1
|
Jing D, Wu W, Huang X, Zhang Z, Chen X, Huang F, Liu J, Zhang Z, Shao Z, Pu F. Transketolase promotes osteosarcoma progression through the YY1-PAK4 axis. FEBS J 2025. [PMID: 39853945 DOI: 10.1111/febs.17375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/18/2024] [Accepted: 12/16/2024] [Indexed: 01/26/2025]
Abstract
Osteosarcoma, a malignant bone tumor that occurs in adolescents, proliferates and is prone to pulmonary metastasis. Osteosarcoma is characterized by high genotypic heterogeneity, making it difficult to identify reliable anti-osteosarcoma targets. The genotype of osteosarcoma may be highly dynamic, but its high dependence on energy remains constant. Fortunately, tumors tend to have relatively consistent metabolic types. Targeting metabolism with anti-tumor therapies is a new strategy for treating tumors. Genes related to carbohydrate metabolism are widely and highly expressed in tumor tissues. Transketolase (TKT), a key enzyme at the non-oxidative stage of the pentose phosphate pathway, is up-regulated in various tumors. In the present study, TKT promoted osteosarcoma cell proliferation non-metabolically. Specifically, TKT bound directly to amino acid residues of Yin Yang 1 (YY1) at amino acids 201-228, stimulating YY1 to bind to the promoter of P21 activated kinase 4 (PAK4) and resulting in PAK4 expression and activation of the phosphoinositide 3-kinase-Akt signaling pathway. Additionally, we designed a peptide, YY1-PEP, based on the exact mechanism of how TKT promotes osteosarcoma. Per in vivo and in vitro experiments, YY1-PEP displayed anti-osteosarcoma properties. The present study provides a new feasible strategy against osteosarcoma progression.
Collapse
Affiliation(s)
- Doudou Jing
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenhao Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuanzuo Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuhua Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Orthopedics, Wuhan No.1 Hospital, China
| |
Collapse
|
2
|
Zhang X, Shi L, Xing M, Li C, Ma F, Ma Y, Ma Y. Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review). Int J Mol Med 2025; 55:15. [PMID: 39513614 PMCID: PMC11573320 DOI: 10.3892/ijmm.2024.5456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Long non‑coding RNA (lncRNA) is a class of non‑coding RNA molecules located in the cytoplasm or nucleus, which can regulate chromosome structure and function by interacting with DNA, RNA, proteins and other molecules; binding to mRNA bases in a complementary manner, affecting the splicing, stabilization, translation and degradation of mRNA; acting as competing endogenous RNA competitively binds to microRNAs to regulate gene expression and participate in the regulation of various vital activities of the body. The PI3K/AKT signalling pathway plays a key role in numerous biological and cellular processes, such as cell proliferation, invasion, migration and angiogenesis. It has been found that the lncRNA/PI3K/AKT axis regulates the expression of cancer‑related genes and thus tumour progression. The abnormal regulation of lncRNA expression in the lncRNA/PI3K/AKT axis is clearly associated with clinicopathological features and plays an important role in regulating biological functions. In the present review, the expression and biological functions of PI3K/AKT‑related lncRNAs both in vitro and in vivo over recent years, were comprehensively summarized and analyzed. Their correlation with clinicopathological features was also evaluated, with the objective of furnishing a solid theoretical foundation for clinical diagnosis and the monitoring of efficacy in digestive system neoplasms. The present review aimed to provide a comprehensive overview of the expression and biological functions of PI3K/AKT‑related lncRNAs in digestive system neoplasms and to assess their correlation with clinicopathological features. This endeavor seeks to establish a solid theoretical foundation for the clinical diagnosis and efficacy monitoring of digestive system tumors.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Lei Shi
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Mengzhen Xing
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Chunjing Li
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Fengjun Ma
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Yuning Ma
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Yuxia Ma
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
3
|
Ma X, Lu T, Yang Y, Qin D, Tang Z, Cui Y, Wang R. DEAD-box helicase family proteins: emerging targets in digestive system cancers and advances in targeted drug development. J Transl Med 2024; 22:1120. [PMID: 39707322 DOI: 10.1186/s12967-024-05930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/30/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer has become one of the major diseases threatening human health in the twenty-first century due to its incurability. In 2022, new cases of esophageal and gastrointestinal cancers accounted for 17.1% of all newly diagnosed cancer cases worldwide. Despite significant improvements in early cancer screening, clinical diagnostics, and treatments in recent years, the overall prognosis of digestive system cancer patients remains poor. The DEAD-box helicase family, a crucial member of the RNA helicase family, participates in almost every aspect of RNA metabolism, including transcription, splicing, translation, and degradation, and plays a key role in the occurrence and progression of various cancers. This article aims to summarize and discuss the role and potential clinical applications of DEAD-box helicase family proteins in digestive system cancers. The discussion includes the latest progress in the occurrence, development, and treatment of esophageal and gastrointestinal tumors; the main functions of DEAD-box helicase family proteins; their roles in digestive system cancers, including their relationships with clinical factors; effects on cancer proliferation, migration, and invasion; and involved signaling pathways; as well as the existing inhibitory strategies targeting DDX family proteins, are discussed. Additionally, outlooks on future research directions are provided.
Collapse
Affiliation(s)
- Xiaochao Ma
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Tianyu Lu
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Yue Yang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Da Qin
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Ze Tang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Youbin Cui
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China.
| | - Rui Wang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| |
Collapse
|
4
|
Xu H, Wang T, Nie H, Sun Q, Jin C, Yang S, Chen Z, Wang X, Tang J, Feng Y, Sun Y. USP36 promotes colorectal cancer progression through inhibition of p53 signaling pathway via stabilizing RBM28. Oncogene 2024; 43:3442-3455. [PMID: 39343961 PMCID: PMC11573713 DOI: 10.1038/s41388-024-03178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Colorectal cancer (CRC) stands as the second most common cause of cancer-related mortality globally and p53, a widely recognized tumor suppressor, contributes to the development of CRC. Ubiquitin-specific protease 36 (USP36), belonging to the deubiquitinating enzyme family, is involved in tumor progression across multiple cancers. However, the underlying molecular mechanism in which USP36 regulates p53 signaling pathway in CRC is unclear. Here, our study revealed that USP36 was increased in CRC tissues and associated with unfavorable prognosis. Functionally, elevated USP36 could promote proliferation, migration, and invasion of CRC cells in vitro and in vivo. Mechanistically, USP36 could interact with and stabilize RBM28 via deubiquitination at K162 residue. Further, upregulated RBM28 could bind with p53 to suppress its transcriptional activity and therefore inactivate p53 signaling pathway. Collectively, our investigation identified the novel USP36/RBM28/p53 axis and its involvement in promoting cell proliferation and metastasis in CRC, which presents a promising therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Hengjie Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Tuo Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Hongxu Nie
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Qingyang Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Chi Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Sheng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Zhihao Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Xiaowei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Junwei Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| | - Yifei Feng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China.
| |
Collapse
|
5
|
Zhang B, Chen X, Song H, Gao X, Ma S, Ji H, Qu H, Xia S, Shang D. Identification of basement membrane-related prognostic model associated with the immune microenvironment and synthetic therapy response in pancreatic cancer: integrated bioinformatics analysis and clinical validation. J Cancer 2024; 15:6273-6298. [PMID: 39513120 PMCID: PMC11540510 DOI: 10.7150/jca.100891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Pancreatic cancer (PC) is a common and highly malignant tumor. Basement membrane (BM) is formed by the crosslinking of extracellular matrix macromolecules and acts as a barrier against tumor cell metastasis. However, the role of BM in PC prognosis, immune infiltration, and treatment remains unclear. This study collected transcriptome and clinical survival data of PC via TCGA, GEO, and ICGC databases. PC patients (PCs) from the First Affiliated Hospital of Dalian Medical University were obtained as the clinical validation cohort. BM-related genes (BMRGs) were acquired from GeneCards and basement membraneBASE databases. A total of 46 differential-expressed BMRGs were identified. Then the BM-related prognostic model (including DSG3, MET, and PLAU) was built and validated. PCs with a low BM-related score had a better outcome and were more likely to benefit from oxaliplatin, irinotecan, and KRAS(G12C) inhibitor-12, and immunotherapy. Immune analysis revealed that BM-related score was positively correlated with neutrophils, cancer-associated fibroblasts, and macrophages infiltration, but negatively correlated with CD8+ T cells, NK cells, and B cells infiltration. PCs from the clinical cohort further verified that BM-related model could accurately predict PCs' outcomes. DSG3, MET, and PLAU were notably up-regulated within PC tissues and linked to a poor prognosis. In vitro experiments showed that DSG3 knockdown markedly suppressed the proliferation, migration, and invasion of PC cells. Molecular docking indicated that epigallocatechin gallate had a strong binding activity with DSG3, MET, and PLAU and may be used as a potential therapeutic agent for PC. In conclusion, this study developed a BM-related model associated with PC prognosis, immune infiltration, and treatment, which provided new insights into PC stratification and drug intervention.
Collapse
Affiliation(s)
- Biao Zhang
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xu Chen
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyi Song
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shurong Ma
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Hongying Ji
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huixian Qu
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilin Xia
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Dong Shang
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Li S, Feng T, Yuan H, Li Q, Zhao G, Li K. DEAD-box RNA helicases in the multistep process of tumor metastasis. Mol Biol Rep 2024; 51:1006. [PMID: 39306810 DOI: 10.1007/s11033-024-09912-9] [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: 06/26/2024] [Accepted: 09/03/2024] [Indexed: 01/04/2025]
Abstract
RNA helicases constitute a large family of proteins that share a catalytic core with high structural similarity. DEAD-box (DDX) proteins belong to the largest RNA helicase subfamily, and DDX members have been implicated in all facets of RNA metabolism, from transcription to translation, miRNA maturation, and RNA delay and degradation. Interestingly, an increasing number of studies have suggested a relationship between DDX proteins and cancer initiation and progression. The expression levels of many DDX proteins are elevated in a majority of cancers, and recent studies have demonstrated that some DDX proteins have a potent positive effect on promoting the metastasis of malignant cells. Metastasis is a complex, multistep cascade process that includes local invasion, intravasation and survival in the circulation, arrest at a distant organ site, extravasation and metastatic colonization; here, we review this process and present the suggested functions and mechanisms of DDX family proteins in particular steps of the invasion‒metastasis cascade.
Collapse
Affiliation(s)
- Shan Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Tianyu Feng
- Department of Laboratory Medicine, Clinical Laboratory Medicine Research Center of West China Hospital, Sichuan Clinical Research Center for Laboratory Medicine, West China Hospital, Sichuan University, Chengdu City, People's Republic of China
| | - Hang Yuan
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Qin Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Gang Zhao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| | - Kai Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| |
Collapse
|
7
|
Huang T, Ren K, Ling X, Li Z, Chen L. Transcription factor Yin Yang 1 enhances epithelial-mesenchymal transition, migration, and stemness of non-small cell lung cancer cells by targeting sonic hedgehog. Mol Cell Biochem 2024:10.1007/s11010-024-05104-y. [PMID: 39261409 DOI: 10.1007/s11010-024-05104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a frequent type of lung cancer. Transcription factor Yin Yang 1 (YY1), an endogenous transcription factor containing zinc finger structure, can accelerate NSCLC progression. However, the impact of YY1 on the stemness of NSCLC cells and the mechanism of promoting NSCLC cell progression is unclear. YY1 and Sonic hedgehog (Shh) expressions were monitored by RT-qPCR, western blot, and immunohistochemistry. Overall survival was tested through Kaplan-Meier analysis. The interaction between YY1 and Shh was confirmed. Then, cell migration, stemness, and epithelial-mesenchymal transition (EMT) were assessed with functional experiments in vitro and in vivo. YY1 and Shh were highly expressed in NSCLC tissues and positively correlated with the poor OS of NSCLC patients. Functional experiments denoted that YY1 or Shh overexpression could accelerate EMT, migration, and stemness of NSCLC cells, and YY1 or Shh knockdown played the opposite role to its overexpression. Mechanism analysis disclosed that Shh, as a target gene of YY1, was positively related to YY1. The rescued experiment manifested that Shh silencing could reverse the induction effect of YY1 overexpression on EMT, migration, and stemness of NSCLC cells. In vivo experiments also confirmed that YY1 could accelerate tumor growth and EMT and weaken apoptosis. YY1 promotes NSCLC EMT, migration, and stemness by Shh, which might be novel diagnostic markers and therapeutic targets for NSCLC therapy.
