1
|
Vatankhah A, Moghaddam SH, Afshari S, Afshari AR, Kesharwani P, Sahebkar A. Recent update on anti-tumor mechanisms of valproic acid in glioblastoma multiforme. Pathol Res Pract 2024; 263:155636. [PMID: 39395298 DOI: 10.1016/j.prp.2024.155636] [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: 08/23/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Glioblastoma multiforme (GBM) is a malignant tumor of the brain that is considered to be incurable. Currently, surgical removal of tumors, chemotherapy with temozolomide, and radiation treatment remain established options for treatment. Nevertheless, the prognosis of those with GBM continues to be poor owing to the inherent characteristics of tumor growth and spread, as well as the resistance to treatment. To effectively deal with the present circumstances, it is vital to do extensive study to understand GBM thoroughly. The following piece provides a concise overview of the most recent advancements in using valproic acid, an antiseizure medication licensed by the FDA, for treating GBM. In this review, we outline the most recent developments of valproic acid in treating GBM, as well as its fundamental mechanisms and practical consequences. Our goal is to provide a greater understanding of the clinical use of valproic acid as a potential therapeutic agent for GBM.
Collapse
Affiliation(s)
- Abulfazl Vatankhah
- School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Sadaf Afshari
- Student Research Committee, Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Sharma A, Raut SS, Shukla A, Gupta S, Singh A, Mishra A. DDX3X dynamics, glioblastoma's genetic landscape, therapeutic advances, and autophagic interplay. Med Oncol 2024; 41:258. [PMID: 39368002 DOI: 10.1007/s12032-024-02525-z] [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: 06/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma is one of the most aggressive and deadly forms of cancer, posing significant challenges for the medical community. This review focuses on key aspects of Glioblastoma, including its genetic differences between primary and secondary types. Temozolomide is a major first-line treatment for Glioblastoma, and this article explores its development, how it works, and the issue of resistance that limits its effectiveness, prompting the need for new treatment strategies. Gene expression profiling has greatly advanced cancer research by revealing the molecular mechanisms of tumors, which is essential for creating targeted therapies for Glioblastoma. One important protein in this context is DDX3X, which plays various roles in cancer, sometimes promoting it or otherwise suppressing it. Additionally, autophagy, a process that maintains cellular balance, has complex implications in cancer treatment. Understanding autophagy helps to identify resistance mechanisms and potential treatments, with Chloroquine showing promise in treating Glioblastoma. This review covers the interplay between Glioblastoma, DDX3X, and autophagy, highlighting the challenges and potential strategies in treating this severe disease.
Collapse
Affiliation(s)
- Arpit Sharma
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shruti S Raut
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Alok Shukla
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Shivani Gupta
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Amit Singh
- Department of Pharmacology, IMS-Banaras Hindu University, Varanasi, 221005, India.
| | - Abha Mishra
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| |
Collapse
|
3
|
Zhao S, Ni K, Xie J, Cheng C, Zhao N, Liu J, Ji W, Wang Q, Zhang P, Liu Y. Exploring the prognostic value of BRMS1 + microglia based on single-cell anoikis regulator patterns in the immunologic microenvironment of GBM. J Neurooncol 2024; 170:101-117. [PMID: 39143438 PMCID: PMC11447114 DOI: 10.1007/s11060-024-04781-5] [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: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Anoikis is a specialized form of programmed cell death induced by the loss of cell adhesion to the extracellular matrix (ECM). Acquisition of anoikis resistance is a significant marker for cancer cell invasion, metastasis, therapy resistance, and recurrence. Although current research has identified multiple factors that regulate anoikis resistance, the pathological mechanisms of anoikis-mediated tumor microenvironment (TME) in glioblastoma (GBM) remain largely unexplored. METHODS Utilizing single-cell RNA sequencing (scRNA-seq) data and employing non-negative matrix factorization (NMF), we identified and characterized TME cell clusters with distinct anoikis-associated gene signatures. Prognostic and therapeutic response analyses were conducted using TCGA and CGGA datasets to assess the clinical significance of different TME cell clusters. The spatial relationship between BRMS1 + microglia and tumor cells was inferred from spatial transcriptome RNA sequencing (stRNA-seq) data. To simulate the tumor immune microenvironment, co-culture experiments were performed with microglia (HMC3) and GBM cells (U118/U251), and microglia were transfected with a BRMS1 overexpression lentivirus. Western blot or ELISA were used to detect BRMS1, M2 macrophage-specific markers, PI3K/AKT signaling proteins, and apoptosis-related proteins. The proliferation and apoptosis capabilities of tumor cells were evaluated using CCK-8, colony formation, and apoptosis assays, while the invasive and migratory abilities of tumor cells were assessed using Transwell assays. RESULTS NMF-based analysis successfully identified CD8 + T cell and microglia cell clusters with distinct gene signature characteristics. Trajectory analysis, cell communication, and gene regulatory network analyses collectively indicated that anoikis-mediated TME cell clusters can influence tumor cell development through various mechanisms. Notably, BRMS1 + AP-Mic exhibited an M2 macrophage phenotype and had significant cell communication with malignant cells. Moreover, high expression of BRMS1 + AP-Mic in TCGA and CGGA datasets was associated with poorer survival outcomes, indicating its detrimental impact on immunotherapy. Upregulation of BRMS1 in microglia may lead to M2 macrophage polarization, activate the PI3K/AKT signaling pathway through SPP1/CD44-mediated cell interactions, inhibit tumor cell apoptosis, and promote tumor proliferation and invasion. CONCLUSION This pioneering study used NMF-based analysis to reveal the important predictive value of anoikis-regulated TME in GBM for prognosis and immunotherapeutic response. BRMS1 + microglial cells provide a new perspective for a deeper understanding of the immunosuppressive microenvironment of GBM and could serve as a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Kaixiang Ni
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Cheng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Ning Zhao
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Ji
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| | - Yuankun Liu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
4
|
Biegański M, Szeliga M. Disrupted glutamate homeostasis as a target for glioma therapy. Pharmacol Rep 2024:10.1007/s43440-024-00644-y. [PMID: 39259492 DOI: 10.1007/s43440-024-00644-y] [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: 06/28/2024] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). Gliomas, malignant brain tumors with a dismal prognosis, alter glutamate homeostasis in the brain, which is advantageous for their growth, survival, and invasion. Alterations in glutamate homeostasis result from its excessive production and release to the extracellular space. High glutamate concentration in the tumor microenvironment destroys healthy tissue surrounding the tumor, thus providing space for glioma cells to expand. Moreover, it confers neuron hyperexcitability, leading to epilepsy, a common symptom in glioma patients. This mini-review briefly describes the biochemistry of glutamate production and transport in gliomas as well as the activation of glutamate receptors. It also summarizes the current pre-clinical and clinical studies identifying pharmacotherapeutics targeting glutamate transporters and receptors emerging as potential therapeutic strategies for glioma.
Collapse
Affiliation(s)
- Mikołaj Biegański
- Immunooncology Students' Science Association, Medical University of Warsaw, Żwirki i Wigury 61, Warszawa, 02-091, Poland
| | - Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, Warszawa, 02-106, Poland.
| |
Collapse
|
5
|
Li J, Zhao L, Wu Z, Huang S, Wang J, Chang Y, Liu L, Jin H, Lu J, Huang C, Xie Q, Huang H, Su Z. SelK promotes glioblastoma cell proliferation by inhibiting β-TrCP1 mediated ubiquitin-dependent degradation of CDK4. J Exp Clin Cancer Res 2024; 43:231. [PMID: 39155374 PMCID: PMC11331741 DOI: 10.1186/s13046-024-03157-x] [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: 02/23/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Glioblastoma (GB) is recognized as one of the most aggressive brain tumors, with a median survival of 14.6 months. However, there are still some patients whose survival time was greater than 3 years, and the biological reasons behind this clinical phenomenon arouse our research interests. By conducting proteomic analysis on tumor tissues obtained from GB patients who survived over 3 years compared to those who survived less than 1 year, we identified a significant upregulation of SelK in patients with shorter survival times. Therefore, we hypothesized that SelK may be an important indicator related to the occurrence and progression of GBM. METHODS Proteomics and immunohistochemistry from GB patients were analyzed to investigate the correlation between SelK and clinical prognosis. Cellular phenotypes were evaluated by cell cycle analysis, cell viability assays, and xenograft models. Immunoblots and co-immunoprecipitation were conducted to verify SelK-mediated ubiquitin-dependent degradation of CDK4. RESULTS SelK was found to be significantly upregulated in GB samples from short-term survivors (≤ 1 year) compared to those from long-term survivors (≥ 3 years), and its expression levels were negatively correlated with clinical prognosis. Knocking down of SelK expression reduced GB cell viability, induced G0/G1 phase arrest, and impaired the growth of transplanted glioma cells in nude mice. Down-regulation of SelK-induced ER stress leads to a reduction in the expression of SKP2 and an up-regulation of β-TrCP1 expression. Up-regulation of β-TrCP1, thereby accelerating the ubiquitin-dependent degradation of CDK4 and ultimately inhibiting the malignant proliferation of the GB cells. CONCLUSION This study discovered a significant increase in SelK expression in GB patients with poor prognosis, revealing a negative correlation between SelK expression and patient outcomes. Further mechanistic investigations revealed that SelK enhances the proliferation of GB cells by targeting the endoplasmic reticulum stress/SKP2/β-TrCP1/CDK4 axis.
Collapse
Affiliation(s)
- Jizhen Li
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No.109, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China
| | - Lingling Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Zerui Wu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Shirui Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Junyu Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Yuanyuan Chang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Li Liu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Jianglong Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Qipeng Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No.109, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China.
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China.
| | - Zhipeng Su
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
6
|
Hasnat MA, Ohmi Y, Yesmin F, Kambe M, Kawamoto Y, Bhuiyan RH, Mizutani M, Hashimoto N, Tsuchida A, Ohkawa Y, Kaneko K, Tajima O, Furukawa K, Furukawa K. Crucial roles of exosomes secreted from ganglioside GD3/GD2-positive glioma cells in enhancement of the malignant phenotypes and signals of GD3/GD2-negative glioma cells. NAGOYA JOURNAL OF MEDICAL SCIENCE 2024; 86:435-451. [PMID: 39355364 PMCID: PMC11439612 DOI: 10.18999/nagjms.86.3.435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 10/03/2024]
Abstract
Neuroectoderm-derived tumors characteristically express gangliosides such as GD3 and GD2. Many studies have reported that gangliosides GD3/GD2 enhance malignant phenotypes of cancers. Recently, we reported that human gliomas expressing GD3/GD2 exhibited enhanced malignant phenotypes. Here, we investigated the function of GD3/GD2 in glioma cells and GD3/GD2-expressing glioma-derived exosomes. As reported previously, transfectant cells of human glioma U251 MG expressing GD3/GD2 showed enhanced cancer phenotypes compared with GD3/GD2-negative controls. When GD3/GD2-negative cells were treated with exosomes secreted from GD3/GD2-positive cells, clearly increased malignant properties were observed. Furthermore, increased phosphorylation of signaling molecules was detected after 5-15 min of exosome treatment, ie, higher tyrosine phosphorylation of platelet-derived growth factor receptor, focal adhesion kinase, and paxillin was found in treated cells than in controls. Phosphorylation of extracellular signal-regulated kinase-1/2 was also enhanced. Consequently, it is suggested that exosomes secreted from GD3/GD2-positive gliomas play important roles in enhancement of the malignant properties of glioma cells, leading to total aggravation of heterogenous cancer tissues, and also in the regulation of tumor microenvironments.
Collapse
Affiliation(s)
- Mohammad Abul Hasnat
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet-3114, Bangladesh
| | - Yuhsuke Ohmi
- Department of Clinical Engineering, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Farhana Yesmin
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Mariko Kambe
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Yoshiyuki Kawamoto
- Department of Immunology, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Momoka Mizutani
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Noboru Hashimoto
- Department of Tissue Regeneration, Tokushima University School of Dentistry, Tokushima, Japan
| | - Akiko Tsuchida
- Laboratory of Glyco-bioengineering, The Noguchi Institute, Tokyo, Japan
| | - Yuki Ohkawa
- Osaka International Cancer Institute, Osaka, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
7
|
Yang Z, Wu H, Wang Z, Bian E, Zhao B. The role and application of small extracellular vesicles in glioma. Cancer Cell Int 2024; 24:229. [PMID: 38951882 PMCID: PMC11218314 DOI: 10.1186/s12935-024-03389-z] [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: 01/08/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Small extracellular vesicles (sEVs) are cell-derived, nanometer-sized particles enclosed by a lipid bilayer. All kinds of biological molecules, including proteins, DNA fragments, RNA, lipids, and metabolites, can be selectively loaded into sEVs and transmitted to recipient cells that are near and distant. Growing shreds of evidence show the significant biological function and the clinical significance of sEVs in cancers. Numerous recent studies have validated that sEVs play an important role in tumor progression and can be utilized to diagnose, stage, grading, and monitor early tumors. In addition, sEVs have also served as drug delivery nanocarriers and cancer vaccines. Although it is still infancy, the field of basic and translational research based on sEVs has grown rapidly. In this review, we summarize the latest research on sEVs in gliomas, including their role in the malignant biological function of gliomas, and the potential of sEVs in non-invasive diagnostic and therapeutic approaches, i.e., as nanocarriers for drug or gene delivery and cancer vaccines.
