1
|
Jia Y, Jia R, Chen Y, Lin X, Aishan N, li H, Wang L, Zhang X, Ruan J. The role of RNA binding proteins in cancer biology: A focus on FMRP. Genes Dis 2025; 12:101493. [PMID: 40271197 PMCID: PMC12017997 DOI: 10.1016/j.gendis.2024.101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 04/25/2025] Open
Abstract
RNA-binding proteins (RBPs) act as crucial regulators of gene expression within cells, exerting precise control over processes such as RNA splicing, transport, localization, stability, and translation through their specific binding to RNA molecules. The diversity and complexity of RBPs are particularly significant in cancer biology, as they directly impact a multitude of RNA metabolic events closely associated with tumor initiation and progression. The fragile X mental retardation protein (FMRP), as a member of the RBP family, is central to the neurodevelopmental disorder fragile X syndrome and increasingly recognized in the modulation of cancer biology through its influence on RNA metabolism. The protein's versatility, stemming from its diverse RNA-binding domains, enables it to govern a wide array of transcript processing events. Modifications in FMRP's expression or localization have been associated with the regulation of mRNAs linked to various processes pertinent to cancer, including tumor proliferation, metastasis, epithelial-mesenchymal transition, cellular senescence, chemotherapy/radiotherapy resistance, and immunotherapy evasion. In this review, we emphasize recent findings and analyses that suggest contrasting functions of this protein family in tumorigenesis. Our knowledge of the proteins that are regulated by FMRP is rapidly growing, and this has led to the identification of multiple targets for therapeutic intervention of cancer, some of which have already moved into clinical trials or clinical practice.
Collapse
Affiliation(s)
- Yunlu Jia
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ruyin Jia
- The Second School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xuanyi Lin
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Nadire Aishan
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Han li
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
2
|
Zhuang Y, Lu F, Wang X, Yao J, Wan Y, Qin S, Cao X, Sheng J. LoC-SERS detection platform based on targeted signal anchoring mechanism, high-sensitivity detection of protein biomarkers in precancerous lesions of gastric cancer. Talanta 2025; 294:128190. [PMID: 40280078 DOI: 10.1016/j.talanta.2025.128190] [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: 02/25/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
In this study, a novel lab-on-a-chip surface-enhanced Raman scattering (LoC-SERS) analysis platform was developed based on chip laboratory for monitoring precancerous lesions of GC. The platform integrated graphene-like Au nano-hexagonal arrays (Au NHAs) as a high-efficiency plasma substrate with the highly integrated microarray chip as an ideal system for sensitive recognition and rapid quantification of matrix metalloproteinase-9 (MMP-9) and interleukin-6 (IL-6) based on the targeted signal anchoring mechanism. Au NHAs were surface modified to form a double stranded structure with aptamer chains and single stranded DNA1 (ssDNA1), Au nanobipyramid with Ag shell (Au NBP@Ag) surface modified single stranded DNA2 (ssDNA2) and Raman signaling molecules. When the target is present, the aptamer specifically captures the target protein and detaches from the Au NHAs surface, exposing ssDNA1 and ssDNA2 to pair and bind. This resulted in the Au NBP@Ag probe carrying the signaling molecule anchoring to the surface of Au NHAs, thereby enhancing the SERS signal. The platform demonstrated excellent detection efficiency and could complete detection tasks within 20 min. The platform can detect MMP-9 and IL-6 at levels down to the pg/mL order of magnitude, demonstrating excellent detection sensitivity. In addition, the platform has the capability of simultaneous dual-target detection, demonstrating good clinical applicability and accuracy when testing real serum samples. Overall, this platform provides an efficient solution for early diagnosis and progression monitoring of precancerous lesions of GC, further providing a scientific basis for clinical decision-making and intervention.