Collapse
Affiliation(s)
- Tonghai Huang
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China.
| | - Kangqi Ren
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Xiean Ling
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Zeyao Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Lin Chen
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| |
Collapse
|
8
|
Cui P, Wang H, Bai Z. Integrated single-cell and bulk RNA-seq analysis identifies a prognostic T-cell signature in colorectal cancer. Sci Rep 2024; 14:20177. [PMID: 39215032 PMCID: PMC11364821 DOI: 10.1038/s41598-024-70422-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Colorectal cancer (CRC) is a major contributor to global morbidity and mortality, necessitating more effective therapeutic approaches. T cells, prominent in the tumor microenvironment, exert a crucial role in modulating immunotherapeutic responses and clinical outcomes in CRC. This study introduces a pioneering method for characterizing the CRC immune microenvironment using single-cell sequencing data. Unlike previous approaches, which focused on individual T-cell signature genes, we utilized overall infiltration levels of colorectal cancer signature T-cells. Through weighted gene co-expression network analysis, Lasso regression, and StepCox analysis, we developed a prognostic risk model, TRGS (T-cell related genes signatures), based on six T cell-related genes. Multivariate Cox analysis identified TRGS as an independent prognostic factor for CRC, showcasing its superior predictive efficacy compared to existing immune-related prognostic models. Immunoreactivity analysis revealed higher Immunophenoscore and lower Tumor Immune Dysfunction and Exclusion scores in the low-risk group, indicating potential responsiveness to immune checkpoint inhibitor therapy. Additionally, patients in the low-risk group demonstrated heightened sensitivity to 5-fluorouracil-based chemotherapy regimens. In summary, TRGS emerges as a standalone prognostic biomarker for CRC, offering insights to optimize patient responses to immunotherapy and chemotherapy, thereby laying the groundwork for personalized tumor management strategies.
Collapse
Affiliation(s)
- Peng Cui
- Department of General Surgery, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xi-Cheng District, Beijing, 100050, People's Republic of China
| | - Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
| | - Zhigang Bai
- Department of General Surgery, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xi-Cheng District, Beijing, 100050, People's Republic of China.
| |
Collapse
|
9
|
Li P, Ma X, Gu X. The essential roles of lncRNAs/PI3K/AKT axis in gastrointestinal tumors. Front Cell Dev Biol 2024; 12:1442193. [PMID: 39161590 PMCID: PMC11330846 DOI: 10.3389/fcell.2024.1442193] [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: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
The role of long noncoding RNA (lncRNA) in tumors, particularly in gastrointestinal tumors, has gained significant attention. Accumulating evidence underscores the interaction between various lncRNAs and diverse molecular pathways involved in cancer progression. One such pivotal pathway is the PI3K/AKT pathway, which serves as a crucial intracellular mechanism maintaining the balance among various cellular physiological processes for normal cell growth and survival. Frequent dysregulation of the PI3K/AKT pathway in cancer, along with aberrant activation, plays a critical role in driving tumorigenesis. LncRNAs modulate the PI3K/AKT signaling pathway through diverse mechanisms, primarily by acting as competing endogenous RNA to regulate miRNA expression and associated genes. This interaction significantly influences fundamental biological behaviors such as cell proliferation, metastasis, and drug resistance. Abnormal expression of numerous lncRNAs in gastrointestinal tumors often correlates with clinical outcomes and pathological features in patients with cancer. Additionally, these lncRNAs influence the sensitivity of tumor cells to chemotherapy in multiple types of gastrointestinal tumors through the abnormal activation of the PI3K/AKT pathway. These findings provide valuable insights into the mechanisms underlying gastrointestinal tumors and potential therapeutic targets. However, gastrointestinal tumors remain a significant global health concern, with increasing incidence and mortality rates of gastrointestinal tumors over recent decades. This review provides a comprehensive summary of the latest research on the interactions of lncRNA and the PI3K/AKT pathway in gastrointestinal tumor development. Additionally, it focuses on the functions of lncRNAs and the PI3K/AKT pathway in carcinogenesis, exploring expression profiles, clinicopathological characteristics, interaction mechanisms with the PI3K/AKT pathway, and potential clinical applications.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
10
|
Yang S, Zhang D, Sun Q, Nie H, Zhang Y, Wang X, Huang Y, Sun Y. Single-Cell and Spatial Transcriptome Profiling Identifies the Transcription Factor BHLHE40 as a Driver of EMT in Metastatic Colorectal Cancer. Cancer Res 2024; 84:2202-2217. [PMID: 38657117 DOI: 10.1158/0008-5472.can-23-3264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/14/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Colorectal cancer is one of the most common malignant tumors in humans, with liver metastasis being the primary cause of mortality. The epithelial-mesenchymal transition (EMT) process endows cancer cells with enhanced metastatic potential. To elucidate the cellular mechanisms driving EMT in colorectal cancer, we analyzed single-cell RNA sequencing data from 11 nonmetastatic primary tumors (TnM) and 11 metastatic primary tumors (TM) from colorectal cancer patients. Compared with the TnM group, the TM samples showed elevated numbers of malignant epithelial cell and cancer-associated fibroblast (CAF) subsets that displayed enrichments of EMT, angiogenesis, and TGFβ signaling pathways. One specific TM-enriched subgroup of malignant epithelial cells underwent EMT to transdifferentiate into CXCL1+ CAFs that subsequently differentiated into SFRP2+ CAFs, which was validated by spatial transcriptomic and pseudotime trajectory analyses. Furthermore, cell-cell communication analysis identified BHLHE40 as a probable key transcription factor driving EMT that was associated with poor prognosis. Finally, in vitro and in vivo experiments functionally substantiated that BHLHE40 promoted the proliferation, invasion, migration, EMT, and liver metastasis of colorectal cancer cells. In summary, this study identified BHLHE40 as a key transcription factor regulating EMT that promotes liver metastasis in colorectal cancer. Significance: Integrated analysis of single-cell RNA sequencing and spatial transcriptomics in metastatic colorectal cancer provides insights into the mechanisms underlying EMT and cancer-associated fibroblast differentiation, which could help improve patient diagnosis and treatment.
Collapse
Affiliation(s)
- Sheng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Dongsheng Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Qingyang Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Hongxu Nie
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yue Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Xiaowei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yuanjian Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| |
Collapse
|
11
|
Peng C, Yang P, Zhang D, Jin C, Peng W, Wang T, Sun Q, Chen Z, Feng Y, Sun Y. KHK-A promotes fructose-dependent colorectal cancer liver metastasis by facilitating the phosphorylation and translocation of PKM2. Acta Pharm Sin B 2024; 14:2959-2976. [PMID: 39027256 PMCID: PMC11252482 DOI: 10.1016/j.apsb.2024.04.024] [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: 01/13/2024] [Revised: 03/01/2024] [Accepted: 04/15/2024] [Indexed: 07/20/2024] Open
Abstract
Excessive fructose diet is closely associated with colorectal cancer (CRC) progression. Nevertheless, fructose's specific function and precise mechanism in colorectal cancer liver metastasis (CRLM) is rarely known. Here, this study reported that the fructose absorbed by primary colorectal cancer could accelerate CRLM, and the expression of KHK-A, not KHK-C, in liver metastasis was higher than in paired primary tumors. Furthermore, KHK-A facilitated fructose-dependent CRLM in vitro and in vivo by phosphorylating PKM2 at Ser37. PKM2 phosphorylated by KHK-A inhibited its tetramer formation and pyruvic acid kinase activity but promoted the nuclear accumulation of PKM2. EMT and aerobic glycolysis activated by nuclear PKM2 enhance CRC cells' migration ability and anoikis resistance during CRLM progression. TEPP-46 treatment, targeting the phosphorylation of PKM2, inhibited the pro-metastatic effect of KHK-A. Besides, c-myc activated by nuclear PKM2 promotes alternative splicing of KHK-A, forming a positive feedback loop.
Collapse
Affiliation(s)
- Chaofan Peng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Peng Yang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Dongsheng Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Chi Jin
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Wen Peng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Tuo Wang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Qingyang Sun
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Zhihao Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Yifei Feng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing 210029, China
| | - Yueming Sun
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Colorectal Institute of Nanjing Medical University, Nanjing 210029, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing 210029, China
| |
Collapse
|
12
|
Zhao J, Yang X, Gong W, Zhang L, Li C, Han X, Zhang Y, Chu X. LINC00908 attenuates LUAD tumorigenesis through DEAD-box helicase 54. Am J Cancer Res 2024; 14:2371-2389. [PMID: 38859824 PMCID: PMC11162691 DOI: 10.62347/hxzm6394] [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: 03/19/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is one of the leading causes of cancer-related death worldwide. We identified a specific long non-coding RNA (LncRNA), LINC00908, which was downregulated in LUAD tissues and associated with good outcome. LINC00908 inhibited glycolysis by regulating the expression of the DEAD-box helicase 54 (DDX54), which was screened by a nine-gene risk signature, where DDX54 showed a positive correlation with several glycolysis-related genes. Experimental verification confirmed that DDX54 regulated nine key glycolytic enzymes, thereby affecting the level of glycolysis in LUAD. Further, the expression of LINC00908 in LUAD tumorigenesis was modulated by a transcription factor, regulatory factor X2 (RFX2). The RFX2/LINC00908/DDX54 axis regulated LUAD tumor growth, migration, invasion, cell apoptosis and glycolysis both in vitro and in vivo. These results demonstrate that this axis may serve as a novel mediator in LUAD progress and offer a novel therapeutic target for more precise diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
- Jiahua Zhao
- Department of Thoracic Surgery, The Sixth Medical Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Xuhui Yang
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Wenwen Gong
- Department of Pharmacy, The Medical Supplies Center of PLA General HospitalBeijing, China
| | - Lin Zhang
- Department of Outpatient Service, 986th Hospital Affilliated to Air Force Medical UniversityXi’an, Shaanxi, China
| | - Chenxi Li
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Xiao Han
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Yang Zhang
- Department of Cardiology, The Second Medical Center, Chinese PLA General HospitalBeijing, China
| | - Xiangyang Chu
- Department of Thoracic Surgery, The First Medical Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| |
Collapse
|
13
|
Shan J, Liang Y, Yang Z, Chen W, Chen Y, Sun K. RNA polymerase I subunit D activated by Yin Yang 1 transcription promote cell proliferation and angiogenesis of colorectal cancer cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:265-273. [PMID: 38682174 PMCID: PMC11058543 DOI: 10.4196/kjpp.2024.28.3.265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/27/2023] [Accepted: 04/25/2023] [Indexed: 05/01/2024]
Abstract
This study aims to explore possible effect of RNA polymerase I subunit D (POLR1D) on proliferation and angiogenesis ability of colorectal cancer (CRC) cells and mechanism herein. The correlation of POLR1D and Yin Yang 1 (YY1) expressions with prognosis of CRC patients in TCGA database was analyzed. Quantitative realtime polymerase chain reaction (qRT-PCR) and Western blot were applied to detect expression levels of POLR1D and YY1 in CRC cell lines and CRC tissues. SW480 and HT- 29 cells were transfected with si-POLR1D or pcDNA3.1-POLR1D to achieve POLR1D suppression or overexpression before cell migration, angiogenesis of human umbilical vein endothelial cells were assessed. Western blot was used to detect expressions of p38 MAPK signal pathway related proteins and interaction of YY1 with POLR1D was confirmed by dual luciferase reporter gene assay and chromatin immunoprecipitation (ChIP). TCGA data showed that both POLR1D and YY1 expressions were up-regulated in CRC patients. High expression of POLR1D was associated with poor prognosis of CRC patients. The results showed that POLR1D and YY1 were highly expressed in CRC cell lines. Inhibition or overexpression of POLR1D can respectively suppress or enhance proliferation and angiogenesis of CRC cells. YY1 inhibition can suppress CRC progression and deactivate p38 MAPK signal pathway, which can be counteracted by POLR1D overexpression. JASPAR predicted YY1 can bind with POLR1D promoter, which was confirmed by dual luciferase reporter gene assay and ChIP. YY1 transcription can up-regulate POLR1D expression to activate p38 MAPK signal pathway, thus promoting proliferation and angiogenesis ability of CRC cells.
