1
|
Wang Q, Sun S, Sun G, Han B, Zhang S, Zheng X, Chen L. Histone modification inhibitors: An emerging frontier in thyroid Cancer therapy. Cell Signal 2025; 131:111703. [PMID: 40044017 DOI: 10.1016/j.cellsig.2025.111703] [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: 11/28/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 04/15/2025]
Abstract
Thyroid cancer (TC) is the most common endocrine cancer and is a serious health concern due to its aggressiveness and high incidence. Histone modifications affect DNA accessibility and gene transcriptional activity by altering the structure of chromatin. Abnormal histone modifications may affect genome stability and disrupt gene expression patterns, leading to many diseases, including cancer. A growing body of research suggests that histone modifications and TC progression are inextricably linked. This article discusses the impact of aberrant histone modification patterns on TC. By targeting specific histone-modifying enzymes, it may be possible to regulate gene expression and inhibit the growth of TC. Finally, we summarize the relevant histone modification inhibitors to better understand the development stage of the use of these drugs to inhibit histone-modifying enzymes in cancer treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shu Sun
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Guojun Sun
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Bing Han
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Song Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Xiaowei Zheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| | - Lu Chen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China.
| |
Collapse
|
2
|
Hou X, Yu T, Chol CH, An J, Zhang L, Sun M, Mecinović J, Gao P, Li L. Identification and insights into the uncommon configuration of novel histone lysine acetyltransferase PCAF and methyltransferase GLP inhibitors from Wikstroemia chamaedaphne by molecular docking, and in vitro assessment. Int J Biol Macromol 2025; 304:140601. [PMID: 39900167 DOI: 10.1016/j.ijbiomac.2025.140601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Biomedically important human histone acetyltransferase PCAF and histone methyltransferase GLP have emerged as interesting epigenetic targets for the development of anticancer chemotherapeutics. In this study, two novel daphnane-type diterpenes, along with ten related known ones were obtained from the flower buds of Wikstroemia chamaedaphne. Compounds 1 and 2, a ring cleavage 1α-alkyldaphnane-type backbone diterpenoids with uncommon H-10 β configuration, were elucidated by the integrated analyses of NMR spectra and experimental and calculated ECD. Compound 3 displays the most potent inhibitory activity against the histone lysine acetyltransferase PCAF and histone methyltransferase GLP. Compounds 1-3 could also bind to GLP and PCAF through hydrogen bonding, alkyl and van der Waals interactions and exhibited a high affinity as supported by the molecular docking results. In addition, molecular dynamics simulations revealed the stability of the PCAF-3 complex during the entire simulation process. Selected daphnane-type diterpenes exhibited potent activity against human BXPC-3 pancreatic cells. The cell cycle arrest experiment revealed that daphnane-type diterpenes could markedly arrest S-phases or G2/M-phase in BXPC-3 cells to inhibit cell proliferation. Overall, the results imply that daphnane-based natural products could be used as promising scaffolds for rational drug design in treatment of human diseases.
Collapse
Affiliation(s)
- Xuefen Hou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tongtong Yu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Choe Hyok Chol
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116027, China
| | - Jingkai An
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116027, China
| | - Meijia Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Pinyi Gao
- College of Pharmaceutical and Biotechnology Engineering, Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Lingzhi Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
3
|
Mowoe MO, Allam H, Nqada J, Bernon M, Gandhi K, Burmeister S, Kotze U, Kahn M, Kloppers C, Dharshanan S, Azween Z, Maimela P, Townsend P, Jonas E, Blackburn JM. Identification and validation of a novel autoantibody biomarker panel for differential diagnosis of pancreatic ductal adenocarcinoma. Front Immunol 2025; 16:1494446. [PMID: 39949781 PMCID: PMC11821970 DOI: 10.3389/fimmu.2025.1494446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/07/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction New biomarkers are urgently needed to detect pancreatic ductal adenocarcinoma (PDAC) at an earlier stage for individualized treatment strategies and to improve outcomes. Autoantibodies (AAbs) in principle make attractive biomarkers as they arise early in disease, report on disease-associated perturbations in cellular proteomes, and are static in response to other common stimuli, yet are measurable in the periphery, potentially well in advance of the onset of clinical symptoms. Methods Here, we used high-throughput, custom cancer antigen microarrays to identify a clinically relevant autoantibody biomarker combination able to differentially detect PDAC. Specifically, we quantified the serological AAb profiles of 94 PDAC, chronic pancreatitis (CP), other pancreatic- (PC) and prostate cancers (PRC), non-ulcer dyspepsia patients (DYS), and healthy controls (HC). Results Combinatorial ROC curve analysis on the training cohort data from the cancer antigen microarrays identified the most effective biomarker combination as CEACAM1-DPPA2-DPPA3-MAGEA4-SRC-TPBG-XAGE3 with an AUC = 85·0% (SE = 0·828, SP = 0·684). Additionally, differential expression analysis on the samples run on the iOme™ array identified 4 biomarkers (ALX1-GPA33-LIP1-SUB1) upregulated in PDAC against diseased and healthy controls. Identified AAbs were validated in silico using public immunohistochemistry datasets and experimentally using a custom PDAC protein microarray comprising the 11 optimal AAb biomarker panel. The clinical utility of the biomarker panel was tested in an independent cohort comprising 223 PDAC, PC, PRC, colorectal cancer (CRC), and HC samples. Combinatorial ROC curve analysis on the validation data identified the most effective biomarker combination to be CEACAM1-DPPA2-DPPA3-MAGEA4-SRC-TPBG-XAGE3 with an AUC = 85·0% (SE = 0·828, SP = 0·684). Subsequently, the specificity of the 11-biomarker panel was validated against other cancers (PDAC vs PC: AUC = 70·3%; PDAC vs CRC: AUC = 84·3%; PDAC vs PRC: AUC = 80·2%) and healthy controls (PDAC vs HC: AUC = 80·9%), confirming that this novel AAb biomarker panel is able to selectively detect PDAC amongst other confounding diseases. Conclusion This AAb panel may therefore have the potential to form the basis of a novel diagnostic test for PDAC.
