1
|
Shi X, Jin L, Meng X, Huo X, Sun Y, Xue L, Wei Y, Wang Y, Yin Z, Zhao Y, Chen L. Transcriptomic analysis identified novel biomarker in invasive placenta accreta spectrum. Placenta 2024; 158:301-309. [PMID: 39549433 DOI: 10.1016/j.placenta.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
INTRODUCTION Placenta accreta spectrum (PAS) disorders pose a grave threat to maternal life due to severe hemorrhage and the heightened risk of peripartum hysterectomy. Consequently, there's a pressing need for circulating biomarkers in clinical settings. MicroRNAs (miRNAs), being stable in peripheral circulation, hold promise as potential biomarkers for PAS. METHODS This study recruited singleton live pregnancies, including cases of invasive PAS, placenta previa (PP), and controls, across three phases. Initially, RNA-seq of peripheral blood identified 6 miRNAs in the screening phase. Subsequently, in the training and validation phases, miR-23a-5p, along with its target genes ASF1B and CHTF8, were validated using qRT-PCR. The diagnostic value of these markers for PAS and adverse outcomes was evaluated using Receiver Operating Characteristic (ROC) curves. RESULTS The results showed miR-23a-5p was down-regulated in PAS, whereas ASF1B and CHTF8 were up-regulated. miR-23a-5p had modest diagnostic efficiency for PAS and adverse outcomes, as the AUC were 0.689 and 0.711 respectively. However, when miR-23a-5p combined with CHTF8, the AUC can improve greatly to 0.869 in PAS diagnosis and 0.856 in prediction of adverse outcomes. DISCUSSION We propose the miR-23a-5p plays a role in PAS pathogenesis through regulating cell proliferation, migration, invasion, apoptosis by targeting various genes. This study confirmed its potential value of miR-23a-5p combined with target gene CHTF8 as novel biomarkers for PAS and adverse outcomes.
Collapse
Affiliation(s)
- Xiaoming Shi
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| | - Ling Jin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
| | - Xinlu Meng
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Xiao Huo
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yan Sun
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yuanyuan Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Zhongnan Yin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Lian Chen
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
2
|
Zhang P, Wang P, Wang Y. ASF1B is an essential prognostic indicator linked to the growth and resistance characteristics of bladder cancer. Tissue Cell 2024; 89:102477. [PMID: 39018712 DOI: 10.1016/j.tice.2024.102477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Anti-silencing function 1 (ASF1) is a conserved histone H3-H4 chaperone protein. ASF1B (Anti-Silencing Function 1B Histone Chaperone), a paralog of ASF1, is involved in tumor metabolism and growth. The regulatory network of ASF1B in cancer is intricate and remains inadequately explored. The objective of this study was to examine the biological role of ASF1B in bladder cancer (BC). METHODS The presence of ASF1B in BC was examined using The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases. In addition, a correlation analysis was performed to evaluate the association between the BC pathway scores and ASF1B. ASF1B expression in BC cells was detected using western blott and RT-PCR. Several investigations were conducted, both within and outside of a living organism, to confirm the involvement of ASF1B in the regulation of biological processes in BC cells. RESULTS Our examination of the database indicates that ASF1B exhibits significant expression levels in BC cells and is potentially strongly associated with the growth of BC cells and the repair of DNA. The expression of ASF1B in BC cells was found to be significantly elevated, as indicated by the results of western blot and RT-PCR. The findings of the cell plate cloning test, edu analysis, flow cytometry, and transwell experiments demonstrated that the inhibition of ASF1B greatly impeded the proliferation and migration of BC cells. After establishing drug-resistant BC cell lines in a lab, suppressing ASF1B gene expression led to a notable reduction in BC cells' resistance to cisplatin. Confirmation was achieved by flow cytometry and western blott assays. Our in vivo findings demonstrated that the suppression of ASF1B resulted in an amelioration of the pathological condition, a decrease in resistance to cisplatin, and an inhibition of the growth of BC in mice.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Urology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Peng Wang
- Ministry of Scientific and Technological Innovation, Yantai Hi-tech Industrial Development Zone, Yantai, Shandong 264000, China
| | - Yirong Wang
- Department of Radiotherapy, Yantaishan Hospital, Yantai, Shandong 264025, China.