Collapse
Affiliation(s)
- Jianfeng Shan
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| | - Yuanxiao Liang
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| | - Zhili Yang
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| | - Wenshan Chen
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| | - Ke Sun
- Department of Colorectal Surgery, Xinchang People’s Hospital, Xinchang, Zhejiang 312500, China
| |
Collapse
|
14
|
Xu J, Zhou Y, He S, Wang Y, Ma J, Li C, Liu Z, Zhou X. Activation of the YY1-UGT2B7 Axis Promotes Mammary Estrogen Homeostasis Dysregulation and Exacerbates Breast Tumor Metastasis. Drug Metab Dispos 2024; 52:408-421. [PMID: 38575184 DOI: 10.1124/dmd.124.001640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Metastasis is the most common pathway of cancer death. The lack of effective predictors of breast cancer metastasis is a pressing issue in clinical practice. Therefore, exploring the mechanism of breast cancer metastasis to uncover reliable predictors is very important for the clinical treatment of breast cancer patients. In this study, tandem mass tag quantitative proteomics technology was used to detect protein content in primary breast tumor tissue samples from patients with metastatic and nonmetastatic breast cancer at diagnosis. We found that the high expression of yin-yang 1(YY1) is strongly associated with poor prognosis in high-grade breast cancer. YY1 expression was detected in both clinical tumor tissue samples and tumor tissue samples from mammary-specific polyomavirus middle T antigen overexpression mouse model mice. We demonstrated that upregulation of YY1 expression was closely associated with breast cancer metastasis and that high YY1 expression could promote the migratory invasive ability of breast cancer cells. Mechanistically, YY1 directly binds to the UGT2B7 mRNA initiation sequence ATTCAT, thereby transcriptionally regulating the inhibition of UGT2B7 expression. UGT2B7 can regulate the development of breast cancer by regulating estrogen homeostasis in the breast, and the abnormal accumulation of estrogen, especially 4-OHE2, promotes the migration and invasion of breast cancer cells, ultimately causing the development of breast cancer metastasis. In conclusion, YY1 can regulate the UGT2B7-estrogen metabolic axis and induce disturbances in estrogen metabolism in breast tumors, ultimately leading to breast cancer metastasis. Disturbances in estrogen metabolism in the breast tissue may be an important risk factor for breast tumor progression and metastasis SIGNIFICANCE STATEMENT: In this study, we propose for the first time a regulatory relationship between YY1 and the UGT2B7/estrogen metabolism axis and explore the molecular mechanism. Our study shows that the YY1/UGT2B7/estrogen axis plays an important role in the development and metastasis of breast cancer. This study further elucidates the potential mechanisms of YY1-mediated breast cancer metastasis and the possibility and promise of YY1 as a predictor of cancer metastasis.
Collapse
Affiliation(s)
- Jiahao Xu
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Ying Zhou
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Shiqing He
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Yinghao Wang
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Jiachen Ma
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Changwen Li
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Zhao Liu
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Xueyan Zhou
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| |
Collapse
|
15
|
Ai LJ, Li GD, Chen G, Sun ZQ, Zhang JN, Liu M. Molecular subtyping and the construction of a predictive model of colorectal cancer based on ion channel genes. Eur J Med Res 2024; 29:219. [PMID: 38576045 PMCID: PMC10993535 DOI: 10.1186/s40001-024-01819-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Colorectal cancer (CRC) is a highly heterogeneous malignancy with an unfavorable prognosis. The purpose of this study was to address the heterogeneity of CRC by categorizing it into ion channel subtypes, and to develop a predictive modeling based on ion channel genes to predict the survival and immunological states of patients with CRC. The model will provide guidance for personalized immunotherapy and drug treatment. METHODS A consistent clustering method was used to classify 619 CRC samples based on the expression of 279 ion channel genes. Such a method was allowed to investigate the relationship between molecular subtypes, prognosis, and immune infiltration. Furthermore, a predictive modeling was constructed for ion channels to evaluate the ion channel properties of individual tumors using the least absolute shrinkage and selection operator. The expression patterns of the characteristic genes were validated through molecular biology experiments. The effect of potassium channel tetramerization domain containing 9 (KCTD9) on CRC was verified by cellular functional experiments. RESULTS Four distinct ion channel subtypes were identified in CRC, each characterized by unique prognosis and immune infiltration patterns. Notably, Ion Cluster3 exhibited high levels of immune infiltration and a favorable prognosis, while Ion Cluster4 showed relatively lower levels of immune infiltration and a poorer prognosis. The ion channel score could predict overall survival, with lower scores correlated with longer survival. This score served as an independent prognostic factor and presented an excellent predictive efficacy in the nomogram. In addition, the score was closely related to immune infiltration, immunotherapy response, and chemotherapy sensitivity. Experimental evidence further confirmed that low expression of KCTD9 in tumor tissues was associated with an unfavorable prognosis in patients with CRC. The cellular functional experiments demonstrated that KCTD9 inhibited the proliferation, migration and invasion capabilities of LOVO cells. CONCLUSIONS Ion channel subtyping and scoring can effectively predict the prognosis and evaluate the immune microenvironment, immunotherapy response, and drug sensitivity in patients with CRC.
Collapse
Affiliation(s)
- Lian-Jie Ai
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Guo-Dong Li
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Gang Chen
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Zi-Quan Sun
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jin-Ning Zhang
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Ming Liu
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
16
|
Zhao J, Guo M, Yan Y, Wang Y, Zhao X, Yang J, Chen J, Chen C, Tang L, Zeng W, Liu Y, Qin M, Zhou Y, Xu L. The miR-7/EGFR axis controls the epithelial cell immunomodulation and regeneration and orchestrates the pathology in inflammatory bowel disease. J Adv Res 2024; 57:119-134. [PMID: 37094666 PMCID: PMC10918346 DOI: 10.1016/j.jare.2023.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
INTRODUCTION The epithelial immunomodulation and regeneration are intrinsic critical events against inflammatory bowel disease (IBD). MiR-7 is well documented as a promising regulator in the development of various diseases including inflammatory diseases. OBJECTIVES This study aimed to assess the effect of miR-7 in intestinal epithelial cells (IECs) in IBD. METHODS MiR-7def mice were given dextran sulfate sodium (DSS) to induce enteritis model. The infiltration of inflammatory cells was measured by FCM and immunofluorescence assay. 5'deletion assay and EMSA assays were performed to study the regulatory mechanism of miR-7 expression in IECs. The inflammatory signals and the targets of miR-7 were analyzed by RNA-seq and FISH assay. IECs were isolated from miR-7def, miR-7oe and WT mice to identify the immunomodulation and regeneration capacity. IEC-specific miR-7 silencing expression vector was designed and administered by the tail vein into murine DSS-induced enteritis model to evaluate the pathological lesions of IBD. RESULTS We found miR-7 deficiency improved the pathological lesions of DSS-induced murine enteritis model, accompanied by elevated proliferation and enhanced transduction of NF-κB/AKT/ERK signals in colonic IECs, as well as decreased local infiltration of inflammatory cells. MiR-7 was dominantly upregulated in colonic IECs in colitis. Moreover, the transcription of pre-miR-7a-1, orchestrated by transcription factor C/EBPα, was a main resource of mature miR-7 in IECs. As for the mechanism, EGFR, a miR-7 target gene, was downregulated in colonic IECs in colitis model and Crohn's disease patients. Furthermore, miR-7 also controlled the proliferation and inflammatory-cytokine secretion of IECs in response to inflammatory-signals through EGFR/NF-κB/AKT/ERK pathway. Finally, IEC-specific miR-7 silencing promoted the proliferation and transduction of NF-κB pathway in IECs and alleviated the pathological damage of colitis. CONCLUSION Our results present the unknown role of miR-7/EGFR axis in IEC immunomodulation and regeneration in IBD and might provide clues for the application of miRNA-based therapeutic strategies in colonic diseases.
Collapse
Affiliation(s)
- Juanjuan Zhao
- School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Yaping Yan
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Ya Wang
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Xu Zhao
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Jing Yang
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Jing Chen
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Chao Chen
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Lin Tang
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Wenhuan Zeng
- Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Yiting Liu
- Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Ming Qin
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China
| | - Lin Xu
- School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou Province, Guizhou, Zunyi 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
17
|
Wu J, Wu Y, Chen S, Guo Q, Shao Y, Liu C, Lin K, Wang S, Zhu J, Chen X, Ju X, Xia L, Wu X. PARP1-stabilised FOXQ1 promotes ovarian cancer progression by activating the LAMB3/WNT/β-catenin signalling pathway. Oncogene 2024; 43:866-883. [PMID: 38297082 DOI: 10.1038/s41388-024-02943-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
Metastasis is an important factor that causes ovarian cancer (OC) to become the most lethal malignancy of the female reproductive system, but its molecular mechanism is not fully understood. In this study, through bioinformatics analysis, as well as analysis of tissue samples and clinicopathological characteristics and prognosis of patients in our centre, it was found that Forkhead box Q1 (FOXQ1) was correlated with metastasis and prognosis of OC. Through cell function experiments and animal experiments, the results show that FOXQ1 can promote the progression of ovarian cancer in vivo and in vitro. Through RNA-seq, chromatin immunoprecipitation sequencing (ChIP-seq), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA), Western blotting (WB), quantitative real-time polymerase chain reaction (qRT‒PCR), immunohistochemistry (IHC), luciferase assay, and ChIP-PCR, it was demonstrated that FOXQ1 can mediate the WNT/β-catenin pathway by targeting the LAMB promoter region. Through coimmunoprecipitation (Co-IP), mass spectrometry (MS), ubiquitination experiments, and immunofluorescence (IF), the results showed that PARP1 could stabilise FOXQ1 expression via the E3 ubiquitin ligase Hsc70-interacting protein (CHIP). Finally, the whole mechanism pathway was verified by animal drug combination experiments and clinical specimen prognosis analysis. In summary, our results suggest that PARP1 can promote ovarian cancer progression through the LAMB3/WNT/β-catenin pathway by stabilising FOXQ1 expression.
Collapse
Affiliation(s)
- Jiangchun Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Siyu Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Yang Shao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chaohua Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Kailin Lin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Simin Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Jun Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xiaojun Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xingzhu Ju
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Lingfang Xia
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Andrabi MQ, Kesavan Y, Ramalingam S. Non-coding RNAs as Biomarkers for Survival in Colorectal Cancer Patients. Curr Aging Sci 2024; 17:5-15. [PMID: 36733201 DOI: 10.2174/1874609816666230202101054] [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: 06/16/2022] [Revised: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 02/04/2023]
Abstract
Colorectal cancer (CRC) has a high incidence and fatality rate worldwide. It ranks second concerning death worldwide. Cancer patients are diagnosed with the disease at a later stage due to the absence of early diagnostic methods, which leads to increased death. With the help of recent advancements in the fields of diagnosis and therapy, the development of novel methods using new targets could be helpful for the long-term survival of CRC patients when CRC is detected early. However, the prognosis for the advanced stage of CRC is abysmal. New biomarkers are emerging as promising alternatives since they can be utilized for early detection of CRC, are simple to use, and non-invasive. Non-coding RNAs (ncRNAs) have been seen to have an aberrant expression in the development of many malignancies, including CRC. In the past two decades, much research has been done on non-coding RNAs, which may be valuable as biomarkers and targets for antitumor therapy. Non-coding RNAs can be employed in detecting and treating CRC. Non-coding RNAs play an essential role in regulating gene expression. This article reviews ncRNAs and their expression levels in CRC patients that could be used as potential biomarkers. Various ncRNAs have been associated with CRC, such as microRNAs, long non-coding RNAs, circular RNAs, etc. The expression of these non-coding RNAs may provide insights into the stages of cancer and the prognosis of cancer patients and therefore proper precautionary measures can be taken to decrease cancer-related deaths.