Collapse
Affiliation(s)
- Metoboroghene O Mowoe
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hisham Allam
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Joshua Nqada
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Marc Bernon
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Karan Gandhi
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Sean Burmeister
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Urda Kotze
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Miriam Kahn
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Christo Kloppers
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Suba Dharshanan
- Recombinant Protein Facility, Sengenics Corporation, Kuala Lumpur, Malaysia
| | - Zafirah Azween
- Recombinant Protein Facility, Sengenics Corporation, Kuala Lumpur, Malaysia
| | - Pamela Maimela
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Paul Townsend
- Faculty of Health Sciences and Sports, University of Stirling, Stirling, United Kingdom
| | - Eduard Jonas
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Pan B, Liu C, Su J, Xia C. Activation of AMPK inhibits cervical cancer growth by hyperacetylation of H3K9 through PCAF. Cell Commun Signal 2024; 22:306. [PMID: 38831454 PMCID: PMC11145780 DOI: 10.1186/s12964-024-01687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/28/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Dysregulation in histone acetylation, a significant epigenetic alteration closely associated with major pathologies including cancer, promotes tumorigenesis, inactivating tumor-suppressor genes and activating oncogenic pathways. AMP-activated protein kinase (AMPK) is a cellular energy sensor that regulates a multitude of biological processes. Although a number of studies have identified the mechanisms by which AMPK regulates cancer growth, the underlying epigenetic mechanisms remain unknown. METHODS The impact of metformin, an AMPK activator, on cervical cancer was evaluated through assessments of cell viability, tumor xenograft model, pan-acetylation analysis, and the role of the AMPK-PCAF-H3K9ac signaling pathway. Using label-free quantitative acetylproteomics and chromatin immunoprecipitation-sequencing (ChIP) technology, the activation of AMPK-induced H3K9 acetylation was further investigated. RESULTS In this study, we found that metformin, acting as an AMPK agonist, activates AMPK, thereby inhibiting the proliferation of cervical cancer both in vitro and in vivo. Mechanistically, AMPK activation induces H3K9 acetylation at epigenetic level, leading to chromatin remodeling in cervical cancer. This also enhances the binding of H3K9ac to the promoter regions of multiple tumor suppressor genes, thereby promoting their transcriptional activation. Furthermore, the absence of PCAF renders AMPK activation incapable of inducing H3K9 acetylation. CONCLUSIONS In conclusion, our findings demonstrate that AMPK mediates the inhibition of cervical cancer growth through PCAF-dependent H3K9 acetylation. This discovery not only facilitates the clinical application of metformin but also underscores the essential role of PCAF in AMPK activation-induced H3K9 hyperacetylation.
Collapse
Affiliation(s)
- Botao Pan
- Foshan Women and Children Hospital, Foshan, 528000, China
| | - Can Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China
| | - Jiyan Su
- Foshan Women and Children Hospital, Foshan, 528000, China
| | - Chenglai Xia
- Foshan Women and Children Hospital, Foshan, 528000, China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China.
| |
Collapse
|
5
|
Histone Modifications Represent a Key Epigenetic Feature of Epithelial-to-Mesenchyme Transition in Pancreatic Cancer. Int J Mol Sci 2023; 24:ijms24054820. [PMID: 36902253 PMCID: PMC10003015 DOI: 10.3390/ijms24054820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignant diseases due to its high invasiveness, early metastatic properties, rapid disease progression, and typically late diagnosis. Notably, the capacity for pancreatic cancer cells to undergo epithelial-mesenchymal transition (EMT) is key to their tumorigenic and metastatic potential, and is a feature that can explain the therapeutic resistance of such cancers to treatment. Epigenetic modifications are a central molecular feature of EMT, for which histone modifications are most prevalent. The modification of histones is a dynamic process typically carried out by pairs of reverse catalytic enzymes, and the functions of these enzymes are increasingly relevant to our improved understanding of cancer. In this review, we discuss the mechanisms through which histone-modifying enzymes regulate EMT in pancreatic cancer.