Collapse
Affiliation(s)
- Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - HaoYuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ZhiWei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ErBao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| |
Collapse
|
8
|
Cui C, Yang T, Wang S, Jia Z, Zhao L, Han X, Sun X, Zong J, Wang S, Chen D. Discussion on the relationship between gut microbiota and glioma through Mendelian randomization test based on the brain gut axis. PLoS One 2024; 19:e0304403. [PMID: 38809931 PMCID: PMC11135782 DOI: 10.1371/journal.pone.0304403] [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: 12/06/2023] [Accepted: 05/11/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND In the realm of Gut-Brain axis research, existing evidence points to a complex bidirectional regulatory mechanism between gut microbiota and the brain. However, the question of whether a causal relationship exists between gut microbiota and specific types of brain tumors, such as gliomas, remains unresolved. To address this gap, we employed publicly available Genome-Wide Association Study (GWAS) and MIOBEN databases, conducting an in-depth analysis using Two-Sample Mendelian Randomization (MR). METHOD We carried out two sets of MR analyses. The preliminary analysis included fewer instrumental variables due to a high genome-wide statistical significance threshold (5×10-8). To enable a more comprehensive and detailed analysis, we adjusted the significance threshold to 1×10-5. We performed linkage disequilibrium analysis (R2 <0.001, clumping distance = 10,000kb) and detailed screening of palindromic SNPs, followed by MR analysis and validation through sensitivity analysis. RESULTS Our findings reveal a causal relationship between gut microbiota and gliomas. Further confirmation via Inverse Variance Weighting (IVW) identified eight specific microbial communities related to gliomas. Notably, the Peptostreptococcaceae and Olsenella communities appear to have a protective effect, reducing glioma risk. CONCLUSION This study not only confirms the causal link between gut microbiota and gliomas but also suggests a new avenue for future glioma treatment.
Collapse
Affiliation(s)
- Chenzhi Cui
- Graduate school, Dalian Medical University, Dalian, Dalian, China
- Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, China
| | - Tianke Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - ShengYu Wang
- Medical Laboratory Technology, College of Medical Laboratory, Dalian Medical University, Dalian, China
| | - Zhuqiang Jia
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Naqu People’s Hospital, Tibet, China
| | - Lin Zhao
- Department of Quality Management, Dalian Municipal Central Hospital, Dalian, China
| | - Xin Han
- Naqu People’s Hospital, Tibet, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohong Sun
- Department of Nursing, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junwei Zong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shouyu Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Chen
- Graduate school, Dalian Medical University, Dalian, Dalian, China
- Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, China
| |
Collapse
|
9
|
Lu E, Zhao B, Yuan C, Liang Y, Wang X, Yang G. Novel cancer-fighting role of ticagrelor inhibits GTSE1-induced EMT by regulating PI3K/Akt/NF-κB signaling pathway in malignant glioma. Heliyon 2024; 10:e30833. [PMID: 38774096 PMCID: PMC11107102 DOI: 10.1016/j.heliyon.2024.e30833] [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: 02/09/2024] [Revised: 04/21/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
Background Glioma is the most common malignant brain tumor of the central nervous system. Despite of the improvement of therapeutic strategy, the prognosis of malignant glioma patients underwent by STUPP strategy is still unexpected. Previous studies have suggested that ticagrelor exerted chemotherapeutic effects by inhibition of epithelial-mesenchymal transition (EMT) in various diseases including tumors. However, whether ticagrelor can exhibit the antitumor efficiency in glioma by affecting the EMT process is still unclear. In this study, we investigated the cancer-fighting role of ticagrelor and demonstrated its chemotherapeutic mechanism in glioma. Materials and methods The MTT assay was performed to detect the cytotoxicity of ticagrelor in glioma cells. We evaluated the expression of Ki67 in glioma cells by immunofluorescence assay after ticagrelor treatment. We conducted wound healing assay and transwell assay to determine the effects of ticagrelor on the migration and invasion of glioma cells. RNA-seq analysis was conducted to examine potential target genes and alternative signaling pathways for ticagrelor treatment. The expression levels of key EMT -related proteins were examined by Western blot experiment. Results Ticagrelor inhibited the proliferation, migration and invasion of glioma cells with a favorable toxicity profile in vitro. Ticagrelor downregulated the expression of GTSE1 in glioma cells. RNA-seq analysis explored that GTSE1 acted as the potential target gene for ticagrelor treatment. Upregulation of GTSE1 antagonized the inhibitory effect of ticagrelor on the invasion of glioma and EMT progression by regulation of PI3K/Akt/NF-κB signaling pathway. And ticagrelor also exhibited the similar chemotherapeutic effect of glioma in vivo. Conclusions Ticagrelor as a potential chemotherapeutic option induced the inhibition of the GTSE1-induced EMT progression by regulation of PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Enzhou Lu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Boxian Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Chao Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| |
Collapse
|
10
|
Fisher JL, Wilk EJ, Oza VH, Gary SE, Howton TC, Flanary VL, Clark AD, Hjelmeland AB, Lasseigne BN. Signature reversion of three disease-associated gene signatures prioritizes cancer drug repurposing candidates. FEBS Open Bio 2024; 14:803-830. [PMID: 38531616 PMCID: PMC11073506 DOI: 10.1002/2211-5463.13796] [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: 02/11/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Drug repurposing is promising because approving a drug for a new indication requires fewer resources than approving a new drug. Signature reversion detects drug perturbations most inversely related to the disease-associated gene signature to identify drugs that may reverse that signature. We assessed the performance and biological relevance of three approaches for constructing disease-associated gene signatures (i.e., limma, DESeq2, and MultiPLIER) and prioritized the resulting drug repurposing candidates for four low-survival human cancers. Our results were enriched for candidates that had been used in clinical trials or performed well in the PRISM drug screen. Additionally, we found that pamidronate and nimodipine, drugs predicted to be efficacious against the brain tumor glioblastoma (GBM), inhibited the growth of a GBM cell line and cells isolated from a patient-derived xenograft (PDX). Our results demonstrate that by applying multiple disease-associated gene signature methods, we prioritized several drug repurposing candidates for low-survival cancers.
Collapse
Affiliation(s)
- Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Vishal H. Oza
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Sam E. Gary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| |
Collapse
|
11
|
Rentzeperis F, Rivera D, Zhang JY, Brown C, Young T, Rodriguez B, Schupper A, Price G, Gomberg J, Williams T, Bouras A, Hadjipanayis C. Recent Developments in Magnetic Hyperthermia Therapy (MHT) and Magnetic Particle Imaging (MPI) in the Brain Tumor Field: A Scoping Review and Meta-Analysis. MICROMACHINES 2024; 15:559. [PMID: 38793132 PMCID: PMC11123314 DOI: 10.3390/mi15050559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024]
Abstract
Magnetic hyperthermia therapy (MHT) is a promising treatment modality for brain tumors using magnetic nanoparticles (MNPs) locally delivered to the tumor and activated with an external alternating magnetic field (AMF) to generate antitumor effects through localized heating. Magnetic particle imaging (MPI) is an emerging technology offering strong signal-to-noise for nanoparticle localization. A scoping review was performed by systematically querying Pubmed, Scopus, and Embase. In total, 251 articles were returned, 12 included. Articles were analyzed for nanoparticle type used, MHT parameters, and MPI applications. Preliminary results show that MHT is an exciting treatment modality with unique advantages over current heat-based therapies for brain cancer. Effective application relies on the further development of unique magnetic nanoparticle constructs and imaging modalities, such as MPI, that can enable real-time MNP imaging for improved therapeutic outcomes.
Collapse
Affiliation(s)
- Frederika Rentzeperis
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Daniel Rivera
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Jack Y. Zhang
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Cole Brown
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Tirone Young
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Benjamin Rodriguez
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
| | - Alexander Schupper
- Department of Neurological Surgery, Mount Sinai Hospital, New York, NY 10029, USA;
| | - Gabrielle Price
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Jack Gomberg
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (F.R.); (D.R.); (J.Y.Z.); (C.B.); (T.Y.); (G.P.); (J.G.)
| | - Tyree Williams
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY 10029, USA;
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA;
| | - Constantinos Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA;
- Center for Image-Guided Neurosurgery, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
12
|
Scheffler P, Fung C, Momjian S, Koessinger D, Häni L, Neidert N, Straehle J, Volz F, Schnell O, Beck J, El Rahal A. Dexamethasone in Patients with Glioblastoma: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:1393. [PMID: 38611071 PMCID: PMC11011080 DOI: 10.3390/cancers16071393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
OBJECTIVE Glioblastomas are the most common primary central nervous system (CNS) tumors. Although modern management strategies have modestly improved overall survival, the prognosis remains dismal, with treatment side effects often impinging on the clinical course. Glioblastomas cause neurological dysfunction by infiltrating CNS tissue and via perifocal oedema formation. The administration of steroids such as dexamethasone is thought to alleviate symptoms by reducing oedema. However, despite its widespread use, the evidence for the administration of dexamethasone is limited and conflicting. Therefore, we aimed to review the current evidence concerning the use and outcomes of dexamethasone in patients with glioblastoma. METHODS We performed a systematic review and meta-analysis according to the PRISMA-P guidelines. We performed a restricted search using the keywords "Dexamethasone" and "Glioblastoma" on PubMed, Web of Science, Cochrane Library, and Academic Search Premier. We included studies reporting on overall survival (OS) and progression-free survival (PFS) in glioblastoma patients receiving higher or lower dexamethasone doses. The risk of bias was assessed using ROBINS-I. We performed a meta-analysis using a random effects model for OS and PFS. RESULTS Twenty-two retrospective studies were included. Higher doses of dexamethasone were associated with poorer OS (hazard ratio 1.62, confidence interval 1.40-1.88) and PFS (1.49, 1.23-1.81). OS remained worse even when studies corrected for clinical status (1.52, 1.38-1.67). CONCLUSION Despite the widespread use of dexamethasone in glioblastoma patients, its use is correlated with worse long-term outcomes. Consequently, Dexamethasone administration should be restricted to selected symptomatic patients. Future prospective studies are crucial to confirm these findings.
Collapse
Affiliation(s)
- Pierre Scheffler
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Christian Fung
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Shahan Momjian
- Department of Neurosurgery, Geneva University Hospital, Faculty of Medicine of Geneva, 1205 Geneva, Switzerland
| | - Dominik Koessinger
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Levin Häni
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Nicolas Neidert
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
- Berta-Ottenstein Programme, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Jakob Straehle
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Florian Volz
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
| | - Amir El Rahal
- Department of Neurosurgery, Medical Center University of Freiburg, 79098 Freiburg, Germany; (P.S.)
- Department of Neurosurgery, Geneva University Hospital, Faculty of Medicine of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
13
|
Salvato I, Marchini A. Immunotherapeutic Strategies for the Treatment of Glioblastoma: Current Challenges and Future Perspectives. Cancers (Basel) 2024; 16:1276. [PMID: 38610954 PMCID: PMC11010873 DOI: 10.3390/cancers16071276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Despite decades of research and the best up-to-date treatments, grade 4 Glioblastoma (GBM) remains uniformly fatal with a patient median overall survival of less than 2 years. Recent advances in immunotherapy have reignited interest in utilizing immunological approaches to fight cancer. However, current immunotherapies have so far not met the anticipated expectations, achieving modest results in their journey from bench to bedside for the treatment of GBM. Understanding the intrinsic features of GBM is of crucial importance for the development of effective antitumoral strategies to improve patient life expectancy and conditions. In this review, we provide a comprehensive overview of the distinctive characteristics of GBM that significantly influence current conventional therapies and immune-based approaches. Moreover, we present an overview of the immunotherapeutic strategies currently undergoing clinical evaluation for GBM treatment, with a specific emphasis on those advancing to phase 3 clinical studies. These encompass immune checkpoint inhibitors, adoptive T cell therapies, vaccination strategies (i.e., RNA-, DNA-, and peptide-based vaccines), and virus-based approaches. Finally, we explore novel innovative strategies and future prospects in the field of immunotherapy for GBM.