Collapse
Affiliation(s)
- Mohammad Qasim Andrabi
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Yasodha Kesavan
- Department of Biotechnology, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| |
Collapse
|
19
|
Chen W, Zhang Q, Dai X, Chen X, Zhang C, Bai R, Chen Y, Zhang K, Duan X, Qiao Y, Zhao J, Tian F, Liu K, Dong Z, Lu J. PGC-1α promotes colorectal carcinoma metastasis through regulating ABCA1 transcription. Oncogene 2023; 42:2456-2470. [PMID: 37400530 DOI: 10.1038/s41388-023-02762-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Colorectal cancer (CRC) is a highly aggressive cancer in which metastasis plays a key role. However, the mechanisms underlying metastasis have not been fully elucidated. Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), a regulator of mitochondrial function, has been reported as a complicated factor in cancer. In this study, we found that PGC-1α was highly expressed in CRC tissues and was positively correlated with lymph node and liver metastasis. Subsequently, PGC-1α knockdown was shown to inhibit CRC growth and metastasis in both in vitro and in vivo studies. Transcriptomic analysis revealed that PGC-1α regulated ATP-binding cassette transporter 1 (ABCA1) mediated cholesterol efflux. Mechanistically, PGC-1α interacted with YY1 to promote ABCA1 transcription, resulting in cholesterol efflux, which subsequently promoted CRC metastasis through epithelial-to-mesenchymal transition (EMT). In addition, the study identified the natural compound isoliquiritigenin (ISL) as an inhibitor that targeted ABCA1 and significantly reduced CRC metastasis induced by PGC-1α. Overall, this study sheds light on how PGC-1α promotes CRC metastasis by regulating ABCA1-mediated cholesterol efflux, providing a basis for further research to inhibit CRC metastasis.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Qiushuang Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Chengjuan Zhang
- Department of Pathology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province, 450003, P. R. China
| | - Ruihua Bai
- Department of Pathology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province, 450003, P. R. China
| | - Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Kai Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Xiaoxuan Duan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China.
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province, 450001, P. R. China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, P. R. China.
| |
Collapse
|
20
|
Chen LJ, Chen X, Niu XH, Peng XF. LncRNAs in colorectal cancer: Biomarkers to therapeutic targets. Clin Chim Acta 2023; 543:117305. [PMID: 36966964 DOI: 10.1016/j.cca.2023.117305] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death in men and women worldwide. As early detection is associated with lower mortality, novel biomarkers are urgently needed for timely diagnosis and appropriate management of patients to achieve the best therapeutic response. Long noncoding RNAs (lncRNAs) have been reported to play essential roles in CRC progression. Accordingly, the regulatory roles of lncRNAs should be better understood in general and for identifying diagnostic, prognostic and predictive biomarkers in CRC specifically. In this review, the latest advances on the potential diagnostic and prognostic lncRNAs as biomarkers in CRC samples were highlighted, Current knowledge on dysregulated lncRNAs and their potential molecular mechanisms were summarized. The potential therapeutic implications and challenges for future and ongoing research in the field were also discussed. Finally, novel insights on the underlying mechanisms of lncRNAs were examined as to their potential role as biomarkers and therapeutic targets in CRC. This review may be used to design future studies and advanced investigations on lncRNAs as biomarkers for the diagnosis, prognosis and therapy in CRC.
Collapse
Affiliation(s)
- Ling-Juan Chen
- Department of Clinical Laboratory, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangdong Province, China
| | - Xiang Chen
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangdong Province, China
| | - Xiao-Hua Niu
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangdong Province, China
| | - Xiao-Fei Peng
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan 511518, Guangdong Province, China.
| |
Collapse
|
21
|
Liu M, Xue G, Liu R, Wang Y, Sheng X, Sun W. Saponin from Platycodi radix inactivates PI3K/AKT signaling pathway to hinder colorectal cancer cell proliferation, invasion, and migration through miR-181c/d-5p/RBM47. Mol Carcinog 2023; 62:174-184. [PMID: 36321407 DOI: 10.1002/mc.23474] [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: 05/16/2022] [Revised: 08/29/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022]
Abstract
Colorectal cancer (CRC) is the third frequent cancer and second leading reason of cancer-related mortality all over the globe. Saponins from Platycodi radix (SPR) and microRNAs (miRNAs) have been reported to regulate CRC cell progression. Real-time quantitative polymerase chain reaction (RT-qPCR) detected miR-181c-5p, miR-181d-5p, and RBM47 expression level. Cell counting kit-8 (CCK-8), 5-ethynyl-20-deoxyuridine (EdU), colony formation, transwell, and wound healing assays validated that miR-181c-5p and miR-181d-5p promote CRC cell proliferation, migration and invasion and SPR exerts opposite effects. Cignal Finder Reporter Array and western blot proved that the activity of PI3K/AKT pathway was decreased by RBM47 overexpression. RNA pulldown, luciferase reporter, and RNA-binding protein immunoprecipitation (RIP) assays proved the interaction between miR-181c/d-5p and RBM47, and RBM47 and PTEN. Rescue experiments were carried out to validate that RBM47 reverses the influence of miR-181c/d-5p on the progression of CRC cells. The stability of PTEN was probed by real-time quantitative polymerase chain reaction in CRC cells treated with Actinomycin D (Act D). To be concluded, SPR inactivates PI3K/AKT signaling pathway to suppress CRC cell proliferation, invasion, and migration via miR-181c/d-5p/RBM47. Elucidating the mechanisms of SPR underlying CRC may offer novel insight into CRC treatment.
Collapse
Affiliation(s)
- Mingkai Liu
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Guiyang Xue
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Rixu Liu
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Yi Wang
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Xiaoqian Sheng
- Department of Clinical Laboratory, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Wei Sun
- Department of General Surgery, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| |
Collapse
|
22
|
Mao S, Xia A, Tao X, Ye D, Qu J, Sun M, Wei H, Li G. A pan-cancer analysis of the prognostic and immunological roles of matrix metalloprotease-1 (MMP1) in human tumors. Front Oncol 2023; 12:1089550. [PMID: 36727076 PMCID: PMC9885257 DOI: 10.3389/fonc.2022.1089550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Objective Cancer remains the leading killer of human health worldwide. It has been shown that matrix metalloproteinase-1(MMP1) is related to poor prognosis in cancers such as BRCA, CESC and COAD. However, systematic pan-cancer analysis about the prognostic and immunological roles of MMP1 has not been explored. Here, the purpose of this study was to investigate the prognostic and immunological roles of MMP1 in pan-cancer and confirm cancer-promoting effect in pancreatic cancer. Methods In our study, bioinformatics were first used to analyze data from multiple databases. Then, several bioinformatics tools were utilized to investigate the role of MMP1 in 33 tumor types. Finally, molecular biology experiments were carried out to prove the cancer-promoting effect of MMP1 in pancreatic cancer. Results MMP1 expression was higher in tumor tissues than in control tissues in most tumor types. High expression of MMP1 was associated with poor overall survival (OS) and disease-free survival (DFS) in some tumor types. Further analysis of MMP1 gene mutation data showed that MMP1 mutations significantly influenced the prognosis of STAD. In addition, MMP1 expression was closely related to cancer-associated fibroblast (CAFs) infiltration in a variety of cancers and played an important role on immune infiltration score, tumor mutational burden (TMB) and microsatellite instability (MSI). Gene Ontology enrichment analysis indicated that these 20 genes were mainly related to extracellular structure organization/extracellular matrix organization/extracellular matrix disassembly/collagen metabolic process in the enriched biological processes. Finally, molecular biology experiments confirmed the cancer-promoting effect of MMP1 in pancreatic cancer. Conclusions Our pan-cancer analysis comprehensively proved that MMP1 expression is related with clinical prognosis and tumor immune infiltration, and MMP1 can become a prognostic and immunological biomarker.
Collapse
Affiliation(s)
- Shuai Mao
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Anliang Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xuewen Tao
- Department of Hepatobiliary Surgery, Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
| | - Dingde Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiamu Qu
- Department of Hepatobiliary Surgery, Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
| | - Meiling Sun
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Haowei Wei
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Guoqiang Li
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China,*Correspondence: Guoqiang Li,
| |
Collapse
|
23
|
Jin C, Wang T, Zhang D, Yang P, Zhang C, Peng W, Jin K, Wang L, Zhou J, Peng C, Tan Y, Ji J, Chen Z, Sun Q, Yang S, Tang J, Feng Y, Sun Y. Acetyltransferase NAT10 regulates the Wnt/β-catenin signaling pathway to promote colorectal cancer progression via ac 4C acetylation of KIF23 mRNA. J Exp Clin Cancer Res 2022; 41:345. [PMID: 36522719 PMCID: PMC9753290 DOI: 10.1186/s13046-022-02551-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND N4-acetylcytidine (ac4C) as a significant RNA modification has been reported to maintain the stability of mRNA and to regulate the translation process. However, the roles of both ac4C and its 'writer' protein N-acetyltransferase 10 (NAT10) played in the disease especially colorectal cancer (CRC) are unclear. At this point, we discover the underlying mechanism of NAT10 modulating the progression of CRC via mRNA ac4C modification. METHODS The clinical significance of NAT10 was explored based on the TCGA and GEO data sets and the 80 CRC patients cohort of our hospital. qRT-PCR, dot blot, WB, and IHC were performed to detect the level of NAT10 and ac4C modification in CRC tissues and matched adjacent tissues. CCK-8, colony formation, transwell assay, mouse xenograft, and other in vivo and in vitro experiments were conducted to probe the biological functions of NAT10. The potential mechanisms of NAT10 in CRC were clarified by RNA-seq, RIP-seq, acRIP-seq, luciferase reporter assays, etc. RESULTS: The levels of NAT10 and ac4C modification were significantly upregulated. Also, the high expression of NAT10 had important clinical values like poor prognosis, lymph node metastasis, distant metastasis, etc. Furthermore, the in vitro experiments showed that NAT10 could inhibit apoptosis and enhance the proliferation, migration, and invasion of CRC cells and also arrest them in the G2/M phase. The in vivo experiments discovered that NAT10 could promote tumor growth and liver/lung metastasis. In terms of mechanism, NAT10 could mediate the stability of KIF23 mRNA by binding to its mRNA 3'UTR region and up-regulating its mRNA ac4c modification. And then the protein level of KIF23 was elevated to activate the Wnt/β-catenin pathway and more β-catenin was transported into the nucleus which led to the CRC progression. Besides, the inhibitor of NAT10, remodelin, was applied in vitro and vivo which showed an inhibitory effect on the CRC cells. CONCLUSIONS NAT10 promotes the CRC progression through the NAT10/KIF23/GSK-3β/β-catenin axis and its expression is mediated by GSK-3β which forms a feedback loop. Our findings provide a potential prognosis or therapeutic target for CRC and remodelin deserves more attention.
