1
|
Betancur MI, Case A, Ilich E, Mehta N, Meehan S, Pogrebivsky S, Keir ST, Stevenson K, Brahma B, Gregory S, Chen W, Ashley DM, Bellamkonda R, Mokarram N. A neural tract-inspired conduit for facile, on-demand biopsy of glioblastoma. Neurooncol Adv 2024; 6:vdae064. [PMID: 38813113 PMCID: PMC11135361 DOI: 10.1093/noajnl/vdae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Abstract
Background A major hurdle to effectively treating glioblastoma (GBM) patients is the lack of longitudinal information about tumor progression, evolution, and treatment response. Methods In this study, we report the use of a neural tract-inspired conduit containing aligned polymeric nanofibers (i.e., an aligned nanofiber device) to enable on-demand access to GBM tumors in 2 rodent models. Depending on the experiment, a humanized U87MG xenograft and/or F98-GFP+ syngeneic rat tumor model was chosen to test the safety and functionality of the device in providing continuous sampling access to the tumor and its microenvironment. Results The aligned nanofiber device was safe and provided a high quantity of quality genomic materials suitable for omics analyses and yielded a sufficient number of live cells for in vitro expansion and screening. Transcriptomic and genomic analyses demonstrated continuity between material extracted from the device and that of the primary, intracortical tumor (in the in vivo model). Conclusions The results establish the potential of this neural tract-inspired, aligned nanofiber device as an on-demand, safe, and minimally invasive access point, thus enabling rapid, high-throughput, longitudinal assessment of tumor and its microenvironment, ultimately leading to more informed clinical treatment strategies.
Collapse
Affiliation(s)
| | - Ayden Case
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Nalini Mehta
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Sean Meehan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Sabrina Pogrebivsky
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Stephen T Keir
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Kevin Stevenson
- Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Barun Brahma
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Simon Gregory
- Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Wei Chen
- Center for Genomic and Computational Biology, Duke University, Durham, Georgia, USA
| | - David M Ashley
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Ravi Bellamkonda
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Nassir Mokarram
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
2
|
Yuan J, Jiang X, Lan H, Zhang X, Ding T, Yang F, Zeng D, Yong J, Niu B, Xiao S. Multi-Omics Analysis of the Therapeutic Value of MAL2 Based on Data Mining in Human Cancers. Front Cell Dev Biol 2022; 9:736649. [PMID: 35111745 PMCID: PMC8803135 DOI: 10.3389/fcell.2021.736649] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Recent studies have reported that T-cell differentiation protein 2 (MAL2) is an important regulator in cancers. Here, we downloaded data from multiple databases to analyze MAL2 expression and function in pan-cancers, especially in ovarian cancer (OC). Gene Expression Profiling Interactive Analysis (GEPIA) databases was used to examine MAL2 expression in 13 types of cancer. Kaplan–Meier plotter database was used to analyze the overall survival rate of MAL2 in pan-cancers. The Catalog of Somatic Mutations in Cancer (COSMIC), cBioPortal, and UCSC databases were used to examine MAL2 mutation in human cancers. Metascape, STRING, and GeneMANIA websites were used to explore MAL2 function in OC. Furthermore, ggplot2 package and ROC package were performed to analyze hub gene expression and undertake receiver operating characteristic (ROC) analysis. Drug sensitivity of MAL2 in OC was examined by the GSCALite database. In order to verify the results from databases above, real-time quantitative polymerase chain reaction (qRT-PCR) and western blotting were conducted to detect the expression of MAL2 in OC cells. CRISPR/Cas9 system was used to knockout the MAL2 gene in the OC cell lines HO8910 and OVCAR3, using specific guide RNA targeting the exons of MAL2. Then, we performed proliferation, colony formation, migration, and invasion assays to investigate the impact of MAL2 in OC cell lines in vivo and in vitro. Epithelial-mesenchymal transition (EMT)-associated biomarkers were significantly altered in vitro via western blotting and qRT-PCR. Taken together, we observed that MAL2 was remarkably dysregulated in multiple cancers and was related to patient overall survival (OS), mutation, and drug sensitivity. Furthermore, experimental results showed that MAL2 deletion negatively regulated the proliferation, migration, invasion, and EMT of OC, indicating that MAL2 is a novel oncogene that can activate EMT, significantly promote both the proliferation and migration of OC in vitro and in vivo, and provide new clues for treatment strategies.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Jiang
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hua Lan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyu Zhang
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Tianyi Ding
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Fan Yang
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Da Zeng
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiahui Yong
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Beibei Niu
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Beibei Niu, ; Songshu Xiao,
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Beibei Niu, ; Songshu Xiao,
| |
Collapse
|
3
|
Guo T, Yuan D, Zhang W, Zhu D, Xiao A, Mao G, Jiang W, Lin M, Wang J. Upregulation of long noncoding RNA XIST has anticancer effects on ovarian cancer through sponging miR-106a. Hum Cell 2021; 34:579-587. [PMID: 33400246 DOI: 10.1007/s13577-020-00469-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022]
Abstract
Ovarian cancer (OC) is a highly malignant tumor. X inactive specific transcript (XIST) was identified as a cancer-related gene, while its therapeutic effect in OC was poorly defined. The present study was designed to investigate the effectual corollary of the lncRNA XIST in OC. RT-qPCR was used to detect the XIST and miR-106a expression levels of OC tissues and cell lines. OC cell apoptosis and proliferation were detected by flow cytometry, colony formation, and CCK-8 assays. Moreover, bioinformatics analysis was used to predict the targeted miRNA of XIST. The dual-luciferase reporter and RNA pull-down assays were then used to verify the interaction between miR-106a and XIST. OC xenograft nude mice were raised to measure tumor growth. Notably, OC tissues and cells exhibited low XIST levels and high miR-106a levels. The XIST upregulation decreased the OVCAR3 and CAOV3 cell proliferation and inversely promoted cell apoptosis. miR-106a targeted the XIST. Also, the miR-106a overexpression reversed the inhibitory effects of XIST on OC cell proliferation and apoptosis. Our in vivo results suggested that XIST was involved in tumor growth deceleration, while the miR-106a reversed the effect. To conclusion, the present study demonstrated that XIST suppressed OC development via sponging miR-106a both in vitro and in vivo.
Collapse
Affiliation(s)
- Ting Guo
- Institute of Clinical Medicine, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Donglan Yuan
- The Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Wei Zhang
- The Department of Infectious Disease, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Dandan Zhu
- The Department of Obstetrics and Gynecology, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Aifang Xiao
- Emergency Department, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Guangyao Mao
- Institute of Clinical Medicine, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Wenjuan Jiang
- Emergency Department, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China
| | - Mei Lin
- Clinical Laboratory, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China.
| | - Jun Wang
- Emergency Department, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, 225300, Jiangsu, China.
| |
Collapse
|