Collapse
Affiliation(s)
- Ilaria Salvato
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg;
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
14
|
Zolotukhin DB, Horkowitz A, Keidar M. Electromagnetic Nature of Distant Interaction of the Atmospheric Pressure Helium Plasma Discharge Tube with Glioblastoma Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13597-13610. [PMID: 38453642 DOI: 10.1021/acsami.4c00619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Atmospheric pressure coaxial gaseous discharge tubes (DTs) with helium have demonstrated potential for in vitro inactivation or sensitization of glioblastoma cancer cells. Here, we study the effect of two configurations of the DT electrode system on its electromagnetic emissivity as well as other physical factors (heating and UV emission) that form in the vicinity of this device. We demonstrate that the configuration of the DT electrodes that concentrates the discharge streamers near the top of the device has a distant (cm scale) deactivation effect on U87-MG glioblastoma cancer cells when irradiated, without measurable UV components in the DT optical emission spectra. This effect persists even through different barriers such as glass, plastic, or quartz Petri dishes but is eliminated when glass or plastic dishes are filled with water. These findings demonstrate the potential for development of noninvasive, physical-based treatment methods of deep-tissue tumors.
Collapse
Affiliation(s)
- Denis B Zolotukhin
- The George Washington University, 800 22nd Street, Northwest, Washington, District of Columbia 20052, United States
- Tomsk State University of Control Systems and Radioelectronics, 40 Lenin Avenue, Tomsk 634050, Russia
| | - Alex Horkowitz
- The George Washington University, 800 22nd Street, Northwest, Washington, District of Columbia 20052, United States
| | - Michael Keidar
- The George Washington University, 800 22nd Street, Northwest, Washington, District of Columbia 20052, United States
| |
Collapse
|
15
|
Li Y, Wang J, Song SR, Lv SQ, Qin JH, Yu SC. Models for evaluating glioblastoma invasion along white matter tracts. Trends Biotechnol 2024; 42:293-309. [PMID: 37806896 DOI: 10.1016/j.tibtech.2023.09.005] [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: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023]
Abstract
White matter tracts (WMs) are one of the main invasion paths of glioblastoma multiforme (GBM). The lack of ideal research models hinders our understanding of the details and mechanisms of GBM invasion along WMs. To date, many potential in vitro models have been reported; nerve fiber culture models and nanomaterial models are biocompatible, and the former have electrically active neurons. Brain slice culture models, organoid models, and microfluidic chip models can simulate the real brain and tumor microenvironment (TME), which contains a variety of cell types. These models are closer to the real in vivo environment and are helpful for further studying not only invasion along WMs by GBM, but also perineural invasion and brain metastasis by solid tumors.
Collapse
Affiliation(s)
- Yao Li
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jian-Hua Qin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Niaoning 116023, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
16
|
Oliveira AGS, Rocha MA, de Azevedo LS, Coelho ATDM, Chagas RCR, Santos HB, Thomé RG, Samuel P, Wolfram E, Kim B, Reis RM, Ribeiro RIMA. Tapirira guianensis is Selectively Cytotoxic, Induces Apoptosis to the Glioblastoma and Decreases Tumor Growth and Angiogenesis in vivo. PLANTA MEDICA 2024; 90:13-24. [PMID: 37832581 DOI: 10.1055/a-2181-2569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Glioblastoma is the most frequent primary malignant brain tumor without effective treatment, which makes this work extremely relevant. The study of the bioactive compounds from medicinal plants plays an important role in the discovery of new drugs.This research investigated the constituents of Tapirira guianensis and its antitumor potential (in vitro and in vivo) in glioblastoma. The T. guianensis extracts were characterized by mass spectrometry. The ethyl acetate partition (01ID) and its fractions 01ID-F2 and 01ID-F4 from T. guianensis showed potential antitumor treatment evidenced by selective cytotoxicity for GAMG with IC50 14.1 µg/mL, 83.07 µg/mL, 59.27 µg/mL and U251 with IC50 25.92 µg/mL, 37.3 µg/mL and 18.84 µg/mL. Fractions 01ID-F2 and 01ID-F4 were 10 times more selective when compared to TMZ and 01ID for the two evaluated cell lines. T. guianensis also reduced matrix metalloproteinases 2 - 01ID-F2 (21.84%), 01ID-F4 (29.6%) and 9 - 01ID-F4 (73.42%), ID-F4 (53.84%) activities, and induced apoptosis mainly through the extrinsic pathway. Furthermore, all treatments significantly reduced tumor size (01ID p < 0,01, 01ID-F2 p < 0,01 and 01ID-F4 p < 0,0001) and caused blood vessels to shrink in vivo. The present findings highlight that T. guianensis exhibits considerable antitumor potential in preclinical studies of glioblastoma. This ability may be related to the phenolic compounds and sesquiterpene derivatives identified in the extracts. This study deserves further in vivo research, followed by clinical investigation.
Collapse
Affiliation(s)
- Ana Gabriela Silva Oliveira
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Marina Andrade Rocha
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Lucas Santos de Azevedo
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | | | - Rafael César Russo Chagas
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Hélio Batista Santos
- Tissue Processing Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Ralph Gruppi Thomé
- Tissue Processing Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Peter Samuel
- Zurich University of Applied Sciences, Department of Life Sciences and Facility Management, Wädenswil, Switzerland
| | - Evelyn Wolfram
- Zurich University of Applied Sciences, Department of Life Sciences and Facility Management, Wädenswil, Switzerland
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal
| | | |
Collapse
|
17
|
Al-Hawary SIS, Alhajlah S, Olegovich BD, Hjazi A, Rajput P, Ali SHJ, Abosoda M, Ihsan A, Oudah SK, Mustafa YF. Effective extracellular vesicles in glioma: Focusing on effective ncRNA exosomes and immunotherapy methods for treatment. Cell Biochem Funct 2024; 42:e3921. [PMID: 38269511 DOI: 10.1002/cbf.3921] [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: 11/02/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024]
Abstract
This comprehensive article explores the complex field of glioma treatment, with a focus on the important roles of non-coding RNAsRNAs (ncRNAs) and exosomes, as well as the potential synergies of immunotherapy. The investigation begins by examining the various functions of ncRNAs and their involvement in glioma pathogenesis, progression, and as potential diagnostic biomarkers. Special attention is given to exosomes as carriers of ncRNAs and their intricate dynamics within the tumor microenvironment. The exploration extends to immunotherapy methods, analyzing their mechanisms and clinical implications in the treatment of glioma. By synthesizing these components, the article aims to provide a comprehensive understanding of how ncRNAs, exosomes, and immunotherapy interact, offering valuable insights into the evolving landscape of glioma research and therapeutic strategies.
Collapse
Affiliation(s)
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqraa, Saudi Arabia
| | - Bokov Dmitry Olegovich
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Pranchal Rajput
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, India
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - Munther Abosoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Ihsan
- Department of Medical Laboratories Techniques, Imam Ja'afar Al-Sadiq University, Iraq
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
18
|
Wang J, Zhang J, Zhang Q, Zhang W, Zhang Q, Jin G, Liu F. TS-2021, a third-generation oncolytic adenovirus that carried Ki67 promoter, TGF-β2 5'UTR, and IL-15 against experimental glioblastoma. J Med Virol 2024; 96:e29335. [PMID: 38149454 DOI: 10.1002/jmv.29335] [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: 07/09/2023] [Revised: 11/21/2023] [Accepted: 12/09/2023] [Indexed: 12/28/2023]
Abstract
Oncolytic virotherapy is a promising therapeutic approach for glioblastoma (GBM) treatment, although the outcomes are partially satisfactory. Hence, more effective strategies are needed urgently to modify therapeutic viruses to enhance their efficiency and safety in killing tumor cells and improve the survival rate of GBM patients. This study generated a new-generation oncolytic adenovirus Ad5 KT-E1A-IL-15 (TS-2021) and evaluated its antitumor efficacy. Ex vivo analyses revealed Ki67 and TGF-β2 co-localized in GBM cells. In addition, TS-2021 selectively replicated in GBM cells, which was dependent on the expression of Ki67 and TGF-β2. The immunocompetent mice model of GBM demonstrated the in vivo efficacy of TS-2021 by inhibiting tumor growth and improving survival proficiently. Notably, TS-2021 effectively reduced MMP3 expression by inactivating the MKK4/JNK pathway, thereby reducing tumor invasiveness. Altogether, the findings of the present study highlight that TS-2021 can effectively target GBM cells expressing high levels of Ki67 and TGF-β2, exerting potent antitumor effects. Additionally, it can improve efficacy and suppress tumor invasiveness by inhibiting the MKK4/JNK/MMP3 pathway. Thus our study demonstrates the efficiency of the novel TS-2021 in the mouse model and provides a potential therapeutic option for patients with GBM.
Collapse
Affiliation(s)
- Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Wenxin Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Qi Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| |
Collapse
|
19
|
Premachandran S, Haldavnekar R, Ganesh S, Das S, Venkatakrishnan K, Tan B. Self-Functionalized Superlattice Nanosensor Enables Glioblastoma Diagnosis Using Liquid Biopsy. ACS NANO 2023; 17:19832-19852. [PMID: 37824714 DOI: 10.1021/acsnano.3c04118] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Glioblastoma (GBM), the most aggressive and lethal brain cancer, is detected only in the advanced stage, resulting in a median survival rate of 15 months. Therefore, there is an urgent need to establish GBM diagnosis tools to identify the tumor accurately. The clinical relevance of the current liquid biopsy techniques for GBM diagnosis remains mostly undetermined, owing to the challenges posed by the blood-brain barrier (BBB) that restricts biomarkers entering the circulation, resulting in the unavailability of clinically validated circulating GBM markers. GBM-specific liquid biopsy for diagnosis and prognosis of GBM has not yet been developed. Here, we introduce extracellular vesicles of GBM cancer stem cells (GBM CSC-EVs) as a previously unattempted, stand-alone GBM diagnosis modality. As GBM CSCs are fundamental building blocks of tumor initiation and recurrence, it is desirable to investigate these reliable signals of malignancy in circulation for accurate GBM diagnosis. So far, there are no clinically validated circulating biomarkers available for GBM. Therefore, a marker-free approach was essential since conventional liquid biopsy relying on isolation methodology was not viable. Additionally, a mechanism capable of trace-level detection was crucial to detecting the rare GBM CSC-EVs from the complex environment in circulation. To break these barriers, we applied an ultrasensitive superlattice sensor, self-functionalized for surface-enhanced Raman scattering (SERS), to obtain holistic molecular profiling of GBM CSC-EVs with a marker-free approach. The superlattice sensor exhibited substantial SERS enhancement and ultralow limit of detection (LOD of attomolar 10-18 M concentration) essential for trace-level detection of invisible GBM CSC-EVs directly from patient serum (without isolation). We detected as low as 5 EVs in 5 μL of solution, achieving the lowest LOD compared to existing SERS-based studies. We have experimentally demonstrated the crucial role of the signals of GBM CSC-EVs in the precise detection of glioblastoma. This was evident from the unique molecular profiles of GBM CSC-EVs demonstrating significant variation compared to noncancer EVs and EVs of GBM cancer cells, thus adding more clarity to the current understanding of GBM CSC-EVs. Preliminary validation of our approach was undertaken with a small amount of peripheral blood (5 μL) derived from GBM patients with 100% sensitivity and 97% specificity. Identification of the signals of GBM CSC-EV in clinical sera specimens demonstrated that our technology could be used for accurate GBM detection. Our technology has the potential to improve GBM liquid biopsy, including real-time surveillance of GBM evolution in patients upon clinical validation. This demonstration of liquid biopsy with GBM CSC-EV provides an opportunity to introduce a paradigm potentially impacting the current landscape of GBM diagnosis.