Collapse
Affiliation(s)
- Chi Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Tuo Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Dongsheng Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Peng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Chuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Wen Peng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Kangpeng Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Jiahui Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Chaofan Peng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Yuqian Tan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Jiangzhou Ji
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Zhihao Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Qingyang Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Sheng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
| | - Junwei Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
| | - Yifei Feng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- The Colorectal Institute of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Si W, Xu X, Wan L, Lv F, Wei W, Xu X, Li W, Huang D, Zhang L, Li F. RUNX2 facilitates aggressiveness and chemoresistance of triple negative breast cancer cells via activating MMP1. Front Oncol 2022; 12:996080. [PMID: 36483054 PMCID: PMC9724742 DOI: 10.3389/fonc.2022.996080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2023] Open
Abstract
Breast cancer remains the most common malignancy in women and constantly threatens the lives of patients worldwide. State-of-the-art renewal has indicated the involvement of RUNX-associated transcription factor 2 (RUNX2) in tumorigenesis and cancer progression, yet the detailed information during breast cancer is largely obscure. Herein, we took advantage of breast cancer cell lines and in vivo tumorigenicity test as well as multifaceted phenotypic analyses (e.g., RNA-sequencing, ChIP and qRT-PCR assay) to verify the pathogenic mechanism of RUNX2 in triple negative breast cancer aggressiveness and chemoresistance. Strikingly, the proliferation, migration, invasion and chemoresistance of resistant cell lines in triple negative breast cancer was effectively suppressed by RUNX2 silencing, and the in vivo tumorigenicity was significantly weakened as well. Furthermore, with the aid of transcriptomic and bioinformatic analyses, we found MMP1 was highly expressed in triple negative breast cancer (TNBC) and showed a strong correlation with the poor prognosis of the patients, which was consistent with the expression pattern of RUNX2. Finally, by conducting ChIP and qRT-PCR assessment, we verified that RUNX2 functioned via directly binding to the specific motifs in the promoter of MMP1 and thus activating the transcriptional process. Collectively, our data demonstrated the facilitating effect of RUNX2 during triple negative breast cancer progression by directly orchestrating the expression of MMP1, which supplied overwhelming new references for RUNX2-MMP1 axis serving as a novel candidate for breast cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Wentao Si
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiaodan Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lijuan Wan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fengxu Lv
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wei Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaojun Xu
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dabing Huang
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Leisheng Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Feifei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
CAF-derived exosomal WEE2-AS1 facilitates colorectal cancer progression via promoting degradation of MOB1A to inhibit the Hippo pathway. Cell Death Dis 2022; 13:796. [PMID: 36123327 PMCID: PMC9485119 DOI: 10.1038/s41419-022-05240-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 01/22/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal components in the tumor microenvironment (TME) and closely involved in tumor progression. However, the precise biological functions and molecular mechanisms of CAFs in the TME have yet to be understood. Here, we demonstrate that WEE2-AS1 is highly expressed in the CAF-derived small extracellular vesicles (sEVs). Moreover, WEE2-AS1 is markedly higher in plasma sEVs of CRC patients than in healthy subjects and its high level predicts advanced pathological staging and poor survival. Then, we conducted a series of in vitro and in vivo experiments. Elevated expression of WEE2-AS1 in sEVs increases CRC cell proliferation in vitro. Importantly, aberrant CAF-sEVsWEE2-AS1 leads to tumor formation and progression in BALB/c nude mice and promotes AOM/DSS-induced tumorigenesis. Mechanistically, WEE2-AS1 functions as a modular scaffold for the MOB1A and E3 ubiquitin-protein ligase praja2 complexes, leading to MOB1A degradation via the ubiquitin-proteasome pathway. The Hippo pathway is then inhibited and more YAP are transported into the nucleus, where they activate downstream gene transcription. Together, our data reveal that CAF-sEVsWEE2-AS1 interacts with MOB1A, promotes degradation of MOB1A, inhibits the Hippo pathway, and facilitates the growth of CRC cells. Hence, exosomal WEE2-AS1 may be a promising therapeutic target and circulating biomarker for CRC diagnosis and prognosis.
Collapse
|
26
|
Wu J, Wu Y, Guo Q, Chen S, Wang S, Wu X, Zhu J, Ju X. SPOP promotes cervical cancer progression by inducing the movement of PD-1 away from PD-L1 in spatial localization. J Transl Med 2022; 20:384. [PMID: 36042498 PMCID: PMC9429754 DOI: 10.1186/s12967-022-03574-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/04/2022] [Indexed: 12/20/2022] Open
Abstract
Background Metastasis is a major obstacle in the treatment of cervical cancer (CC), and SPOP-mediated regulatory effects are involved in metastasis. However, the mechanisms have not been fully elucidated. Methods Proteomic sequencing and SPOP immunohistochemistry (IHC) were performed for the pelvic lymph node (pLN)-positive and non-pLN groups of CC patients. The corresponding patients were stratified by SPOP expression level for overall survival (OS) and relapse-free survival (RFS) analysis. In vitro and in vivo tests were conducted to verify the causal relationship between SPOP expression and CC metastasis. Multiplex immunofluorescence (m-IF) and the HALO system were used to analyse the mechanism, which was further verified by in vitro experiments. Results SPOP is upregulated in CC with pLN metastasis and negatively associated with patient outcome. In vitro and in vivo, SPOP promotes CC proliferation and metastasis. According to m-IF and HALO analysis, SPOP may promote CC metastasis by promoting the separation of PD-1 from PD-L1. Finally, it was further verified that SPOP can achieve immune tolerance by promoting the movement of PD-1 away from PD-L1 in spatial location and function. Conclusion This study shows that SPOP can inhibit the immune microenvironment by promoting the movement of PD-1 away from PD-L1, thereby promoting pLN metastasis of CC and resulting in worse OS and RFS. The SPOP is associated with pelvic lymph node (pLN) metastasis and prognosis in cervical cancer (CC) patients. This paper discusses the potential mechanism of pLN metastasis of CC from the perspective of spatial location. This is a multi-cross study, including clinical data, tissue microarray (TMA), multicolor immunofluorescence (m-IF), spatial immunolocalization, in vitro and in vivo functional and mechanism research fusion, from clinical to basic and clinical research.
Collapse
Affiliation(s)
- Jiangchun Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Siyu Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Simin Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jun Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Xingzhu Ju
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
27
|
Peng J, Pei S, Cui Y, Xia Y, Huang Y, Wu X, Zheng M, Weng M, Han X, Fu H, Yang L, Zhou W, Fu Z, Wang S, Xie H. Comparative analysis of transient receptor potential channel 5 opposite strand-induced gene expression patterns and protein-protein interactions in triple-negative breast cancer. Oncol Lett 2022; 24:259. [PMID: 35765270 PMCID: PMC9219028 DOI: 10.3892/ol.2022.13379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/04/2022] [Indexed: 11/06/2022] Open
Abstract
In patients with triple-negative breast cancer (TNBC), high tumour mutation burden and aberrant oncogene expression profiles are some of the causes of poor prognosis. Therefore, it is necessary to identify aberrantly expressed oncogenes, since they have the potential to serve as therapeutic targets. Transient receptor potential channel 5 opposite strand (TRPC5OS) has been previously shown to function as a novel tumour inducer. However, the underlying mechanism of TRPC5OS function in TNBC remain to be elucidated. Therefore, in the present study TRPC5OS expression was first measured in tissue samples of patients with TNBC and a panel of breast cancer cell lines (ZR-75-1, MDA-MB-453, SK-BR-3, JIMT-1, BT474 and HCC1937) by using qRT-PCR and Western blotting. Subsequently, the possible effects of TRPC5OS on MDA-MB-231 cells proliferation were determined using Cell Counting Kit-8 and 5-Ethynyl-2′-deoxyuridine assays after Lentiviral transfection of MDA-MB-231. In addition, potential interaction partners of TRPC5OS were explored using liquid chromatography-mass spectrometry (LC-MS)/MS. Gene expression patterns following TRPC5OS overexpression were also detected in MDA-MB-231 cells by using High-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) analysis were then used to systematically verify the potential interactions among the TRPC5OS-regulated genes. The potential relationship between TRPC5OS-interacting proteins and gene expression patterns were studied using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) analysis. TRPC5OS expression was found to be significantly higher in TNBC tumour tissues and breast cancer cell lines compared with luminal tumour tissues and ZR-75-1. In addition, the overexpression of TRPC5OS significantly increased cell proliferation. High-throughput sequencing results revealed that 5,256 genes exhibited differential expression following TRPC5OS overexpression, including 3,269 upregulated genes and 1,987 downregulated genes. GO analysis results indicated that the functions of these differentially expressed genes were enriched in the categories of ‘cell division’ and ‘cell proliferation’ regulation. KEGG analysis showed that the TRPC5OS-regulated genes were associated with processes of ‘homologous recombination’ and ‘TNF signalling pathways’. Subsequently, 17 TRPC5OS-interacting proteins were found using LC-MS/MS and STRING analysis. The most important protein among interacting proteins was ENO1 which was associated with glycolysis and regulated proliferation of cancer. In summary, data from the present study suggest that TRPC5OS overexpression can increase TNBC cell proliferation and ENO1 may be a potential target protein mediated by TRPC5OS. Therefore, TRPC5OS may serve as a novel therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jinghui Peng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yangyang Cui
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yiqin Xia
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yue Huang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiaowei Wu
- Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Mingjie Zheng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Miaomiao Weng
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xu Han
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hongtao Fu
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lili Yang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wenbin Zhou
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ziyi Fu
- Breast Disease Laboratory, Women and Children Central Laboratory, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
28
|
MicroRNA-147a Targets SLC40A1 to Induce Ferroptosis in Human Glioblastoma. Anal Cell Pathol 2022; 2022:2843990. [PMID: 35942174 PMCID: PMC9356897 DOI: 10.1155/2022/2843990] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/18/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. Glioblastoma is one of the most common malignant tumors in the brain, and these glioblastoma patients have very poor prognosis. Ferroptosis is involved in the progression of various tumors, including the glioblastoma. This study aims to determine the involvement of microRNA (miR)-147a in regulating ferroptosis of glioblastoma in vitro. Methods. Human glioblastoma cell lines were transfected with the inhibitor, mimic and matched negative controls of miR-147a in the presence or absence of ferroptotic inducers. To knock down the endogenous solute carrier family 40 member 1 (SLC40A1), cells were transfected with the small interfering RNA against SLC40A1. In addition, cells with or without the miR-147a mimic treatment were also incubated with temozolomide (TMZ) to investigate whether miR-147a overexpression could sensitize human glioblastoma cells to TMZ chemotherapy in vitro. Results. We found that miR-147a level was decreased in human glioblastoma tissues and cell lines and that the miR-147a mimic significantly suppressed the growth of glioblastoma cells in vitro. In addition, miR-147a expression was elevated in human glioblastoma cells upon erastin or RSL3 stimulation. Treatment with the miR-147a mimic significantly induced ferroptosis of glioblastoma cells, and the ferroptotic inhibitors could block the miR-147a mimic-mediated tumor suppression in vitro. Conversely, the miR-147a inhibitor prevented erastin- or RSL3-induced ferroptosis and increased the viability of glioblastoma cells in vitro. Mechanistically, we determined that miR-147a directly bound to the 3
-untranslated region of SLC40A1 and inhibited SLC40A1-mediated iron export, thereby facilitating iron overload, lipid peroxidation, and ferroptosis. Furthermore, miR-147a mimic-treated human glioblastoma cells exhibited higher sensitivity to TMZ chemotherapy than those treated with the mimic control in vitro. Conclusion. We for the first time determine that miR-147a targets SLC40A1 to induce ferroptosis in human glioblastoma in vitro.
