-related death, but there is still a lack of effective therapeutic to inhibit tumor metastasis. Therefore, the discovery and study of new tumor metastasis regulators is a prominent measure for cancer diagnosis and treatment. Long non-coding RNA (lncRNA) is a type of non-coding RNAs over 200 bp in length. It has been shown that the abnormally expressed lncRNAs promote tumor metastasis by participating in the epithelial-to-mesenchymal transition (EMT) process, altering the metastatic tumor microenvironment, or changing the extracellular matrix. It is,thus, critical to explore the regulation of lncRNAs expression in cells and the molecular mechanism of lncRNA-mediated cancer metastasis. Simultaneously, it has been shown that lncRNA is one kind of the main components of exosomes, which protects lncRNAs from being rapidly degraded. Meanwhile, the components of exosomes are parent-specific, making exosomal lncRNAs to be potential tumor metastasis markers and therapeutic targets. In view of this, we also summarized the aberrant enrichment of lncRNAs in exosomes and their role in metastatic cancer. The aberrant lncRNAs and exosomal lncRNAs gradually become biomarkers and therapeutic targets for tumor metastatic, and the potential of lncRNAs in therapeutics are studied here. Besides, the lncRNA-related databases, which could greatly facilitate in the study of lncRNAs and exosomal lncRNAs in metastatic of cancer are included in this review.
Collapse
Affiliation(s)
- Yutong Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China; The People's Hospital of Zhang Dian District, Zibo, China
| | - Lijuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Science, Zhengzhou University, Zhengzhou China
| | - Yaping Bi
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Jinyuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Chao Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Xiaojie Si
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Honglin Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Moges Dessale Asmamaw
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China
| | - Qiurong Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China.
| | - Wenchao Chen
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital; Zhengzhou University People's Hospital; Henan University People's Hospital, Zhengzhou China.
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou China.
| |
Collapse
|
17
|
Mirzaei S, Ranjbar B, Tackallou SH. Molecular profile of non-coding RNA-mediated glycolysis control in human cancers. Pathol Res Pract 2023; 248:154708. [PMID: 37536019 DOI: 10.1016/j.prp.2023.154708] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023]
Abstract
The glycolysis is a common characteristic of cancer and it is responsible for providing enough energy to ensure growth. The glycolysis suppression is beneficial in tumor growth reduction. The stimulation/inhibition of glycolysis in cancer is tightly regulated by ncRNAs. The regulation of glycolysis by ncRNAs can influence proliferation and therapy response of tumor. The miRNAs are capable of inactivating enzymes responsible for glycolysis and suppressing signaling networks resulting in glycolysis induction. By regulation of glycolysis, miRNAs can affect therapy response. The lncRNAs and circRNAs follow a same pathway and by targeting glycolysis, they affect progression and therapy response of tumor. Noteworthy, lncRNAs and circRNAs sponge miRNAs in glycolysis mechanism control in tumor cells. Furthermore, ncRNA-mediated regulation of glycolysis mechanism can influence metastasis to organs of body. The ncRNAs regulating glycolysis are reliable biomarkers in cancer patients and more importantly, exosomal ncRNAs due to their presence in body fluids, are minimally-invasive biomarkers.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran
| | | |
Collapse
|
18
|
Hashemi M, Gholami S, Raesi R, Sarhangi S, Mahmoodieh B, Koohpar ZK, Goharrizi MASB, Behroozaghdam M, Entezari M, Salimimoghadam S, Zha W, Rashidi M, Abdi S, Taheriazam A, Nabavi N. Biological and therapeutic viewpoints towards role of miR-218 in human cancers: Revisiting molecular interactions and future clinical translations. Cell Signal 2023:110786. [PMID: 37380085 DOI: 10.1016/j.cellsig.2023.110786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 06/30/2023]
Abstract
Understanding the exact pathogenesis of cancer is difficult due to heterogenous nature of tumor cells and multiple factors that cause its initiation and development. Treatment of cancer is mainly based on surgical resection, chemotherapy, radiotherapy and their combination, while gene therapy has been emerged as a new kind of therapy for cancer. Post-transcriptional regulation of genes has been of interest in recent years and among various types of epigenetic factors that can modulate gene expression, short non-coding RNAs known as microRNAs (miRNAs) have obtained much attention. The stability of mRNA decreases by miRNAs to repress gene expression. miRNAs can regulate tumor malignancy and biological behavior of cancer cells and understanding their function in tumorigenesis can pave the way towards developing new therapeutics in future. One of the new emerging miRNAs in cancer therapy is miR-218 that increasing evidence highlights its anti-cancer activity, while a few studies demonstrate its oncogenic function. The miR-218 transfection is promising in reducing progression of tumor cells. miR-218 shows interactions with molecular mechanisms including apoptosis, autophagy, glycolysis and EMT, and the interaction is different. miR-218 induces apoptosis, while it suppresses glycolysis, cytoprotective autophagy and EMT. Low expression of miR-218 can result in development of chemoresistance and radio-resistance in tumor cells and direct targeting of miR-218 as a key player is promising in cancer therapy. LncRNAs and circRNAs are nonprotein coding transcripts that can regulate miR-218 expression in human cancers. Moreover, low expression level of miR-218 can be observed in human cancers such as brain, gastrointestinal and urological cancers that mediate poor prognosis and low survival rate.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Sadaf Gholami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sareh Sarhangi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | | | - Mitra Behroozaghdam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Wenliang Zha
- Second Affiliated Hospital, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Soheila Abdi
- Department of Physics, Safadasht Branch, Islamic Azad university, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada.