| |
Collapse
|
3
|
Wen HJ, Zhu SY, Yang HG, Guo FY. Investigation on the molecular mechanism of SPA interference with osteogenic differentiation of bone marrow mesenchymal stem cells. Sci Rep 2024; 14:15600. [PMID: 38971916 PMCID: PMC11227578 DOI: 10.1038/s41598-024-66502-2] [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: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
Binding of Staphylococcus aureus protein A (SPA) to osteoblasts induces apoptosis and inhibits bone formation. Bone marrow-derived mesenchymal stem cells (BMSCs) have the ability to differentiate into bone, fat and cartilage. Therefore, it was important to analyze the molecular mechanism of SPA on osteogenic differentiation. We introduced transcript sequence data to screen out differentially expressed genes (DEGs) related to SPA-interfered BMSC. Protein-protein interaction (PPI) network of DEGs was established to screen biomarkers associated with SPA-interfered BMSC. Receiver operating characteristic (ROC) curve was plotted to evaluate the ability of biomarkers to discriminate between two groups of samples. Finally, we performed GSEA and regulatory analysis based on biomarkers. We identified 321 DEGs. Subsequently, 6 biomarkers (Cenpf, Kntc1, Nek2, Asf1b, Troap and Kif14) were identified by hubba algorithm in PPI. ROC analysis showed that six biomarkers could clearly discriminate between normal differentiated and SPA-interfered BMSC. Moreover, we found that these biomarkers were mainly enriched in the pyrimidine metabolism pathway. We also constructed '71 circRNAs-14 miRNAs-5 mRNAs' and '10 lncRNAs-5 miRNAs-2 mRNAs' networks. Kntc1 and Asf1b genes were associated with rno-miR-3571. Nek2 and Asf1b genes were associated with rno-miR-497-5p. Finally, we found significantly lower expression of six biomarkers in the SPA-interfered group compared to the normal group by RT-qPCR. Overall, we obtained 6 biomarkers (Cenpf, Kntc1, Nek2, Asf1b, Troap, and Kif14) related to SPA-interfered BMSC, which provided a theoretical basis to explore the key factors of SPA affecting osteogenic differentiation.
Collapse
Affiliation(s)
- Hong-Jie Wen
- Department of Orthopaedic and Trauma, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Orthopaedic and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China
| | - Shou-Yan Zhu
- Department of Orthopaedic and Trauma, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Orthopaedic and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China
| | - Hua-Gang Yang
- Department of Orthopaedic and Trauma, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Orthopaedic and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China
| | - Feng-Yong Guo
- Department of Orthopaedic and Trauma, The Affiliated Hospital of Yunnan University, Kunming, China.
- Department of Orthopaedic and Trauma, The Second People's Hospital of Yunnan Province, Kunming, China.
| |
Collapse
|
4
|
Zhao Z, Cai Z, Zhang S, Yin X, Jiang T, Shen C, Yin Y, Sun H, Chen Z, Han J, Zhang B. Activation of the FOXM1/ASF1B/PRDX3 axis confers hyperproliferative and antioxidative stress reactivity to gastric cancer. Cancer Lett 2024; 589:216796. [PMID: 38537775 DOI: 10.1016/j.canlet.2024.216796] [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: 10/03/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Nucleosome assembly during DNA replication is dependent on histone chaperones. Recent studies suggest that dysregulated histone chaperones contribute to cancer progression, including gastric cancer (GC). Further studies are required to explore the prognostic and therapeutic implications of histone chaperones and their mechanisms of action in GC progression. Here we identified histone chaperone ASF1B as a potential biomarker for GC proliferation and prognosis. ASF1B was significantly upregulated in GC, which was associated with poor prognosis. In vitro and in vivo experiments demonstrated that the inhibition of ASF1B suppressed the malignant characteristics of GC, while overexpression of ASF1B had the opposite effect. Mechanistically, transcription factor FOXM1 directly bound to the ASF1B-promoter region, thereby regulating its transcription. Treatment with thiostrepton, a FOXM1 inhibitor, not only suppressed ASF1B expression, but also inhibited GC progression. Furthermore, ASF1B regulated the mitochondrial protein peroxiredoxin 3 (PRDX3) transcription in a FOXM1-dependent manner. The crucial role of ASF1B-regulated PRDX3 in GC cell proliferation and oxidative stress balance was also elucidated. In summary, our study suggests that the FOXM1-ASF1B-PRDX3 axis is a potential therapeutic target for treating GC.