Collapse
|
29
|
Upregulated circTMEM59 Inhibits Cell Growth and Metastasis by miR-668-3p/ID4 Axis in Colorectal Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7242124. [PMID: 35656024 PMCID: PMC9155906 DOI: 10.1155/2022/7242124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/06/2022] [Accepted: 04/26/2022] [Indexed: 11/18/2022]
Abstract
The incidence and mortality of colorectal cancer (CRC) are ranked in the top three worldwide in 2020. Abundant studies have reported that circular RNAs (circRNAs) act critical roles in the genesis and development of tumors, including CRC. Nevertheless, the roles and detailed regulation mechanisms of circRNAs that are related to the initiation and development of CRC have not been fully found and clarified. This research primarily revealed that circTMEM59 was greatly downregulated in CRC tissues and cell lines via qRT-PCR. In addition, the decreased expression of circTMEM59 was closely related to adverse clinicopathological characteristics and the shorter survival time of CRC patients. Then, a further study found that the overexpression of circTMEM59 suppressed cell growth and accelerated the cell death of CRC via a series of experiments in vitro and in vivo. Furthermore, circTMEM59 also repressed the metastatic behaviors of CRC cells. Further study revealed that circTMEM59 played the role of competing endogenous RNAs (ceRNAs) by binding to miR-668-3p to increase the expression of inhibitor of DNA binding 4 (ID4) in CRC. In summary, the results of this study clarified the antitumor effects of circTMEM59/miR-668-3p/ID4 axis in CRC progression and provided potential therapeutic targets and clinical prognostic markers for CRC.
Collapse
|
30
|
Liu X, Zhao T, Yuan Z, Ge S. MIR600HG sponges miR-125a-5p to regulate glycometabolism and cisplatin resistance of oral squamous cell carcinoma cells via mediating RNF44. Cell Death Discov 2022; 8:216. [PMID: 35443748 PMCID: PMC9021257 DOI: 10.1038/s41420-022-01000-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
There is increasing evidence that dysregulated long non-coding RNA (lncRNA) is implicated in tumorigenesis and progression. We aim to explore the role of lncRNA MIR600HG in glycometabolism and cisplatin (DDP) resistance of oral squamous cell carcinoma (OSCC) cells via regulating microRNA-125a-5p (miR-125a-5p) and RING finger 44 (RNF44). Expression of MIR600HG, miR-125a-5p, and RNF44 in OSCC clinical samples, cell lines, and DDP-resistant OSCC cells (SCC-9/DDP) was determined. In SCC-9 cells, proliferation, IC50 value of DDP, migration, invasion, and apoptosis were detected; in SCC-9/DDP cells, proliferation, IC50 value of DDP, apoptosis, glucose consumption, and production of lactic acid and ATP were evaluated. The interaction of MR600HG, miR-125a-5p, and RNF44 was verified. MIR600HG and RNF44 were upregulated while miR-125a-5p was downregulated in OSCC tissues and cell lines, and also in SCC-9/DDP cells. In SCC-9 cells, MIR600HG overexpression improved cell growth, metastasis, and inhibited cell susceptibility to DDP; in SCC-9/DDP cells, silencing of MIR600HG promoted apoptosis, improved DDP sensitivity, and inhibited cell glycolysis. Downregulation of miR-125a-5p showed the opposite effect to downregulation of MIR600HG. MIR600HG bound to miR-125a-5p and miR-125a-5p targeted RNF44. Downregulation of miR-125a-5p reversed the improvement of DDP sensitivity and the inhibition of cell glycolysis by downregulated MIR600HG on SCC-9/DDP cells. Downregulating RNF44 reversed the promotion of DDP resistance and cell glycolysis of SCC-9/DDP cells mediated by downregulation of miR-125a-5p. Collectively, our study addresses that MIR600HG downregulation elevates miR-125a-5p and reduces RNF44 expression, thereby improving DDP sensitivity and inhibiting glycolysis in DDP-resistant OSCC cells.
Collapse
Affiliation(s)
- Xingguang Liu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Tengda Zhao
- Department of Oral and Maxillofacial surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhe Yuan
- The Affiliated Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Shaohua Ge
- The Affiliated Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China.
| |
Collapse
|
31
|
Li J, Sun J, Liu Z, Zeng Z, Ouyang S, Zhang Z, Ma M, Kang W. The Roles of Non-Coding RNAs in Radiotherapy of Gastrointestinal Carcinoma. Front Cell Dev Biol 2022; 10:862563. [PMID: 35517505 PMCID: PMC9065280 DOI: 10.3389/fcell.2022.862563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy (RT), or radiation therapy, has been widely used in clinical practice for the treatment of local advanced gastrointestinal carcinoma. RT causes DNA double-strand breaks leading to cell cytotoxicity and indirectly damages tumor cells by activating downstream genes. Non-coding RNA (including microRNAs, long non-coding RNAs (ncRNAs), and circular RNAs) is a type of RNA that does not encode a protein. As the field of ncRNAs increasingly expands, new complex roles have gradually emerged for ncRNAs in RT. It has been shown that ncRNAs can act as radiosensitivity regulators in gastrointestinal carcinoma by affecting DNA damage repair, cell cycle arrest, irradiation-induced apoptosis, cell autophagy, stemness, EMT, and cell pyroptosis. Here, we review the complex roles of ncRNAs in RT and gastrointestinal carcinoma. We also discuss the potential clinical significance and predictive value of ncRNAs in response to RT for guiding the individualized treatment of patients. This review can serve as a guide for the application of ncRNAs as radiosensitivity enhancers, radioresistance inducers, and predictors of response in RT of gastrointestinal carcinoma.
Collapse
|
32
|
Li J, Li Z, Wang S, Bi J, Huo R. Exosomes from human adipose-derived mesenchymal stem cells inhibit production of extracellular matrix in keloid fibroblasts via downregulating transforming growth factor-β2 and Notch-1 expression. Bioengineered 2022; 13:8515-8525. [PMID: 35333672 PMCID: PMC9161879 DOI: 10.1080/21655979.2022.2051838] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/24/2022] Open
Abstract
Keloids are an excessive tissue response to dermal damage, characterized by uncontrolled growth and a high recurrence rate after various treatments. Abnormalities with the extracellular matrix (ECM) are one of the most important contributing factors to the formation of keloids. Although exosomes from human adipose-derived mesenchymal stem cells (adMSC-Exos) have been shown to promote repair and regeneration in wounds, they have seldom been studied for the treatment of keloids. In this study, we aimed to investigate the effects of adMSC-Exos on ECM remodeling in keloids using both in vitro and ex vivo models. The results showed that adMSC-Exos inhibited gene and protein expression of collagen I (COL-1), collagen III (COL-3), fibronectin (FN), and α-smooth muscle actin (α-SMA) in keloid fibroblasts (KFs). Furthermore, using an ex vivo tissue explant model, we found that adMSC-Exos significantly suppressed COL production and disrupted the microvessel stucture. We also demonstrated that adMSC-Exos inhibited the protein expression of Smad3 and Notch-1, and the expression of transforming growth factor β2 (TGF-β2) in KFs, and promoted the expression of TGF-β3. These findings largely explain the mechanisms underlying the inhibition of ECM production in keloids by adMSC-Exos. In conclusion, our results revealed that adMSC-Exos effectively inhibited the production of ECM in keloids, which provides a new potential alternative for the systemic treatment of keloids.
Collapse
Affiliation(s)
- Jing Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyu Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Song Wang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianhai Bi
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ran Huo
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
33
|
He L, Tang L, Wang R, Liu L, Zhu P, Jiang K, Tu G. Long noncoding RNA KB-1980E6.3 promotes breast cancer progression through the PI3K/AKT signalling pathway. Pathol Res Pract 2022; 234:153891. [DOI: 10.1016/j.prp.2022.153891] [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: 02/13/2022] [Revised: 03/26/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
34
|
Chen S, Ning B, Song J, Yang Z, Zhou L, Chen Z, Mao L, Liu H, Wang Q, He S, Zhou Z. Enhanced pentose phosphate pathway activity promotes pancreatic ductal adenocarcinoma progression via activating YAP/MMP1 axis under chronic acidosis. Int J Biol Sci 2022; 18:2304-2316. [PMID: 35414794 PMCID: PMC8990471 DOI: 10.7150/ijbs.69526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Acidic microenvironment is a common physiological phenomenon in tumors, and is closely related to cancer development, but the effects of acidosis on pancreatic adenocarcinoma (PDAC) remains to be elucidated. Methods: Metabonomic assay and transcriptomic microarray were used to detect the changes of metabolites and gene expression profile respectively in acidosis-adapted PDAC cells. Wound healing, transwell and in vivo assay were applied to evaluate cell migration and invasion capacity. CCK8 and colony formation assays were performed to determine cell proliferation. Results: The acidosis-adapted PDAC cells had stronger metastasis and proliferation ability compared with the control cells. Metabonomic analysis showed that acidosis-adapted PDAC cells had both increased glucose and decreased glycolysis, implying a shift to pentose phosphate pathway. The metabolic shift further led to the inactivation of AMPK by elevating ATP. Transcriptomic analysis revealed that the differentially expressed genes in acidosis-adapted cells were enriched in extracellular matrix modification and Hippo signaling. Besides, MMP1 was the most upregulated gene in acidosis-adapted cells, mediated by the YAP/TAZ pathway, but could be reduced by AMPK activator. Conclusion: The present study showed that metabolic reprogramming promotes proliferation and metastasis of acidosis-adapted PDAC cells by inhibiting AMPK/Hippo signaling, thus upregulating MMP1.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Bo Ning
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Jinwen Song
- Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Zihan Yang
- Department of Biomedical Science, City University of Hong Kong, Hong Kong SAR, China
| | - Li Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Zhiji Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Linhong Mao
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Hongtao Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Qingliang Wang
- Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Song He
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Zhihang Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| |
Collapse
|
35
|
Matrix Metalloproteinase-1 (MMP1) Upregulation through Promoter Hypomethylation Enhances Tamoxifen Resistance in Breast Cancer. Cancers (Basel) 2022; 14:cancers14051232. [PMID: 35267540 PMCID: PMC8909089 DOI: 10.3390/cancers14051232] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Cancer recurrence caused by tamoxifen resistance hampers chemotherapy in breast cancer patients. The reasons behind the resistance were investigated by screening epigenetically regulated genes through analysis of methylation data from tamoxifen-resistant MCF-7 cells. MMP1 locus was found to be hypomethylated at a promoter CpG site and its expression was upregulated in the cell line, which was verified by the drug-resistant tumor tissues from breast cancer patients (n = 28). Downregulating MMP1 using a short hairpin RNA inhibited the growth of resistant cells and increased sensitivity to tamoxifen in vitro as well as in a xenografted mouse model in vivo. This study suggests that MMP1 is potentially a target gene to control tamoxifen resistance in breast cancer. Abstract Background: Tamoxifen (tam) is widely used to treat estrogen-positive breast cancer. However, cancer recurrence after chemotherapy remains a major obstacle to achieve good patient prognoses. In this study, we aimed to identify genes responsible for epigenetic regulation of tam resistance in breast cancer. Methods: Methylation microarray data were analyzed to screen highly hypomethylated genes in tam resistant (tamR) breast cancer cells. Quantitative RT-PCR, Western blot analysis, and immunohistochemical staining were used to quantify expression levels of genes in cultured cells and cancer tissues. Effects of matrix metalloproteinase-1 (MMP1) expression on cancer cell growth and drug resistance were examined through colony formation assays and flow cytometry. Xenografted mice were generated to investigate the effects of MMP1 on drug resistance in vivo. Results: MMP1 was found to be hypomethylated and overexpressed in tamR MCF-7 (MCF-7/tamR) cells and in tamR breast cancer tissues. Methylation was found to be inversely associated with MMP1 expression level in breast cancer tissues, and patients with lower MMP1 expression exhibited a better prognosis for survival. Downregulating MMP1 using shRNA induced tam sensitivity in MCF-7/tamR cells along with increased apoptosis. The xenografted MCF-7/tamR cells that stably expressed short hairpin RNA (shRNA) against MMP1 exhibited retarded tumor growth compared to that in cells expressing the control shRNA, which was further suppressed by tam. Conclusions: MMP1 can be upregulated through promoter hypomethylation in tamR breast cancer, functioning as a resistance driver gene. MMP1 can be a potential target to suppress tamR to achieve better prognoses of breast cancer patients.