| |
Collapse
|
19
|
Duan M, Liu Y, Zhao D, Li H, Zhang G, Liu H, Wang Y, Fan Y, Huang L, Zhou F. Gender-specific dysregulations of nondifferentially expressed biomarkers of metastatic colon cancer. Comput Biol Chem 2023; 104:107858. [PMID: 37058814 DOI: 10.1016/j.compbiolchem.2023.107858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/12/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Colon cancer is a common cancer type in both sexes and its mortality rate increases at the metastatic stage. Most studies exclude nondifferentially expressed genes from biomarker analysis of metastatic colon cancers. The motivation of this study is to find the latent associations of the nondifferentially expressed genes with metastatic colon cancers and to evaluate the gender specificity of such associations. This study formulates the expression level prediction of a gene as a regression model trained for primary colon cancers. The difference between a gene's predicted and original expression levels in a testing sample is defined as its mqTrans value (model-based quantitative measure of transcription regulation), which quantitatively measures the change of the gene's transcription regulation in this testing sample. We use the mqTrans analysis to detect the messenger RNA (mRNA) genes with nondifferential expression on their original expression levels but differentially expressed mqTrans values between primary and metastatic colon cancers. These genes are referred to as dark biomarkers of metastatic colon cancer. All dark biomarker genes were verified by two transcriptome profiling technologies, RNA-seq and microarray. The mqTrans analysis of a mixed cohort of both sexes could not recover gender-specific dark biomarkers. Most dark biomarkers overlap with long non-coding RNAs (lncRNAs), and these lncRNAs might have contributed their transcripts to calculating the dark biomarkers' expression levels. Therefore, mqTrans analysis serves as a complementary approach to identify dark biomarkers generally ignored by conventional studies, and it is essential to separate the female and male samples into two analysis experiments. The dataset and mqTrans analysis code are available at https://figshare.com/articles/dataset/22250536.
Collapse
Affiliation(s)
- Meiyu Duan
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Yaqing Liu
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Dong Zhao
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Haijun Li
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Gongyou Zhang
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Hongmei Liu
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China; Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yueying Wang
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Yusi Fan
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China; College of Software, Jilin University, Changchun, Jilin 130012, China.
| | - Lan Huang
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Fengfeng Zhou
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China.
| |
Collapse
|
20
|
Zhan T, Cheng X, Zhu Q, Han Z, Zhu K, Tan J, Liu M, Chen W, Chen X, Chen X, Tian X, Huang X. LncRNA LOC105369504 inhibits tumor proliferation and metastasis in colorectal cancer by regulating PSPC1. Cell Death Discov 2023; 9:89. [PMID: 36894530 PMCID: PMC9998613 DOI: 10.1038/s41420-023-01384-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
There is growing evidence that long non-coding RNAs (lncRNAs) are significant contributors to the epigenetic mechanisms implicated in the emergence, progression and metastasis of the colorectal cancer (CRC), but many remain underexplored. A novel lncRNA LOC105369504, was identified to be a potential functional lncRNA by microarray analysis. In CRC, the expression of LOC105369504 was markedly decreased and resulted in distinct variations in proliferation, invasion, migration and epithelial-mesenchymal transition (EMT) in vivo and in vitro. This study showed that LOC105369504 bound to the protein of paraspeckles compound 1 (PSPC1) directly and regulated its stability using the ubiquitin-proteasome pathway in CRC cells. The suppression of CRC by LOC105369504 could be reversed through PSPC1 overexpression.This study showed that in CRC, LOC105369504 was under-regulated and as a novel lncRNA, LOC105369504 exerted tumor suppressive activity to suppress the proliferation together with metastasis in CRC cells through the regulation of PSPC1. These results offer new perspectives on the lncRNA effect on the progression of CRC.