Collapse
Affiliation(s)
- Yanwen Zhuang
- Institute of Translational Medicine, Medical College, Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, Jiangsu Province, China
| | - Feng Lu
- Affiliated Huishan Hospital of Medical College, Yangzhou University, Wuxi Huishan District People's Hospital, Wuxi, 214187, Jiangsu Province, China
| | - Xiaoyong Wang
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, Jiangsu Province, China
| | - Jie Yao
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, Jiangsu Province, China
| | - Yong Wan
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, Jiangsu Province, China
| | - Shichen Qin
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, Jiangsu Province, China
| | - Xiaowei Cao
- Institute of Translational Medicine, Medical College, Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225001, Jiangsu Province, China
| | - Jinxin Sheng
- Department of General Surgery, Nantong Haimen People's Hospital, Nantong, 226100, Jiangsu Province, China.
| |
Collapse
|
3
|
Katoozian F, Abedi Kichi Z, Sharifi R, Shirvani-Farsani Z. The Expression Analysis of Long Non-coding RNAs Related to Wnt/β-Catenin Signaling in Pancreatic Cancer Patients. Biochem Genet 2025; 63:1605-1619. [PMID: 38594570 DOI: 10.1007/s10528-024-10779-5] [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: 01/22/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024]
Abstract
Background The oncogenic Wnt/β-catenin signaling plays a critical role in carcinogenesis, prognosis, and resistance to therapy. Pancreatic cancer (PC) has high mortality because of its poor prognosis. Several studies have suggested that lncRNAs are directly involved in the development and progression of PC as well as in Wnt/β-catenin signaling. In this study, we investigated and compared the expression of Wnt/β-catenin signaling-related ZFAS1 and HCG11 lncRNAs, and their targets, CTNNB1 and IGF2BP1 genes in the blood of patients with PC and healthy individuals. A total of 47 PC patients and 50 healthy individuals participated in this study. RNA was extracted from the peripheral blood samples of participants, and cDNA was synthesized. The expression level of the selected genes was quantified by real-time PCR. The expression of HCG11 lncRNA and CTNNB1 genes in patients with PC was significantly upregulated compared to healthy individuals, and the expression of the ZFAS1 lncRNA was significantly downregulated. According to the analysis of the ROC curve, the diagnostic powers of ZFAS1 and CTNNB1 in PC were 0.67 and 0.69, respectively. Altogether, the present study suggests a role for ZFAS1 and HCG11 lncRNAs and CTNNB1 and IGF2BP1 in the pathogenesis of pancreatic cancer. Moreover, the peripheral expression of these lncRNAs may be useful as potential biomarkers for PC.
Collapse
Affiliation(s)
- Fatemeh Katoozian
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Abedi Kichi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany
| | - Roya Sharifi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
4
|
Zeng X, Zhang Y, Shapaer T, Abudoukelimu A, Zhao Z, Ma B. IGF2BP3 prefers to regulate alternative splicing of genes associated with the progression of gastric cancer in AGS cells. Discov Oncol 2025; 16:235. [PMID: 39998701 PMCID: PMC11861459 DOI: 10.1007/s12672-025-01880-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Gastric cancer (GC) has become the fifth largest malignant tumor in the world, with a mortality rate ranking fourth. IGF2BP3, as a multifunctional RNA binding protein, is involved in regulating alternative splicing (AS) and m6A modification, and plays a carcinogenic role in the development of gastric cancer, while little is known about the impact of IGF2BP3 on alternative splicing in gastric cancer cells and the underlying mechanism. In this study, we overexpressed IGF2BP3 (IGF2BP3-OE) in gastric cancer AGS cells and obtained transcriptome sequencing data (RNA-seq) to explore the effects of IGF2BP3 on gene expression and AS. The RNA binding profile of IGF2BP3 was utilized to identify how IGF2BP3 binds to and modulate expression and AS patterns of target genes. IGF2BP3-OE resulted in 479 differentially expressed genes (DEGs), majority of which were downregulated. We selected 20 DEGs and validated their expression pattern by RT-qPCR experiment, including ZFAS1, DUSP9, GPX3, IDH2, and H19 that were associated with GC development. More importantly, IGF2BP3-OE significantly modulated AS pattern of thousands of genes, which were enriched in mRNA splicing, cell proliferation, and translation pathways. By integrating the RNA binding profile of IGF2BP3, we found IGF2BP3 binding preferred to modulate the splicing pattern of bound genes, and the overlapped genes were also enriched in mRNA splicing pathways. We validated the AS pattern changes of S100A4 and PLK3 by RT-qPCR. IGF2BP3 probably modulate GC development by regulating AS profile in GC cells. In summary, we explored the dysregulated transcriptome profile that IGF2BP3 affects gene expression and alternative splicing by binding to mRNA, and thus plays a role in the development of GC cells. The IGF2BP3 and identified targets has potential value for GC treatment in future.