Collapse
Affiliation(s)
- Zhou Zhao
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China; Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Zhaolun Cai
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Su Zhang
- State Key Laboratory of Biotherapy and Cancer Center, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaonan Yin
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Tianxiang Jiang
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Chaoyong Shen
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yin
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Sun
- Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Zhixin Chen
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Bo Zhang
- Gastric Cancer Center, Department of General Surgery, Research Laboratory of Tumor Epigenetics and Genomics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Ahmed AA, Chen S, Roman-Escorza M, Angell R, Oxenford S, McConville M, Barton N, Sunose M, Neidle D, Haider S, Arshad T, Neidle S. Structure-activity relationships for the G-quadruplex-targeting experimental drug QN-302 and two analogues probed with comparative transcriptome profiling and molecular modeling. Sci Rep 2024; 14:3447. [PMID: 38342953 PMCID: PMC10859377 DOI: 10.1038/s41598-024-54080-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/08/2024] [Indexed: 02/13/2024] Open
Abstract
The tetrasubstituted naphthalene diimide compound QN-302 binds to G-quadruplex (G4) DNA structures. It shows high potency in pancreatic ductal adenocarcinoma (PDAC) cells and inhibits the transcription of cancer-related genes in these cells and in PDAC animal models. It is currently in Phase 1a clinical evaluation as an anticancer drug. A study of structure-activity relationships of QN-302 and two related analogues (CM03 and SOP1247) is reported here. These have been probed using comparisons of transcriptional profiles from whole-genome RNA-seq analyses, together with molecular modelling and molecular dynamics simulations. Compounds CM03 and SOP1247 differ by the presence of a methoxy substituent in the latter: these two compounds have closely similar transcriptional profiles. Whereas QN-302 (with an additional benzyl-pyrrolidine group), although also showing down-regulatory effects in the same cancer-related pathways, has effects on distinct genes, for example in the hedgehog pathway. This distinctive pattern of genes affected by QN-302 is hypothesized to contribute to its superior potency compared to CM03 and SOP1247. Its enhanced ability to stabilize G4 structures has been attributed to its benzyl-pyrrolidine substituent fitting into and filling most of the space in a G4 groove compared to the hydrogen atom in CM03 or the methoxy group substituent in SOP1247.
Collapse
Affiliation(s)
- Ahmed Abdullah Ahmed
- The School of Pharmacy, University College London, London, WC1N 1AX, UK
- Now at Guy's Cancer Centre, Guy's Hospital, London, SE1 9RT, UK
| | - Shuang Chen
- The School of Pharmacy, University College London, London, WC1N 1AX, UK
| | | | - Richard Angell
- The School of Pharmacy, University College London, London, WC1N 1AX, UK
- Now at Medicines Discovery Institute, Cardiff University, Cardiff, CF10 3AT, UK
| | - Sally Oxenford
- The School of Pharmacy, University College London, London, WC1N 1AX, UK
- Now at Artios Ltd, Cambridge, CB22 3FH, UK
| | | | | | - Mihiro Sunose
- Sygnature Discovery Ltd, BioCity, Nottingham, NG1 1GR, UK
| | - Dan Neidle
- Tax Policy Associates, London, EC1R 0ET, UK
| | - Shozeb Haider
- The School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Tariq Arshad
- Qualigen Therapeutics Inc, Carlsbad, CA, 92011, USA
| | - Stephen Neidle
- The School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
6
|
Xiao F, Zhu H, Guo Y, Zhang Z, Sun G, Xiao Y, Hu G, Huang K, Guo H. CIA-II is associated with lower-grade glioma survival and cell proliferation. CNS Neurosci Ther 2024; 30:e14340. [PMID: 37452510 PMCID: PMC10848044 DOI: 10.1111/cns.14340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/09/2023] [Accepted: 06/24/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND The role of CIA-II has been clarified in several types of tumors; however, whether dysregulated CIA-II expression is also involved in the pathophysiology of lower-grade glioma (LGG) remains undisclosed. METHODS A comprehensive pan-cancer analysis of the expression patterns and prognostic significance of CIA-II in miscellaneous tumors was undertaken. Subsequently, a detailed bioinformatics analysis was executed to identify putative correlations between CIA-II expression and clinical features, prognosis, biological functions, immunological characteristics, genomic alterations, and chemotherapeutics in LGG. In vitro studies were implemented to examine the potential roles of CIA-II in LGG. RESULTS CIA-II expression was found to be abnormally elevated in a variety of tumors, including LGG. Additionally, patients with LGG with higher CIA-II expression owned worse prognosis. Importantly, the results declared that CIA-II expression was an independent prognostic indicator for LGG. Moreover, the expression of CIA-II was tightly interrelated with immune cell infiltration, gene mutations, and chemotherapeutics in LGG. In vitro studies revealed that CIA-II was increased and strongly related to the cell proliferation in LGG. CONCLUSION CIA-II may be an independent prognostic factor and a serviceable therapeutic target in LGG.