Collapse
|
36
|
Jia Z, An J, Liu Z, Zhang F. Non-Coding RNAs in Colorectal Cancer: Their Functions and Mechanisms. Front Oncol 2022; 12:783079. [PMID: 35186731 PMCID: PMC8847166 DOI: 10.3389/fonc.2022.783079] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a common malignancy with high mortality. However, the molecular mechanisms underlying CRC remain unclear. Controversies over the exact functions of non-coding RNAs (ncRNAs) in the progression of CRC have been prevailing for multiple years. Recently, accumulating evidence has demonstrated the regulatory roles of ncRNAs in various human cancers, including CRC. The intracellular signaling pathways by which ncRNAs act on tumor cells have been explored, and in CRC, various studies have identified numerous dysregulated ncRNAs that serve as oncogenes or tumor suppressors in the process of tumorigenesis through diverse mechanisms. In this review, we have summarized the functions and mechanisms of ncRNAs (mainly lncRNAs, miRNAs, and circRNAs) in the tumorigenesis of CRC. We also discuss the potential applications of ncRNAs as diagnostic and prognostic tools, as well as therapeutic targets in CRC. This review details strategies that trigger the recognition of CRC-related ncRNAs, as well as the methodologies and challenges of studying these molecules, and the forthcoming clinical applications of these findings.
Collapse
Affiliation(s)
- Zimo Jia
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Jiaqi An
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Ziyuan Liu
- School of Medicine, Shihezi University, Shihezi, China
| | - Fan Zhang
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| |
Collapse
|
37
|
Lu S, Ding X, Wang Y, Hu X, Sun T, Wei M, Wang X, Wu H. The Relationship Between the Network of Non-coding RNAs-Molecular Targets and N6-Methyladenosine Modification in Colorectal Cancer. Front Cell Dev Biol 2021; 9:772542. [PMID: 34938735 PMCID: PMC8685436 DOI: 10.3389/fcell.2021.772542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
Recent accumulating researches implicate that non-coding RNAs (ncRNAs) including microRNA (miRNA), circular RNA (circRNA), and long non-coding RNA (lncRNAs) play crucial roles in colorectal cancer (CRC) initiation and development. Notably, N6-methyladenosine (m6A) methylation, the critical posttranscriptional modulators, exerts various functions in ncRNA metabolism such as stability and degradation. However, the interaction regulation network among ncRNAs and the interplay with m6A-related regulators has not been well documented, particularly in CRC. Here, we summarize the interaction networks and sub-networks of ncRNAs in CRC based on a data-driven approach from the publications (IF > 6) in the last quinquennium (2016–2021). Further, we extend the regulatory pattern between the core m6A regulators and m6A-related ncRNAs in the context of CRC metastasis and progression. Thus, our review will highlight the clinical potential of ncRNAs and m6A modifiers as promising biomarkers and therapeutic targets for improving the diagnostic precision and treatment of CRC.
Collapse
Affiliation(s)
- Senxu Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Xiangyu Ding
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yuanhe Wang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Shenyang, China
| | - Xiaoyun Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Tong Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China.,Shenyang Kangwei Medical Laboratory Analysis Co. Ltd., Liaoning, China
| | - Xiaobin Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
38
|
Jiang T, Wang H, Liu L, Song H, Zhang Y, Wang J, Liu L, Xu T, Fan R, Xu Y, Wang S, Shi L, Zheng L, Wang R, Song J. CircIL4R activates the PI3K/AKT signaling pathway via the miR-761/TRIM29/PHLPP1 axis and promotes proliferation and metastasis in colorectal cancer. Mol Cancer 2021; 20:167. [PMID: 34922544 PMCID: PMC8684286 DOI: 10.1186/s12943-021-01474-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Accumulating studies have revealed that aberrant expression of circular RNAs (circRNAs) is widely involved in the tumorigenesis and progression of malignant cancers, including colorectal cancer (CRC). Nevertheless, the clinical significance, levels, features, biological function, and molecular mechanisms of novel circRNAs in CRC remain largely unexplored. METHODS CRC-related circRNAs were identified through bioinformatics analysis and verified in clinical specimens by qRT-PCR and in situ hybridization (ISH). Then, in vitro and in vivo experiments were performed to determine the clinical significance of, functional roles of, and clinical characteristics associated with circIL4R in CRC specimens and cells. Mechanistically, RNA pull-down, fluorescence in situ hybridization (FISH), luciferase reporter, and ubiquitination assays were performed to confirm the underlying mechanism of circIL4R. RESULTS CircIL4R was upregulated in CRC cell lines and in sera and tissues from CRC patients and was positively correlated with advanced clinicopathological features and poor prognosis. Functional experiments demonstrated that circIL4R promotes CRC cell proliferation, migration, and invasion via the PI3K/AKT signaling pathway. Mechanistically, circIL4R was regulated by TFAP2C and competitively interacted with miR-761 to enhance the expression of TRIM29, thereby targeting PHLPP1 for ubiquitin-mediated degradation to activate the PI3K/AKT signaling pathway and consequently facilitate CRC progression. CONCLUSIONS Our findings demonstrate that upregulation of circIL4R plays an oncogenic role in CRC progression and may serve as a promising diagnostic and prognostic biomarker for CRC detection and as a potential therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Hongyu Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Lianyu Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hu Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yi Zhang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Jiaqi Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Lei Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Teng Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Ruizhi Fan
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Shuai Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Linsen Shi
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Li Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Renhao Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China. .,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China. .,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
39
|
Xu M, Cui R, Ye L, Wang Y, Wang X, Zhang Q, Wang K, Dong C, Le W, Chen B. LINC00941 promotes glycolysis in pancreatic cancer by modulating the Hippo pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:280-294. [PMID: 34513310 PMCID: PMC8413680 DOI: 10.1016/j.omtn.2021.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/07/2021] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of most lethal cancers and is projected to be the second leading cause of cancer deaths in the United States by 2030. The lack of effective treatment and increased incidence in PDAC encourage a deeper knowledge of PDAC progression. By analyzing a long noncoding RNA (lncRNA) dataset, we found that increased LINC00941 expression led to poor outcomes in PDAC patients. Furthermore, in vitro and in vivo experiments revealed that LINC00941 promoted PDAC cancer cell growth by enhancing aerobic glycolysis. Mechanistically, LINC00941 was found to interact with mammalian STE20-like protein kinase 1 (MST1), which facilitated the protein phosphatase 2A (PP2A)-mediated dephosphorylation of MST1, resulting in Hippo pathway activation and consequently, enhanced glycolysis in PDAC. These results suggest that LINC00941 plays a key role in regulating PDAC tumorigenesis, potentially highlighting novel avenues for PDAC therapy.
Collapse
Affiliation(s)
- Ming Xu
- Department of Gastroenterology, Pudong New Area People’s Hospital, Shanghai 201200, China
| | - Ran Cui
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Lunhe Ye
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Xujing Wang
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Qiqi Zhang
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Kaijing Wang
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Chunxiu Dong
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Wenjun Le
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
- Corresponding author: Wenjun Le, Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200123, China
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
- Corresponding author: Bo Chen, Department of Hepatopancreatobiliary Surgery, Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200123, China.
| |
Collapse
|
40
|
Shi X, Huang X, Chen R, Li Y, Xu Y, Zhang W, Zhu Q, Zha X, Wang J. The transcribed ultraconserved element uc.51 promotes the proliferation and metastasis of breast cancer by stabilizing NONO. Clin Exp Metastasis 2021; 38:551-571. [PMID: 34714466 DOI: 10.1007/s10585-021-10128-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022]
Abstract
Long noncoding RNAs have recently emerged as significant contributors to cancers, including breast cancer (BC). One class of long noncoding RNAs called transcribed ultraconserved regions (T-UCRs) is highly conserved in many species and closely related to diverse physiological and pathological processes. However, the function of T-UCRs in BC remains largely unclear. In this study, we identified uc.51, a T-UCR that is overexpressed in both BC tissues and cell lines and is correlated with larger tumor size. Loss- and gain-of-function assays were performed in vitro and demonstrated that uc.51 promotes the proliferation, migration, and invasion of BC cells. Mechanistically, non-POU domain-containing octamer-binding protein (NONO) was found to physically interact with uc.51 by RNA pulldown followed by mass spectrometry. This interaction was further verified by RNA immunoprecipitation. Moreover, uc.51 positively regulated the expression of NONO, maintained its stability through the ubiquitin-proteasome system, and activated the phosphorylation of CREB. Rescue experiments demonstrated that NONO overexpression compensated for the attenuated influence on BC progression resulting from downregulation of uc.51, indicating that NONO functions downstream of uc.51. In vivo functional experiments also revealed a positive correlation between uc.51 expression and tumor size. Ki-67 and NONO levels in the lv-uc.51-shRNA group were decreased compared with those in the lv-con-shRNA group, according to the immunohistochemical staining results, and a decreased incidence of distant metastasis was observed in the lv-uc.51-shRNA group in the xenograft model. Collectively, our results reveal a substantial role for the uc.51-NONO axis in BC progression and indicate that the uc.51-NONO axis has potential to be a therapeutic target for BC.
Collapse
Affiliation(s)
- Xiaoqing Shi
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Xiaofeng Huang
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Rui Chen
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Yan Li
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Yinggang Xu
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Weiwei Zhang
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Qiannan Zhu
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China
| | - Xiaoming Zha
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210000, China.
| | - Jue Wang
- Department of Breast Disease, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210000, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
41
|
Guo T, Liu D, Peng S, Wang M, Li Y. A Positive Feedback Loop of lncRNA MIR31HG-miR-361-3p -YY1 Accelerates Colorectal Cancer Progression Through Modulating Proliferation, Angiogenesis, and Glycolysis. Front Oncol 2021; 11:684984. [PMID: 34485123 PMCID: PMC8416113 DOI: 10.3389/fonc.2021.684984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022] Open
Abstract
Background Colorectal cancer (CRC) is a common malignant tumor with high metastatic and recurrent rates. This study probes the effect and mechanism of long non-coding RNA MIR31HG on the progression of CRC cells. Materials and Methods Quantitative real-time PCR (qRT-PCR) was used to analyze the expression of MIR31HG and miR-361-3p in CRC tissues and normal tissues. Gain- or loss-of-function assays were conducted to examine the roles of MIR31HG, miR-361-3p and YY1 transcription factor (YY1) in the CRC progression. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and colony formation experiment were conducted to test CRC cell proliferation. CRC cell invasion was determined by Transwell assay. The glucose detection kit and lactic acid detection kit were utilized to monitor the levels of glucose and lactate in CRC cells. The glycolysis level in CRC cells was examined by the glycolytic stress experiment. Western blot was performed to compare the expression of glycolysis-related proteins (PKM2, GLUT1 and HK2) and angiogenesis-related proteins (including VEGFA, ANGPT1, HIF1A and TIMP1) in HUVECs. The binding relationships between MIR31HG and miR-361-3p, miR-361-3p and YY1 were evaluated by the dual-luciferase reporter assay and RNA immunoprecipitation (RIP). Results MIR31HG was up-regulated in CRC tissues and was associated with poorer prognosis of CRC patients. The in-vitro and in-vivo experiments confirmed that overexpressing MIR31HG heightened the proliferation, growth, invasion, glycolysis and lung metastasis of CRC cells as well as the angiogenesis of HUVECs. In addition, MIR3HG overexpression promoted YY1 mRNA and protein level, and forced overexpression of YY1 enhanced MIR31HG level. Overexpressing YY1 reversed the tumor-suppressive effect mediated by MIR31HG knockdown. miR-361-3p, which was inhibited by MIR31HG overexpression, repressed the malignant behaviors of CRC cells. miR-361-3p-mediated anti-tumor effects were mostly reversed by upregulating MIR31HG. Further mechanism studies illustrated that miR-361-3p targeted and negatively regulated the expression of YY1. Conclusion This study reveals that MIR31HG functions as an oncogenic gene in CRC via forming a positive feedback loop of MIR31HG-miR-361-3p-YY1.