Collapse
Affiliation(s)
- Srilakshmi Premachandran
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Rupa Haldavnekar
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Swarna Ganesh
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Sunit Das
- Scientist, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Institute of Medical Sciences, Neurosurgery, University of Toronto, Toronto, Ontario M5T 1P5, Canada
| | - Krishnan Venkatakrishnan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Bo Tan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| |
Collapse
|
20
|
Peleli M, Antoniadou I, Rodrigues-Junior DM, Savvoulidou O, Caja L, Katsouda A, Ketelhuth DFJ, Stubbe J, Madsen K, Moustakas A, Papapetropoulos A. Cystathionine gamma-lyase (CTH) inhibition attenuates glioblastoma formation. Redox Biol 2023; 64:102773. [PMID: 37300955 PMCID: PMC10363444 DOI: 10.1016/j.redox.2023.102773] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
PURPOSE Glioblastoma (GBM) is the most common type of adult brain tumor with extremely poor survival. Cystathionine-gamma lyase (CTH) is one of the main Hydrogen Sulfide (H2S) producing enzymes and its expression contributes to tumorigenesis and angiogenesis but its role in glioblastoma development remains poorly understood. METHODS and Principal Results: An established allogenic immunocompetent in vivo GBM model was used in C57BL/6J WT and CTH KO mice where the tumor volume and tumor microvessel density were blindly measured by stereological analysis. Tumor macrophage and stemness markers were measured by blinded immunohistochemistry. Mouse and human GBM cell lines were used for cell-based analyses. In human gliomas, the CTH expression was analyzed by bioinformatic analysis on different databases. In vivo, the genetic ablation of CTH in the host led to a significant reduction of the tumor volume and the protumorigenic and stemness transcription factor sex determining region Y-box 2 (SOX2). The tumor microvessel density (indicative of angiogenesis) and the expression levels of peritumoral macrophages showed no significant changes between the two genotypes. Bioinformatic analysis in human glioma tumors revealed that higher CTH expression is positively correlated to SOX2 expression and associated with worse overall survival in all grades of gliomas. Patients not responding to temozolomide have also higher CTH expression. In mouse or human GBM cells, pharmacological inhibition (PAG) or CTH knockdown (siRNA) attenuates GBM cell proliferation, migration and stem cell formation frequency. MAJOR CONCLUSIONS Inhibition of CTH could be a new promising target against glioblastoma formation.
Collapse
Affiliation(s)
- Maria Peleli
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23, Uppsala, Sweden; Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winslowsvej 21, 3, 5000, Odense C, Denmark
| | - Ivi Antoniadou
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dorival Mendes Rodrigues-Junior
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23, Uppsala, Sweden
| | - Odysseia Savvoulidou
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winslowsvej 21, 3, 5000, Odense C, Denmark
| | - Laia Caja
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23, Uppsala, Sweden
| | - Antonia Katsouda
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Daniel F J Ketelhuth
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winslowsvej 21, 3, 5000, Odense C, Denmark; Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winslowsvej 21, 3, 5000, Odense C, Denmark
| | - Kirsten Madsen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winslowsvej 21, 3, 5000, Odense C, Denmark; Department of Pathology, Odense University Hospital, J.B Winslowsvej 15, 5000, Odense C, Denmark
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23, Uppsala, Sweden.
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
21
|
Villa GR, Chiocca EA. The Role of Long Noncoding Ribonucleic Acids in Glioblastoma: What the Neurosurgeon Should Know. Neurosurgery 2023; 92:1104-1111. [PMID: 36880757 DOI: 10.1227/neu.0000000000002449] [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: 12/12/2022] [Accepted: 01/12/2023] [Indexed: 03/08/2023] Open
Abstract
A significant proportion of the human transcriptome, long noncoding RNAs (lncRNAs) play pivotal roles in several aspects of glioblastoma (GBM) pathophysiology including proliferation, invasion, radiation and temozolomide resistance, and immune modulation. The majority of lncRNAs exhibit tissue- and tumor-specific expression, lending them to be attractive targets for therapeutic translation. In recent years, unprecedented progress has been made toward our understanding of lncRNA in GBM. In this review, we discuss the function of lncRNAs, including specific lncRNAs that have critical roles in key aspects of GBM pathophysiology, and potential clinical relevance of lncRNAs for patients with GBM.
Collapse
Affiliation(s)
- Genaro Rodriguez Villa
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston , Massachusetts , USA
| | | |
Collapse
|
22
|
Feldheim J, Kessler AF, Feldheim JJ, Schmitt D, Oster C, Lazaridis L, Glas M, Ernestus RI, Monoranu CM, Löhr M, Hagemann C. BRMS1 in Gliomas-An Expression Analysis. Cancers (Basel) 2023; 15:cancers15112907. [PMID: 37296870 DOI: 10.3390/cancers15112907] [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: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The metastatic suppressor BRMS1 interacts with critical steps of the metastatic cascade in many cancer entities. As gliomas rarely metastasize, BRMS1 has mainly been neglected in glioma research. However, its interaction partners, such as NFκB, VEGF, or MMPs, are old acquaintances in neurooncology. The steps regulated by BRMS1, such as invasion, migration, and apoptosis, are commonly dysregulated in gliomas. Therefore, BRMS1 shows potential as a regulator of glioma behavior. By bioinformatic analysis, in addition to our cohort of 118 specimens, we determined BRMS1 mRNA and protein expression as well as its correlation with the clinical course in astrocytomas IDH mutant, CNS WHO grade 2/3, and glioblastoma IDH wild-type, CNS WHO grade 4. Interestingly, we found BRMS1 protein expression to be significantly decreased in the aforementioned gliomas, while BRMS1 mRNA appeared to be overexpressed throughout. This dysregulation was independent of patients' characteristics or survival. The protein and mRNA expression differences cannot be finally explained at this stage. However, they suggest a post-transcriptional dysregulation that has been previously described in other cancer entities. Our analyses present the first data on BRMS1 expression in gliomas that can provide a starting point for further investigations.
Collapse
Affiliation(s)
- Jonas Feldheim
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Almuth F Kessler
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Julia J Feldheim
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Department of Neurosurgery, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Dominik Schmitt
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Department of Nuclear Medicine, University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christoph Oster
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Lazaros Lazaridis
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Ralf-Ingo Ernestus
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Camelia M Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Mario Löhr
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Carsten Hagemann
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| |
Collapse
|
23
|
Behrooz AB, Latifi-Navid H, Nezhadi A, Świat M, Los M, Jamalpoor Z, Ghavami S. Molecular mechanisms of microRNAs in glioblastoma pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119482. [PMID: 37146725 DOI: 10.1016/j.bbamcr.2023.119482] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Glioblastoma (GBM) is human's most prevalent and severe brain cancer. Epigenetic regulators, micro(mi)RNAs, significantly impact cellular health and disease because of their wide range of targets and functions. The "epigenetic symphony" in which miRNAs perform is responsible for orchestrating the transcription of genetic information. The discovery of regulatory miRNA activities in GBM biology has shown that various miRNAs play a vital role in disease onset and development. Here, we summarize our current understanding of the current state-of-the-art and latest findings regarding the interactions between miRNAs and molecular mechanisms commonly associated with GBM pathogenesis. Moreover, by literature review and reconstruction of the GBM gene regulatory network, we uncovered the connection between miRNAs and critical signaling pathways such as cell proliferation, invasion, and cell death, which provides promising hints for identifying potential therapeutic targets for the treatment of GBM. In addition, the role of miRNAs in GBM patient survival was investigated. The present review, which contains new analyses of the previous literature, may lead to new avenues to explore in the future for the development of multitargeted miRNA-based therapies for GBM.
Collapse
Affiliation(s)
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Akram Nezhadi
- Cognitive Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Maciej Świat
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Marek Los
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
24
|
Possible regulation of ganglioside GD3 synthase gene expression with DNA methylation in human glioma cells. Glycoconj J 2023; 40:323-332. [PMID: 36897478 DOI: 10.1007/s10719-023-10108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023]
Abstract
Gangliosides are expressed in nervous systems and some neuroectoderm-derived tumors at high levels and play pivotal roles. However, mechanisms for the regulation of glycosyltransferase genes responsible for the ganglioside synthesis are not well understood. In this study, we analyzed DNA methylation patterns of promoter regions of GD3 synthase (ST8SIA1) as well as mRNA levels and ganglioside expression using human glioma cell lines. Among 5 cell lines examined, 4 lines showed changes in the expression levels of related genes after treatment with 5-aza-dC. LN319 showed up-regulation of St8sia1 and increased b-series gangliosides after 5-aza-dC treatment, and an astrocytoma cell line, AS showed high expression of ST8SIA1 and b-series gangliosides persistently before and after 5-Aza-2'-deoxycytidine treatment. Using these 2 cell lines, DNA methylation patterns of the promoter regions of the gene were analyzed by bisulfite-sequencing. Consequently, 2 regions that were methylated before 5-Aza-2'-deoxycytidine treatment were demethylated in LN319 after the treatment, while those regions were persistently demethylated in AS. These 2 regions corresponded with sites defined as promoter regions by Luciferase assay. Taken together, it was suggested that ST8SIA1 gene is regulated by DNA methylation at the promoter regions, leading to the regulation of tumor phenotypes.
Collapse
|
25
|
Hashimoto N, Kitai R, Fujita S, Yamauchi T, Isozaki M, Kikuta KI. Single-Cell Analysis of Unidirectional Migration of Glioblastoma Cells Using a Fiber-Based Scaffold. ACS APPLIED BIO MATERIALS 2023; 6:765-773. [PMID: 36758146 PMCID: PMC9945112 DOI: 10.1021/acsabm.2c00958] [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] [Indexed: 02/11/2023]
Abstract
Glioblastoma (GBM) is a malignant incurable brain tumor in which immature neoplastic cells infiltrate brain tissue by spreading along nerve fibers. The aim of the study was to compare the migration abilities of glioma cells with those of other cancer cells and elucidate the migratory profiles underlying the differential migration of glioma cells using a fiber-based quantitative migration assay. Here, wound healing and transwell assays were used to assess cell mobility in four cell lines: U87-MG glioblastoma cells, MDA-MB-231 breast cancer cells, HCT116 colorectal cancer cells, and MKN45 gastric cancer cells. We also assessed cell mobility using a fiber model that mimics nerve fibers. Time-lapse video microscopy was used to observe cell migration and morphology. The cytoskeleton arrangement was assessed in the fiber model and compared with that in the conventional cell culture model. The conventional evaluation of cell migration ability revealed that the migration ability of breast cancer and glioblastoma cell lines was higher than that of colon cancer and gastric cancer cell lines. The fiber model confirmed that the glioblastoma cell line had a significantly higher migration ability than other cell lines. Tubulin levels were significantly higher in the glioblastoma cells than in other cell lines. In conclusion, the developed fiber-based culture model revealed the specific migratory profile of GBM cells during invasion.
Collapse
Affiliation(s)
- Norichika Hashimoto
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Fukui General Hospital, 58-16-1 Egami-cho, Fukui-shi, Fukui 910-8561, Japan
| | - Ryuhei Kitai
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Kaga Medical Center, Kaga, Ri 36, Sakumi-machi, Kaga-shi, Ishikawa 922-8522, Japan
| | - Satoshi Fujita
- Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Takahiro Yamauchi
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Makoto Isozaki
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Ken-Ichiro Kikuta
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| |
Collapse
|
26
|
Adekeye AO, Needham D, Rahman R. Low-Density Lipoprotein Pathway Is a Ubiquitous Metabolic Vulnerability in High Grade Glioma Amenable for Nanotherapeutic Delivery. Pharmaceutics 2023; 15:pharmaceutics15020599. [PMID: 36839921 PMCID: PMC9958636 DOI: 10.3390/pharmaceutics15020599] [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: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Metabolic reprogramming, through increased uptake of cholesterol in the form of low-density lipoproteins (LDL), is one way by which cancer cells, including high grade gliomas (HGG), maintain their rapid growth. In this study, we determined LDL receptor (LDLR) expression in HGGs using immunohistochemistry on tissue microarrays from intra- and inter tumour regions of 36 adult and 133 paediatric patients to confirm LDLR as a therapeutic target. Additionally, we analysed expression levels in three representative cell line models to confirm their future utility to test LDLR-targeted nanoparticle uptake, retention, and cytotoxicity. Our data show widespread LDLR expression in adult and paediatric cohorts, but with significant intra-tumour variation observed between the core and either rim or invasive regions of adult HGG. Expression was independent of paediatric tumour grade or identified clinicopathological factors. LDLR-expressing tumour cells localized preferentially within perivascular niches, also with significant adult intra-tumour variation. We demonstrated variable levels of LDLR expression in all cell lines, confirming their suitability as models to test LDLR-targeted nanotherapy delivery. Overall, our study reveals the LDLR pathway as a ubiquitous metabolic vulnerability in high grade gliomas across all ages, amenable to future consideration of LDL-mediated nanoparticle/drug delivery to potentially circumvent tumour heterogeneity.