Collapse
|
6
|
Metformin Induces Apoptosis in Human Pancreatic Cancer (PC) Cells Accompanied by Changes in the Levels of Histone Acetyltransferases (Particularly, p300/CBP-Associated Factor (PCAF) Protein Levels). Pharmaceuticals (Basel) 2023; 16:ph16010115. [PMID: 36678613 PMCID: PMC9863441 DOI: 10.3390/ph16010115] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
Accumulating evidence (mainly from experimental research) suggests that metformin possesses anticancer properties through the induction of apoptosis and inhibition of the growth and proliferation of cancer cells. However, its effect on the enzymes responsible for histone acetylation status, which plays a key role in carcinogenesis, remains unclear. Therefore, the aim of our study was to evaluate the impact of metformin on histone acetyltransferases (HATs) (i.e., p300/CBP-associated factor (PCAF), p300, and CBP) and on histone deacetylases (HDACs) (i.e., SIRT-1 in human pancreatic cancer (PC) cell lines, 1.2B4, and PANC-1). The cells were exposed to metformin, an HAT inhibitor (HATi), or a combination of an HATi with metformin for 24, 48, or 72 h. Cell viability was determined using an MTT assay, and the percentage of early apoptotic cells was determined with an Annexin V-Cy3 Apoptosis Detection Assay Kit. Caspase-9 activity was also assessed. SIRT-1, PCAF, p300, and CBP expression were determined at the mRNA and protein levels using RT-PCR and Western blotting methods, respectively. Our results reveal an increase in caspase-9 in response to the metformin, indicating that it induced the apoptotic death of both 1.2B4 and PANC-1 cells. The number of cells in early apoptosis and the activity of caspase-9 decreased when treated with an HATi alone or a combination of an HATi with metformin, as compared to metformin alone. Moreover, metformin, an HATi, and a combination of an HATi with metformin also modified the mRNA expression of SIRT-1, PCAF, CBP, and p300. However, metformin did not change the expression of the studied genes in 1.2B4 cells. The results of the Western blot analysis showed that metformin diminished the protein expression of PCAF in both the 1.2B4 and PANC-1 cells. Hence, it appears possible that PCAF may be involved in the metformin-mediated apoptosis of PC cells.
Collapse
|
7
|
Bhattacharya A. Epigenetic modifications and regulations in gastrointestinal diseases. EPIGENETICS IN ORGAN SPECIFIC DISORDERS 2023:497-543. [DOI: 10.1016/b978-0-12-823931-5.00005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Wang Y, Han Y, Jin Y, He Q, Wang Z. The Advances in Epigenetics for Cancer Radiotherapy. Int J Mol Sci 2022; 23:ijms23105654. [PMID: 35628460 PMCID: PMC9145982 DOI: 10.3390/ijms23105654] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 02/08/2023] Open
Abstract
Cancer is an important factor threatening human life and health; in recent years, its morbidity and mortality remain high and demosntrate an upward trend. It is of great significance to study its pathogenesis and targeted therapy. As the complex mechanisms of epigenetic modification has been increasingly discovered, they are more closely related to the occurrence and development of cancer. As a reversible response, epigenetic modification is of great significance for the improvement of classical therapeutic measures and the discovery of new therapeutic targets. It has become a research focusto explore the multi-level mechanisms of RNA, DNA, chromatin and proteins. As an important means of cancer treatment, radiotherapy has made great progress in technology, methods, means and targeted sensitization after years of rapid development, and even research on radiotherapy based on epigenetic modification is rampant. A series of epigenetic effects of radiation on DNA methylation, histone modification, chromosome remodeling, RNA modification and non-coding RNA during radiotherapy affects the therapeutic effects and prognosis. Starting from the epigenetic mechanism of tumorigenesis, this paper reviews the latest progress in the mechanism of interaction between epigenetic modification and cancer radiotherapy and briefly introduces the main types, mechanisms and applications of epigenetic modifiers used for radiotherapy sensitization in order to explore a more individual and dynamic approach of cancer treatment based on epigenetic mechanism. This study strives to make a modest contribution to the progress of human disease research.