Collapse
Affiliation(s)
- Ting Zhan
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xueting Cheng
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Qingxi Zhu
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Zheng Han
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Kejing Zhu
- Department of Pharmacy, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Jie Tan
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Men Liu
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Wei Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xiaoli Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xiaohong Chen
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China
| | - Xia Tian
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China.
| | - Xiaodong Huang
- Department of Gastroenterology, WuHan Third Hospital (Tongren hospital of WuHan University), 430060, Wuhan, China.
| |
Collapse
|
21
|
Liu J, Zhan W, Chen G, Yan S, Chen W, Li R. SP1-induced PROX1-AS1 contributes to tumor progression by regulating miR-326/FBXL20 axis in colorectal cancer. Cell Signal 2023; 101:110503. [PMID: 36374774 DOI: 10.1016/j.cellsig.2022.110503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) play pivotal roles in cancers by regulating tumorigenesis and metastasis. LncRNA PROX1-AS1 has been reported to be involved in tumor progression, however, its role in colorectal cancer (CRC) remains ambiguous. Based on TCGA and GTEx databases, we found that the expression of PROX1-AS1 was upregulated in CRC tissues and cells. Bioinformatics analysis revealed that high PROX1-AS1 expression was associated with poor overall survival. Functionally, PROX1-AS1 knockdown suppressed CRC cell proliferation, migration, and invasion in vitro, as well as inhibiting tumor growth in vivo. Mechanistically, PROX1-AS1 was identified to act as a miR-326 sponge by luciferase reporter and RIP assay. Meanwhile, we found that the transcription factor SP1 activated PROX1-AS1/miR-32/FBXL20 axis, thereby promoting CRC progression. Our data demonstrated that PROX1-AS1 served as a promising prognostic biomarker for CRC, and the potential mechanism was unraveled.
Collapse
Affiliation(s)
- Jing Liu
- Department of Imaging, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Wei Zhan
- Department of General Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Gang Chen
- Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Shunkang Yan
- Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Wen Chen
- Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province 550004, China
| | - Rui Li
- Department of Traditional Chinese Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province 550002, China.
| |
Collapse
|
22
|
Fan S, Xing J, Jiang Z, Zhang Z, Zhang H, Wang D, Tang D. Effects of Long Non-Coding RNAs Induced by the Gut Microbiome on Regulating the Development of Colorectal Cancer. Cancers (Basel) 2022; 14:5813. [PMID: 36497293 PMCID: PMC9735521 DOI: 10.3390/cancers14235813] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/29/2022] Open
Abstract
Although an imbalanced gut microbiome is closely associated with colorectal cancer (CRC), how the gut microbiome affects CRC is not known. Long non-coding RNAs (lncRNAs) can affect important cellular functions such as cell division, proliferation, and apoptosis. The abnormal expression of lncRNAs can promote CRC cell growth, proliferation, and metastasis, mediating the effects of the gut microbiome on CRC. Generally, the gut microbiome regulates the lncRNAs expression, which subsequently impacts the host transcriptome to change the expression of downstream target molecules, ultimately resulting in the development and progression of CRC. We focused on the important role of the microbiome in CRC and their effects on CRC-related lncRNAs. We also reviewed the impact of the two main pathogenic bacteria, Fusobacterium nucleatum and enterotoxigenic Bacteroides fragilis, and metabolites of the gut microbiome, butyrate, and lipopolysaccharide, on lncRNAs. Finally, available therapies that target the gut microbiome and lncRNAs to prevent and treat CRC were proposed.
Collapse
Affiliation(s)
- Shiying Fan
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
23
|
Hashemi M, Hajimazdarany S, Mohan CD, Mohammadi M, Rezaei S, Olyaee Y, Goldoost Y, Ghorbani A, Mirmazloomi SR, Gholinia N, Kakavand A, Salimimoghadam S, Ertas YN, Rangappa KS, Taheriazam A, Entezari M. Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance. Pharmacol Res 2022; 186:106535. [DOI: 10.1016/j.phrs.2022.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
|