Collapse
Affiliation(s)
- Adenike O. Adekeye
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - David Needham
- Department of Mechanical Engineering and Material Science, School of Engineering, Duke University, Durham, NC 27708, USA
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence:
| |
Collapse
|
27
|
Cell signaling activation and extracellular matrix remodeling underpin glioma tumor microenvironment heterogeneity and organization. Cell Oncol 2022; 46:589-602. [PMID: 36567397 DOI: 10.1007/s13402-022-00763-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Tumor cells thrive by adapting to the signals in their microenvironment. To adapt, cancer cells activate signaling and transcriptional programs and migrate to establish micro-niches, in response to signals from neighboring cells and non-cellular stromal factors. Understanding how the tumor microenvironment evolves during disease progression is crucial to deciphering the mechanisms underlying the functional behavior of cancer cells. METHODS Multiplex immunohistochemistry, spatial analysis and histological dyes were used to identify and measure immune cell infiltration, cell signal activation and extracellular matrix deposition in low-grade, high-grade astrocytoma and glioblastoma. RESULTS We show that lower grade astrocytoma tissue is largely devoid of infiltrating immune cells and extracellular matrix proteins, while high-grade astrocytoma exhibits abundant immune cell infiltration, activation, and extensive tissue remodeling. Spatial analysis shows that most T-cells are restricted to perivascular regions, but bone marrow-derived macrophages penetrate deep into neoplastic cell-rich regions. The tumor microenvironment is characterized by heterogeneous PI3K, MAPK and CREB signaling, with specific signaling profiles correlating with distinct pathological hallmarks, including angiogenesis, tumor cell density and regions where neoplastic cells border the extracellular matrix. Our results also show that tissue remodeling is important in regulating the architecture of the tumor microenvironment during tumor progression. CONCLUSION The tumor microenvironment in malignant astrocytoma, exhibits changes in cell composition, cell signaling activation and extracellular matrix deposition during disease development and that targeting the extracellular matrix, as well as cell signaling activation will be critical to designing personalized therapy.
Collapse
|
28
|
Tamma R, Ingravallo G, Annese T, d’Amati A, Lorusso L, Ribatti D. Tumor Microenvironment and Microvascular Density in Human Glioblastoma. Cells 2022; 12:cells12010011. [PMID: 36611806 PMCID: PMC9818990 DOI: 10.3390/cells12010011] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is a very aggressive form of cancer affecting the central nervous system. Although it occurs almost exclusively in the brain, glioblastoma can also appear in the brainstem, cerebellum, and spinal cord. It is characterized by high rates of proliferation, invasion, and necrosis. Moreover, GBM is a highly vascularized tumor and presents resistance to therapy. Recent data indicate that GBM cells are surrounded by a microenvironment (TME) which includes a complex network constituted of cellular/extracellular components and vessels able to influence both tumor growth and angiogenesis. In this retrospective study, we evaluated 30 bioptic specimens of adult patients diagnosed with IDH1 wild type GBM taken at the time of the first diagnosis. Each section has been divided into two experimental zones: the tumor side and the healthy surrounding tissue. We performed a series of immunohistochemical stainings with the purpose of evaluating the presence of total and M2 macrophages, CD4+-, CD8+-lymphocytes, and CD34+ microvessels. In addition, we have also evaluated the percentage of cells expressing bcl6 and p53 to determine any possible correlations with TME. Our data showed a significant increase in the total and M2 type macrophages, of CD4+ and CD8+ lymphocytes, and of CD34+ microvessels in the tumoral area respective to the healthy zone. We also confirmed our previous data showing the higher number of p53 and BCL6+ cells in the tumor area with a positive correlation between BCL6 and CD34+ microvessels. In conclusion, the data that came from this work support the important role played by microenvironment components in GBM progression. These results could contribute to the generation of new specific therapies useful in preventing GBM progression.
Collapse
Affiliation(s)
- Roberto Tamma
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (R.T.); (D.R.); Tel.: +39-0805478323 (D.R.); Fax: +39-0805478310 (D.R.)
| | - Giuseppe Ingravallo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, 70124 Bari, Italy
| | - Tiziana Annese
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, 70124 Bari, Italy
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, 70010 Bari, Italy
| | - Antonio d’Amati
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, 70124 Bari, Italy
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, 70124 Bari, Italy
| | - Loredana Lorusso
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, 70124 Bari, Italy
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (R.T.); (D.R.); Tel.: +39-0805478323 (D.R.); Fax: +39-0805478310 (D.R.)
| |
Collapse
|
29
|
McCord M, Bartom E, Burdett K, Baran A, Eckerdt FD, Balyasnikova IV, McCortney K, Sears T, Cheng SY, Sarkaria JN, Stupp R, Heimberger AB, Ahmed A, James CD, Horbinski C. Modeling Therapy-Driven Evolution of Glioblastoma with Patient-Derived Xenografts. Cancers (Basel) 2022; 14:5494. [PMID: 36428586 PMCID: PMC9688760 DOI: 10.3390/cancers14225494] [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: 09/18/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Adult-type diffusely infiltrating gliomas, of which glioblastoma is the most common and aggressive, almost always recur after treatment and are fatal. Improved understanding of therapy-driven tumor evolution and acquired therapy resistance in gliomas is essential for improving patient outcomes, yet the majority of the models currently used in preclinical research are of therapy-naïve tumors. Here, we describe the development of therapy-resistant IDH-wildtype glioblastoma patient-derived xenografts (PDX) through orthotopic engraftment of therapy naïve PDX in athymic nude mice, and repeated in vivo exposure to the therapeutic modalities most often used in treating glioblastoma patients: radiotherapy and temozolomide chemotherapy. Post-temozolomide PDX became enriched for C>T transition mutations, acquired inactivating mutations in DNA mismatch repair genes (especially MSH6), and developed hypermutation. Such post-temozolomide PDX were resistant to additional temozolomide (median survival decrease from 80 days in parental PDX to 42 days in a temozolomide-resistant derivative). However, temozolomide-resistant PDX were sensitive to lomustine (also known as CCNU), a nitrosourea which induces tumor cell apoptosis by a different mechanism than temozolomide. These PDX models mimic changes observed in recurrent GBM in patients, including critical features of therapy-driven tumor evolution. These models can therefore serve as valuable tools for improving our understanding and treatment of recurrent glioma.
Collapse
Affiliation(s)
- Matthew McCord
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kirsten Burdett
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aneta Baran
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Frank D. Eckerdt
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Irina V. Balyasnikova
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Thomas Sears
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shi-Yuan Cheng
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester Minnesota, Rochester, MN 55905, USA
| | - Roger Stupp
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Amy B. Heimberger
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Atique Ahmed
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Charles David James
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Craig Horbinski
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Lou and Jean Malnati Brain Tumor Institute of Northwestern Medicine, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
30
|
Macfarlane FR, Lorenzi T, Painter KJ. The Impact of Phenotypic Heterogeneity on Chemotactic Self-Organisation. Bull Math Biol 2022; 84:143. [PMID: 36319913 PMCID: PMC9626439 DOI: 10.1007/s11538-022-01099-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
The capacity to aggregate through chemosensitive movement forms a paradigm of self-organisation, with examples spanning cellular and animal systems. A basic mechanism assumes a phenotypically homogeneous population that secretes its own attractant, with the well known system introduced more than five decades ago by Keller and Segel proving resolutely popular in modelling studies. The typical assumption of population phenotypic homogeneity, however, often lies at odds with the heterogeneity of natural systems, where populations may comprise distinct phenotypes that vary according to their chemotactic ability, attractant secretion, etc. To initiate an understanding into how this diversity can impact on autoaggregation, we propose a simple extension to the classical Keller and Segel model, in which the population is divided into two distinct phenotypes: those performing chemotaxis and those producing attractant. Using a combination of linear stability analysis and numerical simulations, we demonstrate that switching between these phenotypic states alters the capacity of a population to self-aggregate. Further, we show that switching based on the local environment (population density or chemoattractant level) leads to diverse patterning and provides a route through which a population can effectively curb the size and density of an aggregate. We discuss the results in the context of real world examples of chemotactic aggregation, as well as theoretical aspects of the model such as global existence and blow-up of solutions.
Collapse
Affiliation(s)
- Fiona R Macfarlane
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland.
| | - Tommaso Lorenzi
- Department of Mathematical Sciences "G. L. Lagrange", Politecnico di Torino, Turin, Italy
| | - Kevin J Painter
- Inter-university Department of Regional and Urban Studies and Planning, Politecnico di Torino, Turin, Italy
| |
Collapse
|
31
|
A Head-to-Head Comparison of 18F-Fluorocholine PET/CT and Conventional MRI as Predictors of Outcome in IDH Wild-Type High-Grade Gliomas. J Clin Med 2022; 11:jcm11206065. [PMID: 36294385 PMCID: PMC9605635 DOI: 10.3390/jcm11206065] [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: 08/22/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 01/24/2023] Open
Abstract
(1) Aim: To study the associations between imaging parameters derived from contrast-enhanced MRI (CE-MRI) and 18F-fluorocholine PET/CT and their performance as prognostic predictors in isocitrate dehydrogenase wild-type (IDH-wt) high-grade gliomas. (2) Methods: A prospective, multicenter study (FuMeGA: Functional and Metabolic Glioma Analysis) including patients with baseline CE-MRI and 18F-fluorocholine PET/CT and IDH wild-type high-grade gliomas. Clinical variables such as performance status, extent of surgery and adjuvant treatments (Stupp protocol vs others) were obtained and used to discriminate overall survival (OS) and progression-free survival (PFS) as end points. Multilesionality was assessed on the visual analysis of PET/CT and CE-MRI images. After tumor segmentation, standardized uptake value (SUV)-based variables for PET/CT and volume-based and geometrical variables for PET/CT and CE-MRI were calculated. The relationships among imaging techniques variables and their association with prognosis were evaluated using Pearson’s chi-square test and the t-test. Receiver operator characteristic, Kaplan−Meier and Cox regression were used for the survival analysis. (3) Results: 54 patients were assessed. The median PFS and OS were 5 and 11 months, respectively. Significant strong relationships between volume-dependent variables obtained from PET/CT and CE-MRI were found (r > 0.750, p < 0.05). For OS, significant associations were found with SUVmax, SUVpeak, SUVmean and sphericity (HR: 1.17, p = 0.035; HR: 1.24, p = 0.042; HR: 1.62, p = 0.040 and HR: 0.8, p = 0.022, respectively). Among clinical variables, only Stupp protocol and age showed significant associations with OS and PFS. No CE-MRI derived variables showed significant association with prognosis. In multivariate analysis, age (HR: 1.04, p = 0.002), Stupp protocol (HR: 2.81, p = 0.001), multilesionality (HR: 2.20, p = 0.013) and sphericity (HR: 0.79, p = 0.027) derived from PET/CT showed independent associations with OS. For PFS, only age (HR: 1.03, p = 0.021) and treatment protocol (HR: 2.20, p = 0.008) were significant predictors. (4) Conclusions: 18F-fluorocholine PET/CT metabolic and radiomic variables were robust prognostic predictors in patients with IDH-wt high-grade gliomas, outperforming CE-MRI derived variables.
Collapse
|
32
|
Chryplewicz A, Scotton J, Tichet M, Zomer A, Shchors K, Joyce JA, Homicsko K, Hanahan D. Cancer cell autophagy, reprogrammed macrophages, and remodeled vasculature in glioblastoma triggers tumor immunity. Cancer Cell 2022; 40:1111-1127.e9. [PMID: 36113478 PMCID: PMC9580613 DOI: 10.1016/j.ccell.2022.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/15/2022] [Indexed: 01/10/2023]
Abstract
Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities required for tumor progression and therapeutic resistance. We assessed this concept by combining vascular endothelial growth factor (VEGF) pathway inhibitors that remodel the tumor vasculature with the tricyclic antidepressant imipramine, which enhances autophagy in GBM cancer cells and unexpectedly reprograms immunosuppressive tumor-associated macrophages via inhibition of histamine receptor signaling to become immunostimulatory. While neither drug is efficacious as monotherapy, the combination of imipramine with VEGF pathway inhibitors orchestrates the infiltration and activation of CD8 and CD4 T cells, producing significant therapeutic benefit in several GBM mouse models. Inclusion up front of immune-checkpoint blockade with anti-programmed death-ligand 1 (PD-L1) in eventually relapsing tumors markedly extends survival benefit. The results illustrate the potential of mechanism-guided therapeutic co-targeting of disparate biological vulnerabilities in the tumor microenvironment.