Collapse
Affiliation(s)
- Tao Guo
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Defeng Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shihao Peng
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meng Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyang Li
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
Peng C, Tan Y, Yang P, Jin K, Zhang C, Peng W, Wang L, Zhou J, Chen R, Wang T, Jin C, Ji J, Feng Y, Tang J, Sun Y. Circ-GALNT16 restrains colorectal cancer progression by enhancing the SUMOylation of hnRNPK. J Exp Clin Cancer Res 2021; 40:272. [PMID: 34452628 PMCID: PMC8400830 DOI: 10.1186/s13046-021-02074-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/14/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Recent studies have investigated the role of circular RNAs (circRNAs) as significant regulatory factors in multiple cancer progression. Nevertheless, the biological functions of circRNAs and the underlying mechanisms by which they regulate colorectal cancer (CRC) progression remain unclear. METHODS A novel circRNA (circ-GALNT16) was identified by microarray and qRT-PCR. A series of in vitro and in vivo phenotype experiments were performed to investigate the role of circ-GALNT16 in CRC. The FISH, RNA pulldown assay, RIP assay, RNA sequencing, coimmunoprecipitation, and ChIP were performed to investigate the molecular mechanisms of circ-GALNT16 in CRC progression. RESULTS Circ-GALNT16 was downregulated in CRC and was negatively correlated with poor prognosis. Circ-GALNT16 suppressed the proliferation and metastatic ability of CRC cells in vitro and in vivo. Mechanistically, circ-GALNT16 could bind to the KH3 domain of heterogeneous nuclear ribonucleoprotein K (hnRNPK), which promoted the SUMOylation of hnRNPK. Additionally, circ-GALNT16 could enhance the formation of the hnRNPK-p53 complex by facilitating the SUMOylation of hnRNPK. RNA sequencing assay identified serpin family E member 1 as the target gene of circ-GALNT16 at the transcriptional level. Rescue assays revealed that circ-GALNT16 regulated the expression of Serpine1 by inhibiting the deSUMOylation of hnRNPK mediated by SUMO-specific peptidase 2 and then regulating the sequence-specific DNA binding ability of the hnRNPK-p53 transcriptional complex. CONCLUSIONS Circ-GALNT16 suppressed CRC progression by inhibiting Serpine1 expression through regulating the sequence-specific DNA binding ability of the SENP2-mediated hnRNPK-p53 transcriptional complex and might function as a biomarker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Chaofan Peng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Yuqian Tan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Peng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Kangpeng Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Chuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Wen Peng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Jiahui Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Ranran Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Tuo Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Chi Jin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Jiangzhou Ji
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Yifei Feng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Junwei Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- Nanjing Medical University, Nanjing, China.
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, 210029, Nanjing, People's Republic of China.
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
- Nanjing Medical University, Nanjing, China.
| |
Collapse
|
43
|
Sun J, He D, Fu Y, Zhang R, Guo H, Wang Z, Wang Y, Gao T, Wei Y, Guo Y, Pang Q, Liu Q. A novel lncRNA ARST represses glioma progression by inhibiting ALDOA-mediated actin cytoskeleton integrity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:187. [PMID: 34099027 PMCID: PMC8183030 DOI: 10.1186/s13046-021-01977-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/09/2021] [Indexed: 12/03/2022]
Abstract
Background Glioma is one of the most aggressive malignant brain tumors that is characterized with inevitably infiltrative growth and poor prognosis. ARST is a novel lncRNA whose expression level is significantly decreased in the patients with glioblastoma multiforme. However, the exact mechanisms of ARST in gliomagenesis are largely unknown. Methods The expressions of ARST in the glioma samples and cell lines were analyzed by qRT-PCR. FISH was utilized to detect the distribution of ARST in the glioma cells. CCK-8, EdU and flow cytometry were used to examine cellular viability, proliferation and apoptosis. Transwell and wound-healing assays were performed to determine the migratory and invasive abilities of the cells. Intracranial tumorigenesis models were established to explore the roles of ARST in vivo. RNA pulldown assay was used to examine proteins that bound to ARST. The activities of key enzymes in the glycolysis and production of lactate acid were measured by colorimetry. In addition, RIP, Co-IP, western blot and immunofluorescence were used to investigate the interaction and regulation between ARST, F-actin, ALDOA and cofilin. Results In this study, we reported that ARST was downregulated in the gliomas. Overexpression of ARST in the glioma cells significantly suppressed various cellular vital abilities such as cell growth, proliferation, migration and invasion. The tumorigenic capacity of these cells in vivo was reduced as well. We further demonstrated that the tumor suppressive effects of ARST could be mediated by a direct binding to a glycolytic enzyme aldolase A (ALDOA), which together with cofilin, keeping the polymerization and depolymerization of actin filaments in an orderly dynamic equilibrium. Upregulation of ARST interrupted the interaction between ALDOA and actin cytoskeleton, which led to a rapid cofilin-dependent loss of F-actin stress fibers. Conclusions Taken together, it is concluded that ARST performs its function via a non-metabolic pathway associated with ALDOA, which otherwise modifies the morphology and invasive properties of the glioma cells. This has added new perspective to its role in tumorigenesis, thus providing potential target for glioma diagnosis, therapy, and prognosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01977-9.
Collapse
Affiliation(s)
- Jun Sun
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China.,Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Dong He
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China.,Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yibing Fu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Rui Zhang
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hua Guo
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Zhaojuan Wang
- Department of Physiology, Shandong Medical College, Jinan, 250012, Shandong, People's Republic of China
| | - Yanan Wang
- Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China.,Department of Pathology, Tai-an Municipal Hospital, Jinan, 250012, Shandong, People's Republic of China
| | - Taihong Gao
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yanbang Wei
- Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yuji Guo
- Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
44
|
Li J, Han X, Gu Y, Wu J, Song J, Shi Z, Chang H, Liu M, Zhang Y. LncRNA MTX2-6 Suppresses Cell Proliferation by Acting as ceRNA of miR-574-5p to Accumulate SMAD4 in Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:654746. [PMID: 33869216 PMCID: PMC8044847 DOI: 10.3389/fcell.2021.654746] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has been one of the key causes of cancer deaths worldwide. It has been found that long non-coding RNA (lncRNA) is related to the generation and progression of various cancers (including ESCC). However, there are still many lncRNAs related to ESCC whose functions and molecular mechanisms have not been clearly elucidated. In this study, we first reported that lncRNA MTX2-6 was significantly downregulated in ESCC tissues and cell lines. The decreased expression of MTX2-6 is closely related to larger tumor and worse prognosis of ESCC patients. Through a series of functional experiments, we detected that overexpressed MTX2-6 inhibited cell proliferation and promoted cell apoptosis of ESCC in vitro and in vivo. Further studies showed that MTX2-6 exerts as a competing endogenous RNA (ceRNA) by binding miR-574-5p and elevates the expression of SMAD4 in ESCC. In summary, our results clarify the tumor suppressor roles of MTX2-6/miR-574-5p/SMAD4 axis in the progression of ESCC and provide emerging therapeutic targets for ESCC patients.
Collapse
Affiliation(s)
- Jie Li
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xu Han
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Yan Gu
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Jixiang Wu
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Jianxiang Song
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Zhan Shi
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Huiwen Chang
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Ming Liu
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Yajun Zhang
- Department of Cardiothoracic Surgery, Yancheng Third People's Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng, China.,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| |
Collapse
|
45
|
Li J, Yang P, Chen F, Tan Y, Huang C, Shen H, Peng C, Feng Y, Sun Y. Hypoxic colorectal cancer-derived extracellular vesicles deliver microRNA-361-3p to facilitate cell proliferation by targeting TRAF3 via the noncanonical NF-κB pathways. Clin Transl Med 2021; 11:e349. [PMID: 33784010 PMCID: PMC7967919 DOI: 10.1002/ctm2.349] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hypoxic tumour microenvironment (TME) is a key regulator in cancer progression. However, the communications between hypoxic cells and other components in TME during colorectal cancer (CRC) progression via extracellular vesicles (EVs) remain unclear. METHODS High-throughput sequencing was employed to detect aberrantly expressed microRNAs (miRNAs) in hypoxic EVs. Quantitative real-time PCR was used to confirm and screen preliminarily candidate miRNAs. The effects of EVs derived from hypoxia (<1% O2 ) and miR-361-3p on CRC growth were assessed using CCK-8 assays, colony formation assays, EdU assays, flow cytometric assays and mouse xenograft. Then, the specific mechanisms of miR-361-3p were investigated by RNA immunoprecipitation, luciferase reporter assay, Western blot, chromatin immunoprecipitation, immunohistochemistry and rescue experiments. RESULTS The level of miR-361-3p expression was remarkably elevated in hypoxic EVs and can be transferred to CRC cells. Functional experiments exhibited that hypoxic EVs facilitated cell growth and suppressed cell apoptosis by transferring miR-361-3p of CRC. Hypoxia-inducible factor-1α induced the elevation of miR-361-3p levels in hypoxic EVs. Upregulated miR-361-3p in CRC inhibited cell apoptosis and facilitated cell growth by directly targeting TNF receptor-associated factor 3, which consequently activated the noncanonical NF-κB pathway. Moreover, the high expression of circulating exosomal miR-361-3p was correlated to worse prognosis of CRC patients. CONCLUSIONS Altogether, the abnormality of exosomal miR-361-3p derived from hypoxia acts vital roles in the regulation of CRC growth and apoptosis and can be an emerging prognostic biomarker and a therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Jie Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Peng Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Fangyu Chen
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuqian Tan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Changzhi Huang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Hengyang Shen
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Chaofan Peng
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yifei Feng
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yueming Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
46
|
Anderson G. Tumour Microenvironment: Roles of the Aryl Hydrocarbon Receptor, O-GlcNAcylation, Acetyl-CoA and Melatonergic Pathway in Regulating Dynamic Metabolic Interactions across Cell Types-Tumour Microenvironment and Metabolism. Int J Mol Sci 2020; 22:E141. [PMID: 33375613 PMCID: PMC7795031 DOI: 10.3390/ijms22010141] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
This article reviews the dynamic interactions of the tumour microenvironment, highlighting the roles of acetyl-CoA and melatonergic pathway regulation in determining the interactions between oxidative phosphorylation (OXPHOS) and glycolysis across the array of cells forming the tumour microenvironment. Many of the factors associated with tumour progression and immune resistance, such as yin yang (YY)1 and glycogen synthase kinase (GSK)3β, regulate acetyl-CoA and the melatonergic pathway, thereby having significant impacts on the dynamic interactions of the different types of cells present in the tumour microenvironment. The association of the aryl hydrocarbon receptor (AhR) with immune suppression in the tumour microenvironment may be mediated by the AhR-induced cytochrome P450 (CYP)1b1-driven 'backward' conversion of melatonin to its immediate precursor N-acetylserotonin (NAS). NAS within tumours and released from tumour microenvironment cells activates the brain-derived neurotrophic factor (BDNF) receptor, TrkB, thereby increasing the survival and proliferation of cancer stem-like cells. Acetyl-CoA is a crucial co-substrate for initiation of the melatonergic pathway, as well as co-ordinating the interactions of OXPHOS and glycolysis in all cells of the tumour microenvironment. This provides a model of the tumour microenvironment that emphasises the roles of acetyl-CoA and the melatonergic pathway in shaping the dynamic intercellular metabolic interactions of the various cells within the tumour microenvironment. The potentiation of YY1 and GSK3β by O-GlcNAcylation will drive changes in metabolism in tumours and tumour microenvironment cells in association with their regulation of the melatonergic pathway. The emphasis on metabolic interactions across cell types in the tumour microenvironment provides novel future research and treatment directions.
Collapse
Affiliation(s)
- George Anderson
- Clinical Research Communications (CRC) Scotland & London, Eccleston Square, London SW1V 6UT, UK
| |
Collapse
|