Collapse
Affiliation(s)
- Feng Xiao
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Yun Guo
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Zhe Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Gufeng Sun
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Yao Xiao
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Guowen Hu
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Kai Huang
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| | - Hua Guo
- Department of NeurosurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- Jiangxi Health Commission Key Laboratory of Neurological MedicineNanchangChina
- Institute of NeuroscienceNanchang UniversityNanchangChina
| |
Collapse
|
7
|
Zhao C, Zhou J, Xing J, Yin Q. ASF1B acted as a prognostic biomarker for stomach adenocarcinoma. Medicine (Baltimore) 2023; 102:e35408. [PMID: 38050219 PMCID: PMC10695504 DOI: 10.1097/md.0000000000035408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 09/05/2023] [Indexed: 12/06/2023] Open
Abstract
Stomach adenocarcinoma (STAD) has a high mortality rate due to the lack of highly sensitive biomarkers. Therefore, the search for potential tumor markers is of great value. ASF1B is a prognostic marker for a variety of tumors, while the prognostic value and immune microenvironment of ASF1B in STAD remain unclear, and to be determined. Kaplan-Meier analysis was performed to analyze the prognostic role of ASF1B in STAD. Functional enrichment of ASF1B was explored with GO and KEGG pathway analysis. We also explored the correlation between ASF1B expression and immune infiltration in STAD. ASF1B was significantly upregulated in STAD tissues and high expression of ASF1B indicated a poor overall survival, progression-free survival, and first progression rate in STAD. The functional enrichment analysis of ASF1B and related genes showed high enrichment in the cell cycle and DNA repair, and the ASF1B high expression group was also mainly enriched in pathways such as the cell cycle. Analysis of tumor immune infiltration showed that ASF1B expression was significantly associated with the majority of immune cell infiltration in STAD. Moreover, STAD patients with high ASF1B expression had a higher tumor mutation burden score, microsatellite instability score, PD-1 immunophenoscore, and immune checkpoint expression. Our results suggest that ASF1B was an independent prognostic factor for STAD as well as a potential target for immunotherapy.
Collapse
Affiliation(s)
- Cailing Zhao
- Department of Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jianghao Zhou
- Department of Gastrointestinal Tumor Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jianwei Xing
- Department of General Surgery, Sanya Central Hospital, the Third People’s Hospital of Hainan Province, Sanya, China
| | - Qiushi Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
8
|
Zhang M, Zhang L, Zhou M, Wang E, Meng B, Li Q, Wang X, Wang Y, Li Q. Anti‑silencing function 1B promotes the progression of pancreatic cancer by activating c‑Myc. Int J Oncol 2023; 62:8. [PMID: 36416310 PMCID: PMC9728557 DOI: 10.3892/ijo.2022.5456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
The present study aimed to explore the role of histone chaperone anti‑silencing function 1B (ASF1B) in pancreatic cancer and the underlying mechanism. The biological function of ASF1B was investigated in pancreatic cancer cell lines (PANC‑1 and SW1990) and a mouse xenograft model. Chromatin immunoprecipitation was used to detect the effect of ASF1B on the transcriptional activity of c‑Myc. ASF1B was highly expressed in pancreatic adenocarcinoma (PAAD) samples from The Cancer Genome Atlas. ASF1B expression was positively associated with poor survival rates in patients with PAAD. Silencing of ASF1B in PANC‑1 and SW1990 cells inhibited cell proliferation, migration and invasion, and induced apoptosis. Mechanistically, ASF1B increased H3K56 acetylation (H3K56ac) in a CREB‑binding protein (CBP)‑dependent manner. ASF1B promoted H3K56ac at the c‑Myc promoter and increased c‑Myc expression. In PANC‑1 and SW1990 cells, the CBP inhibitor curcumin and the c‑Myc inhibitor 10058‑F4 reversed the promoting effects of ASF1B on cell proliferation, migration and invasion. In the mouse xenograft model, ASF1B silencing inhibited tumor growth, and was associated with low H3K56ac and c‑Myc expression. ASF1B promoted pancreatic cancer progression by activating c‑Myc via CBP‑mediated H3K56ac.