Collapse
Affiliation(s)
- Agnieszka Chryplewicz
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland
| | - Julie Scotton
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Mélanie Tichet
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Anoek Zomer
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Ksenya Shchors
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Johanna A Joyce
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Department of Oncology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland
| | - Krisztian Homicsko
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Department of Oncology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland.
| |
Collapse
|
33
|
Zhao J, Liu X, Zhou Y, Zheng T, Tian Y. Surface-enhanced Raman scattering technology based on TiO2/Nb2C coated microfluidic chip for monitoring glioma cells invasion in real time. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Almammadov T, Elmazoglu Z, Atakan G, Kepil D, Aykent G, Kolemen S, Gunbas G. Locked and Loaded: β-Galactosidase Activated Photodynamic Therapy Agent Enables Selective Imaging and Targeted Treatment of Glioblastoma Multiforme Cancer Cells. ACS APPLIED BIO MATERIALS 2022; 5:4284-4293. [PMID: 36043987 PMCID: PMC9490748 DOI: 10.1021/acsabm.2c00484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Selective detection and effective therapy of brain cancer, specifically, the very aggressive glioblastoma multiforme (GBM), remains one of the paramount challenges in clinical settings. While radiotherapy combined surgery is proposed as the main treatment course, it has several drawbacks such as complexity of the operation and common development of recurrent tumors in this course of patient care. Unique opportunities presented by photodynamic therapy (PDT) offer promising, effective, and precise therapy against GBM cells along with simultaneous imaging opportunities. However, activatable, theranostic molecular systems in PDT modality for GBM remained scarce. Specifically, even though elevated β-galactosidase (β-gal) activity in glioblastoma cells is well-documented, targeted, activatable therapeutic PDT agents have not been realized. Herein, we report a β-galactosidase (β-gal) activatable phototheranostic agent based on an iodinated resorufin core (RB-1) which was realized in only three steps with commercial reagents in 29% overall yield. RB-1 showed very high singlet oxygen (1O2) quantum yield (54%) accompanied by a remarkable turn-on response in fluorescence upon enzymatic activation. RB-1 was tested in different cell lines and revealed selective photocytotoxicity in U-87MG glioblastoma cells. Additionally, thanks to almost 7% fluorescence quantum yield (ΦF) despite extremely high 1O2 generation yield, RB-1 was also demonstrated as a successful agent for fluorescence imaging of U-87MG cells. Due to significantly lower (β-gal) activity in healthy cells (NIH/3T3), RB-1 stayed in a passive state and showed minimal photo and dark toxicity. RB-1 marks the first example of a β-gal activatable phototheranostic agent toward effective treatment of glioblastoma.
Collapse
Affiliation(s)
- Toghrul Almammadov
- Department
of Chemistry, Koç University, Rumelifeneri Yolu, 34450 Istanbul, Turkey
| | - Zubeyir Elmazoglu
- Department
of Chemistry, Middle East Technical University
(METU), 06800 Ankara, Turkey
| | - Gizem Atakan
- Department
of Chemistry, Middle East Technical University
(METU), 06800 Ankara, Turkey
| | - Dilay Kepil
- Department
of Chemistry, Middle East Technical University
(METU), 06800 Ankara, Turkey
| | - Guzide Aykent
- Department
of Chemistry, Middle East Technical University
(METU), 06800 Ankara, Turkey
| | - Safacan Kolemen
- Department
of Chemistry, Koç University, Rumelifeneri Yolu, 34450 Istanbul, Turkey
- Surface
Science and Technology Center (KUYTAM), Koç University, 34450 Istanbul, Turkey
- Boron
and Advanced Materials Application and Research Center, Koç University, 34450 Istanbul, Turkey
| | - Gorkem Gunbas
- Department
of Chemistry, Middle East Technical University
(METU), 06800 Ankara, Turkey
| |
Collapse
|
35
|
Xie P, Zhang Y, Chen R, Zheng J, Cui G. PTBP3 promotes tumorigenesis of glioblastoma by stabilizing Twist1. Transl Oncol 2022; 25:101520. [PMID: 35987089 PMCID: PMC9411677 DOI: 10.1016/j.tranon.2022.101520] [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: 06/14/2022] [Revised: 07/25/2022] [Accepted: 08/11/2022] [Indexed: 12/04/2022] Open
Abstract
PTBP3 is upregulated in GBM and predicts poor prognosis. PTBP3 promotes proliferation, EMT, migration, and invasion of GBM. PTBP3 stabilizes Twist1 by decreasing its ubiquitination and degradation.
Objective Glioblastoma (GBM) is the most common malignancy tumor of central nervous system. PTBP3 was closely associated with the development of tumor. However, the function and molecular mechanism of PTBP3 in GBM is little known. Methods qPCR and immunoblotting were used to detect PTBP3 expression levels in glioma tissues and cells. CCK8, Edu, flow cytometry, wound healing, and transwell assays were used to examined the function of PTBP3 in GBM. qPCR, Immunoblotting, and ubiquitination assays were performed to identify the mechanism of PTBP3. Results We found that PTBP3 was upregulated in GBM, and high expression of PTBP3 correlated with the poor survival of GBM patients. PTBP3 knockdown reduced proliferation, invasion, and migration of GBM. Conversely, overexpressing PTBP3 has an opposite effect. Moreover, PTBP3 had an effect on the EMT of GBM. More importantly, we found that PTBP3 stabilized Twist1 by decreasing its ubiquitination and degradation. Furthermore, orthotopic xenograft models were used to demonstrate the PTBP3 on the development of GBM in vivo. Conclusion This study proved that PTBP3 promoted tumorigenesis of GBM by stabilizing Twist1, which provided a new therapeutic target for GBM.
Collapse
Affiliation(s)
- Peng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China; Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No.62, Huaihai Road(S.), Huai'an, Jiangsu 223002, P.R. China
| | - Yueqing Zhang
- Department of Neurosurgery, Huai'an Cancer Hospital, No19 shanyang Road, Huai'an, Jiangsu 223200, P.R. China
| | - Rui Chen
- Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No.62, Huaihai Road(S.), Huai'an, Jiangsu 223002, P.R. China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No.62, Huaihai Road(S.), Huai'an, Jiangsu 223002, P.R. China
| | - Gang Cui
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.
| |
Collapse
|
36
|
Norton ES, Whaley LA, Ulloa-Navas MJ, García-Tárraga P, Meneses KM, Lara-Velazquez M, Zarco N, Carrano A, Quiñones-Hinojosa A, García-Verdugo JM, Guerrero-Cázares H. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022; 19:58. [PMID: 35821139 PMCID: PMC9277938 DOI: 10.1186/s12987-022-00354-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive and common type of primary brain tumor in adults. Tumor location plays a role in patient prognosis, with tumors proximal to the lateral ventricles (LVs) presenting with worse overall survival, increased expression of stem cell genes, and increased incidence of distal tumor recurrence. This may be due in part to interaction of GBM with factors of the subventricular zone (SVZ), including those contained within the cerebrospinal fluid (CSF). However, direct interaction of GBM tumors with CSF has not been proved and would be hindered in the presence of an intact ependymal cell layer. Methods Here, we investigate the ependymal cell barrier and its derived extracellular matrix (ECM) fractones in the vicinity of a GBM tumor. Patient-derived GBM cells were orthotopically implanted into immunosuppressed athymic mice in locations distal and proximal to the LV. A PBS vehicle injection in the proximal location was included as a control. At four weeks post-xenograft, brain tissue was examined for alterations in ependymal cell health via immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. Results We identified local invading GBM cells within the LV wall and increased influx of CSF into the LV-proximal GBM tumor bulk compared to controls. In addition to the physical disruption of the ependymal cell barrier, we also identified increased signs of compromised ependymal cell health in LV-proximal tumor-bearing mice. These signs include increased accumulation of lipid droplets, decreased cilia length and number, and decreased expression of cell channel proteins. We additionally identified elevated numbers of small fractones in the SVZ within this group, suggesting increased indirect CSF-contained molecule signaling to tumor cells. Conclusions Our data is the first to show that LV-proximal GBMs physically disrupt the ependymal cell barrier in animal models, resulting in disruptions in ependymal cell biology and increased CSF interaction with the tumor bulk. These findings point to ependymal cell health and CSF-contained molecules as potential axes for therapeutic targeting in the treatment of GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00354-8.
Collapse
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Department of Biology, University of North Florida, Jacksonville, FL, USA
| | - María José Ulloa-Navas
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Patricia García-Tárraga
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Kayleah M Meneses
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
37
|
Adjei‐Sowah EA, O'Connor SA, Veldhuizen J, Lo Cascio C, Plaisier C, Mehta S, Nikkhah M. Investigating the Interactions of Glioma Stem Cells in the Perivascular Niche at Single-Cell Resolution using a Microfluidic Tumor Microenvironment Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201436. [PMID: 35619544 PMCID: PMC9313491 DOI: 10.1002/advs.202201436] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Indexed: 05/03/2023]
Abstract
The perivascular niche (PVN) is a glioblastoma tumor microenvironment (TME) that serves as a safe haven for glioma stem cells (GSCs), and acts as a reservoir that inevitably leads to tumor recurrence. Understanding cellular interactions in the PVN that drive GSC treatment resistance and stemness is crucial to develop lasting therapies for glioblastoma. The limitations of in vivo models and in vitro assays have led to critical knowledge gaps regarding the influence of various cell types in the PVN on GSCs behavior. This study developed an organotypic triculture microfluidic model as a means to recapitulate the PVN and study its impact on GSCs. This triculture platform, comprised of endothelial cells (ECs), astrocytes, and GSCs, is used to investigate GSC invasion, proliferation and stemness. Both ECs and astrocytes significantly increased invasiveness of GSCs. This study futher identified 15 ligand-receptor pairs using single-cell RNAseq with putative chemotactic mechanisms of GSCs, where the receptor is up-regulated in GSCs and the diffusible ligand is expressed in either astrocytes or ECs. Notably, the ligand-receptor pair SAA1-FPR1 is demonstrated to be involved in chemotactic invasion of GSCs toward PVN. The novel triculture platform presented herein can be used for therapeutic development and discovery of molecular mechanisms driving GSC biology.
Collapse
Affiliation(s)
| | - Samantha A. O'Connor
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Jaimeson Veldhuizen
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Costanza Lo Cascio
- Ivy Brain Tumor Center, Barrow Neurological InstituteSt. Joseph's Hospital and Medical Center350 W Thomas RdPhoenixAZ85013USA
| | - Christopher Plaisier
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Shwetal Mehta
- Ivy Brain Tumor Center, Barrow Neurological InstituteSt. Joseph's Hospital and Medical Center350 W Thomas RdPhoenixAZ85013USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287‐9709USA
| |
Collapse
|
38
|
The anti-angiogenic effect of atorvastatin loaded exosomes on glioblastoma tumor cells: An in vitro 3D culture model. Microvasc Res 2022; 143:104385. [DOI: 10.1016/j.mvr.2022.104385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/03/2022] [Accepted: 05/18/2022] [Indexed: 01/10/2023]
|
39
|
Kanwore K, Kanwore K, Adzika GK, Abiola AA, Guo X, Kambey PA, Xia Y, Gao D. Cancer Metabolism: The Role of Immune Cells Epigenetic Alteration in Tumorigenesis, Progression, and Metastasis of Glioma. Front Immunol 2022; 13:831636. [PMID: 35392088 PMCID: PMC8980436 DOI: 10.3389/fimmu.2022.831636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Glioma is a type of brain and spinal cord tumor that begins in glial cells that support the nervous system neurons functions. Age, radiation exposure, and family background of glioma constitute are risk factors of glioma initiation. Gliomas are categorized on a scale of four grades according to their growth rate. Grades one and two grow slowly, while grades three and four grow faster. Glioblastoma is a grade four gliomas and the deadliest due to its aggressive nature (accelerated proliferation, invasion, and migration). As such, multiple therapeutic approaches are required to improve treatment outcomes. Recently, studies have implicated the significant roles of immune cells in tumorigenesis and the progression of glioma. The energy demands of gliomas alter their microenvironment quality, thereby inducing heterogeneity and plasticity change of stromal and immune cells via the PI3K/AKT/mTOR pathway, which ultimately results in epigenetic modifications that facilitates tumor growth. PI3K is utilized by many intracellular signaling pathways ensuring the proper functioning of the cell. The activation of PI3K/AKT/mTOR regulates the plasma membrane activities, contributing to the phosphorylation reaction necessary for transcription factors activities and oncogenes hyperactivation. The pleiotropic nature of PI3K/AKT/mTOR makes its activity unpredictable during altered cellular functions. Modification of cancer cell microenvironment affects many cell types, including immune cells that are the frontline cells involved in inflammatory cascades caused by cancer cells via high cytokines synthesis. Typically, the evasion of immunosurveillance by gliomas and their resistance to treatment has been attributed to epigenetic reprogramming of immune cells in the tumor microenvironment, which results from cancer metabolism. Hence, it is speculative that impeding cancer metabolism and/or circumventing the epigenetic alteration of immune cell functions in the tumor microenvironment might enhance treatment outcomes. Herein, from an oncological and immunological perspective, this review discusses the underlying pathomechanism of cell-cell interactions enhancing glioma initiation and metabolism activation and tumor microenvironment changes that affect epigenetic modifications in immune cells. Finally, prospects for therapeutic intervention were highlighted.