Collapse
Affiliation(s)
- Xiangyue Zeng
- Gastroenterology Department II, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China
| | - Yu Zhang
- Gastroenterology Department II, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China
| | - Tiannake Shapaer
- Gastroenterology Department II, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China
| | - Abulajiang Abudoukelimu
- Gastroenterology Department II, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China
| | - Zeliang Zhao
- Gastroenterology Department II, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China
| | - Binlin Ma
- Breast and Thyroid Surgery Department, The Affiliated Tumor Hospital of Xinjiang Medical University, Ürümqi, 830011, China.
| |
Collapse
|
5
|
Liu X, Ma Z, Zhang X, Li S, An J, Luo Z. Research Progress of Long Non-coding RNA-ZFAS1 in Malignant Tumors. Cell Biochem Biophys 2024; 82:3145-3156. [PMID: 39060915 DOI: 10.1007/s12013-024-01441-3] [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] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Long non-coding RNAs (lncRNAs), although incapable of encoding proteins, play crucial roles in multiple layers of gene expression regulation, epigenetic modifications, and post-transcriptional regulation. Zinc finger antisense 1 (ZFAS1), a lncRNA located in the 20q13 region of the human genome, exhibits dual functions as an oncogene or tumor suppressor in various human malignancies. ZFAS1 plays a crucial role in cancer progression, metastasis, invasion, apoptosis, cell cycle regulation, and drug resistance through complex molecular mechanisms. Additionally, ZFAS1 has a long half-life of over 16 h, demonstrating exceptional stability, and making it a potential biomarker. This review integrates recent studies on the role and molecular mechanisms of ZFAS1 in malignancies and summarizes its clinical significance. By summarizing the role of ZFAS1 in cancer, we aim to highlight its potential as an anti-cancer biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Zhong Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Xianxu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Shicheng Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu Provincial, Lanzhou, 730030, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Zhiqiang Luo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
6
|
Cotino-Nájera S, García-Villa E, Cruz-Rosales S, Gariglio P, Díaz-Chávez J. Resveratrol inhibits Lin28A expression and induces its degradation via the proteasomal pathway in NCCIT cells. Oncol Lett 2024; 28:577. [PMID: 39397804 PMCID: PMC11467847 DOI: 10.3892/ol.2024.14710] [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: 04/29/2024] [Accepted: 08/14/2024] [Indexed: 10/15/2024] Open
Abstract
Lin28A is an oncoprotein overexpressed in several cancer types such as testicular, ovarian, colon, breast and lung cancers. As a pluripotency factor that promotes tumorigenesis, Lin28A is associated with more undifferentiated and aggressive tumors phenotypes. Moreover, Lin28A is a highly stable protein that is difficult to downregulate. The compound resveratrol (RSV) has anticancer effects. The present study aimed to elucidate the mechanisms underlying the downregulation of Lin28A protein expression by RSV in the NCCIT cell line. NCCIT cells were treated with different concentrations of RSV to investigate its effects on Lin28A expression. The mRNA expression levels of Lin28A and ubiquitin-specific protease 28 (USP28) were assessed using reverse transcription-quantitative PCR. Western blot analysis was employed to evaluate the protein levels of Lin28A, USP28 and phosphorylated Lin28A. In addition, in some experiments, cells were treated with a MAPK/ERK pathway inhibitor, and other experiments involved transfecting cells with small interfering RNAs targeting USP28. The results demonstrated that RSV significantly reduced Lin28A expression by destabilizing the protein; this effect was mediated by the ability of RSV to suppress the expression of USP28, a deubiquitinase that normally protects Lin28A from ubiquitination and degradation. Additionally, RSV inhibited phosphorylation of Lin28A via the MAPK/ERK pathway; this phosphorylation event has previously been shown to enhance the stability of Lin28A by increasing its half-life. This resulted in Lin28A degradation through the proteasomal pathway in NCCIT cells. The results provide further evidence of the anticancer activity of RSV, and identified Lin28A and USP28 as promising therapeutic targets. As a stable oncoprotein, downregulating Lin28A expression is challenging. However, the present study demonstrated that RSV can overcome this hurdle by inhibiting USP28 expression and MAPK/ERK signaling to promote Lin28A degradation. Furthermore, elucidating these mechanisms provides avenues for developing targeted cancer therapies.