Collapse
Affiliation(s)
- Min Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Luyang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Minghe Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Enze Wang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Bo Meng
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Qingjun Li
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Xiaoqian Wang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Yunjian Wang
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450008
| | - Qiong Li
- Key Laboratory for Medical Tissue Regeneration of Henan Province, Xinxiang Medical College, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
9
|
Yu GH, Gong XF, Peng YY, Qian J. Anti-silencing function 1B knockdown suppresses the malignant phenotype of colorectal cancer by inactivating the phosphatidylinositol 3-kinase/AKT pathway. World J Gastrointest Oncol 2022; 14:2353-2366. [PMID: 36568946 PMCID: PMC9782623 DOI: 10.4251/wjgo.v14.i12.2353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mounting studies have highlighted the pivotal influence of anti-silencing function 1B (ASF1B) on the malignancy of cancers.
AIM To explore the influence and mechanism of ASF1B in colorectal cancer (CRC).
METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect mRNA expression of ASF1B. Immunohistochemical staining was performed to detect protein expression of ASF1B and Ki67 in tumor tissues. Western blot analysis was used to determine levels of ASF1B and proliferation/epithelial mesenchymal transition (EMT)/stemness-related proteins. In addition, the proliferation of CRC cells was assessed using Cell Counting Kit-8 and 5-Ethynyl-2’-Deoxyuridine assays. The migration and invasion of CRC cells were evaluated using transwell assays. Stemness of CRC cells was tested using the sphere formation assay. To construct a xenograft tumor model, HCT116 cells were introduced into mouse flanks via subcutaneous injection.
RESULTS ASF1B expression was markedly increased in CRC tissues and cells, and it was inversely correlated with overall survival of CRC patients and was positively associated with the tumor node metastasis (TNM) stage of CRC patients. Silencing of ASF1B suppressed proliferation, migration, invasion, stemness and EMT of CRC cells as well as tumorigenesis of xenograft mice. Furthermore, protein levels of P-phosphatidylinositol 3-kinase (p-PI3K) and p-AKT were decreased after silencing of ASF1B in CRC cells. The inhibitory effects of ASF1B knockdown on cell proliferation, stemness and EMT were partly abolished by PI3K activator in CRC cells.
CONCLUSION Silencing of ASF1B inactivated the PI3K/AKT pathway to suppress CRC malignancy in vitro.
Collapse
Affiliation(s)
- Gen-Hua Yu
- Department of Radiation Oncology, Zhebei Mingzhou Hospital, Huzhou 313000, Zhejiang Province, China
| | - Xu-Feng Gong
- Department of Radiation Oncology, Zhebei Mingzhou Hospital, Huzhou 313000, Zhejiang Province, China
| | - Ying-Ying Peng
- Department of Radiation Oncology, Zhebei Mingzhou Hospital, Huzhou 313000, Zhejiang Province, China
| | - Jun Qian
- Department of Colorectal Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, Zhejiang Province, China
| |
Collapse
|
10
|
Zhao Z, Cai Z, Jiang T, Han J, Zhang B. Histone Chaperones and Digestive Cancer: A Review of the Literature. Cancers (Basel) 2022; 14:5584. [PMID: 36428674 PMCID: PMC9688693 DOI: 10.3390/cancers14225584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The global burden of digestive cancer is expected to increase. Therefore, crucial for the prognosis of patients with these tumors is to identify early diagnostic markers or novel therapeutic targets. There is accumulating evidence connecting histone chaperones to the pathogenesis of digestive cancer. Histone chaperones are now broadly defined as a class of proteins that bind histones and regulate nucleosome assembly. Recent studies have demonstrated that multiple histone chaperones are aberrantly expressed and have distinct roles in digestive cancers. OBJECTIVE The purpose of this review is to present the current evidence regarding the role of histone chaperones in digestive cancer, particularly their mechanism in the development and progression of esophageal, gastric, liver, pancreatic, and colorectal cancers. In addition, the prognostic significance of particular histone chaperones in patients with digestive cancer is discussed. METHODS According to PRISMA guidelines, we searched the PubMed, Embase, and MEDLINE databases to identify studies on histone chaperones and digestive cancer from inception until June 2022. RESULTS A total of 104 studies involving 21 histone chaperones were retrieved. CONCLUSIONS This review confirms the roles and mechanisms of selected histone chaperones in digestive cancer and suggests their significance as potential prognostic biomarkers and therapeutic targets. However, due to their non-specificity, more research on histone chaperones should be conducted in the future to elucidate novel strategies of histone chaperones for prognosis and treatment of digestive cancer.
Collapse
Affiliation(s)
- Zhou Zhao
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhaolun Cai
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianxiang Jiang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|