Collapse
Affiliation(s)
- Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Konimpo Kanwore
- Faculty Mixed of Medicine and Pharmacy, Lomé-Togo, University of Lomé, Lomé, Togo
| | | | - Ayanlaja Abdulrahman Abiola
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Xiaoxiao Guo
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Ying Xia
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
40
|
Ayensa-Jiménez J, Doweidar MH, Sanz-Herrera JA, Doblare M. Understanding glioblastoma invasion using physically-guided neural networks with internal variables. PLoS Comput Biol 2022; 18:e1010019. [PMID: 35377875 PMCID: PMC9009781 DOI: 10.1371/journal.pcbi.1010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/14/2022] [Accepted: 03/15/2022] [Indexed: 11/18/2022] Open
Abstract
Microfluidic capacities for both recreating and monitoring cell cultures have opened the door to the use of Data Science and Machine Learning tools for understanding and simulating tumor evolution under controlled conditions. In this work, we show how these techniques could be applied to study Glioblastoma, the deadliest and most frequent primary brain tumor. In particular, we study Glioblastoma invasion using the recent concept of Physically-Guided Neural Networks with Internal Variables (PGNNIV), able to combine data obtained from microfluidic devices and some physical knowledge governing the tumor evolution. The physics is introduced in the network structure by means of a nonlinear advection-diffusion-reaction partial differential equation that models the Glioblastoma evolution. On the other hand, multilayer perceptrons combined with a nodal deconvolution technique are used for learning the go or grow metabolic behavior which characterises the Glioblastoma invasion. The PGNNIV is here trained using synthetic data obtained from in silico tests created under different oxygenation conditions, using a previously validated model. The unravelling capacity of PGNNIV enables discovering complex metabolic processes in a non-parametric way, thus giving explanatory capacity to the networks, and, as a consequence, surpassing the predictive power of any parametric approach and for any kind of stimulus. Besides, the possibility of working, for a particular tumor, with different boundary and initial conditions, permits the use of PGNNIV for defining virtual therapies and for drug design, thus making the first steps towards in silico personalised medicine. In this work, we apply Physically-Guided Neural Networks with Internal Variables (PGNNIV) to the understanding of the Glioblastoma evolution process. We explain the metabolic changes between the proliferative and migrative activity of Glioblastoma cell cultures by using the go or grow activation functions as a pair of internal variables, whose dependence on the oxygen level is unravelled by some building blocks of the whole PGNNIV. Due to its model-free nature, our method is able to identify different classical mechanistic approaches and to outperform cell culture evolution predictions, as we demonstrate in the paper. Unlike Biologically-Informed Neural Networks we can assimilate data obtained from different boundary conditions and under different external stimuli to simulate the tumor progression under arbitrary conditions. We demonstrate this ability by comparing the predictions with different boundary conditions, resulting in different oxygenation conditions. This flexibility enables the use of our proposed method for personalised medical purposes, as the cell culture metabolic information, for a particular tumor, is encapsulated in a sub-network and may be used for arbitrary in silico tests.
Collapse
Affiliation(s)
- Jacobo Ayensa-Jiménez
- Mechanical Engineering Department, School of Engineering and Architecture, University of Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Spain
- Aragón Institute of Health Research (IIS Aragón), Spain
| | - Mohamed H. Doweidar
- Mechanical Engineering Department, School of Engineering and Architecture, University of Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Spain
| | - Jose A. Sanz-Herrera
- Mechanical Engineering Department, School of Engineering, University of Sevilla, Spain
| | - Manuel Doblare
- Mechanical Engineering Department, School of Engineering and Architecture, University of Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Spain
- Aragón Institute of Health Research (IIS Aragón), Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Spain
- * E-mail:
| |
Collapse
|
41
|
Simões RV, Henriques RN, Cardoso BM, Fernandes FF, Carvalho T, Shemesh N. Glucose fluxes in glycolytic and oxidative pathways detected in vivo by deuterium magnetic resonance spectroscopy reflect proliferation in mouse glioblastoma. Neuroimage Clin 2022; 33:102932. [PMID: 35026626 PMCID: PMC8760481 DOI: 10.1016/j.nicl.2021.102932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/23/2022]
Abstract
We performed dynamic glucose enhanced (DGE) 2H-MRS in mouse GBM tumors. Marchenko-Pastur PCA denoising of 2H-MRS spectra improved kinetic quantification. Metabolic kinetics revealed differential glucose pathway fluxes in non-necrotic tumors. Modulation of glucose metabolism reflected tumor heterogeneity (proliferation).
Objectives Glioblastoma multiforme (GBM), the most aggressive glial brain tumors, can metabolize glucose through glycolysis and mitochondrial oxidation pathways. While specific dependencies on those pathways are increasingly associated with treatment response, detecting such GBM subtypes in vivo remains elusive. Here, we develop a dynamic glucose-enhanced deuterium spectroscopy (DGE 2H-MRS) approach for differentially assessing glucose turnover rates through glycolysis and mitochondrial oxidation in mouse GBM and explore their association with histologic features of the tumor and its microenvironment. Materials and methods GL261 and CT2A glioma allografts were induced in immunocompetent mice and scanned in vivo at 9.4 Tesla, harnessing DGE 2H-MRS with volume selection and Marchenko-Pastur PCA (MP-PCA) denoising to achieve high temporal resolution. Each tumor was also classified by histopathologic analysis and assessed for cell proliferation (Ki67 immunostaining), while the respective cell lines underwent in situ extracellular flux analysis to assess mitochondrial function. Results MP-PCA denoising of in vivo DGE 2H-MRS data significantly improved the time-course detection (~2-fold increased Signal-to-Noise Ratio) and fitting precision (−19 ± 1 % Cramér-Rao Lower Bounds) of 2H-labelled glucose, and glucose-derived glutamate-glutamine (Glx) and lactate pools in GL261 and CT2A orthotopic tumors. Kinetic modeling further indicated inter-tumor heterogeneity of glucose consumption rate for glycolysis and oxidation during a defined epoch of active proliferation in both cohorts (19 ± 1 days post-induction), with consistent volumes (38.3 ± 3.4 mm3) and perfusion properties prior to marked necrosis. Histopathologic analysis of these tumors revealed clear differences in tumor heterogeneity between the two GBM models, aligned with metabolic differences of the respective cell lines monitored in situ. Importantly, glucose oxidation (i.e. Glx synthesis and elimination rates: 0.40 ± 0.08 and 0.12 ± 0.03 mM min−1, respectively) strongly correlated with cell proliferation across the pooled cohorts (R = 0.82, p = 0.001; and R = 0.80, p = 0.002, respectively), regardless of tumor morphologic features or in situ metabolic characteristics of each GBM model. Conclusions Our fast DGE 2H-MRS enables the quantification of glucose consumption rates through glycolysis and mitochondrial oxidation in mouse GBM, which is relevant for assessing their modulation in vivo according to tumor microenvironment features such as cell proliferation. This novel application augurs well for non-invasive metabolic characterization of glioma or other cancers with mitochondrial oxidation dependencies.
Collapse
Affiliation(s)
- Rui V Simões
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| | - Rafael N Henriques
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Beatriz M Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Tânia Carvalho
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
42
|
Safaee MM, Wang EJ, Jain S, Chen JS, Gill S, Zheng AC, Garcia JH, Beniwal AS, Tran Y, Nguyen AT, Trieu M, Leung K, Wells J, Maclean JM, Wycoff K, Aghi MK. CD97 is associated with mitogenic pathway activation, metabolic reprogramming, and immune microenvironment changes in glioblastoma. Sci Rep 2022; 12:1464. [PMID: 35087132 PMCID: PMC8795421 DOI: 10.1038/s41598-022-05259-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor with a median survival under two years. Using in silico and in vitro techniques, we demonstrate heterogeneous expression of CD97, a leukocyte adhesion marker, in human GBM. Beyond its previous demonstrated role in tumor invasion, we show that CD97 is also associated with upregulation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/Erk) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathways in GBM. While CD97 knockout decreased Akt activation, CD97 targeting did not alter MAPK/Erk activation, did not slow GBM cell proliferation in culture, and increased levels of glycolytic and oxidative phosphorylation metabolites. Treatment with a soluble CD97 inhibitor did not alter activation of the MAPK/Erk and PI3K/Akt pathways. Tumors with high CD97 expression were associated with immune microenvironment changes including increased naïve macrophages, regulatory T cells, and resting natural killer (NK) cells. These data suggest that, while CD97 expression is associated with conflicting effects on tumor cell proliferative and metabolic pathways that overall do not affect tumor cell proliferation, CD97 exerts pro-tumoral effects on the tumor immune microenvironment, which along with the pro-invasive effects of CD97 we previously demonstrated, provides impetus to continue exploring CD97 as a therapeutic target in GBM.
Collapse
Affiliation(s)
- Michael M Safaee
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Elaina J Wang
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Saket Jain
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Jia-Shu Chen
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Sabraj Gill
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Allison C Zheng
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Joseph H Garcia
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Angad S Beniwal
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Y Tran
- Planet Biotechnology, Inc., Hayward, CA, USA
| | - Alan T Nguyen
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA
| | - Melissa Trieu
- School of Pharmacy, University of California, San Francisco (UCSF), San Francisco, USA
| | - Kevin Leung
- School of Pharmacy, University of California, San Francisco (UCSF), San Francisco, USA
| | - Jim Wells
- School of Pharmacy, University of California, San Francisco (UCSF), San Francisco, USA
| | | | | | - Manish K Aghi
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, USA.
| |
Collapse
|
43
|
Tumor Cell Infiltration into the Brain in Glioblastoma: From Mechanisms to Clinical Perspectives. Cancers (Basel) 2022; 14:cancers14020443. [PMID: 35053605 PMCID: PMC8773542 DOI: 10.3390/cancers14020443] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common and malignant primary brain tumor, defined by its highly aggressive nature. Despite the advances in diagnostic and surgical techniques, and the development of novel therapies in the last decade, the prognosis for glioblastoma is still extremely poor. One major factor for the failure of existing therapeutic approaches is the highly invasive nature of glioblastomas. The extreme infiltrating capacity of tumor cells into the brain parenchyma makes complete surgical removal difficult; glioblastomas almost inevitably recur in a more therapy-resistant state, sometimes at distant sites in the brain. Therefore, there are major efforts to understand the molecular mechanisms underpinning glioblastoma invasion; however, there is no approved therapy directed against the invasive phenotype as of now. Here, we review the major molecular mechanisms of glioblastoma cell invasion, including the routes followed by glioblastoma cells, the interaction of tumor cells within the brain environment and the extracellular matrix components, and the roles of tumor cell adhesion and extracellular matrix remodeling. We also include a perspective of high-throughput approaches utilized to discover novel players for invasion and clinical targeting of invasive glioblastoma cells.
Collapse
|
44
|
Sun D, Li F, Liu L, Yu S, Wang H, Gao X, Liu G, Zhao Y, Qiu G, Jiang X. PSMA3-AS1 induced by transcription factor PAX5 promotes cholangiocarcinoma proliferation, migration and invasion by sponging miR-376a-3p to up-regulate LAMC1. Aging (Albany NY) 2022; 14:509-525. [PMID: 35022330 PMCID: PMC8791211 DOI: 10.18632/aging.203828] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been reported to exhibit a crucial regulatory role in tumor progression, including cholangiocarcinoma (CCA). As a promising lncRNA, proteasome 20S subunit alpha 3 antisense RNA 1 (PSMA3-AS1) is involved in development of various tumors. However, the role and function of PSMA3-AS1 in CCA remain unclear. The aim of this study is to examine the expression, function, mechanism, and clinical significance of PSMA3-AS1 in CCA development. By TCGA database analysis, we found that PSMA3-AS1 was overexpressed in CCA. Consistent with the TCGA analysis, PSMA3-AS1 was significantly overexpressed in CCA tissues and cells by RT-qPCR. Upregulated PSMA3-AS1 was related to lymph node invasion, advanced TNM stage and poor survival, and was an independent risk factor of prognosis for CCA patients. Functionally, CCK-8, EdU and colony formation assays confirmed that upregulated PSMA3-AS1 promoted CCA cell proliferation, whereas downregulated PSMA3-AS1 inhibited proliferation. This result was further confirmed by subcutaneous tumor formation in nude mice. Wound healing and transwell assays confirmed that increased PSMA3-AS1 promoted CCA cell migration and invasion, whereas decreased PSMA3-AS1 inhibited these biological phenotypes. In addition, PSMA3-AS1 promoted the EMT process of CCA by downregulating E-cadherin and upregulating N-cadherin and vimentin. Mechanistically, transcription factor PAX5 bound to the promoter region of PSMA3-AS1 and promoted its transcription. Simultaneously, PSMA3-AS1 primarily localized in the cytoplasm could competitively bind miR-376a-3p to upregulate LAMC1, thereby accelerating CCA progression. This study uncovers that PSMA3-AS1 functions as a cancer-promoting gene in CCA, and PAX5/PSMA3-AS1/miR-376a-3p/LAMC1 axis plays a vital role in CCA development.