Collapse
Affiliation(s)
- Sandra Cotino-Nájera
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of The National Polytechnic Institute, Mexico City 07360, Mexico
| | - Enrique García-Villa
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of The National Polytechnic Institute, Mexico City 07360, Mexico
| | - Samantha Cruz-Rosales
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of The National Polytechnic Institute, Mexico City 07360, Mexico
| | - Patricio Gariglio
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of The National Polytechnic Institute, Mexico City 07360, Mexico
| | - José Díaz-Chávez
- Biomedical Cancer Research Unit, Biomedical Research Institute, National Autonomous University of Mexico/National Cancer Institute, Mexico City 14080, Mexico
- Department of Cellular Biology, Faculty of Sciences, National Autonomous University of Mexico, Mexico City 04510, Mexico
- School of Medicine and Health Sciences, Monterrey Institute of Technology, Mexico City 14380, Mexico
| |
Collapse
|
7
|
Cotino‐Nájera S, García‐Villa E, Cruz‐Rosales S, Gariglio P, Díaz‐Chávez J. The role of Lin28A and Lin28B in cancer beyond Let-7. FEBS Lett 2024; 598:2963-2979. [PMID: 39152528 PMCID: PMC11665955 DOI: 10.1002/1873-3468.15004] [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: 05/11/2024] [Revised: 07/02/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024]
Abstract
Lin28A and Lin28B are paralogous RNA-binding proteins that play fundamental roles in development and cancer by regulating the microRNA family of tumor suppressor Let-7. Although Lin28A and Lin28B share some functional similarities with Let-7 inhibitors, they also have distinct expression patterns and biological functions. Increasing evidence indicates that Lin28A and Lin28B differentially impact cancer stem cell properties, epithelial-mesenchymal transition, metabolic reprogramming, and other hallmarks of cancer. Therefore, it is important to understand the overexpression of Lin28A and Lin28B paralogs in specific cancer contexts. In this review, we summarize the main similarities and differences between Lin28A and Lin28B, their implications in different cellular processes, and their role in different types of cancer. In addition, we provide evidence of other specific targets of each lin28 paralog, as well as the lncRNAs and miRNAs that promote or inhibit its expression, and how this impacts cancer development and progression.
Collapse
Affiliation(s)
- Sandra Cotino‐Nájera
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Enrique García‐Villa
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Samantha Cruz‐Rosales
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Patricio Gariglio
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - José Díaz‐Chávez
- Departamento de Biología Celular, Facultad de CienciasUNAMMexico CityMexico
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones BiomédicasUNAM/Instituto Nacional de CancerologíaMexico CityMexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la SaludMexico
| |
Collapse
|
8
|
Liang H, Li H, Xia N, Chen J, Gao L, Liu H, Lyu P, Guo X, Yang Z. Circulating long noncoding RNA, Zfpm2-As1, and XIST based on medical data analysis are potential plasma biomarkers for gastric cancer diagnosis. Technol Health Care 2024; 32:4919-4928. [PMID: 38820035 PMCID: PMC11612959 DOI: 10.3233/thc-232033] [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: 12/27/2023] [Accepted: 02/19/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) participate in diseases, especially tumorigenesis, including gastric cancer (GC). Although lncRNAs in GC tissues have been extensively studied in previous research, the possible significance of circulating lncRNAs in diagnosing GC is still unknown. OBJECTIVE The present work investigated lncRNAs ZFPM2-AS1 and XIST with high expression in GC tissues proved as potential plasma biomarkers from 20 early GC cases, 100 GC cases, and 90 normal subjects. METHODS The possible correlation between ZFPM2-AS1 and XIST expression levels was analyzed with general characteristics and clinicopathological features. The performance in diagnosis was assessed according to receiver operating characteristic (ROC) analysis. RESULTS According to the results, XIST and ZFPM2-AS1 expression remarkably increased within GC plasma relative to normal subjects (P< 0.01); besides, lncRNA XIST expression after surgery had a tendency of downregulation compared with preoperative levels (P< 0.05). Moreover, the area under ROC curve (AUC) values were 0.62 for ZFPM2-AS1 and 0.68 for XIST, while the pooled AUC value of CA-724 and two lncRNAs was 0.751. CONCLUSION Circulating lncRNAs ZFPM2-AS1 and XIST can serve as the candidate plasma biomarkers used to diagnose GC.