Collapse
Affiliation(s)
| | | | | | - Qiang He
- Correspondence: (Q.H.); (Z.W.); Tel.: +86-431-85619443 (Z.W.)
| | - Zhicheng Wang
- Correspondence: (Q.H.); (Z.W.); Tel.: +86-431-85619443 (Z.W.)
| |
Collapse
|
9
|
Zhou X, Wang N, Zhang Y, Yu H, Wu Q. KAT2B is an immune infiltration-associated biomarker predicting prognosis and response to immunotherapy in non-small cell lung cancer. Invest New Drugs 2021; 40:43-57. [PMID: 34499335 DOI: 10.1007/s10637-021-01159-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Over the past few years, dramatic breakthroughs in the field of tumor immunotherapy with immune checkpoint inhibitors (ICIs) have made a therapeutic revolution for non-small cell lung cancer (NSCLC). While only some patients present a favorable response to this treatment. It is urgent to explore the potential molecular mechanisms underlying the regulation of tumor immune microenvironment in the process of immunotherapy. Lysine acetyltransferase 2B (KAT2B) plays a crucial role in the regulation of gene expression at the post-transcriptional level by acetylation, and is associated with many types of cancer. METHODS RNA-sequencing data, genetic mutation data, and corresponding clinical information were extracted from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, then subjected to immune characteristics, gene expression, survival, genetic alteration, enrichment analyses. RESULTS KAT2B expression correlated positively with infiltrating levels of multiple immune cells and mRNA expression levels of immune checkpoint genes in NSCLC. Furthermore, KAT2B expression was downregulated in tumor tissues, and low KAT2B expression was associated with unsatisfactory efficacy of immune checkpoint blockade (ICB) and poor prognosis of patients with lung adenocarcinoma. Moreover, there were higher somatic genes mutation frequency in patients with low expression of KAT2B. Finally, functional enrichment analysis suggested that KAT2B was mainly linked to the regulation of immune cells and interferon - gamma (IFN-γ) mediated signaling pathways, response to IFN-γ, antigen processing and presentation. CONCLUSION This is the first comprehensive study to disclose that KAT2B is correlated with immune infiltrates and may serve as a novel biomarker predicting prognosis and response to immunotherapy in NSCLC.
Collapse
Affiliation(s)
- Xue Zhou
- Haihe Clinical College of Tianjin Medical University, Tianjin, 300350, China.,Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Ning Wang
- Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Yuefeng Zhang
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Hongzhi Yu
- Department of Respiratory Medicine, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Qi Wu
- Haihe Clinical College of Tianjin Medical University, Tianjin, 300350, China. .,Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China. .,Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
10
|
Jiang R, Zhang Z, Zhong Z, Zhang C. Long-non-coding RNA RUSC1-AS1 accelerates osteosarcoma development by miR-101-3p-mediated Notch1 signalling pathway. J Bone Oncol 2021; 30:100382. [PMID: 34367901 PMCID: PMC8326430 DOI: 10.1016/j.jbo.2021.100382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/07/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023] Open
Abstract
Background Long non-coding RNA (lncRNA) RUSC1-AS1 has been found to modulate several cancers development. In this study, we explored the role of RUSC1-AS1 on osteosarcoma (OS) progression. Methods Quantitative Real-time PCR (qRT-PCR) was conducted to test the relative expression of RUSC1-AS1, Notch1 mRNA and miR-101-3p in OS tissues and adjacent normal tissues. Gain- or loss- of functional assays were carried out to determine the roles of RUSC1-AS1 and miR-101-3p in OS progression both in vitro and in vivo. The expression of E-cadherin, N-cadherin, Vimentin, Snail, Notch1, Ras and ERK was determined by Western blot. Furthermore, the relationships between RUSC1-AS1 and miR-101-3p, Notch1 and miR-101-3p were confirmed through RNA immunoprecipitation (RIP) and dual luciferase reporter gene assay. Results RUSC1-AS1 and Notch1 were up-regulated in OS cells and tissues. Down-regulating RUSC1-AS1 significantly attenuated the proliferative, epithelial-mesenchymal transition (EMT), growth, lung metastasis, migrative and invasive abilities of MG-63 and Saos-2 cells, and aggravated apoptosis, accompanied with down-regulated Notch1-Ras-ERK1/2 in those cells both in vitro and in vivo, while overexpression of RUSC1-AS1 exerted opposite effects. Overexpressing miR-101-3p in OS cells had similar effects as RUSC1-AS1 inhibition. In addition, RUSC1-AS1 functioned as a competing endogenous RNA (ceRNA) to competitively sponge miR-101-3p, thus upregulating Notch1 expression and mediating the malignant behaviors of OS cells. Conclusion RUSC1-AS1 is a novel oncogenic lncRNA in OS through the miR-101-3p-Notch1-Ras-ERK pathway, which might be a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Ziyan Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Zhiwei Zhong
- Department of Pain Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Chao Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, ChangchCun 130033, Jilin, China
| |
Collapse
|