Collapse
Affiliation(s)
- Dongsheng Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Fujun Li
- Department of General Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Lang Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shaobo Yu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Haicun Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Gao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Guanglin Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yuqiao Zhao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Gongcai Qiu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xingming Jiang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
45
|
Younis M, Shaikh S, Shahzad KA. Long non-coding RNA RP5-821D11.7 promotes proliferation, migration, and epithelial-mesenchymal transition in glioma and glioma stem-like cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021:S0079-6107(21)00139-5. [PMID: 34952030 DOI: 10.1016/j.pbiomolbio.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Long noncoding RNA (lncRNA) has been recently revealed as a main regulatory molecule, which implicates many cellular functions. Studies showed that lncRNA abnormally expressed and involved in the progression and tumorigenesis of glioma. Present study identified a novel lncRNA associated with glioma, glioma stem-like cells (GSCs), and then revealed their potential functions. During the screening of lncRNAs, we investigated overexpression of lncRNA RP5-821D11.7 (lncRNA-RP5) in GSCs compared to glioma cells. Lentivirus-mediated shRNA for lncRNA-RP5 was constructed and transfected into glioma cells. Transfected stable glioma cells were transplanted into nude mice and tumor growth was observed. Knockdown of lncRNA-RP5 significantly inhibits proliferation, colony formation, migration and reduces epithelial-mesenchymal transition (EMT) by activating the Wnt/β-catenin pathway. Additionally, the results showed that lncRNA RP5 knockdown enhances cell apoptosis through endoplasmic reticulum stress. Therefore, this study may provide a better understanding about lncRNA-RP5 which revealed that it might be a potential therapeutic target in case of glioma progression and recurrence.
Collapse
Affiliation(s)
- Muhammad Younis
- Center for Infection and Immunity, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Sana Shaikh
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Khawar Ali Shahzad
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| |
Collapse
|
46
|
Cheng Y, Song S, Wei Y, Xu G, An Y, Ma J, Yang H, Qi Z, Xiao X, Bai J, Xu L, Hu Z, Sun T, Wang L, Lu J, Lin Q. Glioma Imaging by O-(2-18F-Fluoroethyl)-L-Tyrosine PET and Diffusion-Weighted MRI and Correlation With Molecular Phenotypes, Validated by PET/MR-Guided Biopsies. Front Oncol 2021; 11:743655. [PMID: 34912706 PMCID: PMC8666958 DOI: 10.3389/fonc.2021.743655] [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: 07/21/2021] [Accepted: 11/11/2021] [Indexed: 12/05/2022] Open
Abstract
Gliomas exhibit high intra-tumoral histological and molecular heterogeneity. Introducing stereotactic biopsy, we achieved a superior molecular analysis of glioma using O-(2-18F-fluoroethyl)-L-tyrosine (FET)-positron emission tomography (PET) and diffusion-weighted magnetic resonance imaging (DWI). Patients underwent simultaneous DWI and FET-PET scans. Correlations between biopsy-derived tumor tissue values, such as the tumor-to-background ratio (TBR) and apparent diffusion coefficient (ADC)/exponential ADC (eADC) and histopathological diagnoses and those between relevant genes and TBR and ADC values were determined. Tumor regions with human telomerase reverse transcriptase (hTERT) mutation had higher TBR and lower ADC values. Tumor protein P53 mutation correlated with lower TBR and higher ADC values. α-thalassemia/mental-retardation-syndrome-X-linked gene (ATRX) correlated with higher ADC values. 1p/19q codeletion and epidermal growth factor receptor (EGFR) mutations correlated with lower ADC values. Isocitrate dehydrogenase 1 (IDH1) mutations correlated with higher TBRmean values. No correlation existed between TBRmax/TBRmean/ADC/eADC values and phosphatase and tensin homolog mutations (PTEN) or O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Furthermore, TBR/ADC combination had a higher diagnostic accuracy than each single imaging method for high-grade and IDH1-, hTERT-, and EGFR-mutated gliomas. This is the first study establishing the accurate diagnostic criteria for glioma based on FET-PET and DWI.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China.,Department of Neurosurgery, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuangshuang Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.,Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yukui Wei
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Geng Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Yang An
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Jie Ma
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Hongwei Yang
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinru Xiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Jie Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Lixin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| | - Zeliang Hu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tingting Sun
- Department of Medicine, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, China International Neuroscience Institute, Beijing, China
| |
Collapse
|
47
|
Chen B, Wang M, Huang R, Liao K, Wang T, Yang R, Zhang W, Shi Z, Ren L, Lv Q, Ma C, Lin Y, Qiu Y. Circular RNA circLGMN facilitates glioblastoma progression by targeting miR-127-3p/LGMN axis. Cancer Lett 2021; 522:225-237. [PMID: 34582975 DOI: 10.1016/j.canlet.2021.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is one of the most devastating cancers and is characterized by rapid cell proliferation and aggressive invasiveness. Legumain (LGMN), a substrate-specific protease, is associated with poor progression of GBM. Circular RNAs (circRNAs) are aberrantly expressed in various cancers and play crucial roles in tumor progression; however, the functional roles of circRNAs originating from LGMN remain largely unknown in GBM. Herein, we found that hsa_circ_0033009 (circLGMN) was the most abundantly expressed circRNA derived from LGMN. CircLGMN was upregulated in high-grade glioma (HGG), and high expression of circLGMN was associated with poor prognosis in patients with glioma. CircLGMN overexpression promoted GBM cell proliferation and enhanced cell invasion. Mechanistically, circLGMN acts as a sponge for miR-127-3p, and prevents miR-127-3p-mediated degradation of LGMN mRNA, ultimately leading to increased LGMN protein expression. Treatment with miR-127-3p mimic suppressed proliferation and reduced invasion of GBM cells overexpressing circLGMN. Moreover, circLGMN overexpression promoted GBM malignancy in vivo, while miR-127-3p overexpression alleviated this effect. Taken together, circLGMN is a novel tumor-promoting circRNA that acts by sponging miR-127-3p, which ultimately leads to LGMN upregulation. Thus, targeting the circLGMN/miR-127-3p/LGMN axis might be a promising strategy for GBM treatment. More importantly, the discovery of the self-regulatory mechanism of LGMN expression by circLGMN, will facilitate further research on LGMN.
Collapse
Affiliation(s)
- Binghong Chen
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Mengying Wang
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China; Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Renhua Huang
- Department of Radiation Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Keman Liao
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Tianwei Wang
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Renhao Yang
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Wenrui Zhang
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China; Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Zhonggang Shi
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China; Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China
| | - Li Ren
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, PR China
| | - Qi Lv
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Chunhui Ma
- Department of Orthopedics, Shanghai General Hospital of Shanghai Jiao Tong University, Shanghai, 200080, PR China
| | - Yingying Lin
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China; Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China.
| | - Yongming Qiu
- Department of Neurosurgery, Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, PR China.
| |
Collapse
|
48
|
Chonan Y, Yamashita T, Sampetrean O, Saya H, Sudo R. Spatial heterogeneity of invading glioblastoma cells regulated by paracrine factors. Tissue Eng Part A 2021; 28:573-585. [PMID: 34841881 DOI: 10.1089/ten.tea.2021.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal type of malignant primary brain tumor in adults. GBM displays heterogeneous tumor cell population comprising glioma-initiating cells (GICs) with stem cell-like characteristics and differentiated glioma cells. During GBM cell invasion into normal brain tissues, which is the hallmark characteristic of GBM, GICs at the invasion front retain stemness, while cells at the tumor core display cellular differentiation. However, the mechanism of cellular differentiation underlying the formation of spatial cellular heterogeneity in GBM remains unknown. In the present study, we first observed spatially heterogeneous GBM cell populations emerged from an isogenic clonal population of GICs during invasion into a 3D collagen hydrogel in a microfluidic device. Specifically, GICs at the invasion front maintained stemness, while trailing cells displayed astrocytic differentiation. The spatial cellular heterogeneity resulted from the difference in cell density between GICs at the invasion front and trailing cells. Trailing GICs at high cell density exhibited astrocytic differentiation via local accumulation of paracrine factors they secreted, while cells at the invasion front of low cell density retained stemness due to the lack of paracrine factors. In addition, we demonstrated that interstitial flow suppressed astrocytic differentiation of trailing GICs by the clearance of paracrine factors. Our findings suggest that intercellular crosstalk between tumor cells is an essential factor in developing the spatial cellular heterogeneity of GBM cells with various differentiation statuses. It also provides insights into the development of novel therapeutic strategies targeting GBM cells with stem cell characteristics at the invasion front.
Collapse
Affiliation(s)
- Yuta Chonan
- Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Tadahiro Yamashita
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Oltea Sampetrean
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Hideyuki Saya
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Ryo Sudo
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| |
Collapse
|
49
|
Barrette AM, Ronk H, Joshi T, Mussa Z, Mehrotra M, Bouras A, Nudelman G, Jesu Raj JG, Bozec D, Lam W, Houldsworth J, Yong R, Zaslavsky E, Hadjipanayis CG, Birtwistle MR, Tsankova NM. Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor Verteporfin in preclinical glioblastoma models. Neuro Oncol 2021; 24:694-707. [PMID: 34657158 DOI: 10.1093/neuonc/noab244] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of GBM growth in disease progression and recurrence. Here we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug's efficacy and survival benefit in GBM models. METHODS Up to eight low-passage patient-derived GBM cell lines with distinct genomic drivers, including three primary/recurrent pairs, were treated with Verteporfin or vehicle to assess in-vitro effects on proliferation, migration, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft models (PDX) were used to assess Verteporfin's brain penetrance and effects on tumor burden and survival. RESULTS Verteporfin treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term Verteporfin treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2 and ITGB1. Finally, long-term Verteporfin treatment appeared non-toxic and conferred survival benefit compared to vehicle in two PDX models: as monotherapy in primary (de-novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation. CONCLUSIONS We demonstrate combined anti-invasive and anti-proliferative efficacy for Verteporfin with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for glioblastoma patients.
Collapse
Affiliation(s)
- Anne Marie Barrette
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Halle Ronk
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tanvi Joshi
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zarmeen Mussa
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meenakshi Mehrotra
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - German Nudelman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joe G Jesu Raj
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dominique Bozec
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Lam
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jane Houldsworth
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raymund Yong
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Zhang P, Ohkawa Y, Yamamoto S, Momota H, Kato A, Kaneko K, Natsume A, Farhana Y, Ohmi Y, Okajima T, Bhuiyan RH, Wakabayashi T, Furukawa K, Furukawa K. St8sia1-deficiency in mice alters tumor environments of gliomas, leading to reduced disease severity. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 83:535-549. [PMID: 34552288 PMCID: PMC8438004 DOI: 10.18999/nagjms.83.3.535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/18/2020] [Indexed: 12/29/2022]
Abstract
Ganglioside GD3/GD2 are over-expressed in various neuroectoderm-derived tumors. Previous studies indicated that GD3 is involved in the enhancement of cancer properties such as rapid growth and increased invasiveness. However, little is known about the functions of GD3/GD2 in glioma cells and glioma microenvironments. To clarify the functions of GD3/GD2 in gliomas, we used a mouse glioma model based on the RCAS/Gtv-a system. At first, we compared the gliomas size between wild-type (WT) and GD3 synthase (GD3S) knockout (KO) mice, showing a less malignant histology and slower tumor growth in GD3S-KO mice than in WT mice. Immunohistochemistry of glioma sections from WT and GD3S-KO mice revealed that reactive microglia/macrophages showed different localization patterns between the two genetic types of mice. CD68+ cells were more frequently stained inside glioma tissues of GD3S-KO mice, while they were stained mainly around glioma tissues in WT mice. The number of CD68+ cells markedly increased in tumor tissues of GD3S-KO mice at 2 weeks after injection of transfectant DF-1 cells. Furthermore, CD68+ cells in GD3S(-/-) glioma tissues expressed higher levels of inducible nitric oxide synthase. We observed higher expression levels of pro-inflammatory cytokine genes in primary-cultured glioma cells of WT mice than in GD3S-KO mice. DNA microarray data also revealed differential expression levels of various cytokines and chemokines in glioma tissues between WT and GD3S-KO mice. These results suggest that expression of GD3S allows glioma cells to promote polarization of microglia/macrophages towards M2-like phenotypes by modulating the expression levels of chemokines and cytokines.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Ohkawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute
| | - Satoko Yamamoto
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Hiroyuki Momota
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Kato
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yesmin Farhana
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Tetsuya Okajima
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Toshihiko Wakabayashi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|