Collapse
Affiliation(s)
- Han Liang
- Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hao Li
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Nan Xia
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingjing Chen
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Linlin Gao
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hao Liu
- Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ping Lyu
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Guo
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziwei Yang
- Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
10
|
Zhang Z, Li Y, Fan L, Wang B, Liu W, Cui J, Tan B. LncRNA THUMPD3-AS1 promotes invasion and EMT in gastric cancer by regulating the miR-1297/BCAT1 pathway. iScience 2023; 26:107673. [PMID: 37705956 PMCID: PMC10495635 DOI: 10.1016/j.isci.2023.107673] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Long noncoding RNA (lncRNA) plays crucial roles in the development of gastric cancer (GC); however, studies of their mechanisms of action are needed to determine their clinical value. The aim of this study is to explore the effects and mechanisms of THUMPD3-AS1 in GC. Elevated levels of THUMPD3-AS1 were observed in GC and demonstrated a significant positive correlation with poor prognosis. Functionally, THUMPD3-AS1 promoted GC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) and induced tumor growth in vivo. THUMPD3-AS1 exerts its regulatory function on BCAT1 through competitive binding with miR-1297. Further investigations confirmed that both THUMPD3-AS1 and miR-1297 interact with BCAT1. These findings suggest that THUMPD3-AS1 promotes GC invasion and EMT by regulating the miR-1297/BCAT1 pathway, indicating that THUMPD3-AS1 may serve as a biomarker and therapeutic target for GC.
Collapse
Affiliation(s)
- Zaibo Zhang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Liqiao Fan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Bingyu Wang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Wenbo Liu
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Jiaxiang Cui
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| | - Bibo Tan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, 12 Health Road, Chang’an District, Shijiazhuang 050011, China
| |
Collapse
|
11
|
Wang L, Xiao K, Dong Z, Meng T, Cheng X, Xu Y. A novel copper-induced cell death-related lncRNA prognostic signature associated with immune infiltration and clinical value in gastric cancer. J Cancer Res Clin Oncol 2023; 149:10543-10559. [PMID: 37291405 PMCID: PMC10423106 DOI: 10.1007/s00432-023-04916-7] [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: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most important malignancies and has a poor prognosis. Copper-induced cell death, recently termed cuproptosis, may directly affect the outcome of GC. Long noncoding RNAs (lncRNAs), possessing stable structures, can influence the prognosis of cancer and may serve as potential prognostic prediction factors for various cancers. However, the role of copper cell death-related lncRNAs (CRLs) in GC has not been thoroughly investigated. Here, we aim to elucidate the role of CRLs in predicting prognosis, diagnosis, and immunotherapy in GC patients. METHODS RNA expression data for 407 GC patients from The Cancer Genome Atlas (TCGA) were gathered, and differentially expressed CRLs were identified. Subsequently, the researchers applied univariate, LASSO, and multivariate Cox regression to construct a prognostic signature consisting of 5 lncRNAs based on the CRLs. Stratified by the median CRLSig risk score, Kaplan-Meier analysis was utilized to compare overall survival (OS) between the high- and low-risk groups. Among the two groups, gene set enrichment analysis (GSEA), tumor microenvironment (TME), drug sensitivity analysis, and immune checkpoint analysis were conducted. In addition, consensus clustering and nomogram analysis were performed to predict OS. Cell experiments and 112 human serum samples were employed to verify the effect of lncRNAs on GC. Furthermore, the diagnostic value of the CRLSig in the serum of GC patients was analyzed by the receiver operating characteristic (ROC) curve. RESULTS A prognostic signature for GC patients was constructed based on CRLs, composed of AC129926.1, AP002954.1, AC023511.1, LINC01537, and TMEM75. According to the K-M survival analysis, high-risk GC patients had a lower OS rate and progression-free survival rate than low-risk GC patients. Further support for the model's accuracy was provided by ROC, principal component analysis, and the validation set. The area under the curve (AUC) of 0.772 for GC patients showed a better prognostic value than any other clinicopathological variable. Furthermore, immune infiltration analysis showed that the high-risk group had greater antitumor immune responses in the tumor microenvironment. In the high-risk subgroup, 23 immune checkpoint genes had significantly higher expression levels than in the low-risk subgroup (p < 0.05). The half-maximal inhibitory concentrations (IC50) of 86 drugs were found to be significantly different in the two groups. Accordingly, the model is capable of predicting the effectiveness of immunotherapy. In addition, the five CRLs in GC serum exhibited statistically significant expression levels. The AUC of this signature in GC serum was 0.894, with a 95% CI of 0.822-0.944. Moreover, lncRNA AC129926.1 was significantly overexpressed in GC cell lines and the serum of GC patients. Importantly, colony formation, wound healing, and transwell assays further confirmed the oncogenic role of AC129926.1 in GC. CONCLUSION In this study, a prognostic signature model consisting of five CRLs was developed to improve OS prediction accuracy in GC patients. The model also has the potential to predict immune infiltration and immunotherapy effectiveness. Furthermore, the CRLSig might serve as a novel serum biomarker to differentiate GC patients from healthy individuals.
Collapse
Affiliation(s)
- Li Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ke Xiao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong Province, China
| | - Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong Province, China
| | - Tao Meng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiaowen Cheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Yuanhong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
12
|
Wang S, Zou C, Lin X, Hu D, Su Y, He H, Zheng X, Zhang L, Huang T, Liao JR, Lin X. RNU12 inhibits gastric cancer progression via sponging miR-575 and targeting BLID. Sci Rep 2023; 13:7523. [PMID: 37160927 PMCID: PMC10169768 DOI: 10.1038/s41598-023-34539-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/03/2023] [Indexed: 05/11/2023] Open
Abstract
Gastric cancer (GC) is one of the major causes of cancer deaths with 5-year survival ratio of 20%. RNU12 is one of long noncoding RNAs (lncRNAs) regulating the tumor progression. However, how RNU12 affecting GC is not clear. qRT-PCR was utilized for determining the RNU12 expression in cell lines, 113 cases of paired gastric cancer (GC) and their adjacent normal gastric tissues. The biofunction alterations of RNU12 were assessed by its overexpression or knockdown in GC cells. MTT and cloning assay were assayed for the cell proliferation, the flow cytometry for the detection of cell cycle and the wound healing assay (WHA) and transwell invasion assay (TIA) for examining the migration and invasion of cells. The expressions of a set of genes related proliferation and migration were investigated with the Western Blotting (WB). RNA immunoprecipitation (RIP), biotinylated RNA pull-down and dual luciferase reporter tests were used to detect the interactions of RNU12 with miR-575/BLID. The in vivo proliferation and migration ability of RNU12 infected cells were determined in zebrafish system. This study revealed that RNU12 inhibited proliferation, invasion and metastasis by sponging of miR-575 and regulating the downstream BLID and modulated EMT of GC cells. The RNU12/miR-575/BLID axis is likely to be the prognosis biomarkers and drug targets of GC.
Collapse
Affiliation(s)
- Shaoli Wang
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Changyan Zou
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xinyi Lin
- Fujian Medical University, Fuzhou, 350122, China
| | - Dan Hu
- Department of Pathology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Ying Su
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Huocong He
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiongwei Zheng
- Department of Pathology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lurong Zhang
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jin-Rong Liao
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China.
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou, 350014, China.
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, 350122, China.
| |
Collapse
|
13
|
Zhang M, Peng S. The association and clinical relevance of phase-separating protein CAPRIN1 with noncoding RNA. Cell Stress Chaperones 2023; 28:125-132. [PMID: 36627508 PMCID: PMC10050270 DOI: 10.1007/s12192-023-01320-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
CAPRIN1, cell cycle-associated protein 1, is an RNA-binding protein in stress granules, P bodies, and messenger RNA transport granules and has a high level of expression in cancer. It promotes the proliferation and invasion of cancer cells and enhances their glycolysis and chemoresistance. In addition, it mediates the formation of intracellular SGs in various ways when exposed to endogenous and exogenous stress. As an RNA-binding protein, it not only directly binds to several mRNAs associated with the cell cycle but also is the target of miRNA, lncRNA, and circRNA. Recently, CAPRIN1 is identified as a phase-separating protein that mediates the liquid-liquid phase separation within tumor cells. Moreover, the formation of CAPRIN1-mediated phase separation is regulated by circRNA and lncRNA. In addition, CAPRIN1 is associated with ubiquitination, which affects the relevant characteristics of cancer cells. This review discusses the different regulatory mechanisms of CAPRIN1 in various tumors and its association with noncoding RNA, suggesting its potential as an oncogenic signal and possibly as a diagnostic indicator in the future. This may provide the multifunctional characteristic insight of CAPRIN1 protein and potential therapeutic target in malignancy with high levels of CAPRIN1.
Collapse
Affiliation(s)
- Mojian Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Shuping Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|